Download PDF
ads:
Caracterização bioquímica, estrutural e funcional de vesículas
secretadas por leveduras
Débora Leite de Oliveira
Orientadores: Marcio Lourenço Rodrigues e Leonardo Nimrichter
Rio de Janeiro
Janeiro/ 2011
Tese de Doutorado apresentada ao Programa de
Pós-graduação em Química Biológica, Instituto de
Bioquímica Médica da Universidade Federal do Rio
de Janeiro, como parte dos requisitos necessários à
obtenção de título de Doutor em Ciências (Química
Biológica).
ads:
Livros Grátis
http://www.livrosgratis.com.br
Milhares de livros grátis para download.
ii
Trabalho realizado no Laboratório de Estudos Integrados em
Bioquímica Microbiana, Departamento de Microbiologia
Geral, Instituto de Microbiologia Prof. Paulo de Góes, Centro
de Ciências da Saúde, Universidade Federal do Rio de Janeiro,
sob orientação dos Professores Marcio Lourenço Rodrigues e
Leonardo Nimrichter.
ads:
iii
FICHA CATALOGRÁFICA
Oliveira, Débora Leite
Caracterização bioquímica, estrutural e funcional de vesículas secretadas por leveduras/
Débora Leite de Oliveira Rio de Janeiro, 2011
X, 149f.
Tese de Doutorado em Ciências (Química Biológica)
Universidade Federal do Rio de Janeiro/ Instituto de Bioquímica Médica, 2011.
Orientadores: Marcio Leorenço Rodrigues e Leonardo Nimrichter
Referências Bibliográficas:
1. Vesículas 2. Secreção não- convencional 3. Cryptococcus neoformans 4. Exossomos
5. Cápsula 6. Saccharomyces cerevisiae
I. Rodrigues, Marcio
II. Nimrichter, Leonardo
III. UFRJ, Instituto de Bioquímica Médica, Doutorado em Ciências (Química Biológica)
IV. Caracterização bioquímica, estrutural e funcional de vesículas secretadas por
leveduras/ Débora
iv
FOLHA DE APROVAÇÃO:
Caracterização bioquímica, estrutural e funcional de vesículas secretadas por leveduras
Débora Leite de Oliveira
Rio de Janeiro, 12 de Janeiro de 2011.
_____________________________________________________________________________
(Dr. Marcio Lourenço Rodrigues, Instituto de Microbiologia Prof. Paulo de Góes, UFRJ)
______________________________________________________________________________
(Dr. Leonardo Nimrichter, Instituto de Microbiologia Prof. Paulo de Góes, UFRJ)
______________________________________________________________________________
(Dra. Débora Foguel, Instituto de Bioquímica Médica, UFRJ)
______________________________________________________________________________
(Dra. Thaís, Instituto de Microbiologia Prof. Paulo de Góes, UFRJ)
______________________________________________________________________________
(Dra. Rosana Puccia, Departamento de Microbiologia, Imunologia e Parasitologia, Unifesp)
v
vi
Agradecimentos
Agradeço pela oportunidade de presenciar um momento de reforma na ciência
nacional, o qual conduzirá a Ciência a lugar de destaque em nossa cultura e sociedade.
Intuitivamente, acredito que as conseqüências sociais desta reforma serão impactantes. É
gratificante saber que a concretização dos meus ideais, aqui colocados nesta tese, participa da
atmosfera de mudança que respiramos hoje no meio científico. Agradeço portanto pela chance
de contribuir com o desenvolvimento científico do Brasil. Obrigada à todos que me ajudaram a
construir esta tese, obrigada sobretudo aos meus orientadores Marcio e Leonardo pela solidez,
cumplicidade e segurança.
Dentre colaboradores, amigos e todos que participaram desta etapa da minha
formação, agradeço francamente a: Allan Guimarães, AndLuiz Souza dos Santos e todo o seu
grupo, Arturo Casadevall, Célio Freire de Lima, Ernesto Nakayasu, Geralda Almeida, Igor de
Almeida, José Roberto Meyer Fernandes, Joshua Nosanchuk, Kildare Miranda, Radames
Cordero, Susana Frases, Vivek Malhotra e Vivian Rumjanek. Agradeço especialmente a toda
equipe do Laboratório de Estudos Integrados em Bioquímica Microbiana, por tornarem a minha
rotina ainda mais prazerosa. Acredito que o aprendizado se de forma mais contundente
quando acompanhado de experiências positivas, portanto, muito obrigada pelos ensinamentos
e pela diversão! Por fim, agradeço ao Instituto de Bioquímica Médica pelo acolhimento e
excelência.
vii
RESUMO:
Caracterização bioquímica, estrutural e funcional de vesículas secretadas por leveduras
Débora Leite de Oliveira
Orientadores: Marcio Lourenço Rodrigues e Leonardo Nimrichter
Os mecanismos pelos quais macromoléculas são transportadas através da parede celular
em fungos ainda são pouco conhecidos. O Cryptococcus neoformans, agente etiológico da
criptococose, apresenta uma cápsula polissacarídica externa ao corpo celular. Foi demonstrado
nesta tese que o polissacarídeo capsular é exportado para o ambiente extracelular em vesículas
membranosas. A caracterização bioquímica destes compartimentos vesiculares revelou a
presença de diferentes moléculas associadas a virulência, incluindo lipídeos, polissacarídeos e
proteínas. O papel da secreção vesicular durante a interação do fungo com a célula hospedeira
também foi avaliado. Foi demonstrado que macrófagos podem incorporar vesículas
extracelulares produzidas pelo C. neoformans. O tratamento de macrófagos com vesículas
fúngicas resultou na modulação da produção de citocinas e óxido nítrico além de interferir com
a capacidade fagocítica e da atividade microbicida. Estes dados estabeleceram que vesículas
secretadas pelo C. neoformans são biologicamente ativas. Com o intuito de elucidar quais vias
do tráfego intracelular estão potencialmente envolvidas na biogênese vesicular a levedura
Saccharomyces cerevisiae foi usada como organismo modelo. Vesículas foram isoladas de cepas
mutadas em diferentes genes que controlam passos importantes na via secretora clássica e na
via de formação de corpos multivesiculares (MVBs). Análises combinadas por microscopia
eletrônica de transmissão, espalhamento de luz, análises cromatográficas e proteômicas
revelaram que mutações em ambas as vias afetam a composição de vesículas extracelulares,
embora nenhuma das mutações analisadas interrompa a secreção de vesículas. Análises
lipídicas mostraram que mutantes com defeitos na secreção pós-Golgi apresentam uma cinética
mais lenta de liberação de vesículas. Estes dados sugerem que fungos produzem vesículas
extracelulares de atividade biológica de alta relevância, em processos que associam eventos de
secreção convencional e não-convencional.
viii
ABSTRACT:
Biochemical, structural and functional characterization of vesicles secreted by yeast cells
Débora Leite de Oliveira
Orientadores: Marcio Lourenço Rodrigues e Leonardo Nimrichter
The mechanisms by which macromolecules are transported across the cell wall of fungi are
still poorly known. Cryptococcus neoformans, the causative agent of cryptococcosis, has a
polysaccharide capsule outside of the cell body. It was demonstrated in this study that capsular
polysaccharides are exported to the extracellular environment within membrane vesicles. The
biochemical composition of these vesicular compartments revealed the presence of different
molecules associated with virulence, including lipids, polysaccharides and proteins. The role of
vesicular secretion during the interaction of fungi with host cells was also addressed. It was
demonstrated that macrophages can incorporate extracellular vesicles produced by C.
neoformans. Treatment of the macrophages with fungal vesicles resulted in modulation of
cytokine and nitric oxide production, phagocytic ability and microbicidal activity. These findings
establish that cryptococcal vesicles are biologically active. To elucidate what intracellular traffic
pathways originate the extracellular vesicles, the yeast Saccharomyces cerevisiae was used as a
model organism. Vesicles were isolated from cells with mutations in different genes involved in
key steps of the secretory or multivesicular body (MVB) formation pathways. Transmission
electron microscopy in combination with dynamic light scattering, chromatographic approaches
and proteomics revealed that mutations in both pathways affected the composition of yeast
extracellular vesicles, but none abrogated vesicle production. Lipid analysis revealed that
mutants with defects in Golgi-related components of the classic secretory pathway had slower
vesicle release kinetics. These results suggest that fungi produce extracellular vesicles with
highly relevant biological activities through cellular processes that combine events of both
conventional and non-conventional secretory pathways.
ix
LISTA DE ABREVIATURAS:
ABC (“ATP Binding Cassette”) – cassette de ligação a ATP
AcbA (“acyl-coenzyme A binding protein) proteína ligadora de acil- coenzima A
CMH ceramida monohexose
CMI (“cell mediated immunity”) – imunidade mediada por célula
CNS (“central nervous system”) – sistema nervosa central
COP (“coat protein”) – proteínas de revestimento
CtxB-FITC subunidade B da toxina do cólera marcada com FITC
CWP (“cell wall protein”) – proteína de parede celular
DAPI (“4',6-diamidino-2-phenylindole”) - 4',6-diamidino-2-fenilindol
DC célula dendrítica
DiI (“dialkylcarbocyanine iodide”) – iodeto de dialquilcarbocianina
DTT (“1,4-Dithioerythritol”) - 1,4-Ditiotreitol
ELISA (“enzyme-linked immunosorbent assay”) – ensaio imunoenzimático
emPAI (“exponentially modified Protein Abundance Index”) - índice de abundância de proteína
modificado exponencialmente
ESCRT (“endosomal sorting complex required for transport”) – complexo de endereçamento
endossomal necessário ao transporte
ESI-MS (“electronspray ionization mass spectrometry”) – ionização por bombardeamento de
electron acoplado a espectometria de massas
FCS (“fetal cow serum”) – soro fetal bovino
FGF2 (“fibroblast growth factor 2”) – fator de crescimento de fibroblasto 2
FITC (“fluorescein isothiocyanate”) – isotiocianato de fluoresceína
GlcCer - glucosilceramida
x
GPI - glicofosfatidilinositol
GRASP (“General receptor for phosphoinositides 1-associated scaffold protein”) – proteína de
arcabouço associada a receptor de fosfoinositídeos 1
GXM - glucuronoxilomanana
HIV (“human immunodeficiency virus”) – vírus da immunodeficiência humana
HPTLC (“high performance thin layer chromatography”) cromatografia em camada fina da alta
performance
Hsp (“heat shock protein”) – proteína de choque térmico
IEM (“immunoelectronmicroscopy”) – microscopia imunoeletrônica
IFN-γ interferon - gamma
IL interleucina
ILVs (“intraluminal vesicles”) – vesículas intraluminais
LDH lactato desidrogenase
L-NIL - N6-(1-Iminoetil)-L-lisina dihidrocloreto
Mab 18B7 anticorpo monoclonal 18B7
MIF (“macrophage migration inhibitory factor”) - fator inibidor de migração de macrófagos
NO (“nitric oxide”) – óxido nítrico
OMVs (“outer membrane vesicles”) – vesículas de membrane externa
PC (“phosphatidylcholine”)- fosfatidilcolina
PRR (“pattern recognition receptors”) receptores de reconhecimento de padrões
PVP - polivinilpirrolidona
RE retículo endoplasmático
rf índice de retenção
TEM (“transmission electron microscopy”) – microscopia eletrônica de transmissão
TGF- β (“transforming growth factor beta”) fator de transformação de crescimento beta
xi
TGN (“trans Golgi network”) – rede trans Golgi
TNF-α (“tumor necrosis factor alpha”) – fator de necrose tumoral alpha
ts (“temperature- sensitive”) - sensível a temperatura
t- SNARE (“trans SNAP (Soluble NSF Attachment Protein) Rceptor”) receptor para SNAP trans
UFC unidade formadora de colônia
Vps (“vacuolar sorting protein”) – proteína de endereçamento para vacúolo
v- SNARE (“vesicle SNAP (Soluble NSF Attachment Protein) Rceptor”) receptor para SNAP
vesicular
WT (“wild type”) – tipo selvagem
xii
ÍNDICE
I-Introdução……………………………………………………………………………………………………………………………….1
1. Mecanismos clássicos de secreção: aspectos históricos………………………………………………….1
2. A parede celular fúngica…………………………………………………………………………………………………7
3. Mecanismos não-convencionais de secreção…………………………………………………………………8
4. Cryptococcus neoformans: patógeno e modelo de estudo……………………………………………13
5. Resposta immune anti-C. neoformans………………………………………………………………………….22
II- Objetivos…………………………………………………………………………………………………………………………...27
III- Materiais e Métodos…………………………………………………………………………………………………………..29
IV- Resultados………………………………………………………………………………………………………………………….47
Anexo 1………………………………………………………………………………………………………………………..56
Anexo 2……………………………………………………………………………………………………………………..…69
Anexo 3……………………………………………………………………………………………………………………..…78
Anexo 4………………………………………………………………………………………………………………………..88
V- Discussão…………………………………………………………………………………………………………………………..117
VI- Referências Bibliográficas…………………………………………………………………………………………………126
VII- Anexo 5…………………………………………………………………………………………………………………………...148
1
I Introdução:
1. Mecanismos clássicos de secreção: aspectos históricos
O trabalho pioneiro que descreveu o transporte e secreção de proteínas em células
eucariotas foi desenvolvido por George Palade e conferiu a ele o prêmio Nobel de Fisiologia em
1974. Havia sido descoberto através de estudos morfológicos e de fracionamento celular que as
proteínas atravessam uma complexa rede de membranas intracelulares até serem secretadas e
que vesículas de transporte estabelecem a comunicação entre estas membranas (Palade, 1975).
Este trabalho levantou diversas questões sobre como essas vesículas eram criadas, como
atingiam o compartimento correto e como os diferentes compartimentos mantinham sua
identidade a despeito da constante fusão de vesículas de transporte (Schekman, 2002). Coube a
Randy Sheckman e colaboradores o desafio de elucidar os mecanismos envolvidos em transporte
de vesículas e secreção. Para este estudo, foi escolhida a levedura Saccharomyces cerevisiae
como organismo modelo devido a facilidade de manipulação genética. A partir de uma triagem
em mutantes sensíveis a temperatura (ts, do termo em inglês “temperature-sensitive”) foi
possível identificar 23 genes envolvidos na secreção de proteínas (Novick, et al. 1980). Esta
técnica é utilizada no estudo de genes essenciais pois o fenótipo mutante é expresso somente a
37
o
C, uma vez que a proteína apresenta mutações pontuais que causam a perda de estrutura e
portanto de função a 37
o
C. Tais mutantes apresentavam acúmulo de vesículas citoplasmáticas
na temperatura de restrição (Novick, et al. 1980). Mais do que isso, diferentes mutantes
apresentavam diferentes aspectos morfológicos, pois presumivelmente acumulavam vesículas
em diferentes pontos da via de secreção (Figura 1).
2
Figura 1. Micrografias eletrônicas de mutantes sec de S. cerevisiae em temperatura restritiva. Os
mutantes apresentam acúmulo de vesículas de transporte (A), extensão de RE (B) e expansão do
Complexo de Golgi (Bb “Barkeley Body”). Símbolos: ves, vesículas; va, vacúolo; er, retículo
endoplasmático; n, núcleo; nm, membrana nuclear; np, poro nuclear. Imagem reproduzida de Novick, et
al. 1980.
Proteínas destinadas à membrana plasmática ou ao ambiente extracelular engajam a
chamada via biossintética secretória (Bonifacino & Glick, 2004). Aquelas endereçadas ao vacúolo
e endossomos compartilham os estágios iniciais desta via (Figura 2). Tais proteínas apresentam
uma seqüência sinal para síntese no retículo endoplasmático (RE) na sua extremidade N-
terminal. Após a tradução acoplada ao transporte para o lúmen do RE, as proteínas são
transportadas ao Complexo de Golgi por carreadores vesiculares (Figura 2) (Bonifacino &
Lippincott-Schwartz, 2003). Após receberem modificações pós-transcricionais são endereçadas
ao destino final (por exemplo, membrana plasmática ou endossomos) a partir da rede trans-Golgi
(TGN) (Figura 2).
O brotamento de vesículas do compartimento doador bem como a incorporação seletiva de
proteínas cargo requer o recrutamento de “proteínas de revestimento” (coat proteins) as quais
compõem complexos supramoleculares capazes de deformar a membrana do compartimento e
induzir o brotamento (Figura 3) (Kirchhausen, 2000). Os principais complexos de revestimento
que operam na via secretória são COPII, o qual reveste vesículas que brotam do RE e se fundem
3
ao Cis-Golgi (Waters, et al. 1991; Barlowe, et al. 1994), COPI, responsável pelo transporte intra-
Golgi e pelo transporte retrogrado Golgi-ER e Clatrina, que reveste vesículas que brotam da rede
trans-Golgi (TGN) e da membrana plasmática (Figura 2) (Letourneur, 1994). Depois de formada, a
vesícula libera o complexo de revestimento que estará disponível no citosol para formação de
uma nova vesícula (Figura 3, passo 4). A perda do complexo se pela ação de enzimas
acessórias (Bonifacino & Glick, 2004).
Figura 2. Vias de tráfego intracelular e os principais compartimentos envolvidos. As setas indicam os
possíveis caminhos percorridos por vesículas de transporte. As cores indicam a localização dos complexos
de revestimento COPII (azul), COPI (vermelho) e clatrina (amarelo) (Adaptado de: Bonifacino & Glick,
2004). A via biossintética- secretora carreia moléculas sintetizadas no RE e modificadas no Golgi para o
ambiente extracelular ou para demais compartimentos intracelulares. A via endocítica carreia moléculas a
partir da superfície celular para os lisossomos/vacúolo e vesículas que brotam do Golgi também podem
engajar a via endocítica.
Diversas classes de proteínas funcionam no contato inicial entre as membranas da vesícula e
do compartimento aceptor e são coletivamente chamadas de proteínas de ancoramento
(tethering proteins”). O principal tipo de reconhecimento se através de proteínas
transmembrana contendo um longo domínio super-hélice que reconhecem Rab-GTPases
4
específicas (conhecidas como proteínas Ypt em leveduras) ligadas à membrana das vesículas
(Figura 3, passo 5) (Segev, 2001; Jahn, et al. 2003).
Figura 3. Mecanismos de brotamento e fusão de vesículas de transporte. As proteínas de revestimento
formam um complexo na membrana do compartimento doador (1). O complexo de revestimento induz
brotamento na membrana e a vesícula se forma contendo proteínas cargo solúveis e transmembrana vem
como a v-SNARE específica deste compartimento (2) A cisão da vesícula ocorre com auxílio de proteínas
acessórias (3). A vesícula perde o revestimento em conseqüência da inativação de GTPases existentes no
complexo de revestimento e da ação de outras enzimas (4). A vesícula é guiada ao compartimento
aceptor, provavelmente através de componentes do citoesqueleto. O reconhecimento do compartimento
aceptor ocorre pela ligação de uma Rab-GTPase específica da vesícula a um “fator de reconhecimento”
presente na membrana alvo (5). A ligação de v-SNARE a t-SNARE também é fundamental para o
reconhecimento (6) além de mediar a fusão das membranas (7). Após a fusão as proteínas SNAREs se
dissociam e v-SNARE é reciclada para o compartimento doador através do mesmo mecanismo (Adaptado
de Bonifacino & Glick, 2004).
A fusão de membranas requer mecanismos redundantes de reconhecimento para garantir a
alta fidelidade do processo (Bonifacino & Glick, 2004). Por isso, proteínas SNAREs também
operam no reconhecimento do compartimento receptor (Figura 3, passos 6 e 7). Foi proposto
que vesículas destinadas a um dado compartimento carreiam em sua membrana uma v-SNARE
específica, a qual reconhece uma t-SNARE cognata na membrana do compartimento receptor
(Rothman, 1994). Estas proteínas são ancoradas as membranas pela porção C-terminal, possuem
5
“motivos SNAREs” repetidos de 60-70 aminoácidos que auxiliam a formação de uma super-hélice
e apresentam o N-terminal voltado para o citosol (Bock, et al. 2001). Estudos estruturais
mostraram que a ligação entre v- e t-SNAREs forma um feixe de 4 hélices bastante estável
(Sutton, et al. 1998), sendo um monômero de v-SNARE e um trímero de t-SNARE. Além do
reconhecimento, SNAREs medeiam fusão de membranas, processo energeticamente
desfavorável pois requer a remoção de moléculas de água entre as duas superfícies hidrofílicas
das membranas. A formação do feixe de super-hélices parece suprir a energia livre necessária
para fundir duas bicamadas lipídicas (Figura 4) (Weber, et a. 1998). Uma vez que houve a fusão, o
complexo cis-SNARE (v- e t- SNAREs associadas na mesma membrana) deve se dissociar, processo
que requer o recrutamento de proteínas NSamF e α-SNAP e hidrólise de ATP (Sutton, et al. 1998;
Rice & Brunger, 1999). v-SNAREs são então empacotadas em novas vesículas e recicladas ao
compartimento anterior (Bonifacino & Glick, 2004).
Figura 4. A. Mecanismo de fusão de vesículas de transporte
mediado por proteínas SNAREs. Um forte pareamento entre
SNAREs força a íntima aposição das duas bicamadas lipídicas de
modo que as moléculas de água sejam expelidas da interface. B.
Estrutura cristal das duas super-lices complexadas (v- e t-
SNAREs) (adaptado de Sutton, et al. 1998).
6
Como se pode perceber, o funcionamento correto da via biossintética-secretória depende de
uma extensa maquinaria molecular que deve garantir (i) que as moléculas-cargo sejam
selecionadas das residentes no compartimento doador, (ii) que as vesículas de transporte se
formem, (iii) que as vesículas cheguem ao compartimento aceptor e (vi) que a maquinária de
transporte seja reciclada ao compartimento doador (Stephens & Pepperkok, 2001). A via
endocítica segue as mesmas regras utilizando uma maquinária molecular totalmente distinta. A
endocitose capta macromoléculas no fluído extracelular a partir da formação de vesículas
revestidas por Clatrina na membrana plasmática (Figura 2). Estas podem ser encaminhadas aos
endossomos de reciclagem e retornam a membrana plasmática. Alternativamente, as vesículas
são encaminhadas ao endossomo inicial, o qual sofre maturação e acidificação do conteúdo
enquanto recebe hidrolases e H
+
-ATPaes lisossomais advindas do Complexo de Golgi. Neste
estágio, o compartimento endocítico é chamado endossomo tardio e funde-se ao lisossomo/
vacúolo para completar a degradação do conteúdo endocitado (Figura 2) (Kornfeld & Mellman,
1989).
Em células fúngicas a via secretória confronta-se com uma barreira adicional: a parede
celular. Células vegetais enfrentam o mesmo problema, entretanto, sabe-se que uma estrutura
oriunda da membrana plasmática atravessa a parede celular permitindo a troca de
macromoléculas entre células vizinhas. Tal estrutura denomina-se plasmodesmo (Hyun & Uddin,
2010). Em procariotos os mecanismos de secreção através da parede são amplamente
conhecidos e envolvem ltiplos genes (SECA, SECY, SECE E SECG) e diversas proteínas
acessórias (Driessen, et al. 1998). Em bactérias gram negativas, seis diferentes mecanismos de
secreção foram identificados (sistemas de secreção do tipo I ao VI). Estes sistemas operam
7
liberando proteínas no espaço extracelular e no caso de patógenos, injetando toxinas na célula
hospedeira (Bingle et al. 2008; Cianciotto, 2005; Craig & Li, 2008). Em eucariotos, embora a
maioria dos estudos de mecanismos de trágefo tenham se desenvolvido em leveduras,
pouquíssimo é conhecido sobre o transporte de macromoléculas através a rígida estrutura da
parede celular de fungos.
2. A parede celular fúngica:
A parede celular é um envoltório de superfície que confere rigidez, forma e proteção contra
estresse físico e lise osmótica, além de contribuir com a regulação da permeabilidade celular
(Kapteyn, et al. 1999). A parede celular fúngica é formada basicamente por β1-3 e β1-6 glucanas,
além de polímeros de β1-4-N-acetilglucosamina denominados quitina e proteínas altamente
glicosiladas conhecidas como CWP (Cell Wall Proteins) (Klis, et al 2002). A maioria das CWPs é
transportada para superfície em ligação à âncora de glicofosfatidilinositol (GPI), posteriormente
sofrem clivagem e incorporação à parede celular através de ligação a uma porção remanescente
da âncora (Muller, et al. 1997). Embora a estrutura da parede celular seja rígida, é fato que essa
estrutura é suficientemente plástica para permitir o crescimento celular induzido pela pressão de
turgor (Levin, 2005). Para isso, a parede celular sofre constante remodelamento, o qual é
acompanhado pelo endereçamento de vesículas secretórias contendo componentes da parede
celular e enzimas como glucanases e quitinases (Novick & Schekman, 1979, 1983; Levin, 2005).
Este remodelamento é um processo finamente regulado e depende da polimerização de actina
(Drubin & Nelson, 1996). Alguns fungos podem apresentar ainda melanina na parede celular, a
qual confere maior proteção contra os estresses ambientais (Gomes & Nosanchuk, 2003). Esse
8
pigmento é também determinante para a porosidade da parede (Jacobson & Ikeda, 2005). A
melanina é sintetizada a partir de compostos fenólicos (Gomez & Nosanchuk, 2003) por
mecanismos descritos nas próximas seções.
Moléculas aparentemente atípicas são freqüentemente encontradas na parede celular de
fungos, como proteínas de choque térmico (hsp), glicoesfingolipídeos e histonas (Chaffin, et al.
1998; Rodrigues, et al. 2000; Nosanchuck et al. 2003; Nimrichter, et al. 2005). Tal diversidade
molecular sugere que existam mecanismos não-convencionais de endereçamento destas
moléculas para fora da célula.
3. Mecanismos não-convencionais de secreção:
A secreção de proteínas a partir do eixo RE-Golgi é extremamente acurada e eficiente e
carreia a maioria das proteínas destinadas ao espaço extracelular. Estas apresentam o peptídeo
sinal que dirige a tradução acoplada ao transporte para o interior do RE, conforme mencionado
anteriormente. Entretanto, diversas proteínas sabidamente encontradas no ambiente
extracelular não apresentam o peptídeo sinal, sendo sintetizadas em ribossomos livres no citosol.
Exemplos dessas proteínas são: fator-α de leveduras, FGF2 (fibroblast growth factor 2), MIF
(macrophage migration inhibitory factor), AcbA (acyl-coenzyme A binding protein) e IL-
(revisado em Nickel & Rabouille, 2009). Os processos relacionados ao transporte de tais
proteínas para o ambiente extracelular são coletivamente conhecidos como secreção não-
convencional de proteínas. Neste sentido, diferentes mecanismos estão implicados. O fator-α,
por exemplo, utiliza um transportador transmembrana da família ABC (ATP-binding cassette)
(McGrath & Varshavsky, 1989). A citocina IL1- β é abundantemente secretada durante a resposta
9
inflamatória, uma vez que caspase 1 é ativada e cliva a forma precursora de IL1- β gerando a
proteína madura (Andrei, et al. 1999). Tem sido proposto que IL1-β é translocada para o interior
de lisossomos secretórios e carreada para o espaço extracelular após a secreção lisossomal
(Andrei, et al. 1999; Andrei, et al. 2004). Todavia, mecanismos de secreção dependentes de
microvesículas também devem estar implicados. Foi descrito que vesículas extracelulares
denominadas exossomos contém IL1-β na forma ativa (Qu, et al. 2007).
Exossomos são vesículas liberadas no espaço extracelular que apresentam entre 40-100 nm e
possuem origem endossomal (Figura 5). A composição lipídica dos exossomos é uma de suas
características marcantes, sendo estes enriquecidos em colesterol, esfingolipídios e
glicerofosfolipídios contendo ácidos graxos de cadeia longa (Simons & Raposo, 2009). Exossomos
correspondem a vesículas intraluminais (ILVs intraluminal vesicles) do compartimento
endossomal conhecido como corpo multivesicular (MVB multivesicular body). MVBs são
compartimentos intermediários da via endocítica que se fundem ao lisossomo/vacúolo para
degradação de seu conteúdo (Simons & Raposo, 2009). Alternativamente, MVBs podem fundir-se
com a membrana plasmática resultando na exocitose das ILVs (Figura 5), então denominadas
exossomos (Schorey & Bhatnagar, 2008). A secreção de exossomos é um mecanismo de secreção
não-convencional descrito em diversos tipos celulares como reticulócitos (Pan, et al. 1985),
linfócitos B (Raposo, et al. 1996), células dendríticas (Zitvogel, et al. 1998) e neurônios (Marzesco,
et al. 2005).
A formação das ILVs ocorre a partir de invaginações da membrana limitante do MVB e o
endereçamento para MVB envolve monoubiquitinação da proteína cargo (Figura 6)(Katzmann, et
al. 2004). Entretanto, algumas proteínas que não possuem sítio de ubiquitinação são
10
endereçadas ao MVB pois são preferencialmente segregadas em microdomínios de membrana
do tipo lipid rafts (de Gassart, et al. 2003). A formação de três complexos hetero-oligoméricos
precede a biogênese do MVB: ESCRT-I, ESCRT-II e ESCRT-III (endosomal sorting complex required
for transport). Estes complexos são formados majoritariamente por proteínas Vps (vacuolar
sorting protein). Mais de 60 produtos gênicos envolvidos no tráfego de proteínas entre Complexo
de Golgi e vacúolo foram identificados em leveduras, sendo o subset Vps classe E responsável
pelo endereçamento ao MVB (Hurley & Emr, 2006). Mutantes vps- classe E exibem acúmulo de
membranas endossomais e defeitos na formação das ILVs (Bowers & Stevens, 2005). Uma vez
que os complexos ESCRT são montados sobre a face citosólica do endossomo, ocorre o
brotamento de vesículas internas (Figura 6). A dissociação da maquinária ESCRT ocorre pela
ação da proteína Vps4 (Yeo, et al. 2003). Estudos de proteômica revelam a presença de diversas
proteínas citosólicas nestas vesículas, tais como actina, cofilina e piruvato quinase, o que é
topologicamente viável, uma vez que invaginações internas podem carrear aleatoriamente o
conteúdo citoplasmático para o interior das ILVs (Schorey & Bhatnagar, 2008).
Diversos estudos têm investigado a função de exossomos em diferentes modelos celulares.
Primeiramente, foi proposto que este mecanismo estaria implicado na eliminação de proteínas
Figura 5. Exossomos são liberados após a fusão
exocítica de MVBs com a membrane plasmática.
O experimento de pulso e caça com BSA
conjugado a partículas de ouro revelou que 20
minutos após a endocitose exossomos contendo
BSA-Gold foram liberados (Simons & Raposo,
2009). Símbolos: PM, membrana plasmática;
MVB, corpos multivesiculares.
11
obsoletas durante a maturação e diferenciação de reticulócitos (Johnstone, et al. 1984). Em
células dendríticas e linfócitos B, os exossomos participam da apresentação de antígenos
(Raposo, et al. 1996, Zitvogel, et al. 1998). Este mecanismo de apresentação parece ser
especialmente relevante no desenvolvimento da imunidade anti-tumoral (Wolfers, et al. 2001).
Foi demonstrado ainda que exossomos carreiam RNA mensageiros, os quais são transportados
para células vizinhas onde sofrem tradução (Valadi, et al. 2007), estabelecendo um novo modelo
de troca de material genético. Além disso, acredita-se que a liberação de exossomos é um dos
mecanismos de vigilância imunológica de patógenos intracelulares, uma vez que células
infectadas com Mycobacterium sp. (Bhatnagar, et al. 2007b; Beatty, et al. 2000), Salmonella
typhimurium (Bhatnagar, et al. 2007a) ou Toxoplasma gondii (Bhatnagar, et al. 2007a) produzem
exossomos contendo padrões moleculares destes patógenos, os quais são capazes de induzir
uma resposta pro-inflamatória em macrófagos não-infectados.
Recentemente foi descrito que células vegetais são capazes de produzir vesículas
extracelulares semelhantes a exossomos, a despeito da presença de parede celular (Regente et
al. 2009). Análises genômicas comparativas revelaram que alguns gêneros do domínio Archaea
apresentam proteínas homólogas ao complexo ESCRT-III, as quais atuam no remodelamento de
membranas e formação de vesículas (Makarova, et al. 2010). De fato, vesículas extracelulares
similares a exossomos foram isoladas de Sulfolobales e de algumas espécies de Thermococci
durante a resposta a estresse e a infecções virais (Ellen, et al. 2009; Soler, et al. 2008). Bactérias
também desenvolveram mecanismos de secreção vesicular, os quais parecem ser potentes
mecanismos de virulência em bactérias patogênicas (Kuehn & Kesty, 2005). Em bactérias Gram
negativas, a biogênese destas vesículas ocorre a partir da membrana externa à parede celular e
12
são chamadas OMVs (outer membrane vesicles). Fatores de virulência como β-lactamase,
fosfolipase C e fosfatase alcalina são encontradas nas OMVs de Pseudomonas aeroginosa
(Bomberguer, et al. 2009). Outras bactérias como Helicobacter pylori e Vibrio cholera também
secretam fatores de virulência via OMVs (Yonezawa, et al. 2009; Roy, et al. 2010). Por outro lado,
bactérias Gram positivas como Staphylococcus aureus e Bacillus anthracis também secretam
vesículas extracelulares, entretanto sua via de biogênese não é conhecida (Lee, et al. 2009;
Rivera, et al. 2010).
Figura 6. Papel dos complexos ESCRT na formação de MVBs. O receptor de superfície hipotético é
monoubiquitinado e ocorre formação do endossomo. Diversos componentes dos complexos ESCRT- I, -II e
-III se reúnem na face citosólica do endossomo e são responsáveis por promoverem invaginações de
membrane que levam a formação das ILVs. Complexos ESCRT também são responsáveis por manter a
especificidade das proteínas-alvo destinadas a esta via. (Adaptado de: Alam & Sundquist, 2007).
Outro mecanismo de secreção não-convencional proposto recentemente é dependente da
proteína GRASP (General receptor for phosphoinositides 1-associated scaffold protein) (Kinseth,
13
et al. 2007). Originalmente foi proposto que a GRASP localiza-se na matriz do Aparato de Golgi,
funcionando na manutenção da organização das cisternas (Barr, et al. 1997; Shorter, et al. 1999).
Entretanto diferentes trabalhos sugerem que GRASP apresenta uma segunda função em
mecanismos de secreção não-convencional (Kinseth, et al. 2007; Duran, et al. 2010, Manjithaya,
et al. 2010). Foi demonstrado que em Dictyostelium discoideum a proteína ligadora de acil-
coenzima A (AcbA) é secretada por meios não convencionais (Kinseth, et al. 2007). Fora da célula,
AcbA é clivada e gera um peptídeo que sinaliza para indução de esporulação (Anjard & Loomins,
2005). Mutantes deficientes na expressão da proteína GrpA (ortólogo de GRASP de mamíferos)
são incapazes de secretar AcbA e de induzir esporulação (Kinseth, et al. 2007). Posteriormente,
esse evento foi avaliado em leveduras e alguns outros componentes desta via não-convencional
de secreção foram identificados, como os genes relacionados à autofagia ATG5, ATG7, ATG8 e
ATG12; a t-SNARE de membrana plasmática SSO1 e os componentes da via de formação de MVB
VPS 23 E VPS4 (componentes dos complexos ESCRT-I e III, respectivamente) (Duran, et al. 2010).
Corroborando com estes dados, a proteína GRASP também parece ser importante na secreção
do homólogo de AcbA em Pichia pastoris (Manjithaya, et al. 2010). Acredita-se que a secrecão de
AcbA envolva a formação de autofagossomas, os quais convergem com a via de formação de
MVBs sendo então exocitados juntamente com os exossomos. Entretanto o papel da proteína
GRASP neste processo permanece obscuro (Duran, et al. 2010).
4. Cryptococcus neoformans: patógeno e modelo de estudo
C. neoformans é um fungo da ordem Basidiomycota, leveduriforme, agente etiológico da
criptococose. A principal característica morfológica deste fungo é a presença de uma cápsula
14
polissacarídica que circunda toda a célula a qual pode ser facilmente evidenciada por coloração
negativa usando tinta Nanquim (Figura 7). As cepas de C. neoformans são agrupadas em duas
variedades, de acordo com características sorológicas, bioquímicas e genéticas sendo estas: C.
neoformans var. neoformans (sorotipo D) e C. neoformans var. grubii (sorotipo A). O complexo
Cryptococcus inclui ainda outra espécie patogênica, o C. gattii (dividida nos sorotipos B e C)
(Franzot, et al. 1998, Steenbergen & Casadevall, 2003). C. gattii ocorre em áreas subtropicais e
pode infectar indivíduos imunocompetentes. o C. neoformans é um microrganismo
cosmopolita saprófito que causa infecções oportunistas em indivíduos deficientes em imunidade
celular.
A criptococose é contraída pela inalação de esporos ou leveduras dessecadas
(Chayakulkeeree & Perfect, 2006) e se estabelece inicialmente como uma pneumonia
assintomática. Estima-se que 99% da população apresenta anticorpos contra proteínas de C.
neoformans sem apresentar histórico clínico de infecção (Chen, et al. 1999). Acredita-se que
eventualmente a infecção pode permanecer em um estado de latência, e uma vez que o
hospedeiro se torna imunocomprometido a infecção assintomática pode ser reativada, causando
doença (Casadevall & Perfect, 1998). Embora o C. neoformans seja capaz de infectar a maioria
dos hospedeiros imunocompetentes, o desenvolvimento da criptococose ocorre em poucos
Figura 7. Microscopia óptica de campo claro de
uma levedura de C. neoformans contrastada com
tinta Nanquim em magnificação de 100x. O halo
branco em torno do corpo celular é o espaço de
repulsão do nanquim ocupado pela cápsula.
15
indivíduos, pois depende do estado imunológico do hospedeiro (Steenbergen & Casadevall,
2003). Estima-se que ocorra um milhão de casos de criptococose por ano em indivíduos HIV-
positivos, dos quais 600.000 são fatais (Park, et al. 2009). Em regiões mais carentes de assistência
médica, a sobrevivência média desses pacientes após o diagnostico da criptococose é de menos
de um mês (French, et al. 2002). Outros grupos de pacientes susceptíveis tem surgido como
aqueles submetidos a tratamentos com drogas anti-neoplásicas, submetidos a transplante de
órgãos, e hemodiálise (Casadevall & Perfect, 1998). Uma vez que a infecção pulmonar não é
contida, o microorganismo ganha a corrente sanguínea, se dissemina e atinge o sistema nervoso
central (CNS) causando um quadro grave de meningoencefalite criptocócica, o qual envolve
inflamação no espaço subaracnóide e no parênquima cerebral (Steenbergen & Casadevall 2003).
Para entender de que maneira ocorre esta doença, é importante conhecer as características
do C. neoformans que o tornam um patógeno. Neste sentido, existem fatores de virulência bem
definidos que seguem os requisitos estabelecidos no postulado molecular de Koch (Falkow,
1988). Estas são moléculas produzidas pelo C. neoformans cuja inativação por silenciamento ou
mutação do(s) gene(s) responsável(is) por sua expressão gera atenuação da virulência em
modelo animal. A complementação desses mutantes com as seqüências gênicas originais gera
restauração da virulência (Steenbergen & Casadevall 2003). Dentre estes fatores, destaca-se o
polissacarídeo capsular majoritário conhecido como glucuronoxilomanana (GXM).
A cápsula do C. neoformans é composta em 90% por GXM, molécula que possui peso
molecular variando entre 1.7 e 7 MDa (McFadden, et al. 2006). Esta molécula consiste em uma
cadeia linear de α-(1,3)-manana contendo um resíduo de ácido β-(1,2)-glucurônico a cada tríade
de manose. Resíduos de manose também podem ser 6-O-acetilados e receberem substituições
16
de β-(1,2) ou β-(1,4)-xilose (Cherniak and Sundstrom, 1994; Cherniak et al., 1988; McFadden et
al., 2006). A razão molar entre xilose: manose: ácido glucurônico varia de acordo com o sorotipo,
sendo 1:3:1; 2:3:1; 3:3:1 e 4:3:1 para os sorotipos D, A, B e C, respectivamente (Cherniak and
Sundstrom, 1994). A Figura 8 apresenta um esquema representativo da estrutura linear da GXM.
Além da GXM, a cápsula é constituída em 8% pelo polissacarídeo denominado galactoxilomanana
(GalXM) (Bose et al., 2003; Vaishnav et al., 1998), o qual apresenta uma cadeia linear de α-(1,6)-
galactana contendo cadeias laterais oligossacarídicas curtas de α-(1,3)- e α-(1,4)-manana, além
de grupos laterais de β-(1,2)- e β-(1,3)- xilose (Bose et al., 2003; McFadden et al., 2006; Vaishnav
et al., 1998). Especula-se que manoproteínas compõem minoritariamente a cápsula do C.
neoformans. Todavia, sua função na arquitetura capsular é pouco conhecida (Zaragosa, et al.
2009).
A maioria dos polissacarídeos microbianos que compõem a parede celular (tais como quitina
e glucanas) são sintetizados por enzimas presentes na membrana plasmática e durante a
biossíntese os polímeros são diretamente transportados para o espaço periplasmático (Leal-
Morales et al., 1994;
Valdivia & Schekman, 2003; Ortiz &
Novick, 2006). Ao contrário, a GXM é
sintetizada no ambiente intracelular (Feldmesser, et al. 2001) e transportada através de
mecanismos pouco conhecidos. Acredita-se que o transporte de GXM envolva o produto do gene
Figura 8. Um dos possíveis
motivos repetitivos de GXM. As
posições das substituições, bem
como o número de substituições,
variam de acordo com o
sorotipo.
17
CAP59, entretanto sua função não foi esclarecida (Garcia-Rivera, et al. 2004). A mutação do gene
SAV1, o qual codifica uma Rab-GTPase fundamental para o transporte a partir de TGN, levou ao
acúmulo de vesículas citoplasmáticas contendo GXM em C. neoformans, demonstrando que o
polissacarídeo é sintetizado no Aparato de Golgi (Yoneda & Doering, 2006). Além disso, o
tratamento de C. neoformans com Brefeldina A (um inibidor da via biossintética secretora) levou
a diminuição do diâmetro capsular, confirmando que a síntese de GXM envolve elementos do
Golgi (Hu, et al. 2007). A simples fusão de vesículas de transporte a membrana plasmática
permite que a GXM atinja o espaço periplasmático, entretanto não explica de que maneira o
polissacarídeo de alto peso molecular atravessa a parede celular. Estruturas semelhantes a
vesículas atravessando o espaço periplasmático foram observadas por criofratura e por TEM
(Takeo, et al. 1973; Rodrigues, et al. 2000), o que pode ser um indício de que C. neoformans
apresenta um mecanismo sofisticado de transporte de macromoléculas através da parede
celular.
Diversos estudos mostraram que a cápsula é um importante fator de virulência (Zaragoza, et
al. 2009). Os primeiros genes estão implicados na arquitetura capsular foram CAP59, CAP64,
CAP60 e CAP10. O primeiro a ser identificado foi o CAP59, uma vez que sua deleção por
recombinação homóloga resultou em um fenótipo acapsular e na perda da virulência em modelo
murino (Chang & Kwon-Chung, 1994). Embora sua função não seja totalmente esclarecida, sabe-
se que a proteína codificada pelo CAP59 apresenta homologia com uma α-1,3-manosiltransferase
(Sommer, et al. 2003). Além disso, CAP59 apresenta um provável domínio transmembrana e
estudos de ultraestrutura revelaram papel no transporte intracelular de GXM (Garcia-Rivera et
al., 2004). As deleções de CAP64, CAP60 e CAP10 também resultaram em fenótipo acapsular e
18
perda de virulência (Chang et al. 1996, Chang & Kwon-Chung, 1998, Chang & Kwon-Chung, 1999),
entretanto pouco se sabe sobre a função destas proteínas. Cap10p possui homologia de
seqüência com uma xilosiltransferase (Klutts et al., 2007) e quando conjugada a GFP apresenta-
se distribuída pelo citoplasma em grupos que assemelham-se a vesículas citoplasmáticas (Chang
& Kwon-Chung, 1999).
A cápsula es envolvida em diversos mecanismos de patogenicidade, incluindo proteção
contra fagocitose, modulação da resposta imunológica, propagação do fungo em tecidos
hospedeiros, e causa de danos de diversa natureza (Zaragoza, et al. 2009). É proposto que a
cápsula polissacarídica mascara estruturas da parede celular que seriam associadas a receptores
de reconhecimento de padrões (PRRs) (Vecchiarelli, 2000). A fagocitose do C. neoformans é
dependente de opsoninas como anticorpos (Kozel, et al. 1998) e iC3b (Kozel & Pfrommer, 1986).
A capacidade de sobreviver no interior de células fagocíticas é um mecanismo central na
patogênese do C. neoformans que parece ter sido adquirido ao longo da evolução através do
contato com predadores ameboides no meio ambiente (Feldmesser, et al. 2001). De fato, o C.
neoformans é considerado um patógeno intracelular facultativo capaz de explorar o ambiente
intracelular para replicação (Feldmesser, et al. 2000) (Figura 10). A cápsula polissacarídica parece
desempenhar um papel chave no parasitismo intracelular, uma vez que mutantes acapsulares
não são capazes de se replicar dentro de células fagocíticas (Feldmesser, et al. 2000). O escape
do C. neoformans durante a interação com fagócitos também está associado a um mecanismo de
extrusão, no qual ocorre replicação no interior de macrófagos e liberação dos fungos para o
espaço extracelular sem que haja danos, tanto para o fungo quanto para a célula hospedeira
(Alvarez & Casadevall, 2006; Ma, et al. 2006). Um mutante acapsular do C. neoformans não sofre
19
extrusão; o revestimento do mesmo com GXM exógena, entretanto, resulta na recuperação da
capacidade de extrusão da cepa selvagem (Alvarez & Casadevall, 2006). Além disso, foi
observado por video-microscopia que o C. neoformans é capaz de infectar macrófagos
diretamente a partir de células adjacentes infectadas, sem se expor ao espaço extracelular,
através de mecanismo conhecido como propagação célula-célula (Alvarez & Casadevall, 2007;
Ma, 2007). A infecção de macrófagos pelo C. neoformans leva ao acúmulo de vesículas
citoplasmáticas contendo GXM, o que interfere na função das células hospedeiras tornado-as
menos responsivas (Tucker & Casadevall, 2002).
É notável que durante o cultivo in vitro e na infecção pulmonar ocorre o crescimento
capsular (Littman, 1958; Rivera, et al. 1998). Este fenômeno é relevante durante a infecção, uma
vez que se observou em modelo in vivo que cepas incapazes de induzir o aumento diâmetro
capsular promovem uma eficiente resposta inflamatória nos pulmões e a infecção é contida
(Blackstock & Murphy, 1997; Blackstock et al., 1999). Na última década, o mecanismo de
crescimento capsular foi amplamente estudado. Acreditava-se que a cápsula crescia como
conseqüência do acúmulo de GXM na face externa da parede celular (Pierini & Doering, 2001).
Entretanto, foi demonstrado através de marcação covalente da cápsula que o crescimento se
de forma distal, isto é, as fibras de polissacarídeo são secretadas para o ambiente extracelular e
então reincorporadas as fibras pré-existentes (Figura 9) (Zaragoza, et al. 2006). Esta re-
incorporação se através de ligações iônicas entre as fibras e cátions divalentes presentes no
meio (Nimrichter, et al. 2007). De fato, altas concentrações de GXM são detectadas na forma
livre em meio de cultura (Frases, et al. 2008). In vivo, o polissacarídeo extracelular solúvel se
20
acumula na corrente sanguínea provocando diminuição da migração de leucócitos para os sítios
de infecção (Vecchiarelli, 2000).
Figura 9. Observação da cápsula do C. neoformans durante incubação em meio de indução de
crescimento capsular. As estruturas capsulares foram coradas inicialmente em verde por reação com o
componente C3 do sistema complemento, seguindo-se coloração com anticorpo monoclonal anti GXM
(vermelho). Este dado sugere que as fibras de GXM mais antigas permanecem junto ao corpo celular e
que a cápsula cresce de forma distal (Zaragoza, et al. 2006).
A síntese de melanina também tem sido associada à virulência no C. neoformans. Mutantes
albinos gerados por mutagênese randômica apresentaram perda de virulência (Williamson,
1997). A melanina é um pigmento sintetizado pela enzima lacase através da polimerização
oxidativa de compostos fenólicos como L-DOPA, dopamina, epinefrina e noreponefrina
(Williamson, 1997). A melanina encontra-se na parede celular, exercendo um papel protetor
contra estresse oxidativo, nitrosativo e contra agentes antimicrobianos (Polacheck, 1991;
Doering, et al. 1999). A lacase foi definida como fator de virulência uma vez que a deleção do
gene levou a perda de virulência e a subseqüente complementação do gene restaurou a
virulência do C. neoformans (Williamson, 1997). Acredita-se que o neurotropismo do C.
neoformans pode ser parcialmente explicado pela capacidade do fungo de utilizar
neurotransmissores tais quais epinefrina e dopamina como substratos para síntese de melanina
(Steenbergen & Casadevall, 2003). Foi descrito que a secreção de lacase é modulada in vivo,
21
ocorrendo preferencialmente no cérebro e sendo inibida nos pulmões, o que facilitaria a função
deste fator no neurotropismo (Waterman, et al. 2007).
Fosfolipase B1 também foi caracterizada como fator de virulência no C. neoformans (Cox, et
al. 2001). Esta enzima possui atividade de fosfolipase B, lisofosfolipase e transciclase e todas
estas atividades foram abolidas na cepa mutante deletada no gene PLB1 (Cox, et al. 2001).
Estudos in vivo tanto em camundongos quanto em coelhos demonstraram que a cepa plb1
apresenta perda significativa de virulência quando comparada a virulências das cepas selvagem e
reconstituída (Cox, et al. 2001). Além disso, foi demonstrado que isolados clínicos de C.
neoformans apresentam atividade significativamente maior de PLB1 que isolados do meio
ambiente (Ghannoum, 2000). Um modelo de infecção murina revelou que uma correlação
entre os níveis de atividade de fosfolipase secretada (baixa, intermediária ou alta) e a virulência
das cepas correspondentes (Ghannoum, 2000). O mecanismo de virulência associado a PLB1
secretada deve envolver a hidrólise de fosfolipídios nas membranas das células hospedeiras. Cox
e colaboradores demonstraram que a cepa plb1
apresenta dificuldade de replicação no interior
de monócitos, o que pode estar relacionado à incapacidade de degradar a membrana do
fagossomo (Cox, et al. 2001). Além disso, um dos principais surfactantes do pulmão é um
substrato de PLB1: dipalmitoil-fosfatidilcolina. A degradação do surfactante pode contribuir no
estabelecimento da infecção (Cox, et al. 2001).
Outra importante molécula definida como determinante de virulência do C. neoformans é a
glucosilceramida (GlcCer) (Rittershaus, et al. 2006). Este glicoesfingolipídio é uma molécula de
superfície sendo encontrada majoritariamente na parede celular (Rodrigues, et al. 2000). A
deleção da enzima glucosilceramida sintase I (GCSI) levou a perda de virulência pelo C.
22
neoformans, a qual foi restaurada com a complementação do gene (Rittershaus, et al. 2006).
Sabe-se que a GlcCer é altamente imunogênica (Rodrigues, et al. 2000) e que anticorpos
monoclonais anti-GlcCer são capazes de proteger camundongos contra doses letais de C.
neoformans (Rodrigues, et al. 2007). A ligação de anticorpos aos sítios de brotamento na parede
celular inibe diretamente a replicação (Rodrigues, et al. 2000). Acredita-se que a GlcCer participa
do controle do ciclo celular em pH fisiológico, porém o mecanismo de patogenicidade ainda não
está totalmente elucidado (Rittershaus, et al. 2006). Através do mesmo princípio de deleção e
complementação genética, descreveu-se que a enzima urease também é um fator de virulência
no C. neoformans (Cox, et al. 2000). Urease catalisa a degradação de uréia a amônia e
carbamato, alcalinizando localmente o ambiente, entretanto sua contribuição para o mecanismo
de patogenicidade não é conhecida (Casadevall & Perfect, 1998).
Todos os fatores de virulência descritos acima têm em comum o fato de serem secretados
para o ambiente extracelular, onde vão agir destruindo componentes do hospedeiro (Cox, et al.
2001), protegendo o C. neoformans de ataques do sistema imune (Williamson, 1999, Zaragoza, et
al. 2008) ou modulando negativamente a imunidade do hospedeiro (Vecchiarelli, 2000).
5. Resposta imune anti- C. neoformans:
Diversos mecanismos tanto da imunidade inata quanto da adaptativa estão relacionados a
proteção contra a criptococose. Além de barreiras físicas como pele e mucosa do trato
respiratório, o sistema complemento e células fagocíticas efetoras exercem papel fundamental
na resposta não- específica a infecção (revisado em Voelz & May, et al. 2010).
23
O C. neoformans é capaz de ativar tanto a via clássica (mediada por anticorpo) quanto a via
alternativa do Complemento (via hidrólise espontânea de C3 e deposição na superfície
microbiana) (Kozel, et al. 1998 b). Observações de modelos animais revelaram que cobaios
depletados de proteínas do complemento (de C3 a C9) não são aptos a eliminar a infecção por C.
neoformans (Diamond, et al. 1973; Diamond, et al. 1974). Ambas a vias convergem para a
formação do complexo C3- convertase (C4b2a para via clássica e C3bBb para via alternativa) que
leva a hidrólise de C3 em C3a e C3b. C3b age como uma potente opsonina facilitando a
fagocitose via receptores do complemento (CR1, CR3 ou CR4) (Levitz, et al. 1997) e ao mesmo
tempo compõe o complexo C5 convertase que cliva C5 em C5a e C5b. C5a age juntamente com
C3a como um mediador da inflamação e quimioatraente de células fagocíticas efetoras. C5b
inicia a formação do complexo de ataque a membrana (C5b, C6, C7, C8, C9) (Dessauer, et al.
1984), entretanto o C. neoformans é resistente a lise celular mediada por este complexo
(Shapiro, et al. 2002). Camundongos deficientes em C5 são mais suceptíveis a injeção intravenosa
de C. neoformans e morrem três vezes mais rápido de pneumonia aguda do que camundongos
que expressam C5 (Rhodes, 1985; Rhodes, et al. 1980). Cobaios deficientes em C4 apresentaram
o mesmo tempo de sobrevivência dos selvagens após infecção por C. neoformans, indicando que
a via alternativa do complemento tem maior importância na proteção anti- C. neoformans
(Diamond, et al. 1973). Entretanto foi demonstrado que a via clássica é necessária para que haja
uma cinética ótima de opsonização (Diamond, et al. 1974).
A fagocitose do C. neoformans pode ocorrer via receptores do complemento, receptores Fc e
através do reconhecimento direto de componentes de superfície. A GXM é capaz de se ligar aos
receptores TLR4 e CD14 (Shoham, et al. 2001; Barbosa, et al. 2007) promovendo internalização.
24
Dímeros TLR2/TLR1 e TLR2/TLR6 também reconhecem GXM, entretanto não se sabe se estes
receptores são capazes de mediar a fagocitose (Fonseca, et al. 2010). Receptores de manose de
células dendríticas (DCs dendritic cells) e receptores CD18 se ligam a manoproteínas da cápsula
(Pietrella, et al. 2005; Dong & Murphy, 1997).
Neutrófilos parecem contribuir fortemente com a resposta anti- C. neoformans. Desafios com
C. neoformans induzem uma rápida migração de células polimorfonucleares, as quais promovem
uma intensa resposta oxidativa local (Miller & Mitchell, 1991). Foi observado que uma intensa
neutropenia em camundongos está associada a um prolongado período de sobrevivência a
infecção pulmonar pelo C. neoformans, sugerindo que possivelmente os neutrófilos exercem
uma função imunoregulatória além de antimicrobiana (Mednick, et al. 2003).
O C. neoformans desenvolveu estratégias únicas para sobreviver no interior de macrófagos,
sendo considerado um patógeno intracelular facultativo (Feldmesser, et al. 2001) (Figura 10).
Diferentemente de outros patógenos como Listeria monocytogenes e Shigella flexneri, o C.
neoformans sobrevive no interior de fagolisossomos e não necessita de mecanismos de escape
para colonização do citoplasma (Levitz, et al. 1999; Southwick & Purich, 1996). Mais do que isso,
o C. neoformans não promove inibição da fusão fagossomo- lisossomo a exemplo da Legionella
pneumophila, (Horwitz, et al. 1983) nem tampouco interfere com a maturação e acidificação do
fagossomo, como se observa durante a infecção por Histoplasma capsulatum e Micobacterium
spp (Strasser, et al. 1999; Sturgill-Koszycki, et al. 1994). Na realidade, o C. neoformans replica-se
no interior de fagolisossomos e elevações no pH deste compartimento levam a redução de sua
taxa de proliferação (Levitz, et al. 1997). Em última análise, a proliferação do C. neoformans
acarreta em lise celular (Del Poeta, 2004; Feldmesser et al. 2000) (Figura 10). Alternativamente,
25
mecanismos de extrusão discutidos anteriormente nesta tese permitem que a levedura ganhe o
espaço extracelular novamente (Alvarez & Casadevall, 2006; Ma, et al. 2006). O ambiente
intracelular de macrófagos é claramente benéfico para este microorganismo e acredita-se que
ele permaneça durante anos colonizando este ambiente em um estado de latência (Garcia-
Hermoso, et al. 1999; Goldman, et al. 2001). Além disso, é possível que o C. neoformans se utilize
de um mecanismo de “Cavalo de Tróia” para atravessar a barreira hematoencefálica, sendo
carreados por monócitos para o interior do sistema nervosa central (Charlier, et al. 2009).
Figura 10. Representação esquemática de um macrófago infectado pelo C. neoformans. Após a
fagocitose ocorre a fusão fagolisossomal. A membrana do fagolisossomo se rompe e o acúmulo de
vesículas citoplasmáticas contendo GXM. O C. neoformans se replica no interior do compartimento
fagolisossomal. Após a lise celular, as leveduras são liberadas no meio extracelular (Adaptado de:
Feldmesser, et al. 2001).
Na infecção por C. neoformans, células dendríticas (DCs) funcionam como as principais
células apresentadoras de antígenos, as quais iniciam uma imunidade adaptativa celular
26
protetora (Bauman, et al. 2000). DCs induzem a maturação de linfócitos T de maneira mais
eficiente de macrófagos alveolares ou peritoniais (Osterholzer, et al. 2009).
Tanto o componente humoral quanto o celular são importantes na resposta immune
adaptativa ao C. neoformans. A eficiência da resposta humoral é dependente da especificidade
do anticorpo (Mukherjee, et al. 1995). Evidencias de modelos murinos mostram ainda que o
efeito protetor de anticorpos relaciona-se a imunidade mediada por célula T. Camundongos
deficientes em células CD4 positivas ou IFN- γ não são protegidos contra infecção através da
administração passiva de IgG1 protetor, enquanto camundongos deficientes em células CD8
positivas, células NK ou C3 são protegidos (Beenhouwer, et al. 2001; Shapiro, et al. 2002; Yuan,
et al. 1997). Portanto, a imunidade adaptativa humoral faz parte da defesa do hospedeiro e está
integrada a demais elementos do sistema imune inato e adaptativo.
A característica comum dos fatores de risco associados ao desenvolvimento da
criptococose (por exemplo infecção por HIV, tratamento com corticosteróides, leucemia) é a
perda da funcionalidade plena da imunidade adaptativa celular (CMI cell mediated immunity).
A CMI age no controle da infecção através do efeito citotóxico direto ou regulando a função de
fagócitos e células NK (Levitz, et al. 1994). Os efeitos regulatórios da CMI dependem do status
imunológico do indivíduo infectado. Citocinas do tipo Th1 estão associadas a imunidade
protetora enquanto do tipo Th2 estão associadas a suceptibilidade (Hoag, et al. 1997). O
aumento na produção de IFN-γ e de TNF-α associam-se ao controle da infecção (Flesch, et al.
1989; Kawakami, et al. 1995; Milam, et al. 2007), enquanto camundongos deficientes na
produção de IFN-γ apresentam aumento da carga fúngica (Arora, et al. 2005). Foi demonstrado
recentemente que a citocina IL-17 produzida por células Th17 também exerce importante papel
27
na sobrevivência de camundongos infectados com C. neoformans (Kleinschek, et al. 2006; Muller,
et al. 2007). Por outro lado, citocinas do tipo Th2 como IL-4 e IL-13 associam-se a uma redução
na capacidade de combater o C. neoformans (Blackstock & Murphy, 2004; Kawakami, et al.
1999). A progressão da infecção em indivíduos HIV-positivos correlaciona-se com perda da
resposta Th1 (Altfed, et al. 2000) e com aumento do perfil de citocinas do tipo Th2 em indivíduos
transplantados (Singh, et al. 2008). Portanto, o balanço Th1-Th2-Th17 é essencial para o controle
da infecção.
28
II Objetivos:
Objetivo Geral:
Os mecanismos pelos quais macromoléculas atravessam a parede celular de fungos ainda são
pouco compreendidos. Esta é portanto uma questão central na biologia celular de organismos
dotados de parede celular. Nesse contexto, esta tese visa caracterizar as estruturas celulares
carreadoras de macromoléculas, bem como a maquinária molecular necessária para que o
transporte através da parede celular ocorra. A caracterização de moléculas transportadas por tal
via e a função deste mecanismo de secreção também foram abordados utilizando S. cerevisiae
como organismo modelo de biologia celular e em modelo de infecção celular pela levedura
patogênica C. neoformans.
Objetivos específicos:
1) Compreender como moléculas tais como GXM e CMH produzidas pelo C. neoformans
atravessam a parede celular e atingem o espaço extracelular. Caracterizar a existência de
ultraestruturas semelhantes a vesículas visualizadas na parede celular e isolar tais
estruturas dos sobrenadantes de cultura de C. neoformans.
2) Através de crioultramicrotomia, caracterizar ultraestruturas intracelulares envolvidas no
transporte de GXM. Avaliar a existência de subpopulações de vesículas que diferem
quanto a composição.
29
3) Determinar o impacto das vesículas secretadas pelo C. neoformans na interação com
macrófagos murinos, incluindo a produção de citocinas e indução de atividade
microbicida.
4) Avaliar que genes estão potencialmente envolvidos na formação/liberação de vesículas
extracelulares. Isolar, caracterizar morfologica e bioquimicamente vesículas
extracelulares isoladas de diferentes mutantes de vias do tráfego intracelular utilizando S.
cerevisiae como organismo modelo.
30
III - Materiais e Métodos:
A. Cultivo de leveduras:
As cepas de C. neoformans utilizadas nesse trabalho foram: ATCC 24067 (sorotipo D), B3501
(sorotipo D), H99 (sorotipo A, isolado clínico), HEC3393 (sorotipo A, isolado clínico) e Cap67
(sorotipo D, mutante acapsular). As células foram mantidas meio Sabouraud, composto por 20%
de dextrose e 10% de peptona. Para o isolamento de vesículas as células foram crescidas em1L
de Meio Mínimo, composto por Dextrose (15 mM), MgSO
4
(10 mM), KH
2
PO
4
(29.4 mM), glicina
(13 mM), and tiamina (3 µM). As células foram cultivadas a temperatura ambiente por 72 horas
sob agitação.
As cepas de S. cerevisiae usadas neste estudo foram RSY255, RSY113, SEY6210 e BY4741, as
quais apresentam fenótipo selvagem (WT). Diferentes mutantes de vias de tráfego intracelular
também foram utilizados, os quais estão listados na Tabela I do Artigo IV e serão descritos a
seguir. As cepas RSY782, SF2642-1D, e RSY954 são respectivamente, mutantes temperatura-
restritivos dos genes essenciais sec1-1, sec4-2 e bos1-1/sec32-1. Estes estão relacionados a via
secretória clássica. As cepas EEY6-2 and EEY9 correspondem a mutantes deletados nos genes
vps23 e snf7/vps32, respectivamente, os quais estão relacionados a via de secreção de
exossomos. A cepa Grh1 é defectiva na expressão da proteína homologa a GRASP Grh1p.
Células WT e mutantes foram pré-inoculadas em meio Sabouraud, por 24 horas a temperatura
ambiente e sob agitação. Para o isolamento de vesículas as células foram crescidas em 1L de
meio Sabouraud por 48h a temperatura ambiente e sob agitação. Para os mutantes temperatura-
restritivos, após 48h de crescimento as células foram centrifugadas e transferidas para 1L de
31
Sabouraud e incubadas por 16h a 37
o
C. Diferentes tempos de incubação a 37
o
C (1, 3 e 16h)
foram utilizados no experimento de cinética de secreção do mutante sec4-2. Controles de
viabilidade celular na temperatura de restrição foram feitos através da contagem de unidades
formadoras de colônia (UFC).
B. Isolamento de vesículas:
Após o crescimento celular nas condições descritas acima, as células foram centrifugadas a
5.000 x g a 4
o
C por 15 minutos. Os sedimentos foram descartados e os sobrenadantes coletados
e novamente centrifugados sucessivamente a 10.000 x g e 15.000 x g a 4
o
C por 15 minutos,
garantindo a retirada de células e fragmentos celulares. Os mesmos sobrenadantes foram
filtrados em uma membrana de policarbonato com poro correspondente a 0.8µm e
posteriormente concentrados aproximadamente 20 vezes usando um sistema de ultrafiltração
em membrana com cutoff de 100 kDa. Os sobrenadantes concentrados foram então submetidos
a ultracentrifugação a 100.000 x g por 1h a 4
o
C. Os sedimentos gerados foram lavados 2 vezes
em PBS, sendo seqüencialmente ressuspensos e ultracentrifugados novamente. Para remover
GXM extravesicular as preparações de vesículas de C. neoformans foram passadas em uma
coluna cromatográfica de afinidade, na qual o anticorpo monoclonal anti GXM Mab18B7 foi
mobilizado em partículas de Sepharose. As frações não ligadas a coluna foram coletadas. O
material contido no sedimento obtido por centrifugação a 100.000 x g foi analisado por
diferentes métodos descritos a seguir. Em todas as análises quantitativas de componentes
vesiculares as preparações foram normalizadas pelo número de células através de contagem em
câmara de Neubauer. Nos estudos de interação com macrófagos, vesículas de C. neoformans
32
foram quantificadas e normalizadas através da dosagem de esterol pelo kit fluorimétrico Amplex
Red (Molecular Probes). Em diferentes ensaios foram detectados entre 6 e 10 µg de esterol a
partir de 2 L de cultura de C. neoformans, sugerindo que 0.5 fg de esterol são recuperados por
célula. Esta aparente baixa eficiência é esperada já que as vesículas supostamente se rompem no
sobrenadante de cultura para liberação de componentes secretados.
C. Análise de Lipídeos Neutros:
Os sedimentos obtidos após a ultracentrifugação a 100000xg foram submetidos a secagem
por centrifugação a vácuo e posteriormente ressuspensos em uma mistura de CHCl
3
: metanol:
H
2
O na proporção 8/4/3 v/v/v. As misturas foram vigorosamente agitadas. Usualmente, duas
fases se formam. A fase superior corresponde a fração aquosa e contém proteínas e açúcares,
além de outros componentes hidrofílicos. A fase inferior corresponde a fração orgânica e contém
os lipídeos neutros. A fase inferior foi levada a secagem em atmosfera de N
2
e submetida a
partição de Folch, a qual consiste a partição das moléculas extraídas em uma mistura de CHCl
3
:
metanol: KCl 0.25% na proporção 8:4:3. A fase inferior foi então submetida a análise por
cromatografia em camada fina de alta performance (HPTLC). Para análise de esteróis, os estratos
lipídicos foram aplicados em placas de sílica e separados usando um sistema de solventes
contendo hexano: éter: ácido acético na proporção 40: 20: 1 v/v/v. As placas foram borrifadas
com uma solução contendo 0.05% de FeCl
3
, 5% de ácido acético e 5% de ácido sulfúrico. As
placas foram aquecidas a 100 C por 5 minutos e as bandas equivalentes a esteróis surgiram em
cor violeta. Como controle foi usado um padrão de ergosterol (Sigma) e as bandas que migraram
como mesmo índice de retenção (rf) que o padrão foram identificadas. A presença de
33
glucosilceramida (CMH) foi avaliada usando o sistema de solventes CHCl
3
: metanol: H
2
O na
proporção 65: 25: 4 v/v/v. As placas foram reveladas borrifando uma solução de orcinol sulfúrico
seguido de incubação a 100 C por 3 minutos.
A fase inferior da partição de Folch também foi analisada por espectrometria de massas em
modo de ionização por eletrospray (ESI-MS), utilizando o instrumento Finnigan LCQ DUO ion trap
(Thermo Electron, San Jose, CA). As amostras foram diluídas em CHCl
3
:metanol 1:1, v/v,
contendo 10mM de iodeto de lítio e injetadas no equipamento de ESI-MS operando em fluxo de
10µl/minuto. A análise foi realizada em modo positivo. As voltagens da fonte e do capilar foram
respectivamente de 4.5 kV e 3V. A temperatura do capilar foi mantida a 200
o
C. Os espectros
foram coletados em um intervalo de massa sobre carga (m/z) correspondendes a 300 a 800. A
energia de dissociação na fonte utilizada foi de 25 V. Os experimentos de ativação colisional
induzida (MS/MS) foram realizados com percentual de energia entre 20 - 60% (1-3 eV). Todos os
espectros foram processados usando Xcalibur software (Thermo Electron). Os experimentos aqui
descritos foram realizados em equipamento disponível no Instituto de Ciências Biológicas da
Universidade de São Paulo.
D. Análise de Fosfolipídios:
A análise de fosfolipídios foi feita a partir de vesículas isoladas de C. neoformans. O
sedimento de contendo vesículas foi preparado como descrito acima e os lipídeos resultantes
foram ressupensos em 90% de metanol. Alíquotas de 100 µl foram injetadas em cromatógrafo
líquido equipado com uma coluna cromatográfica de fase reversa (Supleco Ascentis Express, C18,
150x2.1mm), equilibrada em solução de metanol: H
2
O, 95:5, v/v, contendo 1mM de acetato de
34
amônio. As amostras foram eluídas em concentrações crescentes de misturas de clorofórmio-
água, 500/0,2, v/v, contendo acetato de amônio 1 mM. As frações eluídas foram coletadas
automaticamente (intervalos de 1 minuto) e submetidas a análise por ESI-MS conectada a um
espectrômetro de massas do tipo quadrupolo/tempo de vôo (QTOF-MS, PE Sciex API III
+
),
operando no modo de varredura do pico parentalcom m/z de 184,1 (característico de
fosfatidilcolina). Dessa forma, a câmara de colisão foi preenchida com argônio e o detector foi
programado setado para transmitir os sinais oriundos do pico 184.1 m/z. Para confirmar as
assinalações feitas a partir dos dados de LC/MS/MS, e para melhor definir as cadeias alifáticas da
fosfatidilcolina na amostra, espectros de íons positivos e negativos foram coletados a partir da
injeção direta de frações coletadas durante LC/MS/MS. As condições experimentais foram
otimizadas utilizando padrões de di-palmitoil-fosfatidilcolina, heptadecanoil-liso-fosfatidilcolina e
di-heptanoilfosfatidilcolina. Estes experimentos foram feitos utilizando métodos originalmente
descritos por Jensen, et al. (Jensen, et al. 1986) e, nessa tese, foram realizados com a
colaboração do Dr. Kym Faull (Pasarow Mass Spectrometry Laboratory, Semel Institute for
Neuroscience and Human Behavior and the Department of Psychiatry and Biobehavioral
Sciences, David Geffen School of Medicine, University of California, Los Angeles, EUA).
E. Detecção de GXM por ELISA:
Para inferir se as vesículas isoladas do sobrenadante de cultura do C. neoformans continham
GXM, foi usado ELISA de captura. As amostras de vesículas correspondem a fração aquosa após a
extração lipídica. Placas de 96 poços foram revestidas com 1 µg/ml do anticorpo monoclonal
Mab 12A1, uma IgM específica para GXM. Após 1h de incubação a placa foi bloqueada com PBS
1% BSA incubando por 1h a temperatura ambiente. As placas foram lavadas 3 vezes com PBS-
35
0.05% Tween. GXM purificada foi usada como curva-padrão. Curva-padrão e amostras foram
incubadas por 1h a temperatura ambiente e novamente lavadas 3 vezes com PBS-0.05% Tween.
As placas foram então incubadas com 1 µg/ml do anticorpo de detecção, Mab18B7, uma IgG de
camundongo anti-GXM. Após uma hora de incubação a 25 C as placas foram novamente lavadas
3 vezes e incubadas com 1 µg/ml do anticorpo secundário anti-IgG de camundongo acoplado a
fosfatase alcalina por 1h. Após nova etapa de lavagem as placas foram incubadas com o Tampão
Glicina-pH 9.0 contendo 0,2%de cloreto de zinco e cloreto de magnésio, bem como 0.1% do
substrato p-nitrofenil-fosfato dissódico hexahidratado. Os resultados foram lidos a 405 nm
utilizando um espectrofotômetro.
F. Análise da detecção de vesículas em suspensões de células não viáveis:
Para averiguar se a secreção de vesículas é um fenômeno fisiológico e portanto dependente
da viabilidade celular ou se seria fruto de artefatos oriundos de células mortas, a presença de
lipídeos marcadores de vesículas nos sedimentos 100.000 x g foi avaliada nas seguintes
condições: i) Cultura de C. neoformans foi crescida em Meio Mínimo por 72h; ii) Após o
crescimento por 72h células foram tratadas com 100mM de azida de sódio por 1h a 25 C e iii)
após 72h de crescimento células foram incubadas a 50
o
C por 1h. As preparações de vesículas
foram normalizadas pelo número de células nas três condições, através de contagem em Câmara
de Neubauer. A viabilidade celular foi acompanhada inoculando-se células controle, tratadas
com NaN
3
e incubadas a 50
o
C em placas de Ágar Sabouraud. Após as incubações, as células
foram removidas e as vesículas foram isoladas dos sobrenadantes, conforme descrito acima. Os
sedimentos de vesículas foram submetidos a análise de lipídeos neutros descrita previamente.
36
G. Ensaio de formação extracelular de vesículas:
A fim de avaliar se as vesículas se formariam de forma randômica como resultado de
agregação aleatória de lipídeos e GXM liberados pelas células, o mutante acapsular do C.
neoformans, Cap67, foi crescido em Meio Mínimo. A este meio, foi adicionada GXM a 15 µg/ml,
concentração previamente estabelecida em sobrenadantes da cepa capsulada. Após 72h de
crescimento sob esta condição, vesículas foram isoladas do sobrenadante. Em paralelo células
da cepa Cap67 foram crescidas na ausência de GXM, a qual foi adicionada na concentração de 15
µg/ml após a remoção das células. Os sobrenadantes contendo GXM foram ultracentrifugados e
o sedimento 100000 x g foi lavado três vezes e submetido a cromatografia de afinidade em
resina de sefarose acoplada ao Mab18B7. A GXM presente nas vesículas, bem como nos
sobrenadantes foi então quantificada por ELISA de captura. Sobrenadante de Cap67 foi usado
como controle negativo.
H. Ensaio de reincorporação de GXM:
A cepa acapsular de C. neoformans Cap67 foi crescida por 48h em Meio Mínimo, lavada 3
vezes em PBS e ajustada para 10
6
células. Estas, foram incubadas em 100µl de suspensão
vesicular purificada da cepa capsulada HEC3393. Em paralelo, a concentração de GXM desta
mesma amostra foi dosada por ELISA e correspondeu a 10 µg/ml. A suspensão de células e
vesículas foi incubada por 12h a 25
o
C e então as células foram lavadas extensivamente em PBS e
fixadas em PBS 4% paraformaldeído. As células foram lavadas 3 vezes em PBS, bloqueadas por 1h
em PBS 5% BSA e posteriormente incubadas com o anticorpo anti-GXM MAb18B7 a 1µg/ml por
1h. Após 3 lavagens em PBS, as células foram incubadas com o anticorpo secundário anti- IgG
37
conjugado a FITC. Laminas foram montadas e as leveduras foram observadas em um microscópio
de fluorescência Axioplan 2 (Zeiss, Alemanha). As imagens foram adquiridas usando uma câmera
digital Color View SX e processadas usando o software analySIS (Soft Image System). Como
controle, o Mab18B7 foi substituído por uma IgG irrelevante. Como controle positivo as células
foram incubadas com GXM solúvel, purificada como descrito previamente (Nimrichter, et al.
2007).
I. Aferição de tamanho de cápsula:
Para induzir a expressão de cápsula no C. neoformans, células foram incubadas em meio
Sabouraud dez vezes diluído em 50mM de MOPS pH 7.3, conforme descrito anteriormente
(Zaragoza & Casadevall, 2004). Após incubação por 6 e 24h a 37
o
C sob agitação, as células foram
centrifugadas a 2000 x g por 10 minutos e fixadas com PBS 4% de paraformaldeído. Após a
fixação as células foram lavadas com PBS e a cápsula foi evidenciada com tinta nanquim. O
tamanho da cápsula foi medido por microscopia óptica de campo claro. O tamanho da cápsula é
definido como a distância entre a parede celular e a borda externa da cápsula. Esta medida
morfométrica foi determinada usando o software ImageJava (http://rsb.info.nih.gov/ij/). Todos
as medidas foram feitos em triplicatas e foram estatisticamente validados pelo teste t Student’s.
J. Microscopia Eletrônica de Transmissão (TEM):
TEM foi usada para visualizar vesículas tanto de C. neoformans quanto de S. cerevisiae. Foram
analisadas preparações de vesículas isoladas e vesículas associadas a leveduras durante a
infecção de tecido pulmonar in vivo. Para as preparações a partir de vesículas isoladas, os
38
sedimentos obtidos após as lavagens a 100000xg foram fixadas em 2% de glutaraldeído em 0.1M
de cacodilato a temperatura ambiente por 2h e em seguida foram incubados por 16h em 4% de
formaldeído, 1% de glutaraldeído em PBS. Posteriormente as amostras foram incubadas por 90
minutos em 2% de OsO
4
, desidratadas serialmente em etanol e embebidas em Spurr. Cortes
ultrafinos foram obtidos em um Reichert Ultracut e contrastados em acetato de uranila 0.5%.
Amostras foram observadas em um microscópio eletrônico de transmissão JEOL 1200EX
operando à 80kV.
Para detecção de vesículas in vivo, foram feitas preparações de tecido pulmonar de
camundongos infectados com C. neoformans da cepa ATCC 24067. Camundongos C57BL/6 foram
infectados com 1x10
4
leveduras de forma intratraqueal e em seguida sacrificados com 2h, 48h ou
7 dias de infecção. O processamento para TEM foi feito conforme previamente descrito
(Feldmesser, et al. 2001).
K. Crioultramicrotomia e imuno detecção:
Este procedimento foi baseado em estudos pioneiros da década de 70 (Tokuyasu, 1973).
Células foram fixadas em paraformaldeído 4%, glutaraldeído 0.2% e ácido pícrico 1% diluídos em
0.1M de cacodilato de sódio pH 7.2. Após 60 minutos a temperatura ambiente as células foram
lavadas 3 vezes, infiltradas em gelatina, cortadas em cubos de 1mm e infiltradas em
polivinilpirrolidona 25% (PVP) e sacarose 2.3M por 16h a 4
o
C. Os blocos embebidos em PVP
foram montados em um suporte e congelados rapidamente por imersão em N
2
líquido.
Criossecções foram obtidas a -130
o
C em um crioultramicrotomo Ultracut UCT (Reichert). Os
cortes foram coletados com uma alça e transferida para grades de níquel revestidas com
39
películas de Formivar. Para a imunomarcação, as grades foram bloqueadas com PBS 1% BSA por
1h. As grades foram então incubadas com o anticorpo primário. O anti-GXM de camundongo
Mab18B7 foi usado a 1µg/ml, enquanto anticorpos anti-CMH foram isolados de soro de
pacientes como previamente descrito (Rodrigues, et al. 2000). As grades foram lavadas em PBS
por 3 vezes de 10 minutos e posteriormente incubadas com anti- IgG de camundongo ou anti-
Humano conjugados a partículas de ouro de 15 nm. Após uma série de lavagens em PBS e H
2
O
destilada as grades foram deixadas por 15 minutos em 2% de acetate de uranila aquosa e
transferida para uma gota de 0.75% de metilcelulose. Após a secagem das grades, as amostras
foram observadas em um microscópio eletrônico de transmissão JEOL 1200EX operando a 80kV.
L. Fracionamento de vesículas em gradiente de densidade:
Vesículas de C. neoformans foram obtidas por ultracentrifugação como descrito
anteriormente. Para identificar diferentes subpopulações de vesículas um gradiente de Optiprep
foi preparado como previamente descrito (Cantin, et al. 2008). O gradiente foi preparado
variando de 18% a 6% em incrementos de 1.2%. Ao todo 11 frações de 300 µl foram diluídas em
PBS a partir da solução estoque de 60% de Optiprep. As vesículas foram aplicadas sobre o
gradiente e ultracentrifugadas em um rotor de ângulo móvel, SW50 (Beckman) a 250000xg por
75 minutos. Onze frações foram coletadas a partir da parte superior e cada fração foi analisada
quanto a presença de e esterol e GXM.
Para identificar vesículas produzidas durante a infecção in vitro de macrófagos, o sedimento
100000xg obtido como no protocolo descrito anteriormente e foi submetido a um gradiente de
sacarose. Após a purificação das vesículas, estas foram homogeneizadas em uma solução com
40
concentração de 85% (peso/volume) de sacarose em TBS, gerando uma concentração final de
42.5% de sacarose. Esta suspensão foi transferida para um tubo de ultracentrífuga e uma solução
de 35% de sacarose foi aplicada sobre a anterior, seguido de uma última camada de 5% de
sacarose em TBS. O gradiente foi centrifugado por 18h a 200000xg e frações de 0.25 ml foram
coletadas a partir do topo do gradiente. As frações foram extensivamente dialisadas contra TBS,
secadas por centrifugação a vácuo e preparadas para análise lipídica em HPLC e detecção de
GXM, conforme descrito acima.
M. Cultivo de macrófagos murinos:
As linhagens celulares de macrófagos murinos RAW 264.7 e J774.16 (obtidas a partir da
ATCC), foram cultivadas em meio mínimo essencial de Dulbecco (DMEM) suplementado com 10%
de soro fetal bovino (FCS), 2mM de L- glutamina, 1mM de piruvato de sódio, aminoácidos não-
essenciais (Gibco-Invitrogen cat. N. 11360), 10mM de HEPES e 50 mM de 2-β-mercaptoetanol. As
células foram mantidas a 37
o
C em atmosfera de 7.5% de CO
2
e sob condições livres de LPS.
N. Incorporação de componentes vesiculares por macrófagos:
Células RAW264.7 foram plaqueadas em placas de 24 poços contendo lamínulas na
proporção de 1x10
5
células por poço. Amostras de vesículas foram coradas com o fluoróforo
lipofílico dialquilcarbocianina (DiI) (Invitrogen, V22885). Suspensões de vesículas foram
normalizadas para 2 µg/ml de esterol e suplementadas com DiI em uma concentração final de
3µM. Após 30 minutos a temperatura ambiente, a suspensão de vesículas foi ultracentrifugada a
100000xg por 1h e lavada 3 vezes em PBS. As vesículas coradas com DiI foram então incubadas
41
com os macrófagos por 30 minutos a 37
o
C, 7.5% CO
2
. Como controle incubamos meio estéril
com DiI, ultracentrifugamos e lavamos 3 vezes, sob as mesmas condições que a suspensão de
vesículas. Os macrófagos foram então fixados com paraformaldeído 4% em PBS por 5 minutos a
25
o
C e bloqueados com PBS 5% BSA por 1 hora a temperatura ambiente. Para visualização da
membrane plasmática foi utilizada a subunidade B da toxina do cólera marcada com FITC (CtxB-
FITC), a qual foi incubada com as células bloqueadas por 1h na concentração de 1 µg/ml. CtxB-
FITC liga-se a GM1, gangliosídeo majoritariamente presente na membrane plasmática. As células
foram lavadas 3 vezes com PBS e o núcleo foi evidenciado com 4’,6-diamidino-2-fenilindol (DAPI)
a 10 µg/ml. As amostras foram montadas em lâminas contendo Meio de Montagem (50% de
glicerol e 50 mM de N-propil galato em PBS) e foram visualizadas em microscópio confocal Leica
AOBS com laser de varredura (Mannheim, Alemanha). Foi utilizada lente objetiva com aumento
de 63x em óleo de imersão. Imagens tridimensionais foram obtidas a partir da reconstrução de
eixo z utilizando o software ImageJ (NIH [http://rsb.info.nih.gov/ij/]). Alternativamente, células
foram incubadas com vesículas coradas com DiI, bloqueadas com PBS 5%BSA e incubadas por 1h
com soro de pacientes com criptococose. Como controle negativo foi usado soro de indivíduos
sadios. Os soros foram diluídos 100 vezes em PBS 1% BSA. Após 3 lavagens as amostras foram
incubadas com anticorpo anti- IgG humano conjugado a AlexaFluor488 (Invitrigen). As amostras
foram novamente lavadas, o núcleo foi corado com 10µg/ml de DAPI e lâminas foram montadas
em Meio de Montagem. As amostras foram visualizadas em um microscópio de fluorescência
Axioplan 2 (Zeiss, Alemanha), as imagens adquiridas usando a câmera digital Color View SX e
processadas com o software analySIS (Soft Image System).
42
O. Ensaio de Atividade de Lactato Desidrogenase (LDH):
Para avaliar se a exposição de macrófagos às vesículas gera dano celular, a atividade da
enzima citoplasmática LDH foi medida nos sobrenadantes de cultura. Células da linhagem
RAW264.7 foram plaqueadas na densidade de 1x10
6
células/ ml em placas de 24 poços e
estimuladas com suspensão de vesículas à 2µg/ml de esterol por 16h. Alíquotas do sobrenadante
foram coletadas e suplementadas com NADH e piruvato de sódio ajustados para concentrações
finais de 0.3 e 4.7 mM, respectivamente. Uma cinética de decaimento da absorbância a 340 nm
pelo NADH consumido foi feita do tempo 0 à 10 minutos em um espectrofotômetro. O controle
positivo foi gerado adicionando 10% de Triton X-100 às culturas e o controle negativo foi usado
sobrenadante de células não- estimuladas incubadas em meio por 16h. O experimento foi feito
em triplicatas e estatisticamente analisado pelo teste t Student’s.
P. Produção de citocinas e NO por macrófagos estimulados com vesículas:
Células RAW foram plaqueadas em placas de 96 poços a 1x105 células/ poço. As
monocamadas foram lavadas duas vezes com meio sem soro, o qual foi substituído por 1% de
Nutridoma-SP (Boehringer Mannheim), e incubadas com diferentes diluições de vesículas por
16h a 37
o
C, 7.5% CO
2
. Como controle positivo foi usado 1µg/ml de LPS. Os sobrenadantes foram
coletados e as citocinas interleucina 10 (IL-10), Fator de Transformação do Crescimento β (TGF-β)
e Fator de Necrose Tumoral α (TNF-α) foram testadas por ELISA (R&D systems). Os níveis de
óxido nítrico (NO) no sobrenadante foram medidos pelo método de Griess (Green, et al. 1982). O
inibidor específico da enzima NO- sintase induzida (iNOS), N6-(1-Iminoetil)-L-lisina dihidrocloreto
(L-NIL) (Cayman Chemical), foi usado para confirmar que a enzima é especificamente induzida
43
durante o processo de ativação descrito. Para estes experimentos as células foram estimuladas
com vesículas por 16h na presença ou ausência de 3µM de L-NIL. Para averiguar se a resposta
aos estímulos se pela presença de LPS contaminante, experimentos controles foram feitos na
presença de 10µg/ml de Polimixina B e a produção de NO foi avaliada. Todos os experimentos
foram feitos em triplicatas e analisados estatisticamente pelo ANOVA seguido de pos-teste de
Bonferroni.
Q. Determinação do índice de fagocitose por Citometria de Fluxo:
Para avaliar se estímulo com vesículas altera a capacidade fagocítica de macrófagos frente ao
C. neoformans, foi utilizado o protocolo baseado no método de Chaka (Chaka, et al. 1995). As
leveduras foram incubadas com isotiocianato de fruoresceína (FITC) a 0.5 mg/ml em PBS por 10
minutos. Os macrófagos foram previamente estimulados por 16h com preparações de vesículas
de diferentes cepas de C. neoformans na concentração de 1 µM de esterol. Após lavar
extensivamente com PBS, of fungos foram incubados com células RAW por 3h na proporção 5:1
fungo/ célula, seguido de sucessivas lavagens para retirar os fungos não aderidos. A intensidade
de fluorescência dos macrófagos é função direta da associação com fungos marcados com FITC.
As células infectadas foram então deaderidas, fixadas com PBS 4% paraformaldeído, lavadas e
analisadas em citômetro de fluxo FACScalibur (BD Biosciences). Os dados foram analisados
usando o software winMDI2.9 e estatisticamente validados pelo teste de variança ANOVA
seguido do pos-teste de Bonferroni. Para visualizar o aspecto geral das células, lamínulas de
células infectadas com fungos marcados com FITC foram preparadas em paralelo e observadas
44
por epifluorescência (Axioplan 2- Zeiss, Alemanha). As imagens foram adquiridas em câmera
digital Color View SX e processadas pelo software analySIS.
R. Determinação da atividade microbicida de macrófagos estimulados com vesículas:
Monocamadas de macrófagos foram estimuladas por 16h com preparações de vesículas de
diferentes cepas de C. neoformans em concentração correspondente a 0.4 µg/ml de esterol. O
sobrenadante foi removido e a monocamada foi lavada com DMEM sem soro. Células fúngicas
foram ressuspensas em DMEM e adicionadas as monocamadas celulares na razão 5:1
fungo/célula. Após incubação por 3h a 37
o
C, 7.5% CO
2
, as amostras foram lavadas 3 vezes em
PBS para remover os fungos não aderidos e meio fresco foi adicionado para continuar as
incubações por 2h ou 5h (total de 5h e 8h de incubação) a 37
o
C, 7.5% CO
2
. Após as incubações,
as células foram novamente lavadas e lisadas com H
2
O destilada gelada e estéril. Macrófagos
lisados contendo células ngicas foram imediatamente plaqueados em agar Sabouraud para
contagem de UFC. Os controles consistem em macrófagos não estimulados. Todos os
experimentos foram feitos em triplicatas e analisados estatisticamente pelo teste de variança
ANOVA seguido de pos-teste de Bonferroni.
S. Análise do diâmetro de vesículas por Espalhamento de Luz:
Vesículas em suspensão permanecem em movimento Browniano, o qual pode ser traduzido
em flutuações no espalhamento de luz quando em fase líquida. Essa propriedade pode ser
medida pela técnica de Espalhamento Dinâmico de Luz (dynamic light scattering, DSL), e revela
informações sobre diâmetro e heterogeneidade da amostra (Eisenman, et al. 2009). Medidas de
45
tamanho de vesículas foram feitas em um analisador de partículas 90Plus/BIMAS Multi Angle
Particle Sizing (Brookhaven Instruments) como descrito previamente (Eisenman, et al. 2009).
T. Identificação de proteínas por LC- MS/MS:
A caracterização proteômica de vesículas de diferentes mutantes de S. cerevisiae foi feita
com base na metodologia recentemente descrita para análise de vesículas fúngicas
(Albuquerque, et al. 2008; Rodrigues, et al. 2008). As vesículas purificadas foram ressuspensas
em 40ml de NH4HCO3 a 400 mM contendo 8M de uréia e as pontes dissulfeto foram reduzidas
através da adição de 10ml de ditiotreitol (DTT) a 50 mM, seguido de incubação por 15 minutos a
50 C. Resíduos de cisteína foram alquilados pela adição de 100 mM de iodoacetamina (10 ml),
seguido de 15 minutos de incubação a temperatura ambiente sob proteção da luz. A
concentração final de uréia foi ajustada para 1M e a mistura foi suplementada com 4 mg de
tripsina (Promega). A digestão acorreu por 16h a 37
o
C. Os peptídeos resultantes foram
submetidos a cromatografia de fase reversa em colunas C18 (POROS R2 50, Applied Biosystems).
Para isto, as amostras foram homogeneizadas em 5% de acetonitrila (ACN) contendo 0.5% de
ácido fórmico (FA) e foram por fim aplicadas as colunas C18. Os peptídeos foram eluídos em um
gradiente linear (5-40% CAN/0.1%FA) durante 200 minutos e diretamente analisados por ESI-MS
(LTQXL/ETD, Thermo Fisher). O Nanospray foi fixado a 1.35 kV em uma pressão de 0.2 psi N
2
. Os
espectros foram coletados entre 400-1700 m/z e os 10 íons mais abundantes foram submetidos
por duas vezes a dissociação induzida por colisão (CID), com energia de colisão normalizada para
35%. Todos os espectros MS/MS de peptídeos entre 800-3500 Da, com mais de 10 contagens e
com pelo menos 15 fragmentos foram convertidos em arquivos DTA utilizando o programa
46
Bioworks v. 3.3.1 (ThermoFisher). Arquivos DTA foram submetidos a pesquisa usando
TurboSequest (Eng, et al. 1994) e base de dados de proteínas de S. cerevisiae
(www.yeastgenome.org) combinado a análise de sequencias de queratina humana e tripsina
(www.ncbi.nlm.nih.gov/protein). Proteínas assinaladas foram re-filtradas com total de Xcorr $
3.5. A taxa de falso positivo (false-discovery rate, FDR) foi estimada como descrito previamente
(Liu, et al. 2004). Proteínas com peptídeos em comum foram agrupadas para avaliar o índice de
redundância. Determinação semi-quantitativa da abundância relativa de proteínas foi feita com
base na contagem spectral de cada proteína (Liu, et al. 2004). Este método foi ainda validado
pelo cálculo do índice de abundância de proteína modificado exponencialmente (emPAI), o qual
foi deduzido segundo a metodologia proposta por Ishihama et. al. (Ishihama, et al. 2005). Foi
usado cutoff de 10 espectros para validar assinalações positivas, resultando em um total de 127
proteínas sequenciadas e validadas.
U. Sistemas de predição e análise de interações funcionais baseadas nos dados de
proteoma:
Diagramas de Venn foram construídos utilizando a ferramenta Venny, disponível em
http://bioinfogp.cnb.csic.es/tools/venny/index.html. Interações funcionais entre proteínas foram
caracterizadas usando o programa Osprey Network Visualization (version 1.2.0), utilizando a base
de dados de S. cerevisiae disponibilizada pelo próprio software. Análise de predição de proteínas
contendo possíveis sítios de ligação a âncoras de GPI foram feitas pelo programa FragAnchor
(http://navet.ics.hawaii.edu/,fraganchor/NNHMM/NNHMM.html) (Poisson, et al. 2007). A
predição de proteínas contendo sítio de clivagem do peptídeo sinal para síntese no retículo
47
endoplasmático (RE) foi feita através do programa SignalP 3.0
(http://www.cbs.dtu.dk/services/SignalP/), conforme descrito previamente (Bendtsen, et al.
2004).
48
VI Resultados:
A seção de resultados apresentados nessa tese consiste em dados publicados em 4 artigos
científicos em revistas revisadas internacionalmente. Estes trabalhos encontram-se em anexo
(Anexos 1 a 4) e serão mencionados no decorrer do texto. Esclarece-se, portanto, que a descrição
abaixo corresponde a uma versão resumida dos resultados, cuja descrição detalhada está
disponível nas publicações citadas (Anexos).
1) A secreção de polissacarídeo capsular em vesículas é uma solução eucariótica para o
problema do transporte de macromoléculas através da parede celular.
Evidências prévias da literatura sugeriam que o polissacarídeo capsular de C. neoformans,
bem como o esfingolipídio imunogênico GlcCer, poderiam ser transportados para o espaço
extracelular em carreadores vesiculares (Rodrigues, et al. 2000; Sakaguchi, et al. 1993). O
presente trabalho (Anexo 1) comprovou pela primeira vez a existência de vesículas extracelulares
secretadas por um fungo.
Vesículas em associação com a parede celular puderam ser observados por TEM (Figura 1A-C,
Anexo 1). Estruturas vesiculares também foram observadas por TEM durante infecção in vivo, em
cortes de tecido pulmonar de camundongos infectados (Figura 1, D e E). Estas micrografias
sugeriram que vesículas eram capazes de atravessar a parede celular, e portanto deveriam ser
encontradas no espaço extracelular. Para testar tal hipótese, foi utilizado um protocolo clássico
de isolamento de exossomos baseado em etapas de centrifugações diferenciais e
49
ultracentrifugações. O sedimento originado da ultracentrifugação a 100000xg foi submetido a
preparação para TEM. As preparações mostraram se tratar de vesículas dotadas de bicamada
lipídica, relativamente heterogêneas em termos de tamanho e eletrondensidade, como pode-se
observar na Figura 1 F-H.
A composição lipídica dessas vesículas foi analisada por HPTLC e por ESI-MS/MS. Foi possível
detectar a presença dos dois esteróis majoritários do C. neoformans, ergosterol e obtusifoliol,
bem como do principal cerebrosídeo do C. neoformans, N-2-hidroxioctadecanoil-1-β-D-
glucopiranosil-9-metil-4,8-esfingadienina (Anexo 1, Figura 2). Fosfatidilcolina (PC) é o principal
fosfolipídio constituinte de membranas no C. neoformans (Rawat et al., 1984). Por isso,
analisamos amostras de vesículas por LC-MS/MS operando no modo de busca do íon precursor,
ajustado para o fragmento 184.1 m/z, o qual corresponde a PC. Foi possível identificar três
diferentes espécies de PC que variam quanto a composição do ácido graxo, sendo estas:
C
44
H
81
NO
8
P (782.570 Da); C
44
H
83
NO
8
P (784.586 Da); C
44
H
85
NO
8
P (786.601 Da) (Anexo 2, Figura
4B). Para garantir que a caracterização lipídica não incluiria frações de membranas não-
vesiculares, controles de integridade vesicular foram feitos por TEM. Vesículas apresentaram
bicamada lipídica e tamanho variando de 50 a 300 nm, como estabelecido previamente (Anexo 2,
Figura 4A).
Foi visto também que a secreção vesicular é dependente de viabilidade celular, sendo
portanto um fenômeno biológico ativo. Mesmo apresentando mais de 99% de viabilidade celular
após 72h de cultivo, era necessário comprovar que as vesículas não eram oriundas da minoria de
células mortas. Para isso, foi desenvolvido um experimento de pulso e caça, utilizando
precursores radioativos da ceramida uma vez que GlcCer foi caracterizada como marcador de
50
vesículas (Anexo 1, Figura 2 e Figura 3B). A detecção de [
3
H]GlcCer nas frações vesiculares indica
que este mecanismo de secreção requer células metabolicamente ativas (Anexo 1, FiguraFigura
3A). Ainda assim as vesículas poderiam ser oriundas de extravasamento citoplasmático de
organelas, uma vez que a [
3
H]GlcCer seria incorporada na maioria das membranas intracelulares.
Para excluir essa hipótese, o perfil lipídico de vesículas isoladas sob condições normais de cultivo
foi comparado ao perfil de vesículas isoladas de células mortas. Vesículas foram isoladas do
sobrenadante de cultura de células mortas por inibição metabólica (10mM de NaN
3
), onde se
observou 83% de morte celular, ou por aquecimento (1h, 50
o
C), método que resultou em 99%
de morte celular. Em ambos os casos observou-se uma redução expressiva da quantidade de
lipídeos no sedimento 100000xg (Anexo 1, Figura 3C).
Para avaliar se as vesículas lipídicas carreariam o polissacarídeo formador de cápsula, foram
feitos imunoensaios para detecção de GXM no sedimento obtido por centrifugação a 100000xg
com o anticorpo monoclonal anti-GXM Mab18B7. Foi demonstrado por imuno-microscopia
eletrônica de transmissão, bem como por ELISA, que de fato a GXM se encontra no interior das
estruturas vesiculares (Anexo 1, Figura 4A e 4B). Para confirmar que a GXM detectada por ELISA
era de fato intravesicular, a amostra de vesículas foi sucessivamente passada por uma coluna de
afinidade acoplada ao Mab18B7. Alíquotas foram retiradas após cada passagem e observou-se
por ELISA que os níveis de GXM mantiveram-se constantes, enquanto os níveis de GXM livre
purificada caem em cada passagem (Anexo 1, Figura 4C). Sobrenadantes de cultura de C.
neoformans são extremamente ricos em GXM livre. Para excluir a possibilidade de vesículas
formarem-se randomicamente pela agregação de lipídeos liberados no sobrenadante de cultura
e desta maneira englobarem a GXM livre, vesículas foram isoladas do mutante acapsular Cap67
51
em diferentes condições. Este experimento baseou-se no fato de que o Cap67 é capaz de
produzir vesículas, entretanto estas não contém GXM. O mutante acapsular foi crescido em meio
mínimo, meio mínimo suplementado com GXM e em uma terceira condição o meio foi
suplementado com GXM após o cultivo celular. Em ambas as condições a GXM adicionada não foi
encontrada na fração vesicular (Anexo 1, Figura 4D).
Foi visto também que uma correlação direta entre o tamanho da cápsula e a secreção
vesicular de GXM. Uma vez que a cápsula foi induzida, observou-se por ELISA um concomitante
aumento do conteúdo vesicular de GXM (Anexo 1, Figura 5). Além disso, pode-se observar que o
mutante acapsular foi capaz de reincorporar a GXM vesicular (Anexo 1, Figura 6), formando uma
cápsula evidenciada por imunofluorescência. Estes resultados agregam função biológica ao
mecanismo de secreção vesicular.
2) Crioultramicrotomia do C. neoformans e fracionamento vesicular revelaram associação
íntima entre lipídeos e GXM.
Protocolos para IEM (immunoelectron microscopy) em células fúngicas em geral envolvem
etapas como degradação enzimática da parede celular que podem gerar artefatos, os quais
limitam a interpretação das observações (Yamaguchi, et al. 2005). A fim de obter preparações
preservadas de C. neoformans para IEM, aplicamos o protocolo de crioultramicrotomia. Foi
possível obter preparações extremamente bem preservadas, sobretudo em relação as organelas
membranosas e a parede celular (Anexo 2 Figura 1). Inclusive compartimentos nunca antes
descritos em C. neoformans foram visualizados por esta técnica, como vesículas citoplasmáticas
52
semelhantes a endossomos e organelas eletrondensas semelhantes a acidossomos (Anexo 2,
Figura 1B e 2).
A marcação intracelular para imuno-microscopia eletrônica de transmissão com Mab18B7 foi
na maioria das vezes encontrada em associação com bicamadas lipídicas (Anexo 2, Figura 3). Tais
estruturas membranosas assemelham-se a vesículas carreadoras de GXM, corpos multi-
vesiculares e membranas reticulares posicionadas na periferia celular (Anexo 2, Figura 3). Neste
estudo, foi possível observar tanto a presença de vesículas intracelulares contendo GXM quanto
de vesículas intracelulares não contendo GXM (Anexo 2, Figura 1C, 2). Nesse contexto,
resolvemos avaliar se a distribuição de GXM nas vesículas extracelulares obedecia a mesma
regra. O fracionamento das vesículas em gradiente de densidade revelou a existência de
subpopulações de vesículas que contém e que não contém GXM (Anexo 2, Figura 5). Esta
heterogeneidade de conteúdo sugere a existência de mais de uma via intracelular de produção
de vesículas.
3) Vesículas extracelulares secretadas pelo C. neoformans modulam a função de
macrófagos.
A fim de entender a influência da secreção de vesículas no processo de infecção celular,
primeiramente foi demonstrado que vesículas são produzidas durante a infecção in vitro de
macrófagos. A interação de fungos opsonizados com Mab18B7 ocorreu por 1h na proporção 5:1
fungo: célula. Após a lavagem, as monocamadas de células J774 foram incubadas por 16h e
então, após a confirmação microscópica da lise celular, os sobrenadantes de cultura foram
recolhidos e avaliados para presença de componentes vesiculares. Devido a maior complexidade
53
do sistema, as vesículas foram fracionadas em gradiente de sacarose e frações contendo GlcCer e
GXM foram detectadas (Anexo 1, Figura 7).
Para avaliar se macrófagos incorporam componentes vesiculares, as células foram incubadas
com vesículas isoladas e marcadas com o fluoróforo DiI, o qual intercala-se em membranas. Foi
visto por microscopia confocal que membranas vesiculares se encontram no interior dos
macrófagos após 30 minutos de incubação (Anexo 3, Figura 1A). Além das membranas, proteínas
imunogênicas sabidamente presentes nas vesículas (Rodrigues, et al. 2008) também se
encontram no interior de macrófagos em co-localização com a marcação de DiI (Anexo 3, Figura
1B). Foi demonstrado também, através do ensaio de atividade de LDH, que a despeito da
presença de diversos fatores de virulência nas vesículas (Rodrigues, 2008), estas não causam lise
celular (Anexo 3, Figura 1C).
Este trabalho demonstrou que vesículas secretadas pelo C. neoformans são capazes de
modular a produção de NO e citocinas por células RAW (Anexo 3, Figura 2 e 3). A composição
vesicular, sobretudo no que tange ao conteúdo de GXM, tem influência direta sobre quais
citocinas serão elevadas ou diminuídas e sobre a intensidade de produção de NO (Anexo 3,
Figura 2 e 3). A atividade fagocítica de macrófagos estimulados com vesículas bem como a
atividade microbicida também são significativamente aumentados (Anexo 3, Figura 4 e 5).
Fungos marcados com FITC foram usados num ensaio de citometría de fluxo para quantificar o
índice de fagocitose. Observou-se um aumento relativo de até 74% na população de macrófagos
positivos para FITC quando os fagócitos foram pré-estimulados com vesículas (Anexo 3, Figura
4A). Microscopias de fluorescência da mesma amostra confirmaram essa observação, uma vez
que macrófagos estimulados com vesículas apresentaram um número evidentemente maior de
54
fungos internalizados (Anexo 3, Figura 4B). Após 5h e 8h de infecção, foi quantificado o número
de fungos viáveis no interior de macrófagos através da técnica de UFC. Foi possível concluir que
macrófagos pré-incubados com vesículas isoladas apresentaram aumento da capacidade
fungicida e mais do que isso, essa aumento foi ainda mais expressivo quando as células foram
tratadas com vesículas de CAP67, as quais não contém GXM (Anexo 3, Figura 5).
4) Caracterização de vesículas extracelulares de S. cerevisiae: evidencias para participação
de diferentes vias de tráfego na biogênese vesicular.
S. cerevisiae foi a levedura escolhida como modelo de estudo uma vez que é o organismo em
que mais se compreende os mecanismos de tráfego intracelular. Mutantes com defeitos em
diferentes pontos das vias de tráfego foram selecionados com base em dados prévios da
literatura e vesículas foram isoladas e caracterizadas a partir destas diferentes cepas listadas na
Tabela 1 do anexo 4. Primeiramente foi feita a caracterização das vesículas quanto ao tamanho
por Espalhamento de Luz Dinâmico e quanto ao aspecto morfológico por TEM (Anexo 4, Figura
1). Foram isoladas vesículas de dois mutantes de vias de tráfego, o sec4-2 ts e o snf7
. Apenas o
mutante sec4-2 ts apresentou alteração significativa do diâmetro vesicular (Anexo 4, Figura 1B)
enquanto na TEM de vesículas ambos os mutantes mostraram-se morfologicamente semelhantes
ao tipo selvagem (WT) (Anexo 4, Figura 1A).
55
Figura 11. Etapas do tráfego intracelular bloqueadas por diferentes mutações. Cinco mutantes foram
utilizados no estudo de proteômica. O mutante bos1-1 ts acumula vesículas que brotam do RE. O mutante
sec4-2 ts acumula vesículas que brotam da TGN. O mutante sec1-1 ts acumula vesículas estágio de fusão
com a membrane plasmática. O mutante vps23
apresenta defeito no endereçamento de moléculas para
os MVBs. O mutante snf7
apresenta problemas na formação de vesículas internas dos MVBs.
A análise proteômica das vesículas de S. cerevisiae foi feita com 3 cepas selvagens e 5
diferentes mutantes (Anexo 4, Tabela 1 e Figura 11). Um resumo das proteínas encontradas,
classificadas por função biológica encontra-se na Figura 2A bem como na Tabela S1 do anexo 4.
Um universo tão diverso de proteínas encontradas dentro de um mesmo compartimento pode
parecer contraditório. Entretanto 90% das proteínas caracterizadas apresentam algum tipo de
interação funcional com alguma outra dentro do proteoma vesicular (Anexo 4, Figura 2B). A
caracterização proteômica incluiu estudos de predição do sítio de clivagem do peptídeo sinal
para síntese no RE e de sequencias para ancoramento de GPI. De acordo com esses dados de
bioinformática, 4% das proteínas apresentam seqüência para ancoramento de GPI, contra 1% no
proteoma total da célula. Também observou-se uma concentração de aproximadamente 4 vezes
56
de proteínas sintetizadas no RE em comparação com o proteoma total da célula (Anexo 4, Figura
3). O perfil qualitativo do proteoma não variou significativamente entre células selvagens e
mutantes, como representado nos diagramas de Venn (Anexo 4, Figura 4). O mutante snf7
foi o
que apresentou a maior alteração (72% de similaridade com o WT). Diante disso, resolvemos
avaliar se proteínas individuais seriam alteradas quantitativamente. Para tanto, calculamos o
valor de emPAI (exponentially modified protein abundance index) para cada proteína de cada
amostra. Dentre estas, 57.5% das proteínas foram significativamente alteradas em sua
abundância, as quais estão listadas na Tabela S2 do anexo 4. Gráficos de correlação comparando
a abundância relativa de proteínas de cada mutante com seu respectivo WT foram montados e o
cálculo da regressão linear revelou que o mutante snf7
é o que mais se difere do WT em
relação a composição proteômica das vesículas (Anexo 4, Figura 5).
Análises das frações lipídicas dos mutantes sec4-2 ts e snf7
mostraram que uma redução
expressiva da secreção total de vesículas no mutante sec 4-2 ts, enquanto que no mutante snf7
a secreção não tenha sido quantitativamente alterada (Anexo 4, Figura 6A e B). O mutante sec 4-
2 ts acumula esterol no interior celular (Anexo 4, Figura 6C), o que está de acordo com sua
capacidade diminuída de liberar vesículas contendo esterol. A diminuição da secreção de
vesículas no mutante sec 4-2 ts foi evidente após o acompanhamento da cinética de secreção a
qual se estagnou após 3h no mutante enquanto que no WT teve o pico de secreção em 18h. A
análise de um outro mutante envolvendo mecanismos de secreção não-convencional (Figura 7
do Anexo 4) revelou que a deleção do gene de GRASP causa redução no conteúdo de esterol das
frações vesiculares e acúmulo intracelular de esterol, assim como visto para o mutante sec4-2 ts.
57
Anexo 1:
EUKARYOTIC CELL, Jan. 2007, p. 48–59 Vol. 6, No. 1
1535-9778/07/$08.00ϩ0 doi:10.1128/EC.00318-06
Copyright © 2007, American Society for Microbiology. All Rights Reserved.
Vesicular Polysaccharide Export in Cryptococcus neoformans Is a
Eukaryotic Solution to the Problem of Fungal Trans-Cell
Wall Transport
Marcio L. Rodrigues,
1
Leonardo Nimrichter,
1
‡De´bora L. Oliveira,
1
Susana Frases,
2
Kildare Miranda,
3
Oscar Zaragoza,
2
Mauricio Alvarez,
2
Antonio Nakouzi,
2
Marta Feldmesser,
2,4
and Arturo Casadevall
2,4
*
Laborato´rio de Estudos Integrados em Bioquı´mica Microbiana, Instituto de Microbiologia Professor Paulo de Go´es,
Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941590, Brazil
1
; Department of Microbiology and
Immunology
2
and Division of Infectious Diseases
4
of the Department of Medicine, Albert Einstein College of
Medicine, 1300 Morris Park Ave., Bronx, New York 10461; and Laborato´rio de
Ultraestrutura Celular Hertha Meyer, Instituto de Biofı´sica Carlos Chagas Filho,
Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941590, Brazil
3
Received 6 October 2006/Accepted 6 November 2006
The mechanisms by which macromolecules are transported through the cell wall of fungi are not known. A
central question in the biology of Cryptococcus neoformans, the causative agent of cryptococcosis, is the
mechanism by which capsular polysaccharide synthesized inside the cell is exported to the extracellular
environment for capsule assembly and release. We demonstrate that C. neoformans produces extracellular
vesicles during in vitro growth and animal infection. Vesicular compartments, which are transferred to the
extracellular space by cell wall passage, contain glucuronoxylomannan (GXM), a component of the crypto-
coccal capsule, and key lipids, such as glucosylceramide and sterols. A correlation between GXM-containing
vesicles and capsule expression was observed. The results imply a novel mechanism for the release of the major
virulence factor of C. neoformans whereby polysaccharide packaged in lipid vesicles crosses the cell wall and the
capsule network to reach the extracellular environment.
Cryptococcus neoformans is a yeast-like pathogenic fungus that
is the etiologic agent of human cryptococcosis. Infection is usually
asymptomatic and restricted to the lung in immunocompetent
individuals, but fungal cells can disseminate to other organs and
cause cryptococcal meningoencephalitis, a common syndrome in
immunosuppressed patients (29, 39). Significant progress in our
understanding of how C. neoformans causes disease has been
made in the last decade, but many aspects of cryptococcal patho-
genesis remain poorly understood.
C. neoformans represents a unique model in cell biology stud-
ies because it is the only eukaryotic pathogen with a polysaccha-
ride capsule, a structure that is essential for virulence (4, 19). The
major capsular polysaccharide glucuronoxylomannan (GXM) has
an average mass ranging from 1.7 ϫ 10
6
to 7 ϫ 10
6
daltons (22)
and is released extracellularly during infection, inducing a number
of deleterious effects to the host (39). GXM also represents a
potential vaccine component and is the target of therapeutic an-
tibodies that are currently in clinical trial (3, 8, 20, 27).
The fungal cell wall is a compact, albeit dynamic, structure that
plays important roles in several biological processes that deter-
mine cell shape, morphogenesis, reproduction, cell-cell and cell-
matrix interactions, and osmotic and physical protection (25).
Although the relevance of the cell wall to fungal physiology and
pathogenesis is clear, the mechanisms by which large molecules
(molecular weight, Ͼ1 million) cross this rigid structure to reach
the extracellular environment are largely unknown. In C. neofor-
mans, it has been suggested that capsule components containing
epitopes recognized by a monoclonal antibody (MAb) to GXM
are synthesized intracellularly and exported through the cell wall,
possibly inside membrane vesicles (11). These results were vali-
dated in a recent study using a secretion mutant of C. neoformans
in which post-Golgi secretory vesicles containing GXM were ac-
cumulated in the plasma membrane region (40). Based on these
observations, one could imagine that surface components of C.
neoformans, including the capsule, are synthesized in the cyto-
plasm and exported to the exterior of the cell in secretory vesicles
that traverse the cell wall. Indeed, C. neoformans does produce
glucosylceramide (GlcCer)-containing vesicles that are trans-
ferred to the cell wall (1, 25, 30). Since C. neoformans efficiently
releases exocellular capsular material, we hypothesized the exis-
tence of extracellular secreted vesicles containing GlcCer and
capsule components.
In the present work, we describe for the first time that a fungal
cell can produce extracellular vesicles that are secreted across the
cell wall. Supernatants of C. neoformans cultures contained vesi-
cles with bilayered membranes. Lipid analysis revealed that key
fungal lipids, such as GlcCer, ergosterol, and a novel sterol, are
present in these membranes. By different approaches, we dem-
onstrated that GXM is packaged inside the vesicles, which cross
the cell wall and the capsule network to reach the extracellular
environment. A correlation between capsule growth and detec-
* Corresponding author. Mailing address: Department of Microbi-
ology and Immunology, Albert Einstein College of Medicine, 1300
Morris Park Avenue, Bronx, NY 10461. Phone: (718) 430-2215. Fax:
(718) 430-8968. E-mail: [email protected].
† Present address: Servicio de Micologı´a, Centro Nacional de Mi-
crobiologı´a, Instituto de Salud Carlos III, Ctra Majadahonda-Pozuelo
km 2, 28220 Majadahonda, Madrid, Spain.
‡ M.L.R., L.N., and D.L.O. contributed equally to this work.
Published ahead of print on 17 November 2006.
48
tion of vesicle-associated GXM was observed, suggesting that C.
neoformans can release the polysaccharide from the vesicles and
incorporate it into the cell surface. Accordingly, acapsular cells
used vesicle-associated GXM to become encapsulated. GXM-
containing vesicles were produced during macrophage infection,
suggesting a role in pathogenesis. These findings illustrate a new
phenomenon in fungi with potential relevance for such diverse
areas as capsule assembly and pathogenesis and reveal new in-
sights into how secreted molecules reach the extracellular envi-
ronment.
MATERIALS AND METHODS
Culture conditions. The cryptococcal isolates used in this study comprised
strains ATCC 24067 (serotype D; American Type Culture Collection), H99
(serotype A; clinical isolate), HEC3393 (serotype A; clinical isolate), and Cap67
(acapsular mutant). In the different analyses performed, similar data were ob-
tained with encapsulated strains. Except for the presence of GXM, results for
assays using acapsular cells followed the same profile obtained with HEC3393,
H99, and ATCC 24067 cells. For vesicle purification, C. neoformans cells were
inoculated into 1,000-ml Erlenmeyer flasks containing 400 ml of a minimal
medium composed of dextrose (15 mM), MgSO
4
(10 mM), KH
2
PO
4
(29.4 mM),
glycine (13 mM), and thiamine-HCl (3 M). Fungal cells were cultivated for 3
days at 30°C with shaking. The viability of C. neoformans cells after this period
of cultivation was analyzed by propidium iodide staining.
Isolation of vesicles. Fungal cells were separated from culture supernatants by
centrifugation at 4,000 ϫ g for 15 min at 4°C. The supernatants were collected
and again centrifuged at 15,000 ϫ g (4°C) to remove smaller debris. The pellets
were discarded, and the resulting supernatant was concentrated approximately
20-fold using an Amicon ultrafiltration system (cutoff, 100 kDa). To ensure the
removal of cells and cellular debris, the concentrated culture fluid was again
centrifuged as described above and the resulting supernatant was then centri-
fuged at 100,000 ϫ g for1hat4°C. The supernatants were then discarded, and
the pellets were washed by five sequential resuspension and centrifugation steps,
each consisting of 100,000 ϫ g for1hat4°Cwith 0.1 M Tris-buffered saline
(TBS). The pellets were then resuspended in fixative solution for electron mi-
croscopy analysis. Alternatively, 100,000 ϫ g pellets were fractionated by affinity
chromatography and sucrose density centrifugation or extracted with organic
solvents as detailed below.
TEM. Transmission electron microscopy (TEM) was used to visualize vesicles
isolated from supernatants and those that were cell associated, in vitro and in
vivo. The pellets obtained after washing and centrifugation at 100,000 ϫ g were
fixed in 2% glutaraldehyde in 0.1 M cacodylate at room temperature for2hand
then incubated overnight in 4% formaldehyde, 1% glutaraldehyde, and 0.1%
phosphate-buffered saline (PBS). The samples were incubated for 90 min in 2%
osmium, serially dehydrated in ethanol, and embedded in Spurr’s epoxy resin.
Thin sections were obtained on a Reichert Ultracut and stained with 0.5% uranyl
acetate and 0.5% lead citrate. Samples were observed in a JEOL 1200EX trans-
mission electron microscope operating at 80 kV. For immunogold labeling with
antibodies to GXM, the vesicles were fixed in 0.1 M sodium cacodylate buffer
(pH 7.2) containing 4% paraformaldehyde, 0.2% glutaraldehyde, and 1% picric
acid, infiltrated in 25% polyvinylpyrrolidone and 2.1 M sucrose, and rapidly
frozen by immersion in liquid nitrogen. Cryosections were obtained in a tem-
perature range of Ϫ70 to Ϫ90°C using an Ultracut cryo-ultramicrotome
(Reichert). After being blocked in PBS-bovine serum albumin and 50 mM
NH
4
Cl, the cryosections were incubated overnight in the presence of a mouse
monoclonal antibody to GXM (MAb 18B7, 1 g/ml). The antibody is a mouse
immunoglobulin G1 (IgG1) with high affinity for GXM of different cryptococcal
serotypes and has been extensively characterized previously (3). After incubation
with 15-nm (particle size) immunogold-labeled anti-mouse IgG, specimens were
observed in a JEOL 1200EX transmission electron microscope operating at 80 kV.
For detection of cryptococcal vesicles in vivo, murine infection and lung tissue
preparation for transmission electron microscopy were carried out as described
previously (10, 12, 26). Briefly, anesthetized C57BL/6 mice (National Cancer
Institute, Bethesda, MD) were infected intratracheally via a midline neck inci-
sion with 10
4
or 10
6
organisms of C. neoformans strain ATCC 24067 and then
sacrificed 2 h, 48 h, or 7 days after infection. The analysis of vesicle production
in vitro was performed using strain Cap67 as previously described (30). Human
antibodies to GlcCer were used for lipid detection at the cell wall following
previously described experimental conditions (30).
Removal of nonvesicular GXM from 100,000 ؋ g pellets. To remove extrave-
sicular GXM and putative aggregates from vesicle preparations, 100,000 ϫ g pellets
were purified by passage through a column packed with an antibody-bound resin.
MAb 18B7 was coupled to cyanogen bromide-activated Sepharose according to the
manufacturer’s protocols (Sigma, Richmond, CA). Briefly,1goftheresin was
suspended in 1 mM HCl, washed, and resuspended in carbonate buffer, pH 8.0.
MAb 18B7 was dissolved in 5 ml of the same buffer, at 1 mg/ml, and incubated
overnight with the resin. At this pH, MAb 18B7 retained its biological properties,
such as antigen binding and integrity of chains (not shown). Coupling efficiency was
ϳ90%, as confirmed by spectrophotometric determinations at 280 nm. After being
washed with carbonate buffer and blocked with 0.2 M glycine, the resin was sequen-
tially washed with 0.1 M acetate (pH 4.0) and Tris (pH 8.0) buffers for final resus-
pension in PBS. Each vesicle suspension (500 l) was mixed with the antibody-
containing resin (100 l) and incubated for 1 h (37°C) with shaking. The unbound
fraction was recovered by centrifugation. This process was repeated three times, and
the GXM contents in different fractions were analyzed by capture enzyme-linked
immunosorbent assay (ELISA) as described below.
Sucrose gradient. Sucrose density centrifugation was performed as described by
Gutwein et al. (15). The vesicle suspension (0.5 ml) obtained after ultracentrifuga-
tion and purification with the antibody-coupled resin was mixed with an equivalent
volume of 85% (wt/vol) sucrose (in TBS), generating a final concentration of 42.5%
sucrose. This suspension was transferred to an ultracentrifuge tube, and a step
gradient was prepared by overlaying the original suspension with 35% sucrose,
followed by a final layer of 5% sucrose in TBS. The gradient was centrifuged for 18 h
at 200,000 ϫ g, and fractions of 0.25 ml were collected from the top to the bottom
of the gradient. Fractions were then extensively dialyzed against TBS, dried under
vacuum centrifugation, and suspended in 100 l of absolute methanol, resulting in
the immediate formation of a precipitate. The solution was then extracted with 200
l of chloroform for1hat25°C. The organic fraction was recovered by centrifuga-
tion and analyzed by different methods for lipid identification. The residual material
that was not soluble in the chloroform-methanol mixture was then dried under a
nitrogen stream, resuspended in TBS, and assayed for the presence of GXM, as
described later in this section.
Lipid analysis. The pellets obtained from centrifugation of cell supernatants at
100,000 ϫ g were first suspended in methanol, and then two volumes of chloro-
form were added. The mixture was vigorously vortexed and centrifuged to dis-
card precipitates, dried by vacuum centrifugation, and partitioned according to
Folch et al. (13). The lower phase, containing neutral lipids, was recovered for
analysis by high-performance thin-layer chromatography (HPTLC). For sterol
analysis, the lipid extract was loaded into HPTLC silica plates (Si 60F254s;
LiChrospher, Germany) and separated using a solvent system containing hexane-
ether-acetic acid (80:40:2, vol/vol/vol) solvent. The plate was sprayed with a
solution of 50 mg ferric chloride (FeCl
3
) in a mixture of 90 ml water, 5 ml acetic
acid, and 5 ml sulfuric acid. After being heated at 100°C for 3 to 5 min, the sterol
spots were identified by the appearance of a red-violet color. The presence of
GlcCer was evaluated by separating lipids in Folch’s lower phase by HPTLC
using chloroform-methanol-water (65:25:4, vol/vol/vol) (13). Monohexosylcer-
amides were visualized after the plates were sprayed with orcinol-sulfuric acid
reagent, followed by heating at 150°C (1).
The lower phase obtained after Folch’s partition was also analyzed by elec-
trospray ionization mass spectrometry (ESI-MS) using a Finnigan LCQ DUO
ion trap instrument (Thermo Electron, San Jose, CA). Samples were diluted in
chloroform-methanol (1:1, vol/vol), containing 10 mM lithium iodide, and intro-
duced into ESI-MS at a 10-l/min flow rate with the assistance of an infusion
micropump (Harvard Apparatus, Cambridge, MA). Analyses were carried out in
the positive mode. The source and capillary voltages were 4.5 kV and 3 V,
respectively. The capillary temperature was kept at 200°C. Spectra were collected
at a 300- to 800-m/z range. Source-induced dissociation was obtained at 25 V. Ion
trap collision-induced dissociation (ESI-MS/MS) experiments were carried out at
20 to 60% (1 to 3 eV) normalized relative collision energy. All spectra were
processed using Xcalibur software (Thermo Electron).
Lipids in fractions from the sucrose gradient were analyzed by high-perfor-
mance liquid chromatography (HPLC) in a Waters 600 liquid chromatograph
(New York, NY), using an Alltech C
18
column (Deerfield, IL) (250- by 4.6-mm
dimensions, 5-m particle size). A mixture of chloroform and methanol (9:1,
vol/vol) was used as the mobile phase, and lipid components were detected by
absorbance at 280 nm in a Waters 486 detector. The profile of elution of lipids
from sucrose gradient fractions was compared with a standard of purified cryp-
tococcal GlcCer (30).
Radioactive labeling of vesicles. C. neoformans (strain ATCC 24067) was
grown in the presence of
L-[
3
H]serine (20 Ci/mmol) (American Radiolabeled
Chemicals, Inc., St. Louis, MO) and [9,10-
3
H]palmitic acid (50 Ci/mmol)
(DuPont NEN, Boston, MA). These ceramide precursors were added to the
VOL. 6, 2007 GXM-CONTAINING VESICLES IN C. NEDFORMANS 49
culture medium described in “Culture conditions” to generate final amounts of
radioactivity corresponding to 2.0 Ci/ml ([
3
H]serine) and 1.0 Ci/ml
([
3
H]palmitic acid). The culture was incubated for 3 days at 30°C, and then
fungal cells were removed by centrifugation. Vesicles were obtained as described
above and suspended in methanol (100 l). Chloroform (200 l) was added, and
the mixture was centrifuged to discard precipitates. The lipid extract was loaded
into HPTLC plates and separated using chloroform-methanol-water (65:25:4,
vol/vol/vol) as the solvent system. Molecules with migration rates corresponding
to that of GlcCer, identified by comparison with an iodine-stained standard of
glucocerebroside (Avanti Polar Lipids, Alabaster, AL), were scraped off the
plate, suspended in scintillation liquid, and counted for radioactivity. Negative
controls included 100,000 ϫ g pellets from sterile medium containing the radio-
active precursors and vesicle-depleted culture supernatants.
Fungal killing and lipid detection. To evaluate whether cryptococcal vesicles
were physiologically released or were simply an artifact of dead cells, we evalu-
ated the presence of lipid markers in 100,000 ϫ g pellets of living and dead cells.
Cryptococci were treated with 10 mM sodium azide in PBS for 60 min at 25°C or,
alternatively, suspended in PBS and heated at 50°C for the same period. Control
cells (viable) were suspended in PBS and incubated at 25°C for 60 min. Cell
viability was evaluated by inoculating control or treated yeasts onto Sabouraud
dextrose agar and counting CFU. After incubation under the conditions de-
scribed above, yeasts were removed as described previously in this section and
the supernatants ultracentrifuged at 100,000 ϫ g. The resulting pellets were
extracted with mixtures of chloroform-methanol at 2:1 (for GlcCer analysis) or
9:1 (for sterol analysis). Extracts were analyzed by HPTLC following the condi-
tions described in “Lipid analysis.”
Serological assays for GXM detection. The profiles of GXM distribution in the
fractions obtained after sucrose density centrifugation were analyzed using a
capture ELISA (2). Briefly, each well of a 96-well polystyrene plate was coated
with a goat anti-mouse IgM. After removal of unbound antibodies, a solution of
MAb 12A1, an IgM MAb with specificity for GXM, was added to the plate, and
this step was followed by blocking with 1% bovine serum albumin. The ELISA
was used to analyze vesicles after treatment with methanol and chloroform as
described in “Lipid analysis.” Purified GXM was used as a positive control. The
samples were incubated in the plate overnight at 4°C. The plates were then
washed five times with a solution of TBS supplemented with 0.1% Tween 20,
followed by incubation with MAb 18B7 for 1 h. The plate was again washed and
incubated with an alkaline phosphatase-conjugated goat anti-mouse IgG1 for
1 h. Reactions were developed after the addition of p-nitrophenyl phosphate
disodium hexahydrate, followed by reading at 405 nm with a Multiscan MS
(Labsystem, Helsinki, Finland). The antibodies in this assay were all used at a
concentration of 1 g/ml.
Extracellular formation of GXM-containing vesicles. To explore the possibil-
ity that cryptococcal vesicles were formed as a result of random aggregation of
lipids and GXM in the extracellular environment, minimal medium was supple-
mented with GXM (15 g/ml) and inoculated with Cap67 cells, which are known
to produce vesicles (see Fig. 1, 3, and 5). After growth for 72 h at 30°C,
supernatants were collected and evaluated for GXM-containing vesicles. Alter-
natively, acapsular cells were grown in the absence of GXM and the polysaccha-
ride was added to culture supernatants at the same concentration after removal
of yeast cells, followed by incubation for1hat30°C. GXM-containing superna-
tants were centrifuged at 100,000 ϫ g, and the pellets were purified in Sepharose-
bound MAb 18B7. GXM in supernatants and 100,000 ϫ g pellets was then
quantified by capture ELISA. The negative control consisted of GXM determi-
nations in regular Cap67 cultures.
Capsule induction. Capsule expression in C. neoformans was induced as de-
scribed previously (42). Briefly, C. neoformans was inoculated in Sabouraud
broth diluted 10 times in 50 mM MOPS (morpholinepropanesulfonic acid), pH
7.3. After incubation for 6 or 24 h at 37°C with shaking, the cells were recovered
by centrifugation for 10 min at 2,000 ϫ g and fixed in 2% paraformaldehyde in
TBS. The fixative agent was removed by washing the cells with PBS, and after
India ink staining, capsule expression was analyzed microscopically. Capsule
sizes, defined as the distances between the cell wall and the outer border of the
capsule, were determined using ImageJ software, elaborated and provided by the
National Institutes of Health (NIH) (http://rsb.info.nih.gov/ij/). All experiments
were performed in triplicate sets and the results analyzed using Student’s t test.
Supernatant-associated vesicles produced as described above were then purified
by affinity chromatography and different centrifugation steps as described pre-
viously in this section. The GXM concentrations in 100,000 ϫ g fractions under
the different conditions of capsule induction were normalized to the number of
cells in the culture after each condition of stimulation, as measured in a
Neubauer chamber.
GXM binding by acapsular cells. Acapsular C. neoformans cells (strain Cap67,
10
6
cells) were suspended in 100 l of a purified vesicular suspension, with a
GXM concentration corresponding to 10 g/ml. The suspension was incubated
for 12 h at 25°C and extensively washed with PBS, followed by fixation with 4%
paraformaldehyde. The cells were further blocked for1hinPBS-bovine serum
albumin and incubated with MAb 18B7 (1 g/ml) for1hatroom temperature,
followed by a fluorescein isothiocyanate-labeled goat anti-mouse IgG (Fc-spe-
cific) antibody (Sigma). Yeast cells were finally observed with an Axioplan 2
(Zeiss, Germany) fluorescence microscope. Images were acquired using a Color
View SX digital camera and processed with the software system analySIS (Soft
Image System). In control systems, MAb 18B7 was replaced by irrelevant anti-
bodies. In addition, vesicle preparations were simply removed from the experi-
mental system (negative control) or replaced by soluble, nonvesicular GXM
(positive control), purified as described elsewhere (6).
Production of GXM-containing vesicles during the in vitro infection of mac-
rophages with C. neoformans. The ability of cryptococci to produce GXM-con-
taining vesicles during the infection of host cells was evaluated using the J774.16
cell line, which has been extensively used to study C. neoformans-mouse macro-
phage interactions. Macrophage-like cells were cultured in Dulbecco’s modified
Eagle’s medium, supplemented with 10% fetal calf serum, 10% NCTC109 cells,
and 1% nonessential amino acids, at 37°C in a 5% CO
2
atmosphere. Cultures
were also supplemented with 100 U/ml gamma interferon and 1 g/ml lipopoly-
saccharide. The cells were grown to confluence in 75-cm
2
flasks and then infected
with C. neoformans (10 yeast cells per host cell) in the presence of 10 g/ml of
MAb 18B7, which binds to the capsular polysaccharide and is opsonic (3). After
1 h of incubation at 37°C, free C. neoformans cells were removed by washing
them. Based on the observation that J774.16 cells lyse after hosting intracellular
replication of cryptococci (38), washed infected cells were incubated for 18 h at
37°C. Host cell lysis and cryptococcal intracellular replication were observed
microscopically. Fluids from infected cultures were then analyzed for vesicles as
described above.
RESULTS
Production of extracellular vesicles by C. neoformans. Sev-
eral lines of evidence suggested us that secretion of macromol-
ecules by fungi could rely on vesicular transport (11, 14, 30–32,
35, 36, 40), and consequently, we designed experiments to
search for vesicles in fungal cells and culture supernatants. In
acapsular cells, putative vesicular bodies were observed in as-
sociation with the cell wall (Fig. 1A to C), providing additional
evidence for the existence of an extracellular vesicular trans-
port mechanism. To address whether vesicles could be ob-
served during infection by encapsulated cryptococci, the pres-
ence of vesicles in vivo was evaluated by electron microscopy of
C57BL/6 mice infected intratracheally with C. neoformans.
TEM analysis demonstrated extracellular vesicular structures
near the edge of the capsule 2 h after infection (Fig. 1D) as
well as hypolucent vesicular structures in the cryptococcal cell
wall (Fig. 1E). Similar results were observed when mice were
killed 48 h or 7 days after infection (data not shown).
These results implied that cryptococcal vesicles could pass
through the cell wall to be released to the extracellular envi-
ronment. Consequently, we hypothesized that these structures
would be found in culture supernatants. Transmission electron
microscopy of material recovered by concentration of C. neo-
formans culture supernatants followed by differential sedimen-
tation revealed the presence of vesicular bodies (Fig. 1F to H).
Vesicle size and electron density varied considerably, but es-
sentially, all of them corresponded to spherical bodies with
evident bilayered membranes.
GlcCer and sterols are present in vesicular lipid extracts.
The pellets obtained after centrifugation of culture superna-
tants at 100,000 ϫ g were extracted with different mixtures of
organic solvents, and these preparations were analyzed by
50 RODRIGUES ET AL. EUKARYOT.CELL
HPTLC. In different vesicle preparations, bands with migra-
tion rates corresponding to the GlcCer standard were detected
in lipid extracts from both acapsular and encapsulated C. neo-
formans cells (Fig. 2A, inset). Vesicular extracts from both
encapsulated and acapsular isolates also present molecules
with R
f
values corresponding to that of ergosterol, the principal
fungal sterol. In order to confirm that sterols and GlcCer are in
fact the molecules detected by HPTLC analysis, vesicle lipids
from encapsulated cells were examined by ESI (MS and MS/
MS) in positive mode. Although several molecular ions were
detected (Fig. 2), the analysis was focused on sterols and
GlcCer, the major molecules detected in HPTLC analysis.
The principal cerebroside produced by C. neoformans is
N-2Ј-hydroxyoctadecanoyl-1--
D-glucopyranosyl-9-methyl-4,8-
sphingadienine, as demonstrated by our group (30). The full
spectra (scanned from 300 to 800 m/z) of neutral lipids ob-
tained after Folch’s partition in fact suggested the presence of
GlcCer with Li
ϩ
adducts. Monolithiated peaks at m/z 734 ([M ϩ
Li
ϩ
]
ϩ
) and 762 ([M ϩ Li
ϩ
]
ϩ
) were observed and revealed a
profile similar to what has been described by our group and others
for fungal GlcCer (1, 24, 30). These molecular ions correspond to
N-2Ј-hydroxyhexadecanoyl- and N-2Ј-hydroxyoctadecanoyl-1--
D-glucopyranosyl-9-methyl-4,8-sphingadienine, as confirmed af-
ter ESI-MS/MS analysis (Fig. 2B and C). Loss of water ([M Ϫ
H
2
O ϩ Li
ϩ
]
ϩ
) generated peaks at m/z 716 and 744. Abundant
lithiated aglycon ([M Ϫ hexose ϩ Li
ϩ
]
ϩ
) peaks with m/z at 572
and 600 corresponded, respectively, to the loss of 162 units
from the molecular ions at m/z 734 and 762. The variation of 28
units among these peaks indicates the presence of two species
of hydroxylated fatty acids, containing 16 or 18 carbons. The
presence of an abundant ion peak at m/z 480 in both spectra,
corresponding to the loss of OH C
16
and OH C
18
fatty acids
([M Ϫ FA ϩ Li
ϩ
]
ϩ
), confirmed this structural diversity.
Ergosterol and obtusifoliol are the major sterols produced
by C. neoformans (18). The major peaks at m/z 397 and 435
(Fig. 2) supported, respectively, the occurrence of a [M ϩ
H
ϩ
]
ϩ
ion corresponding to ergosterol and a [M ϩ Li
ϩ
]
ϩ
peak
compatible with a molecule differing from obtusifoliol by 2
FIG. 1. TEM of vesicles in acapsular (A to C) and encapsulated (D to H) C. neoformans cells. The occurrence of vesicles in association with
the cell wall of acapsular cryptococci (A and B) or in the extracellular environment (C) is evident after in vitro growth. Vesicle-like structures were
also observed in the lung following murine pulmonary infection (D and E). Putative vesicles near the edge of the capsule (D) or in the cryptococcal
cell wall (E) were observed 2 h after infection. Bars, 100 nm (A to C) and 500 nm (D and E). Arrows point to vesicles, and asterisks are on the
cryptococcal cell wall. (F to H) The pellets obtained by ultracentrifugation were isolated by differential centrifugation, purified from GXM by
affinity chromatography, and analyzed by TEM. Extracellular vesicles with bilayered membranes and different profiles of electron density were
observed. Bars, 100 nm (F and G) and 50 nm (H).
V
OL. 6, 2007 GXM-CONTAINING VESICLES IN C. NEDFORMANS 51
units of mass. For structural elucidation, these ions were sub-
mitted to an ESI-MS/MS scan (Fig. 2D and E). The molecular
ion at m/z 397 ([M ϩ H]
ϩ
) gave rise to fragments at m/z
285 ([C
20
H
29
O]
ϩ
), 302 ([C
22
H
38
]
ϩ
), 190 ([C
14
H
22
]
ϩ
), 173
([C
13
H
17
]
ϩ
), and 154 ([C
11
H
22
]
ϩ
), compatible with the sterol
profile of fragmentation proposed by Scallen et al. (33) and
with a commercial standard of ergosterol (data not shown).
The fragmentation of the molecular ion at m/z 435 ([M ϩ Li]
ϩ
)
yielded daughter ions at m/z 417 ([M Ϫ H
2
O ϩ Li
ϩ
]
ϩ
), 407
([C
28
H
48
OLi]
ϩ
), 267 ([C
29
H
32
Li]
ϩ
), and 186 ([C
13
H
24
Li]
ϩ
).
Minor ions at m/z 391, 351, 337, 311, 295, 284, and 199 were
also observed. All of these peaks were compatible with a
monolithiated molecular ion corresponding to an obtusifoliol-
like molecule lacking the double bond between carbons 8 and
FIG. 2. Lipid analysis of C. neoformans vesicles. (A) Lipid extracts were prepared as described in Materials and Methods and analyzed by
ESI-MS and HPTLC. Vesicle lipids with migration rates corresponding to GlcCer and ergosterol standards (c) were detected by HPTLC (inset)
in preparations from both encapsulated (a) and acapsular (b) C. neoformans cells. ESI-MS analysis demonstrated the presence of molecular ions
corresponding to GlcCer (734 and 762) and sterols (435 and 397) besides other still-unidentified molecules (m/z values in gray). Fragmentation
analysis of molecular ions at m/z 734 (B) and 762 (C) revealed ionized species with m/z values typical of lithiated fungal cerebrosides. Ceramide
structures with C
16
(structure in panel B) and C
18
(structure in panel C) fatty acids were detected. Fragmentation analysis of molecular ions at m/z
397 (D) and 435 (E) revealed the presence of ergosterol (structure in panel D) and 4,14-dimethylergosta-24(24
1
)-en-3-ol (structure in panel E),
an obtusifoliol-like structure.
52 RODRIGUES ET AL. E
UKARYOT.CELL
9. This structure was therefore identified as 4,14-dimethyl-
ergosta-24(24
1
)-en-3-ol.
Vesicle production requires viable C. neoformans cells. After
growth of C. neoformans cells for 72 h, the viability of the cryp-
tococcal population was very close to 100%, as determined by
propidium iodide staining (data not shown). However, even with
high indices of viability, the possibility that the vesicles originated
from a minor fraction of dead cells could not be discarded. In this
context, we first evaluated the ability of C. neoformans cells to use
radioactive precursors of ceramide to produce [
3
H]ceramide-con
-
taining vesicles, based on the evidence that GlcCer is present in
vesicle extracts. The detection of [
3
H]GlcCer in vesicle fractions
from strain ATCC 24067 (Fig. 3A) suggested that C. neoformans
cells metabolically incorporated the radioactive precursors and
secreted their corresponding products in vesicular preparations.
The insignificant levels of [
3
H]GlcCer detection in sterile media
containing the radioactive precursors and in 100,000 ϫ g super-
natants supported this hypothesis. In addition, GlcCer-containing
vesicles being transferred from the plasma membrane to the cell
wall were detected by immunogold labeling with antibodies to
GlcCer, followed by analysis by TEM (Fig. 3B). Wall-associated
extracellular vesicular bodies were also observed in these prepa-
rations.
The possibility that the vesicles could be the products of
dead cells was also considered, given that lipids are present in
the membranes of all organelles. Thus, we compared lipid
profiles obtained from regular vesicle fractions with those ob-
tained from dead cells. To avoid false-negative results, several
methods of fungal killing were evaluated, including metabolic
inhibition (sodium azide treatment) and protein denaturation
(mild heat exposure). C. neoformans cells were grown under
the conditions used for vesicle production, and then the super-
natant was collected and used for vesicle purification. Cell
viability decreased by 83 or 99%, respectively, after treatment
with azide or heat (Fig. 3C). The treated yeast cells were
removed as described in Materials and Methods and the su-
pernatants centrifuged at 100,000 ϫ g. GlcCer was detected in
100,000 ϫ g fractions from living but not heat- or azide-treated
cells, as determined by HPTLC using a purified standard of
GlcCer. Sterol analysis revealed that compounds with migration
rates corresponding to an ergosterol standard were detected in
100,000 ϫ g fractions from supernatants of living and azide-
treated cells but not in preparations from heat-killed cryptococci.
We speculate that the detection of sterols in vesicles from azide-
treated cells could be the result of stress-associated secretory
activity during metabolic inhibition. Also, the sterol contents of
FIG. 3. Vesicle production requires cell viability. (A) The addition of radioactive ceramide precursors to the culture medium results in the
detection of significant levels of radioactive GlcCer in vesicle preparations. In 100,000 ϫ g pellets from sterile medium and in 100,000 ϫ g
supernatants of grown cells, significant levels of radioactive GlcCer were not detected (P Ͻ 0.001). (B) GlcCer-containing vesicles are apparently
transferred from the plasma membrane to the cell wall (a) and then secreted (b). Scale bars represent 100 nm. Arrows point to vesicles, and
asterisks are on the cryptococcal cell wall. (C) Killing of cryptococci with sodium azide or heating demonstrates a decrease in cell viability of 83%
(sodium azide) and 99% (heat), as determined by comparison with CFU counts obtained with untreated yeasts. Lipid analysis of 100,000 ϫ g
fractions of the supernatants obtained after fungal killing revealed the detection of GlcCer in fractions from living but not heat- or azide-treated
cells. Compounds with migration rates corresponding to an ergosterol standard were detected in 100,000 ϫ g fractions from supernatants of living
and azide-treated cells but not in preparations from heat-killed cryptococci. CPM, counts per minute.
VOL. 6, 2007 GXM-CONTAINING VESICLES IN C. NEDFORMANS 53
100,000 ϫ g fractions from living and azide-treated cells were
analyzed by densitometry, showing that the detection of sterol in
the former was 2.5-fold higher (data not shown). In summary,
these results suggest that the vesicles described here are produced
and excreted by living C. neoformans cells rather than released
from dead cells. The results obtained using the different strains
currently studied were very similar.
GXM is contained inside cryptococcal vesicles. The pres-
ence of GXM in vesicular bodies was evaluated by immuno-
gold labeling of purified vesicles with MAb 18B7, followed by
TEM analysis (Fig. 4A). Antibody binding to purified vesicles
was concentrated largely in the vesicular matrix. The presence
of GXM was confirmed by the fact that the antibody reacted
strongly with the vesicles’ contents (obtained by solvent-medi-
ated lysis) in ELISAs (Fig. 4B). As an additional control, the
supernatant obtained before vesicle lysis was also assayed, and
the content of GXM in this preparation was around 10-fold
lower than the content of GXM in the vesicle pellet. The most
straightforward interpretation of these results is that GXM is
contained primarily in the intravesicular compartment but that
some material is found in solution, probably as a result of
vesicle damage and/or disruption in handling. To confirm the
premise that GXM was intravesicular, we removed the extra-
cellular GXM by sequential passage of the vesicle preparation
over a column of Sepharose-bound MAb 18B7 (Fig. 4C). In
this experiment, the vesicle fraction and a vesicle-depleted
supernatant were consecutively passed through the antibody-
containing resin. While the GXM content in the supernatant
seemed to decrease after each step of incubation with the
resin, it stabilized when the vesicle preparation was used.
Hence, we conclude that the vesicles contain GXM.
To exclude the possibility that GXM-containing vesicles
were formed extracellularly by random incorporation of the
polysaccharide into liposomes instead of secretion, the mini-
FIG. 4. GXM is present in purified vesicles. (A) Immunogold labeling of purified vesicles with MAb 18B7 revealed a preferential intravesicular
distribution of GXM, as observed in different vesicular bodies (a to c). A higher magnification of the boxed area shown in panel c demonstrates
the occurrence of bilayered membrane (d). Bars, 150 (a), 180 (b), 120 (c), and 30 (d) nm. (B) The presence of GXM inside the vesicles was
confirmed by polysaccharide detection in purified 100,000 ϫ g fractions from culture supernatants (white bar). Supernatants obtained from these
suspensions were assayed as a control of vesicle integrity, and in fact, GXM was detected at low levels in these preparations (black bar), suggesting
that some of these structures may be disrupted. (C) GXM content of vesicle suspensions and vesicle-depleted supernatants after serial passage
through a column composed of Sepharose-bound MAb 18B7. Sequential passages of vesicle-depleted supernatant through this column result in
a rapid loss of reactivity, while the GXM content of the vesicular preparation is relatively constant. (D) GXM-containing vesicles are not formed
extracellularly, since the addition of the polysaccharide to culture supernatants during or after growth of Cap67 cells is not followed by its detection
in purified 100,000 ϫ g fractions.
54 RODRIGUES ET AL. E
UKARYOT.CELL
mal medium was supplemented with GXM and inoculated with
Cap67 cells. After 72 h of growth at 30°C, Cap67 supernatants
were collected for vesicle purification. Alternatively, acapsular
cells were grown in the absence of GXM, and the polysaccha-
ride was added to culture supernatants after the removal of
yeast cells. Vesicle purification in these systems followed by
polysaccharide determination demonstrated the absence of
significant amounts of GXM in the 100,000 ϫ g fractions (Fig.
4D), suggesting that the association of GXM and vesicles was
not a simple artifact of polysaccharide incorporation into lipo-
somes or polysaccharide-facilitated formation of lipid vesicles.
Interestingly, no significant loss of GXM in supernatants was
observed when the polysaccharide was added to the culture
after fungal growth. When the medium was supplemented with
GXM before inoculation of acapsular cells, however, only one-
third of the total amount of the polysaccharide initially added
was detected in supernatants. This result could be linked to the
well-known ability of Cap67 cells to incorporate GXM into
their cell walls. In fact, immunofluorescence analysis with MAb
18B7 revealed that after growth of Cap67 cells in the presence
of GXM, their surface was stained by the antibody to polysac-
charide (data not shown).
GXM content in vesicles correlates with capsule synthesis.
If GXM were packaged inside vesicles for extracellular trans-
port, we hypothesized that the conditions that promoted
capsule growth would also be associated with an increase in
supernatant vesicles. To evaluate the possible correlation be-
tween capsule synthesis and vesicle secretion, cryptococci were
stimulated to produce capsule, and supernatants were evalu-
ated for vesicle content. Vesicles were obtained by ultracen-
trifugation at 100,000 ϫ g, and pellets were resuspended in
TBS for GXM analysis by capture ELISA. GXM concentra-
tions in 100,000 ϫ g fractions under the different conditions of
capsule induction were normalized to the number of cells in
the culture under each condition of stimulation. In parallel,
yeast cells were collected by centrifugation, washed in PBS,
and fixed in 2% paraformaldehyde for microscopic observation
after India ink staining. After 24 h of incubation of cryptococci
in diluted Sabouraud broth, capsule expression was approxi-
mately twofold higher than in cells incubated for 6 h (Fig. 5A).
Similarly, the content of GXM in the vesicle fraction after 24 h
was twofold higher than that observed after incubation of cryp-
tococci for 6 h (Fig. 5B).
Incorporation of vesicular GXM into the surface of acapsu-
lar cryptococci. The correlation between capsule growth and
GXM content in the vesicle fraction suggested that C. neofor-
mans must have mechanisms to extract the polysaccharide
from vesicles. To evaluate this hypothesis, acapsular C. neofor-
FIG. 5. Association of capsule expression with supernatant accumulation of GXM-containing vesicles. (A) Capsule expression was higher in
yeast cells incubated for 24 h in capsule-inducing media. (B) The profile of GXM detection in ultracentrifugation pellets was very similar to that
observed for determinations of capsule size (inset), suggesting a correlation between capsule expression and the production of vesicles containing
GXM. GXM concentrations in 100,000 ϫ g fractions under the different conditions of capsule induction were normalized to the number of cells
in the culture under each experimental condition.
V
OL. 6, 2007 GXM-CONTAINING VESICLES IN C. NEDFORMANS 55
mans cells were incubated in the presence of purified vesicles
and then evaluated by immunofluorescence with MAb 18B7. A
strong fluorescent reaction of the antibody with acapsular cells
was observed after incubation with the vesicle suspension (Fig.
6), suggesting that C. neoformans can release GXM from ve-
sicular bodies and uses it for capsule growth. Control yeasts,
which had not been incubated with MAb 18B7 or vesicle prep-
arations, presented very weak levels of fluorescence. Yeast
cells incubated with nonvesicular purified GXM also became
encapsulated (data not shown), as extensively described in
previous studies.
Detection of GXM-containing vesicles from regular cultures
or infected macrophages after sucrose gradient separation.
Given the size diversity of the vesicles demonstrated in Fig. 1,
a more detailed analysis of vesicle production by C. neoformans
during its regular growth was performed using ultracentrifuga-
tion, followed by flotation on a step sucrose gradient (15).
After ultracentrifugation for 18 h, 12 fractions were obtained.
HPLC analysis revealed the presence of peaks with retention
times similar to that of a standard GlcCer in all fractions (Fig.
7A). The area of each peak varied depending on the sucrose
concentration in each fraction. Analysis of the presence of
GXM in sucrose gradient fractions was performed using a
capture ELISA. The results shown in Fig. 7B indicate that the
FIG. 6. Acapsular cells of C. neoformans bind GXM from extracel-
lular vesicles. Cap67 cells were incubated in the presence of purified
vesicles and then analyzed by immunofluorescence with MAb 18B7.
Control cells, which were not incubated with MAb 18B7, are shown in
the upper panels (a and b). Yeast cells that were incubated in the
presence of the vesicular preparation reacted strongly with the anti-
body to GXM, as shown in the lower panels (c and d). The left panels
(a and c) represent cryptococci analyzed under differential interfer-
ence contrast, while the right panels (b and d) show images in the
fluorescence mode. Bar, 2 m.
FIG. 7. Analysis of vesicle fractions obtained after ultracentrifugation and sucrose gradient separation. Fractions from supernatants of regular
cultures were extracted with chloroform-methanol mixtures and analyzed by HPLC. In each fraction, a single peak with a retention time (Rt)
corresponding to a GlcCer standard was detected. Analysis of the same fractions (solid line) or preparations obtained from infected macrophages
(dashed line) by capture ELISA revealed different profiles of GXM distribution, although the polysaccharide was always expressively detected in
the region of the gradient presenting the highest density. Lipid and polysaccharide analyses were performed at least three times, always presenting
similar profiles. O.D. 405 nm, optical density at 405 nm.
56 RODRIGUES ET AL. EUKARYOT.CELL
maximum indices of polysaccharide detection were measured
in the most concentrated sucrose fractions, suggesting that
packaging of GXM into lipid membranes results in highly
dense vesicles. The influence of experimental conditions on
GXM distribution in gradient fractions was analyzed by chang-
ing sucrose concentrations. Using different conditions, the
highest content of GXM was always found in the most con-
centrated sucrose fractions (data not shown).
Based on the property that macrophages (J774.16 cells)
infected with C. neoformans become lysed after prolonged
periods of incubation (38), supernatants of yeast-infected mac-
rophage cells were collected for vesicle purification. After sep-
aration of these fluids by ultracentrifugation associated with
sucrose gradient, the presence of GXM was analyzed in each
fraction. By comparison of the profile observed in infection-
derived vesicles with that observed in regular cultures (Fig.
7B), two additional regions containing high levels of polysac-
charide were observed in preparations obtained from infected
macrophages. These results support the idea that C. neofor-
mans produces GXM-containing vesicles inside host cells or,
alternatively, induces the production of host-derived mem-
brane domains filled with polysaccharide, as previously sug-
gested (38).
DISCUSSION
The secretion of macromolecules (molecular weight, Ͼ1
million) by fungal cells is a puzzling topic. To reach the extra-
cellular environment, secreted molecules must cross the cell
wall molecular network, a porous but very rigid complex. In C.
neoformans, it was recently demonstrated that GXM is traf-
ficked within cytoplasmic secretory vesicles (40), supporting a
model that capsular materials are synthesized in the Golgi and
targeted to the plasma membrane for exocytosis. However, it
remains unknown how the vesicles would reach the extracel-
lular environment to be used for capsule expression. In this
regard, we present evidence that the major virulence factor of
C. neoformans is secreted by a novel mechanism involving the
release of membrane vesicles through the cell wall. Vesicle
secretion is apparently not exclusively related to GXM traffic,
since lipid-containing extracellular vesicles were observed in
acapsular cells.
The possibility that the cell wall of C. neoformans is perme-
able to the passage of intact vesicular structures was supported
by microscopic analysis of acapsular cells cultivated in vitro and
encapsulated cells from infected mice. Putative vesicular bod-
ies in association with the cell wall and in the extracellular
milieu were observed in both acapsular and encapsulated cells,
suggesting that C. neoformans can use vesicular transport to
secrete different compounds. Microscopic analysis of pellets
obtained after differential centrifugation of culture superna-
tants revealed intact vesicles ranging in size from 60 to 300 nm.
Despite this heterogeneity, the vesicles had the common ap-
pearance of being rounded and defined by a lipid bilayered
membrane. Differences in electron density were observed, sug-
gesting heterogeneity in vesicular contents. The sizes of the
vesicles were consistent with those shown in Fig. 1 and with the
size predicted from the early electron microscopic studies that
showed intracellular GXM in what appeared to be vesicular
structures (11, 14, 40). In those studies, the vesicular structures
were 100 to 300 nm in diameter, which is within the size range
of the vesicles described here.
Glycosphingolipids, sterols, and glycosylphosphatidylinosi-
tol-anchored proteins form detergent-insoluble lipid rafts on
the plasma membrane (23). They are required for the process-
ing of surface proteins in yeasts, making part of the vesicles
that link the reticuloendothelial system to the Golgi to the
plasma membrane (34). In C. neoformans, it has been sug-
gested that GlcCer-containing vesicles migrate from the cell
membrane to the cell wall (1, 25, 30). In the present study, lipid
analysis by mass spectrometry revealed that a glycosphingo-
lipid (GlcCer) and sterols are components of extracellular ves-
icles, supporting the idea that they are enriched in lipid rafts.
The physiologic production of GlcCer as a vesicle component
was strengthened by the observation that [
3
H]glucosylceramide
was metabolically incorporated in vesicular fractions. Lipids
were not detected in supernatants from dead cells, strongly
suggesting that C. neoformans physiologically secretes vesicles.
The visualization of GlcCer-containing vesicle-like structures
in both cell wall and extracellular spaces of C. neoformans cells
supports the view that these structures are active products of
fungal cells. The relevance of these findings to the pathogen-
esis and control of cryptococcosis is supported by different
studies, which demonstrated that GlcCer induces the produc-
tion of antimicrobial antibodies during human infection (30)
and is the target of defensins from insect and plant cells (37).
More recently, it was demonstrated that GlcCer expression
regulates cryptococcal virulence and is essential for fungal
growth in neutral/alkaline pH in vitro and in vivo (28).
Sterol derivatives, including ergosterol and an obtusifoliol-
like molecule, were also characterized as lipid components of
vesicle membranes. Ergosterol and obtusifoliol have previously
been described as major sterol components of C. neoformans
(18), but to our knowledge, this is the first demonstration of
4,14-dimethylergosta-24(24
1
)-en-3-ol in cryptococcal mem
-
branes. The presence of other classes of hydrophobic com-
pounds is evidently expected, and indeed, additional molecular
ions were observed in ESI-MS analysis. The lack of detection
of these molecules by HPTLC could be due to either small
amounts of each individual lipid component or nonappropri-
ated conditions of separation and staining. The fact that other
lipids are clearly relevant to the physiology and pathogenesis of
C. neoformans (16, 17, 21) justifies the characterization of
other components of vesicle membranes.
GXM, the principal capsular polysaccharide of C. neofor-
mans, was detected in vesicle preparations by serological ap-
proaches. The possibility that polysaccharide aggregates were
contaminating 100,000 ϫ g fractions was discarded, since ves-
icle preparations were purified in a MAb 18B7-bound resin. A
definitive association between GXM and vesicles was obtained
by immunogold labeling of the vesicles with an anti-GXM
antibody followed by TEM analysis, which revealed that the
polysaccharide is indeed surrounded by extracellular mem-
brane domains. This information was supported by the facts
that vesicle formation was not an artifact of polysaccharide-
mediated effects on lipids (Fig. 4D) and that the polysaccha-
ride and GlcCer, a lipid marker of cryptococcal vesicles, were
concomitantly detected in fractions from a sucrose gradient. In
this analysis, the polysaccharide content is directly related to
VOL. 6, 2007 GXM-CONTAINING VESICLES IN C. NEDFORMANS 57
vesicular density, which is in agreement with the high viscosity
described for GXM (22).
McFadden and coworkers recently provided experimental
evidence suggesting that capsule growth involves the produc-
tion of GXM fibers that are released from, rather than at-
tached to, the cell (22). In this model, capsule construction
would depend on the self-association of GXM fibers, in which
newly synthesized molecules would be secreted into the extra-
cellular environment and further incorporated throughout the
capsule by becoming entangled in existing capsular material. A
new model of capsule growth was recently proposed whereby
the capsule grows by apical extension (41). Vesicular transport
to the capsular edge, followed by polysaccharide unloading and
polysaccharide self-assembly into a capsule, would provide a
potential mechanism for apical growth. Such a mechanism may
have an inherent error rate such that some vesicles could be
released to the extracellular space without being unloaded and
could accumulate in the culture supernatant. Alternatively,
material destined for capsule or extracellular secretion may be
found in different vesicular fractions such that the vesicles
recovered represent deliberate extracapsular polysaccharide
transport. We, therefore, hypothesized that C. neoformans
could release capsular polysaccharides inside vesicles, which
would be further lysed by still-unknown mechanisms. The fact
that the detection of GXM in 100,000 ϫ g fractions is higher
when capsule expression is more efficient in C. neoformans
supports this idea. The observation that acapsular mutants can
bind GXM from purified vesicles indicates that C. neoformans
is indeed able to lyse vesicles and use its internal content,
possibly through the activity of exocellular lipases (5).
Our current results and previous reports (11, 14, 40) indicate
that vesicles are synthesized intracellularly and transferred to
the cell surface, from which they are secreted to the extracel-
lular environment. It remains unknown how vesicles with an
average diameter of 160 nm cross the fungal cell wall, a highly
complex and dynamic structure (25). One possible explanation
comes from the observation that the cell wall of Saccharomyces
cerevisiae presents regular pores of around 200 nm, which can
be increased to 400 nm under stress conditions (7). Therefore,
despite being a rigid and complex structure, the fungal cell wall
could allow the passive passage of particles in the range of 60
to 300 nm, dimensions that could accommodate the vesicles
described here. Another mechanism for extracellular transport
could involve motor proteins. In Aspergillus fumigatus, a 180-
kDa polypeptide recognized by antimyosin antibodies was
found to be concentrated in cell wall and plasma membrane
fractions of conidia (9). Immunoelectron microscopy with an
antimyosin antiserum revealed that the antigen is distributed
mainly under the plasma membrane region, although it is also
clearly detected in different layers of the cell wall. The char-
acterization of cell wall motor proteins in fungal cells will
possibly contribute to the understanding of how vesicles are
transported to the extracellular milieu. Our current conclu-
sions from electron microscopy analyses of acapsular and en-
capsulated C. neoformans cells point to the presence of vesicles
in different compartments of the cell surface, including the
capsule network of ATCC 24067 cells.
The immunological activity of GXM (for a review, see ref-
erence 39) and lipids (1, 24, 25, 30) has been demonstrated in
different studies. Consequently, the delivery of GXM in vesi-
cles to target host tissues could involve a twin payload of
immunomodulating molecules that can interfere with immune
function. Our current results and previous reports suggest that
C. neoformans secretes capsular components during the infec-
tion of different host cells. In macrophages infected by cryp-
tococci, GXM is released by ingested yeast cells, followed by
damage to the phagosomal membrane and cytoplasmic accu-
mulation of polysaccharide-containing vesicles (38). In the cur-
rent study, the distributions of GXM in fractions obtained
from ultracentrifugation associated with a sucrose gradient
markedly differed when culture fluids of C. neoformans cells
and supernatants from infected macrophages were compared.
When macrophage-derived supernatants were analyzed, an ad-
ditional peak of polysaccharide detection was observed, which
could be explained by the production of vesicles of different
natures during the interaction of C. neoformans cells with these
macrophages, as previously suggested by Tucker and Casade-
vall (38). The currently presented data also indicate that C.
neoformans produces extracellular vesicles in vivo, supporting
the idea that vesicular transport is relevant for polysaccharide
release during infection. This result may have an impact on the
understanding of the immunomodulatory activity of GXM,
since its secretion during infection is accompanied by the
release of other immunologically active molecules, such as
GlcCer and other still-uncharacterized molecules.
In summary, we report the existence of membrane vesicles
containing capsular polysaccharide in C. neoformans cultures
and host cells infected with this pathogen. In contrast to pro-
karyotes, C. neoformans capsular polysaccharide is synthesized
in the cell body and transported extracellularly for capsule
assembly by a mechanism that involves the production of ves-
icles. Hence, our results and other reports (11, 14, 31, 32, 40)
suggest that the eukaryotic solution to the problem of capsular
assembly takes advantage of a sophisticated trans-cell wall
vesicular transport secretory mechanism that is not available in
prokaryotes. The discovery that polysaccharide is packaged in
vesicles containing immunologically active lipids has the po-
tential to revolutionize our views of capsule assembly, extra-
cellular polysaccharide shedding, and the mechanisms by which
GXM mediates immunosuppression.
ACKNOWLEDGMENTS
M.L.R. was the recipient of an International Fellowship for Latin
America and is supported by grants from Conselho Nacional de Des-
envolvimento Cientı´fico e Tecnolo´gico (CNPq-Brazil), Fundac¸a˜o Uni-
versita´ria Jose´ Bonifa´cio (FUJB-Brazil), and Fundac¸a˜o de Amparo a
Pesquisa do Estado do Rio de Janeiro (FAPERJ-Brazil). K.M. is
supported by Programa de Nanocieˆncia e Nanotecnologia, MCT-
CNPq. L.N. is supported by CNPq-Brazil. A.C. is supported by NIH
grants AI033142, AI033774, AI052733, and HL059842.
We thank Eliandro Lima, Yvonne Kress, and the Albert Einstein
College of Medicine Analytical Imaging Facility staff for help with
electron microscopy. We also thank Igor C. Almeida and Sirlei Daffre
for support with mass spectrometry analysis. We are indebted to Jorge
Jose´ Bastos Ferreira for helpful discussions.
D.L.O. is a M.S. student at Instituto de Bioguimica Medica, UFRJ.
REFERENCES
1. Barreto-Bergter, E., M. R. Pinto, and M. L. Rodrigues. 2004. Structure and
biological functions of fungal cerebrosides. An. Acad. Bras. Cienc. 76:67–84.
2. Casadevall, A., J. Mukherjee, and M. D. Scharff. 1992. Monoclonal antibody
based ELISAs for cryptococcal polysaccharide. J. Immunol. Methods 154:
27–35.
3. Casadevall, A., W. Cleare, M. Feldmesser, A. Glatman-Freedman, D. L.
58 RODRIGUES ET AL. EUKARYOT.CELL
Goldman, T. R. Kozel, N. Lendvai, J. Mukherjee, L. A. Pirofski, J. Rivera,
A. L. Rosas, M. D. Scharff, P. Valadon, K. Westin, and Z. Zhong. 1998.
Characterization of a murine monoclonal antibody to Cryptococcus neofor-
mans polysaccharide that is a candidate for human therapeutic studies.
Antimicrob. Agents Chemother. 42:1437–1446.
4. Chang, Y. C., and K. J. Kwon-Chung. 1994. Complementation of a capsule-
deficient mutation of Cryptococcus neoformans restores its virulence. Mol.
Cell. Biol. 14:4912–4919.
5. Chen, S. C., L. C. Wright, R. T. Santangelo, M. Muller, V. R. Moran, P. W.
Kuchel, and T. C. Sorrell. 1997. Identification of extracellular phospholipase
B, lysophospholipase, and acyltransferase produced by Cryptococcus neofor-
mans. Infect. Immun. 65:405–411.
6. Cherniak, R., L. C. Morris, B. C. Anderson, and S. A. Meyer. 1991. Facili-
tated isolation, purification, and analysis of glucuronoxylomannan of Cryp-
tococcus neoformans. Infect. Immun. 59:59–64.
7. de Souza Pereira, R., and J. Geibel. 1999. Direct observation of oxidative
stress on the cell wall of Saccharomyces cerevisiae strains with atomic force
microscopy. Mol. Cell. Biochem. 201:17–24.
8. Dromer, F., J. Salamero, A. Contrepois, C. Carbon, and P. Yeni. 1987.
Production, characterization, and antibody specificity of a mouse monoclo-
nal antibody reactive with Cryptococcus neoformans capsular polysaccharide.
Infect. Immun. 55:742–748.
9. Esnault, K., B. el Moudni, J.-P. Bouchara, D. Chabasse, and G. Tronchin.
1999. Association of a myosin immunoanalogue with cell envelopes of
Aspergillus fumigatus conidia and its participation in swelling and germina-
tion. Infect. Immun. 67:1238–1244.
10. Feldmesser, M., A. Casadevall, Y. Kress, G. Spira, and A. Orlofsky. 1997.
Eosinophil-Cryptococcus neoformans interactions in vivo and in vitro. Infect.
Immun. 65:1899–1907.
11. Feldmesser, M., Y. Kress, and A. Casadevall. 2001. Dynamic changes in the
morphology of Cryptococcus neoformans during murine pulmonary infection.
Microbiology 147:2355–2365.
12. Feldmesser, M., Y. Kress, P. Novikoff, and A. Casadevall. 2000. Cryptococcus
neoformans is a facultative intracellular pathogen in murine pulmonary in-
fection. Infect. Immun. 68:4225–4237.
13. Folch, J., M. Lees, and G. H. Sloane Stanley. 1957. A simple method for the
isolation and purification of total lipids from animal tissues. J. Biol. Chem.
226:497–509.
14. Garcı´a-Rivera, J., Y. C. Chang, K. J. Kwon-Chung, and A. Casadevall. 2004.
Cryptococcus neoformans CAP59 (or Cap59p) is involved in the extracellular
trafficking of capsular glucuronoxylomannan. Eukaryot. Cell 3:385–392.
15. Gutwein, P., S. Mechtersheimer, S. Riedle, A. Stoeck, D. Gast, S. Joumaa, H.
Zentgraf, M. Fogel, and D. P. Altevogt. 2003. ADAM10-mediated cleavage
of L1 adhesion molecule at the cell surface and in released membrane
vesicles. FASEB J. 17:292–294.
16. Heung, L. J., A. E. Kaiser, C. Luberto, and M. Del Poeta. 2005. The role and
mechanism of diacylglycerol-protein kinase C1 signaling in melanogenesis by
Cryptococcus neoformans. J. Biol. Chem. 280:28547–28555.
17. Heung, L. J., C. Luberto, A. Plowden, Y. A. Hannun, and M. Del Poeta. 2004.
The sphingolipid pathway regulates Pkc1 through the formation of diacyl-
glycerol in Cryptococcus neoformans. J. Biol. Chem. 279:21144–21153.
18. Ibrahim, A. S., H. Sanati, and M. A. Ghannoum. 1996. Lipids of Cryptococ-
cus neoformans, p. 156–167. In R. Prasad and M. A. Ghannoum (ed.), Lipids
of pathogenic fungi. CRC Press, Boca Raton, FL.
19. Kozel, T. R. 1995. Virulence factors of Cryptococcus neoformans. Trends
Microbiol. 3:295–299.
20. Larsen, R. A., P. G. Pappas, J. Perfect, J. A. Aberg, A. Casadevall, G. A.
Cloud, R. James, S. Filler, and W. E. Dismukes. 2005. Phase I evaluation of
the safety and pharmacokinetics of murine-derived anticryptococcal anti-
body 18B7 in subjects with treated cryptococcal meningitis. Antimicrob.
Agents Chemother. 49:952–958.
21. Mare, L., R. Iatta, M. T. Montagna, C. Luberto, and M. Del Poeta. 2005.
APP1 transcription is regulated by inositol-phosphorylceramide synthase
1-diacylglycerol pathway and is controlled by ATF2 transcription factor in
Cryptococcus neoformans. J. Biol. Chem. 280:36055–36064.
22. McFadden, D. C., M. De Jesus, and A. Casadevall. 2006. The physical
properties of the capsular polysaccharides from Cryptococcus neoformans
suggest features for capsule construction. J. Biol. Chem. 281:1868–1875.
23. Mun˜iz, M., and H. Riezman. 2000. Intracellular transport of GPI-anchored
proteins. EMBO J. 19:10–15.
24. Nimrichter, L., M. D. Cerqueira, E. A. Leita˜o, K. Miranda, E. S. Nakayasu,
S. R. Almeida, I. C. Almeida, C. S. Alviano, E. Barreto-Bergter, and M. L.
Rodrigues. 2005. Structure, cellular distribution, antigenicity, and biological
functions of Fonsecaea pedrosoi ceramide monohexosides. Infect. Immun.
73:7860–7868.
25. Nimrichter, L., M. L. Rodrigues, E. G. Rodrigues, and L. R. Travassos. 2005.
The multitude of targets for the immune system and drug therapy in the
fungal cell wall. Microbes Infect. 7:789–798.
26. Novikoff, A. B., and P. M. Novikoff. 1977. Cytochemical contributions to
differentiating GERL from the Golgi apparatus. Histochem. J. 9:525–551.
27. Oscarson, S., M. Alpe, P. Svahnberg, A. Nakouzi, and A. Casadevall. 2005.
Synthesis and immunological studies of glycoconjugates of Cryptococcus neo-
formans capsular glucuronoxylomannan oligosaccharide structures. Vaccine
23:3961–3972.
28. Rittershaus, P. C., T. B. Kechichian, J. C. Allegood, A. H. Merrill, Jr., M.
Hennig, C. Luberto, and M. Del Poeta. 2006. Glucosylceramide synthase is
an essential regulator of pathogenicity of Cryptococcus neoformans. J. Clin.
Investig. 116:1651–1659.
29. Rodrigues, M. L., C. S. Alviano, and L. R. Travassos. 1999. Pathogenicity of
Cryptococcus neoformans: virulence factors and immunological mechanisms.
Microbes Infect. 1:293–301.
30. Rodrigues, M. L., L. R. Travassos, K. R. Miranda, A. J. Franzen, S. Rozental,
W. de Souza, C. S. Alviano, and E. Barreto-Bergter. 2000. Human antibodies
against a purified glucosylceramide from Cryptococcus neoformans inhibit
cell budding and fungal growth. Infect. Immun. 68:7049–7060.
31. Sakaguchi, N. 1993. Ultrastructural study of hepatic granulomas induced by
Cryptococcus neoformans by quick-freezing and deep-etching method. Vir-
chows Arch. B 64:57–66.
32. Sakaguchi, N., T. Baba, M. Fukuzawa, and S. Ohno. 1993. Ultrastructural
study of Cryptococcus neoformans by quick-freezing and deep-etching
method. Mycopathologia 121:133–141.
33. Scallen, T. J., A. K. Dhar, and E. D. Loughran. 1971. Isolation and charac-
terization of C-4 methyl intermediates in cholesterol biosynthesis after treat-
ment of rat liver in vitro with cholestan-3 beta, 5 alpha,6 beta-triol. J. Biol.
Chem. 246:3168–3174.
34. Su¨tterlin, C., T. L. Doering, F. Schimmoller, S. Schroder, and H. Riezman.
1997. Specific requirements for the ER to Golgi transport of GPI-anchored
proteins in yeast. J. Cell Sci. 110:2703–2714.
35. Takeo, K., I. Uesaka, K. Uehira, and M. Nishiura. 1973. Fine structure of
Cryptococcus neoformans grown in vitro as observed by freeze-etching. J.
Bacteriol. 113:1442–1448.
36. Takeo, K., I. Uesaka, K. Uehira, and M. Nishiura. 1973. Fine structure of
Cryptococcus neoformans grown in vivo as observed by freeze-etching. J.
Bacteriol. 113:1449–1454.
37. Thevissen, K., D. C. Warnecke, I. E. Francois, M. Leipelt, E. Heinz, C. Ott,
U. Zahringer, B. P. Thomma, K. K. Ferket, and B. P. Cammue. 2004.
Defensins from insects and plants interact with fungal glucosylceramides.
J. Biol. Chem. 279:3900–3905.
38. Tucker, S. C., and A. Casadevall. 2002. Replication of Cryptococcus neofor-
mans in macrophages is accompanied by phagosomal permeabilization and
accumulation of vesicles containing polysaccharide in the cytoplasm. Proc.
Natl. Acad. Sci. USA 99:3165–3170.
39. Vecchiarelli, A. 2000. Immunoregulation by capsular components of Crypto-
coccus neoformans. Med. Mycol. 38:407–417.
40. Yoneda, A., and T. L. Doering. 2006. A eukaryotic capsular polysaccharide is
synthesized intracellularly and secreted via exocytosis. Mol. Biol. Cell 17:
5141–5152.
41. Zaragoza, O., A. Telzak, R. A. Bryan, E. Dadachova, and A. Casadevall.
2006. The polysaccharide capsule of the pathogenic fungus Cryptococcus
neoformans enlarges by distal growth and is rearranged during budding. Mol.
Microbiol. 59:67–83.
42. Zaragoza, O., and A. Casadevall. 2004. Experimental modulation of capsule
size in Cryptococcus neoformans. Biol. Proced. Online 6:10–15.
VOL. 6, 2007 GXM-CONTAINING VESICLES IN C. NEDFORMANS 59
70
Anexo 2:
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/copyright
Author's personal copy
Cryptococcus neoformans cryoultramicrotomy and vesicle fractionation
reveals an intimate association between membrane lipids
and glucuronoxylomannan
Débora L. Oliveira
a
, Leonardo Nimrichter
a
, Kildare Miranda
b
, Susana Frases
c,d,1
, Kym F. Faull
e
,
Arturo Casadevall
c,d,2
, Marcio L. Rodrigues
a,
*
,2
a
Laboratorio de Estudos Integrados em Bioquimica Microbiana, Instituto de Microbiologia Professor Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro
21941590, Brazil
b
Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941590, Brazil
c
Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
d
Departments of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
e
Pasarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry and Biobehavioral Sciences, David Geffen School
of Medicine, University of California, Los Angeles, CA 90095, USA
article info
Article history:
Received 3 August 2009
Accepted 2 September 2009
Available online 10 September 2009
Keywords:
Cryptococcus neoformans
GXM transport
Secretory vesicles
Cryoimmunoelectronmicroscopy
abstract
Cryptococcus neoformans is an encapsulated pathogenic fungus. The cryptococcal capsule is composed of
polysaccharides and is necessary for virulence. It has been previously reported that glucuronoxyloman-
nan (GXM), the major capsular component, is synthesized in cytoplasmic compartments and transported
to the extracellular space in vesicles, but knowledge on the organelles involved in polysaccharide synthe-
sis and traffic is extremely limited. In this paper we report the GXM distribution in C. neoformans cells
sectioned by cryoultramicrotomy and visualized by transmission electron microscopy (TEM) and poly-
saccharide immunogold staining. Cryosections of fungal cells showed high preservation of intracellular
organelles and cell wall structure. Incubation of cryosections with an antibody to GXM revealed that
cytoplasmic structures associated to vesicular compartments and reticular membranes are in close prox-
imity to the polysaccharide. GXM was generally found in association with the membrane of intracellular
compartments and within different layers of the cell wall. Analysis of extracellular fractions from cryp-
tococcal supernatants by transmission electron microscopy in combination with serologic, chromato-
graphic and spectroscopic methods revealed fractions containing GXM and lipids. These results
indicate an intimate association of GXM and lipids in both intracellular and extracellular spaces consis-
tent with polysaccharide synthesis and transport in membrane-associated structures.
Ó 2009 Elsevier Inc. All rights reserved.
1. Introduction
The most prominent feature of the fungal pathogen Cryptococ-
cus neoformans is the presence of a capsule surrounding the yeast
cell body. This structure is composed mainly of polysaccharides,
including glucuronoxylomannan (GXM) and galactoxylomannan
(GalXM) (Zaragoza et al., 2009). The cryptococcal capsule is crucial
for fungal cell survival in different situations, including environ-
mental interactions with predators (Steenbergen et al., 2001)or
evasion of the immune system during human and animal infec-
tions (Monari et al., 2008; Vecchiarelli, 2007). GXM is considered
the most important virulence factor of C. neoformans (McClelland
et al., 2005) and, for this reason, it is also the most well studied
structural component.
GXM biosynthesis involves several enzymatic steps, starting
with sugar polymerization using nucleotide-activated sugar mono-
mers as precursors (Wills et al., 2001; Zaragoza et al., 2009). In con-
trast to cell wall polysaccharides like chitin and glucans, which are
most probably synthesized at plasma membrane level, GXM epi-
topes are found at intracellular sites (Feldmesser et al., 2001;
Garcia-Rivera et al., 2004; Yoneda and Doering, 2006), that pre-
sumably reflect the sites of initial assembly. Mutation in the gene
coding for a Rab/GTPase ortologue of the Saccharomyces cerevisiae
Sec4p caused accumulation of cytoplasmic GXM-containing vesi-
cles (Yoneda and Doering, 2006), raising the possibility that newly
synthesized polysaccharides might be exported to the cell surface
by vesicular traffic. Since Sec4p is involved in post-Golgi secretion
1087-1845/$ - see front matter Ó 2009 Elsevier Inc. All rights reserved.
doi:10.1016/j.fgb.2009.09.001
* Corresponding author.
E-mail addresses: [email protected], [email protected] (M.L.
Rodrigues).
1
Pressent address: Instituto Nacional de Metrologia, Normalizacao e Qualidade
Industrial, 50 Ave. NS das Gracas, Xerem 25259-020 Duque de Caxias, RJ, Brazil.
2
These authors share senior authorship.
Fungal Genetics and Biology 46 (2009) 956–963
Contents lists available at ScienceDirect
Fungal Genetics and Biology
journal homepage: www.elsevier.com/locate/yfgbi
Author's personal copy
events, it was deduced that GXM was at least partially synthesized
at the Golgi apparatus (Yoneda and Doering, 2006). GXM-contain-
ing vesicles are released to the extracellular milieu (Rodrigues
et al., 2008, 2007) in a process that apparently involves the SEC6
gene (Panepinto et al., 2009). Little is known about the biogenesis
of extracellular GXM-containing vesicles. Although different stud-
ies suggest a role of genes related to conventional post-Golgi secre-
tion in vesicle release (Panepinto et al., 2009; Yoneda and Doering,
2006), proteomic analysis of isolated C. neoformans vesicles reveled
similarities with mammalian exosomes (Rodrigues et al., 2008).
Our knowledge on the steps of GXM biosynthesis and cellular
traffic is limited by the paucity of information on its intracellular
distribution. Previous ultrastructural analysis of C. neoformans cells
by immunoelectron microscopy (IEM) revealed that antibodies
raised against GXM recognize intracellular sites (Feldmesser
et al., 2001; Garcia-Rivera et al., 2004; Yoneda and Doering,
2006, 2009). Conventional IEM procedures, however, include sev-
eral steps of dehydration and embedding with different resins,
which markedly affects preservation of the cell envelope and intra-
cellular structures and organelles, and generates structural arti-
facts. Such subcellular modifications usually include loss of
cytoplasmic content, poor preservation of membranous structures
and cell wall shrinkage (Yamaguchi et al., 2005).
Cryoimmunogold labeling has been successfully used in differ-
ent studies of the C. neoformans biology (Rodrigues et al., 2008,
2007, 2000). Using this technique, in which cells are promptly
fixed, frozen and sectionated, the artifacts that can plague conven-
tional IEM are reduced or avoided, and the result is superior pres-
ervation of subcellular structures, such as the nucleus, vacuoles
and mitochondria. In addition, cryosectioning has been suggested
to be the electron microscopy technique that is best suited for pre-
serving cellular epitopes (Slot and Geuze, 2007).
In a previous study by our group, we used a cryoultramicrotomy
protocol to study the cellular distribution of a glycolipid antigen in
C. neoformans (Rodrigues et al., 2000). Well preserved cells were
obtained, which allowed us to examine in detail different cellular
structures, including the cell wall and membranous structures. In
the present work, that method was used in association with vesicle
isolation and lipid analysis for the study of the distribution of GXM
in C. neoformans. Our data revealed that the polysaccharide is asso-
ciated with lipid matrices and previously unknown subcellular and
extracellular structures, indicating that GXM traffic in C. neofor-
mans involves still unclear cellular events that may include uncon-
ventional steps of secretion.
2. Material and methods
2.1. Fungal cells
C. neoformans strain H99 was grown in minimal medium com-
posed of dextrose (15 mM), MgSO
4
(10 mM), KH
2
PO
4
(29.4 mM),
glycine (13 mM), and thiamine-HCl (3
l
M). Fungal cells were cul-
tivated with shaking in Erlenmeyer flasks at 30 °C for 72 h. Cells
were collected by centrifugation and then processed for IEM.
2.2. Electron microscopy
Electron microscopy procedures were based on the studies by
Tokuyasu (1973) and Rodrigues and colleagues (2000)). The cells
were fixed in 0.1 M sodium cacodylate buffer (pH 7.2) containing
4% paraformaldehyde, 0.2% glutaraldehyde, and 1% picric acid for
60 min at room temperature. The cells were then washed three
times in PBS, infiltrated in gelatin, cut into cubes of 1 mm and infil-
trated in 25% polyvinylpyrrolidone (PVP) and 2.3 M sucrose over-
night in a cold room. The polyvinylpyrrolidone embedded blocks
were then mounted on cryoultramicrotome stubs and flash frozen
by immersion in liquid nitrogen. After trimming, ultrathin cryo-
sections were obtained at a temperature of À130 °C using an
Ultracut UCT cryoultramicrotome (Reichert). Flat ribbons of sec-
tions were shifted from the knife edge with an eyelash and picked
up in a wire loop filled with a drop of 1% (w/v) methyl cellulose,
2.3 M sucrose in PBS buffer. Sections were thawed on the pickup
droplet and transferred, sections downwards, to Formvar carbon-
coated nickel grids.
For immunolabelling, grids were passed over a series of droplets
of washing and were blocked in PBS-bovine serum albumin 1%.
After blocking the cryosections were incubated overnight in the
presence of 1
l
g/ml of the murine monoclonal antibody (mAb) to
GXM 18B7, washed again and incubated for 1 h with 15 nm (parti-
cle size) gold-labeled anti-mouse IgG. In control systems, the cryo-
sections were incubated with PBS instead of the mAb 18B7,
followed by the gold-labeled anti-mouse IgG. After a final washing
series in PBS and distilled water, the sections were left for 15 min
on 2% uranyl acetate (aqueous) and transferred to 0.75% methyl
cellulose droplets. After 30 s, the grids were looped out, the excess
viscous solution was drained away and the sections were allowed
to dry. After drying, specimens were observed in a JEOL 1200EX
transmission electron microscope operating at 80 kV.
Transmission electron microscopy was used to analyze the
morphology and the integrity of extracellular vesicles isolated from
culture supernatants. Pellets obtained after centrifugation of cell-
free supernatants at 100,000g (see vesicle isolation and fractionation
below) were fixed with 2% glutaraldehyde in 0.1 M cacodylate at
room temperature for 2 h, and then incubated overnight in 4%
formaldehyde, 1% glutaraldehyde, and 0.01 M PBS. The samples
were incubated for 90 min in 2% osmium, serially dehydrated in eth-
anol, and embedded in Spurrs epoxy resin. Thin sections were
obtained on a Reichart Ultracut UCT and stained with 0.5% uranyl
acetate and 0.5% lead citrate. Samples were observed in a JEOL
1200EX transmission electron microscope operating at 80 kV.
2.3. Vesicle isolation and fractionation
Isolation of extracellular vesicles was based on a protocol de-
scribed by Rodrigues et al. (2007). Cell-free culture supernatants
were obtained by sequential centrifugation at 5000 and 15,000g
(15 min, 4 °C) for removal of cells and debris. These supernatants
were concentrated approximately 20-fold using an Amicon ultrafil-
tration system (cutoff 100 kDa). The concentrate was again
sequentially centrifuged at 4000 and 15,000g (15 min, 4 °C) and
the remaining supernatant was then ultracentrifuged at 100,000g
for 1 h at 4 °C. The supernatant was discarded, and the pellet was
washed by five sequential suspension and centrifugation steps,
each consisting of 100,000g for 1 h at 4 °C with 0.1 M Tris-buffered
saline. To remove extravesicular GXM contamination, vesicles
were subjected to passage through a column packed with cyano-
gen bromide-activated Sepharose coupled to a monoclonal anti-
body to GXM, as described previously (Rodrigues et al., 2007).
Ultracentrifugation pellets were finally fractionated in Optiprep
gradients, as previously described for the analysis of virions and
mammalian exosomes (Cantin et al., 2008). Optiprep gradients
were prepared in PBS as 11 steps of 300
l
l in 1.2% increments
ranging from 6% to 18%. Vesicles were layered onto the top of
the gradient and ultracentrifuged at 250,000g for 75 min. Eleven
fractions of the gradient were collected from top to bottom. Each
fraction was then analyzed for the presence of sterols and GXM.
2.4. Lipid analysis
The pellets obtained from centrifugation of cell supernatants at
100,000g were first suspended in methanol, and then two volumes
D.L. Oliveira et al. / Fungal Genetics and Biology 46 (2009) 956–963
957
Author's personal copy
of chloroform were added. The mixture was vigorously vortexed
and centrifuged to separate immediately formed polysaccharide-
containing precipitates. The supernatant was removed, dried with
a nitrogen stream, and partitioned according to Folch et al. (1957).
The lower phase, containing neutral lipids, was recovered and
dried by vacuum centrifugation. The dry residue was then pre-
pared for phospholipid analysis by combined liquid chromatogra-
phy–tandem mass spectrometry (LC/MS/MS) or sterol analysis by
thin-layer chromatography (TLC). For sterol analysis, the lipid ex-
tract was loaded into TLC silica plates (Si 60F254s; LiChrospher,
Germany) and separated using a solvent system containing
hexane:ether:acetic acid (80:40:2, vol:vol:vol) solvent. The plate
was sprayed with a solution of 50 mg ferric chloride (FeCl
3
)ina
mixture of 90 ml water, 5 ml acetic acid, and 5 ml sulfuric acid.
Sterol spots were identified by the appearance of a purple color
after heating the plates at 100 °C for 3–5 min. For phospholipid
identification, the dry residue was suspended in 90% methanol.
After centrifugation, aliquots of the supernatant (typically 100
l
l)
were injected onto a reverse phase HPLC column (Supleco
Ascentis
Ò
Express, C18, 150 Â 2.1 mm) equilibrated in buffer A
(methanol/water, 95/5, v/v, containing 1 mM ammonium acetate),
and eluted (100
l
l/m) with an increasing concentration of buffer B
(chloroform/water, 500/0.2, v/v, containing 1 mM ammonium ace-
tate; min/%B, 0/0, 5/0, 55/100). The effluent from the column was
split, with a portion (about 70%) directed to a fraction collector
(1 min fractions) and the remainder (about 30%) passed to an Ion-
spray
Ò
source connected to a triple quadrupole mass spectrometer
(PE Sciex API III
+
) operating in the precursor (parent) scan mode in
which Q1 was scanned (0.3 Da step, 6.7 s/scan, orifice 50 V), the
collision chamber was flooded with argon gas (collision gas thick-
ness setting at 120), and Q3 was set to transmit ions at m/z
184.1 Da.
To confirm assignments made from the LC/MS/MS data, and to
further define the composition of the phosphocholine lipids in
the sample, positive and negative ion mass spectra were recorded
off-line by direct injection of aliquots of fractions collected during
LC/MS/MS. Fractions 24–29 were pooled, and aliquots (20
l
l) were
injected into a stream of methanol/chloroform (4/1, v/v; 50
l
l/m)
entering the Ionspray
Ò
source of the mass spectrometer, before
and following the addition of formic acid (1
l
l, positive ion mode)
and triethylamine (1
l
l, negative ion mode). These experiments
were done with the mass spectrometer operating in either the nor-
mal (Q1 scan range m/z 200–1500, orifice 70 V, 4.6 s/scan), or the
tandem mass spectrometric modes (precursor and fragment scan
modes). In particular, negative ion fragment scans at high orifice
voltage (120) and relatively high collision gas density (CGT setting
150), were used to assign phosphocholine fatty acid substituents
after the addition of triethylamine. These experiments were done
using methods originally described by Jensen et al. (1986). Putative
parent ions for these experiments were assigned from the results
of the LC/MS/MS parent ion data, and confirmed by the results
from the direct injection analyses. The parent ions were assigned
as 16 Da lighter than that determined experimentally, with the loss
of 15 Da predicted to result from source fragmentation at high ori-
fice voltage (loss of one nitrogen methyl group), and 1 Da loss
resulting from abstraction of the proton attached to the phospho-
rous oxygen under basic conditions. Data was analyzed with
instrument-supplied software (MacSpec version 3.3), and optimal
conditions for the experiments were established using authentic
di-palmitoylphosphatidyl choline, heptadecanoyl-lyso-phosphati-
dyl choline and di-heptanoylphosphatidyl choline standards.
2.5. GXM capture ELISA
A 96-well polystyrene plate was coated with the capture IgM
mAb 2D10 (10
l
g/ml) for 1 h and then blocked with 1% bovine ser-
um albumin for. The plates were then incubated with GXM frac-
tions from vesicle samples, obtained by precipitation with a
mixture of chloroform and methanol (see item lipid analysis). After
three washing cycles with TBS 0,05% Tween 20, the plates were
incubated with an IgG1 to GXM (mAb 18B7, 2
l
g/ml) for 1 h at
room temperature. Positive reactions were developed by incuba-
tion with an alkaline phosphatase-conjugated anti-IgG1 followed
by reaction with p-nitrophenyl phosphate disodium hexahydrate
for 30 min. Absorbance values were measured at 405 nm in a
microplate reader.
3. Results and discussion
Electron microscopy procedures for the study of the C. neofor-
mans biology are complex because this fungal species is sur-
rounded by a polysaccharide capsule, which is attached to a thick
cell wall. The combination of a large capsule and thick cell wall
can slow fixation process resulting in cellular degradation. Hence,
obtaining high quality micrographs of cryptococcal cells can take
some trial and error to identify the best conditions for fixation.
In this context, the commonly used protocols for immunogold
analysis of C. neoformans usually include particular steps, such as
partial digestion of the cell wall (Yoneda and Doering, 2006), to en-
sure a more efficient process of embedding and consequent preser-
vation of intracellular compartments. Unfortunately, these
procedures are often accompanied by loss of surface components
and, consequently, produce micrographs that convey limited infor-
mation on the cryptococcal cellular anatomy (Yamaguchi et al.,
2005). The advantages of using cryosectioning techniques have al-
ready been demonstrated in the model yeast S. cerevisiae, where
protocols based on the method of Tokuyasu (1973) resulted in high
resolution images with well preserved cellular structures and effi-
cient immunogold labeling (Griffith et al., 2008). The general pro-
cedures to prepare biological samples according to this method
include fixation, infiltration/cryoprotection and rapid freezing in li-
quid nitrogen.
In the current study cryoultramicrotomy allowed the genera-
tion of well-preserved C. neoformans sections (Fig. 1). Intracellular
compartments such as the nucleus, vacuoles, mitochondria, cell
wall and capsule were clearly distinguishable. Even in the absence
of a post-fixation step with OsO
4
, lipid bilayers were well pre-
served and easily discernible, as evidenced in nuclear compart-
ments and mitochondrial lamellas (Fig. 1). In addition to the
better known organelles, other subcellular structures were evident,
including reticular membrane clusters close to the plasma mem-
brane (Fig. 1C, black arrows). These compartments showed mor-
phological similarities to structures derived from the
endoplasmic reticulum, although their peripheral position inside
the cell suggested early endosomes (Gould and Lippincott-Sch-
wartz, 2009). A membranous compartment that resembled the
endoplasmic reticulum was also visible in the proximity of nucleus
(Fig. 1C). Finally, vesicular structures manifesting electron dense
contents were also apparent (Fig. 1B). These structures had similar-
ities to intermediate/late endosomal compartments called acido-
somes (Allen et al., 1993) and also to mammalian melanosomes
at different maturation stages (Raposo and Marks, 2007). These
compartments, which were consistently observed in our prepara-
tions, are shown in more detail in Fig. 2. Similar intracellular struc-
tures that contained GXM were recently described by Yoneda and
Doering using a secretion-deficient mutant of C. neoformans
(Yoneda and Doering, 2009). However, the electron dense com-
partments observed in our model do not display such intense
GXM labeling (Fig. 2), as described in that study for C. neoformans
cells. In our model, some of the intracellular compartments also
contained internal vesicles (Fig. 1B and C) that show similarities
958 D.L. Oliveira et al. / Fungal Genetics and Biology 46 (2009) 956–963
Author's personal copy
to the so-called multivesicular bodies (MVBs) found in mammalian
cells (Gruenberg and Stenmark, 2004). The existence of MVBs in C.
neoformans was previously suggested using different techniques
(Rodrigues et al., 2008; Takeo et al., 1973), although differences
in morphology and electron density were noticed by comparing
the currently described results with those from previous studies.
Intracellular sites manifesting immunogold staining with mAb
to GXM were usually found in association with membranous struc-
tures (Fig. 3). Quantification of immunogold labeling in intracellu-
lar compartments revealed that approximately 70% of the
antibody-binding sites were membrane-associated. On the basis
of this reactivity we tentatively assigned these areas as sites of
GXM synthesis/transport. Membrane compartments resembling
vesicles carrying GXM were repeatedly observed (Fig. 3), as well
as structures resembling mammalian MVBs (Fig. 3C) and periphe-
ral reticular clusters resembling endosomes (Fig. 3E). Since GXM
staining was often found in close association with the lipid bilayer,
we hypothesize that polysaccharide-binding proteins could be in-
serted in these lipid bilayers functioning as transient GXM anchors,
which still remains to be investigated. Considering that the size of
the complex gold-labeled antibody to GXM may in some instances
exceed the diameter of those structures, we cannot rule out the
possibility that they are located in neighboring intracellular re-
gions, although previous data indeed demonstrate that GXM asso-
ciated with secretory vesicles (Casadevall et al., 2009; Feldmesser
et al., 2001; Garcia-Rivera et al., 2004; Rodrigues et al., 2007;
Yoneda and Doering, 2006).
The intracellular, electron dense compartments described by
Yoneda and Doering (2009) were called sav1 bodies, since they
were most prominently observed in C. neoformans cells with
Fig. 1. Anatomy of the C. neoformans ultrastructure. C. neoformans cells were grown to stationary phase and prepared for EM as described in Section 2. Several organelles were
distinguished, including nucleus (N), vacuoles (V), mitochondria (M), cell wall (CW) and a compacted capsule (Cap). The existence of lamellar structures with unknown
cellular roles (black arrows) is suggested, as well as of vesicular electron dense compartments (white arrows). Black arrow heads indicate putative endoplasmic reticulum.
Unlabeled cells (A and B) and cells stained for the presence of GXM (C) are shown.
D.L. Oliveira et al. / Fungal Genetics and Biology 46 (2009) 956–963
959
Author's personal copy
Fig. 2. Electron dense organelles are present in C. neoformans. The boxed area shown in A is shown in higher magnification in B. Arrows indicate the electron dense bodies.
The composition of these electron dense structures is unknown. Cell surface labeling represents reactivity of cryptococcal components with mAb18B7.
Fig. 3. Cryptococcal GXM is found in association with vesicles and membranous structures. Cells were labeled with GXM binding mAb 18B7. GXM is generally detected in
association with lipid bilayers. In A and D, the arrowhead indicates that GXM can be localized inside vesicles that also present an electron dense content. In B and F, GXM
seems to be localized inside small cytoplasmic vesicle carriers. In F GXM is also presented inside the vacuole (V). In C, an unknown structure containing internal vesicles also
displays mAb18B7 labeling, as well as reticular unknown structures close to the plasma membrane displayed in E.
960 D.L. Oliveira et al. / Fungal Genetics and Biology 46 (2009) 956–963
Author's personal copy
defective expression of sav1p, a putative small GTPase involved in
post-Golgi secretion. The compartments observed in our model are
apparently distinct from the sav1 bodies, since they did not mani-
fest significant reactivity with an antibody to GXM, despite mor-
phological similarities (Figs. 2 and 3A and D). The presence of
GXM in vacuoles (Fig. 3F) could be related to secretion mechanisms
initiated by endocytosis or to mechanisms of GXM recycling. In
fact, the GXM catabolic process is poorly understood and so far
hydrolytic enzymes able to cleave GXM have not been reported
in C. neoformans (Bose et al., 2003; Zaragoza et al., 2009).
Conventional procedures for TEM usually generate artificially
compacted cell wall structures, probably because of dehydration
steps. In our model, as well as in a previous study, cryoultramicr-
otomy procedures allowed the observation of structures that illus-
trate the thickness and complexity of the cell wall. As suggested in
previous studies with fungal cells (Soragni et al., 2001), clear dis-
tinctions between regions of different electron densities were ob-
served (Figs. 1B,C and 3F), which may be related to the
description of secondary cell walls during stationary growth of C.
neoformans (Farkas et al., 2009). Most of the cellular sites recog-
nized by the mAb to GXM were found in association with the cell
wall or within the capsular region (Fig. 1C), corroborating with pre-
vious observations (Yoneda and Doering, 2006).
GXM has been proposed to be exported to the extracellular
space in secretory vesicles, suggesting an association of lipids
and polysaccharides in C. neoformans (Rodrigues et al., 2007). This
notion is supported by our current observations in the cryptococcal
cytoplasm. Glucosylceramide and sterols were previously
Fig. 4. Analysis of phosphatidylcholine (PC) in extracellular vesicles. (A) Transmission electron microscopy showing vesicle morphology in 100,000g fractions isolated from
culture supernatants. (B) Characterization of PC in vesicle fractions: (a) total ion current LCMSMS chromatogram obtained following injection of the crude vesicle preparation
onto a reverse phase column as described in Section 2. The mass spectrometer was operated in the MS/MS precursor (parent) scan mode in which the m/z value of parent ions
that give rise to the m/z 184.1 fragment are recorded, (b–d) electrospray ionization mass spectra of the three peaks shown in Figure A (Peaks 1–3, MS/MS positive ion
precursor (parent) scan mode, precursors of m/z 184.1) and (e) negative ion electrospray mass spectrum of the fragments derived from an m/z 766.7 parent ion under MS/MS
conditions after addition of triethylamine as described in Section 2. Calculated values: C
44
H
81
NO
8
P 782.570 Da; C
44
H
83
NO
8
P 784.586 Da; C
44
H
85
NO
8
P 786.601 Da; C
18
H
31
O
2
279.232 Da.
D.L. Oliveira et al. / Fungal Genetics and Biology 46 (2009) 956–963
961
Author's personal copy
characterized as membrane components of the C. neoformans ves-
icles (Rodrigues et al., 2007), but other lipid components of GXM-
containing vesicles are still unknown. To evaluate the relationship
of lipids with GXM export, we first tested whether extracellular
vesicle samples contained GXM by ELISA. Vesicles obtained from
encapsulated cells reacted with the antibody to GXM, while vesi-
cles from an acapsular mutant manifested a much lower reactivity
(data not shown). These results were in agreement with those de-
scribed by Rodrigues and co-workers (2007) and support the
hypothesis that the major capsular polysaccharide of C. neoformans
associates with lipids for extracellular export. Since phospholipids
are major membrane constituents in most cells, but also in extra-
cellular vesicles of the fungal pathogen Histoplasma capsulatum
(Albuquerque et al., 2008), we evaluated whether the crude frac-
tion of GXM-containing vesicles contained this class of molecules.
To ensure that lipid characterization did not include membrane
fractions from non-vesicular structures, the integrity and morphol-
ogy of 100,000g fractions isolated from culture supernatants was
analyzed by TEM. Vesicles in these fractions showed typical bilay-
ered membranes that generally formed round compartments in the
size range of 50–300 nm (Fig. 4A), which is in agreement with pre-
vious reports on the morphological characterization of fungal
extracellular vesicles (Rodrigues et al., 2008; Eisenman et al., in
press). These fractions were then used for phospholipid extraction.
Cryptococcal phospholipids include phosphatidylinositol, lyso-
phosphatidyl ethanolamine, cardiolipin, glycophospholipids, lyso-
phosphatidyl choline, phosphatidic acid, phosphatidyl
ethanolamine and phosphatidyl choline (PC) (Rawat et al., 1984).
PC was the major phospholipid component of C. neoformans cellu-
lar membranes (Rawat et al., 1984), so this molecule was selected
as a potential candidate component of cryptococcal vesicles. Sam-
ples were subjected to precursor ion scanning of the m/z 184.1
fragment. This ion corresponds to phosphorylcholine, and there-
fore represents a molecular marker for the presence of PC. Control
samples consisted of LCMS analysis of the solvent alone, which
showed no significant peaks. From the vesicle samples, three
strong peaks were observed eluting between 24 and 29 min
(Fig. 4B, panel a). These peaks revealed parent masses of 782.7,
784.6 and 786.8 Da (Fig. 4B, panel b–d). LCMS analysis of two dif-
ferent samples produced essentially identical results (data not
shown). Fractions eluting between 24 and 29 min were pooled
and examined by direct flow-injection MS and MS/MS. Fragmenta-
tion of the peak at m/z 782.7 generated a strong signal at m/z 184
(data not shown), confirming the occurrence of PC. To analyze fatty
acid components, negative MS/MS analysis was performed after
addition of triethylamine. Analysis of the peaks at m/z 782.7 and
784.6 revealed the presence of related fragments at m/z 279.2
(Fig. 4B, panel e) and 281.0 (not shown), consistent with the pres-
ence of, respectively, fatty acids corresponding to C18:2 and C18:1.
The peak at m/z 786.8 did not yield a significant response probably
because of limitations in amount of sample (data not shown).
In our study, intracellular GXM-containing vesicles were
repeatedly observed (Fig. 3), as well as several unlabeled vesicular
structures (Figs. 1C and 2). In this context, we evaluated whether
the distribution of GXM in extracellular vesicles could reflect the
results observed in cellular structures. Fractionation of vesicle
samples by gradient centrifugation followed by lipid and polysac-
charide
analysis
revealed a previously unknown compositional
profile of extracellular fungal vesicles (Fig. 5). Sterol analysis was
used to track membrane-containing fractions, since ergosterol
has been characterized as an easily detectable component of vesic-
ular membranes (Rodrigues et al., 2007). Phospholipid analysis in
gradient fractions was not pursued due to poor sensitivity for lipid
detection in chromatographic methods.
Ergosterol was detected in all fractions obtained after ultracen-
trifugation. GXM-containing fractions of different densities
(fractions 4–6 and 10), as well as sterol-containing fractions that
showed virtually no reactivity with an antibody to GXM (fractions
1–2 and 7–9), were observed. In some cases, the amounts of
GXM and sterols in gradient fractions were directly correlated
Fig. 5. Fractionation of C. neoformans extracellular vesicles. A crude preparation of extracellular vesicles was fractionated in a 6–18% Optiprep velocity gradient. Eleven
fractions were recovered and individually tested for the presence of GXM by ELISA and ergosterol by thin-layer chromatography (TLC). TLC bands (sterol detection) were
quantified by densitometry. Sterol distribution in fractions (dotted line) and GXM content (bars) are shown.
962 D.L. Oliveira et al. / Fungal Genetics and Biology 46 (2009) 956–963
Author's personal copy
(e.g. fractions 4–6), but fractions with lower ergosterol content and
high GXM density were also observed (e.g. fraction 10). These re-
sults are consistent with previously noted heterogeneity in vesicle
size and composition (Albuquerque et al., 2008; Rodrigues et al.,
2008) and indicate that some vesicles are preferentially associated
with polysaccharide transport while others contain no GXM, as
noted in prior immunogold labeling studies (Rodrigues et al.,
2007). The observation of extracellular vesicle fractions with such
different properties indicate that the cellular events required for
vesicle biogenesis are multiple and complex, as previously sug-
gested by morphological analysis of cryptococcal extracellular ves-
icles (Rodrigues et al., 2008). Our results add to the evolving theme
an intimate and intriguing association between lipid and polysac-
charide components in C. neoformans capsule (Nicola et al., 2009;
Sebolai et al., 2007, 2008).
In this work we report the use of two methodological ap-
proaches for studying the process of polysaccharide synthesis
and transport: (1) a cryosectioning protocol for C. neoformans,
which allowed a distinguished resolution of the fungal cytoplasmic
content and surface structures, and (2) a technique for fractionat-
ing vesicles that separates them into GXM- and non-GXM contain-
ing groups. The cryosectioning EM method allowed us to discern in
more detail the distribution of GXM inside the cell. The vesicle
fractionation approach should allow finer resolution studies of ves-
icle composition and content by biochemical and mass spectrome-
try analysis. The results of these studies revealed new details for
the mechanisms required for polysaccharide traffic and raise new
questions about this complex cellular process. Of particular inter-
est was the association of GXM with membrane structures sug-
gesting that this polysaccharide is synthesized in close proximity
to lipids, possibly by anchored enzymes. The possibility of combin-
ing cryoultramicrotomy with other less invasive methods such as
high pressure freezing followed by freeze substitution and electron
tomography could potentially generate new information for the
study of still unknown cell biology attributes of cell-wall contain-
ing pathogens.
Acknowledgements
A.C. was supported by NIH Grants AI33774, HL59842, AI33142,
and AI52733. M.L.R., L.N. and K.M. were supported by Grants from
the Brazilian agencies CNPq and FAPERJ. We thank Leslie Gunther
and the Analytical Imaging Facility team at Albert Einstein College
of Medicine for their assistance with the electron microscopy
experiments. We also thank Jorge José B. Ferreira for helpful dis-
cussions. DLO is a PhD student at Instituto de Bioquímica Médica,
UFRJ.
References
Albuquerque, P.C. et al., 2008. Vesicular transport in Histoplasma capsulatum:an
effective mechanism for trans-cell wall transfer of proteins and lipids in
ascomycetes. Cell Microbiol. 10, 1695–1710.
Allen, R.D. et al., 1993. Acidosomes: recipients of multiple sources of membrane and
cargo during development and maturation. J. Cell Sci. 106 (Pt 1), 411–422.
Bose, I. et al., 2003. A yeast under cover: the capsule of Cryptococcus neoformans.
Eukaryot. Cell 2, 655–663.
Cantin, R. et al., 2008. Discrimination between exosomes and HIV-1: purification of
both vesicles from cell-free supernatants. J. Immunol. Methods 338, 21–30.
Casadevall, A. et al., 2009. Vesicular transport across the fungal cell wall. Trends
Microbiol. 17, 158–162.
Eisenman, H.C., Frases, S., Nicola, A.M., Rodrigues, M.L., Casadevall. A., Vesicle-
associated melanization in Cryptococcus neoformans. Microbiology, in press,
doi:10.1099/mic.0.032854-0.
Farkas, V. et al., 2009. Secondary cell wall formation in Cryptococcus neoformans as a
rescue mechanism against acid-induced autolysis. FEMS Yeast Res. 9, 311–320.
Feldmesser, M. et al., 2001. Dynamic changes in the morphology of Cryptococcus
neoformans during murine pulmonary infection. Microbiology 147, 2355–2365.
Folch, J. et al., 1957. A simple method for the isolation and purification of total
lipides from animal tissues. J. Biol. Chem. 226, 497–509.
Garcia-Rivera, J. et al., 2004. Cryptococcus neoformans CAP59 (or Cap59p) is involved
in the extracellular trafficking of capsular glucuronoxylomannan. Eukaryot. Cell
3, 385–392.
Gould, G.W., Lippincott-Schwartz, J., 2009. New roles for endosomes: from vesicular
carriers to multi-purpose platforms. Nat. Rev. Mol. Cell Biol. 10, 287–292.
Griffith, J. et al., 2008. A cryosectioning procedure for the ultrastructural analysis
and the immunogold labelling of yeast Saccharomyces cerevisiae. Traffic 9,
1060–1072.
Gruenberg, J., Stenmark, H., 2004. The biogenesis of multivesicular endosomes. Nat.
Rev. Mol. Cell Biol. 5, 317–323.
Jensen, N.J. et al., 1986. Fast atom bombardment and tandem mass spectrometry of
phosphatidylserine and phosphatidylcholine. Lipids 21, 580–588.
McClelland, E.E. et al., 2005. Coping with multiple virulence factors: which is most
important? PLoS Pathog. 1, e40.
Monari, C. et al., 2008. Capsular polysaccharide induction of apoptosis by intrinsic
and extrinsic mechanisms. Cell Microbiol. 10, 2129–2137.
Nicola, A.M. et al., 2009. Lipophilic dye staining of Cryptococcus neoformans
extracellular vesicles and capsule. Eukaryot. Cell. 8, 1373–1380.
Panepinto, J. et al., 2009. Sec6-dependent sorting of fungal extracellular exosomes
and laccase of Cryptococcus neoformans. Mol. Microbiol. 71, 1165–1176.
Raposo, G., Marks, M.S., 2007. Melanosomes dark organelles enlighten endosomal
membrane transport. Nat. Rev. Mol. Cell Biol. 8, 786–797.
Rawat, D.S. et al., 1984. Lipid composition of Cryptococcus neoformans.
Microbiologica 7, 299–307.
Rodrigues, M.L. et al., 2008. Extracellular vesicles produced by Cryptococcus
neoformans contain protein components associated with virulence. Eukaryot
Cell. 7, 58–67.
Rodrigues, M.L. et al., 2007. Vesicular polysaccharide export in Cryptococcus
neoformans is a eukaryotic solution to the problem of fungal trans-cell wall
transport. Eukaryot. Cell. 6, 48–59.
Rodrigues, M.L. et al., 2000. Human antibodies against a purified glucosylceramide
from Cryptococcus neoformans inhibit cell budding and fungal growth. Infect
Immunol. 68, 7049–7060.
Sebolai, O.M. et al., 2007. 3-Hydroxy fatty acids found in capsules of Cryptococcus
neoformans. Can. J. Microbiol. 53, 809–812.
Sebolai, O.M. et al., 2008. The influence of acetylsalicylic acid on oxylipin migration
in Cryptococcus neoformans var. neoformans UOFS Y-1378. Can. J. Microbiol. 54,
91–96.
Slot, J.W., Geuze, H.J., 2007. Cryosectioning and immunolabeling. Nat. Protoc. 2,
2480–2491.
Soragni, E. et al., 2001. A nutrient-regulated, dual localization phospholipase A(2) in
the symbiotic fungus Tuber borchii. EMBO J. 20, 5079–5090.
Steenbergen, J.N. et al., 2001. Cryptococcus neoformans interactions with amoebae
suggest an explanation for its virulence and intracellular pathogenic strategy in
macrophages. Proc. Nat. Acad. Sci. USA 98, 15245–15250.
Takeo, K. et al., 1973. Fine structure of Cryptococcus neoformans grown in vitro as
observed by freeze-etching. J. Bacteriol. 113, 1442–1448.
Tokuyasu, K.T., 1973. A technique for ultracryotomy of cell suspensions and tissues.
J.
Cell
Biol. 57, 551–565.
Vecchiarelli, A., 2007. Fungal capsular polysaccharide and T-cell suppression: the
hidden nature of poor immunogenicity. Crit. Rev. Immunol. 27, 547–557.
Wills, E.A. et al., 2001. Identification and characterization of the Cryptococcus
neoformans phosphomannose isomerase-encoding gene, MAN1, and its impact
on pathogenicity. Mol. Microbiol. 40, 610–620.
Yamaguchi, M. et al., 2005. Safe specimen preparation for electron microscopy of
pathogenic fungi by freeze-substitution after glutaraldehyde fixation. Nippon
Ishinkin Gakkai Zasshi 46, 187–192.
Yoneda, A., Doering, T.L., 2006. A eukaryotic capsular polysaccharide is synthesized
intracellularly and secreted via exocytosis. Mol. Biol. Cell 17, 5131–5140.
Yoneda, A., Doering, T.L., 2009. An unusual organelle in Cryptococcus neoformans
links luminal pH and capsule biosynthesis. Fungal Genet. Biol. 46, 682–687.
Zaragoza, O. et al., 2009. The capsule of the fungal pathogen Cryptococcus
neoformans. Adv. Appl. Microbiol. 68, 133–216 (Chapter 4).
D.L. Oliveira et al. / Fungal Genetics and Biology 46 (2009) 956–963
963
79
Anexo 3:
INFECTION AND IMMUNITY, Apr. 2010, p. 1601–1609 Vol. 78, No. 4
0019-9567/10/$12.00 doi:10.1128/IAI.01171-09
Copyright © 2010, American Society for Microbiology. All Rights Reserved.
Extracellular Vesicles from Cryptococcus neoformans
Modulate Macrophage Functions
De´bora L. Oliveira,
1
Ce´lio G. Freire-de-Lima,
2
Joshua D. Nosanchuk,
3,4
Arturo Casadevall,
3,4
Marcio L. Rodrigues,
1
and Leonardo Nimrichter
1
‡*
Laborato´rio de Estudos Integrados em Bioquímica Microbiana, Instituto de Microbiologia Professor Paulo de Go´ es,
1
and
Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro,
2
Rio de Janeiro, Brazil; Department of
Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461
3
; and Department of
Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461
4
Received 16 October 2009/Returned for modification 20 November 2009/Accepted 2 February 2010
Cryptococcus neoformans and distantly related fungal species release extracellular vesicles that traverse the
cell wall and contain a varied assortment of components, some of which have been associated with virulence.
Previous studies have suggested that these extracellular vesicles are produced in vitro and during animal
infection, but the role of vesicular secretion during the interaction of fungi with host cells remains unknown.
In this report, we demonstrate by fluorescence microscopy that mammalian macrophages can incorporate
extracellular vesicles produced by C. neoformans. Incubation of cryptococcal vesicles with murine macrophages
resulted in increased levels of extracellular tumor necrosis factor alpha (TNF-), interleukin-10 (IL-10), and
transforming growth factor (TGF-). Vesicle preparations also resulted in a dose-dependent stimulation of
nitric oxide production by phagocytes, suggesting that vesicle components stimulate macrophages to produce
antimicrobial compounds. Treated macrophages were more effective at killing C. neoformans yeast. Our results
indicate that the extracellular vesicles of C. neoformans can stimulate macrophage function, apparently
activating these phagocytic cells to enhance their antimicrobial activity. These results establish that crypto-
coccal vesicles are biologically active.
Cryptococcus neoformans is an encapsulated yeast that
causes disease in diverse species, including humans. Infection
is most commonly acquired by inhalation of environmental
propagules. C. neoformans rarely causes disease in immuno-
competent individuals, but patients with immunological disor-
ders can develop disseminated and neural cryptococcosis (63).
Extracellular microbial products have been amply demon-
strated to modulate the interaction between host cells and
pathogens. Many virulence factors and immunogens are re-
leased in their soluble forms by fungal cells to the extracellular
space (4, 9, 16, 19, 37, 49, 53, 60, 62, 65, 67). C. neoformans, for
instance, constitutively secretes large amounts of its capsular
polysaccharide glucuronoxylomannan (GXM) (61). Disease
progress is associated with detection of GXM, which is a po-
tent modulator of the immune response (reviewed in reference
81). Other secreted virulence-related factors include galactoxy-
lomannan (GalXM) (14), phospholipases (16), and urease (12,
62). In addition to acting as virulence factors, culture super-
natant components are immunogenic, conferring protection
against C. neoformans infection (51, 53).
Phagocytes are particularly important effector cells in the
control of systemic mycoses (54). The interaction of C. neofor-
mans with phagocytes, including macrophages, monocytes,
dendritic cells, and neutrophils, has been widely studied (23,
32, 43, 46, 50, 59, 68, 77). Cryptococcal GXM is antiphagocytic
(34) and a powerful immunomodulator (45, 79). C. neoformans
capsule size directly correlates with the efficacy of phagocytosis
in vitro (6, 15, 82). Phagocytosis of C. neoformans can result in
either fungal killing (24, 30) or survival (2, 3, 39–41, 71, 80).
Killing of C. neoformans apparently involves the production of
oxidative species (24), while the mechanisms of fungal escape
include phagosome extrusion, cell-to-cell spread, and phago-
somal permeabilization (2, 3, 40, 41, 71). Capsular polysaccha-
rides and melanin are known to modulate the interaction of C.
neoformans with phagocytes in favor of the fungus (27, 39, 47,
48, 71, 72, 74, 76), but the role of other structures in the
outcome of yeast phagocytosis is virtually unknown.
A number of recent studies have shown that GXM, GalXM,
pigments, proteins, and lipids are trafficked in vesicles that
traverse the cell wall (7, 14, 20, 56, 57, 62, 64, 65). Extracellular
vesicles are also produced by the pathogens Candida albicans,
C. parapsilosis, Sporothrix schenckii, and Histoplasma capsula-
tum, as well as by the model yeast Saccharomyces cerevisiae (1),
suggesting that extracellular vesicle secretion is a general prop-
erty of fungal cells. Secreted vesicles are heterogeneous. For
instance, vesicles secreted by C. neoformans were classified into
four different groups based on morphology and electron den-
sity (64). Additionally, vesicle diameter ranges from 30 to 400
nm, with the majority having dimensions of 100 to 150 nm (20,
64, 65). The combined use of serology, biochemistry, proteo-
mics, and lipidomics led to the identification of 2 polysaccha-
rides, phospholipids, 4 neutral lipids, and 76 proteins as extra-
cellular vesicle components secreted by C. neoformans, which
* Corresponding author. Mailing address: Laborato´rio de Estudos
Integrados em Bioquímica Microbiana, Instituto de Microbiologia
Prof. Paulo de Go´es, Avenida Carlos Chagas Filho 373, Cidade Uni-
versita´ria, Centro de Cieˆncias da Sau´de, Bloco E, sub-solo, Rio de
Janeiro RJ 21941902, Brazil. Phone: 55 21 25626740. Fax: 55 21
25608344. E-mail: [email protected].
† Supplemental material for this article may be found at http://iai
.asm.org/.
‡ L.N. and M.L.R. share senior authorship on this article.
Published ahead of print on 9 February 2010.
1601
by on March 18, 2010 iai.asm.orgDownloaded from
means that at least 81 different molecules are released to the
extracellular milieu by vesicular secretion (14, 57, 64). It is
likely that this number is an underestimate resulting from the
difficulty of proteomic studies in vesicles from highly encapsu-
lated cryptococcal cells, since a higher number of vesicular
proteins were characterized in other fungi. For example, in H.
capsulatum, proteomics and lipidomics of extracellular vesicles
revealed an even more complex composition, including 283
proteins and 17 different phospholipids (1).
In this study, we evaluated the influence of extracellular
vesicles on the fate of C. neoformans after phagocytosis by
mouse macrophages. Our results show that fungal vesicles are
biologically active and stimulate macrophages. Moreover, our
results demonstrate that vesicles from an acapsular mutant
strain were more effective in eliciting macrophage activation
and augmenting fungal killing than vesicles from encapsulated
strains. Taken together, our findings suggest that fungal secre-
tory vesicles have the potential to influence the interaction of
C. neoformans with host cells.
MATERIALS AND METHODS
C. neoformans growth conditions. For vesicle isolation, C. neoformans strains
HEC3393 (serotype A, human isolate), B3501 (serotype D), and Cap 67 (sero-
type D acapsular strain, generated in a B3501 background [28]) were cultivated
for 72 h in a minimal medium composed of dextrose (15 mM), MgSO
4
(10 mM),
KH
2
PO
4
(29.4 mM), glycine (13 mM), and thiamine-HCl (3 M). Fungal cells
were cultivated at room temperature with continuous shaking. For macrophage
infection assays, fungal cells were cultivated in Sabouraud liquid medium for 48 h
at room temperature with continuous shaking. All media were prepared with
apyrogenic water, and glassware was rendered sterile and pyrogen free by heating
to 190°C for 4 h (18).
Macrophage cultures. The RAW 264.7 murine macrophage cell line (obtained
from the American Type Culture Collection [ATCC]) was cultivated in complete
Dulbecco’s minimal essential medium (DMEM) supplemented with 10% fetal
calf serum (FCS), 2 mM
L-glutamine, 1 mM sodium pyruvate, 10 mg ml
Ϫ1
gentamicin, minimal essential medium (MEM) nonessential amino acids (cata-
log no. 11360; Gibco-Invitrogen), 10 mM HEPES, and 50 mM 2-beta-mercap-
toethanol. The cells were maintained at 37°C in a 7.5% CO
2
atmosphere. The
macrophage cultures and the experiments described below were all prepared
under lipopolysaccharide-free conditions.
Vesicle isolation. Isolation of extracellular vesicles was done using the protocol
described by Rodrigues et al. (65). After growth until the stationary phase (ϳ10
7
to 10
8
cells/ml), two liters of cell-free culture supernatants was obtained by
sequential centrifugation at 5,000 and 15,000 ϫ g (15 min at 4°C). The super-
natants were concentrated by approximately 20-fold using an Amicon ultrafil-
tration system (cutoff, 100 kDa). The concentrate was again sequentially centri-
fuged at 4,000 and 15,000 ϫ g (15 min at 4°C). The remaining supernatant was
then ultracentrifuged at 100,000 ϫ g for1hat4°C. The supernatant was
discarded, and the pellet was washed by five sequential suspension and centri-
fugation steps, each consisting of 100,000 ϫ g for1hat4°Cwith 0.1 M Tris-
buffered saline. To remove extravesicular GXM contamination, vesicles were
subjected to passage through a column packed with cyanogen bromide-activated
Sepharose coupled to a monoclonal antibody to GXM, as described previously
(65). Fractions that were not bound to the antibody-containing column were
again centrifuged at 100,000 ϫ g. The presence of extracellular vesicles in the
pellets produced by centrifugation at 100,000 ϫ g was checked by chemical
methods, as previously described (65). The quantification of vesicle fractions was
performed based on the presence of sterols in their membranes by the use of a
quantitative fluorimetric Amplex Red sterol assay kit (Molecular Probes). In
different assays, average values of 6 to 10 g of sterol were detected as the final
product of 2-liter cultures. The total sterol obtained after ultracentrifugation
suggests a recovery of 0.5 fg per yeast cell. The apparent low efficiency during
vesicle isolation was expected, since they could naturally disrupt in the superna-
tant. Vesicle suspensions were then adjusted to 30 to 50 g of sterol/ml of
phosphate-buffered saline (PBS) for use in the subsequent experiments.
Visualization of incorporation of vesicular components by macrophages.
RAW 264.7 cells were plated onto the wells of a 24-well plate covered with glass
coverslips (5 ϫ 10
5
cells per well). Vesicle samples were stained with the li-
pophilic fluorophore dialkylcarbocyanine iodide (DiI) (catalog no. V22885; In-
vitrogen) as described previously (55). Briefly, vesicle suspensions were normal-
ized to a sterol concentration corresponding to 2 g/ml and supplemented with
a DiI solution (final concentration, 3 M). After 30 min at room temperature,
the vesicle suspension was ultracentrifuged at 100,000 ϫ g for 1 h and washed
three times as described above. DiI-labeled vesicles were suspended in PBS to
form 2 g/ml sterol solutions and then incubated with the macrophages for 30
min at 37°C in a 7.5% CO
2
atmosphere. Controls included culture medium
incubated with DiI alone and washed with PBS under the same conditions.
Vesicle-treated macrophages were then fixed with 4% paraformaldehyde in PBS
for 5 min and blocked with PBS–5% bovine serum albumin (BSA) for1hat
room temperature. Plasma membranes and nuclei were labeled with cholera
toxin subunit B (CtxB) (1 g/ml) and 4Ј,6-diamidino-2-phenylindole (DAPI) (10
g/ml), respectively, as described previously (75). Samples were then placed in
mounting medium (50% glycerol–50 mM N-propyl gallate–PBS) over glass slides
and visualized with a Leica AOBS laser scanning confocal microscope (Mann-
heim, Germany) with a 63ϫ oil immersion optic. z-axis series and single scan
images were obtained. The x- and z-axis sections were finally reconstructed using
ImageJ software (NIH [http://rsb.info.nih.gov/ij/]). Alternatively, cells incubated
with DiI-stained vesicles were sequentially blocked in PBS containing 5% BSA
and incubated for1hatroom temperature with pooled serum samples from 10
individuals diagnosed with cryptococcosis. As a negative control, cells were
incubated with normal human serum samples from healthy volunteers with no
previous diagnosis of any systemic mycosis. Pooled serum samples were used at
a 1:100 dilution in PBS–1% BSA. After washing, cells were incubated with Alexa
Fluor 488-labeled anti-human immunoglobulin antibody (Invitrogen). Samples
were then washed, placed in mounting medium as described above, and visual-
ized with an Axioplan 2 fluorescence microscope (Zeiss, Germany). Images were
acquired using a Color View SX digital camera and processed with the analySIS
(Soft Image System) software system.
Cell viability. To evaluate whether the exposure of macrophage-like cells to
vesicle samples would lead to cell lysis, cell supernatants were assayed for the
presence of lactate dehydrogenase (LDH) activity. LDH is a cytoplasmic enzyme
retained by viable cells with intact plasma membranes. Membrane damage re-
sults in LDH release to culture supernatants. RAW 264.7 monolayers (1 ϫ 10
6
cells/well) were stimulated for 16 h with vesicle suspension (2 g/ml sterol). After
this period, aliquots of the supernatant were collected and supplemented with
NADH and pyruvate to final concentrations of 0.3 and 4.7 mM, respectively,
followed by 10 min of incubation at room temperature. Based on the property
that NADH, and not NAD, absorbs strongly at 340 nm, the decrease in absor-
bance (A
340
) was measured in a spectrophotometer. Positive and negative con-
trols consisted, respectively, of a RAW 264.7 Triton X-100 (10%) lysate and
supernatants of nontreated cells after 16 h of cultivation. All experiments were
performed in triplicate sets and statistically analyzed by Student’s t test.
Production of cytokines and nitric oxide (NO) after exposure of macrophages
to vesicle samples. RAW monolayers were washed twice in serum-free DMEM,
placed in medium supplemented with various concentrations of fungal vesicles,
and incubated for 16 h at 37°C (7.5% CO
2
atmosphere). In these experiments, we
used 40 ng of vesicular sterol to activate 10
5
macrophages, suggesting that 0.4 pg
of sterol is enough to activate a macrophage. As a positive control, macrophages
were stimulated with 1 g/ml lipopolysaccharide (LPS). Supernatants were then
collected and assayed for interleukin-10 (IL-10), transforming growth factor
(TGF-), and tumor necrosis factor alpha (TNF-) content by enzyme-linked
immunosorbent assay (ELISA) according to the manufacturer’s protocol (R&D
Systems). Alternatively, NO levels were measured in supernatants as previously
described (25). N6-(1-Iminoethyl)-
L-lysine dihydrochloride (L-NIL) (Cayman
Chemical), the specific inhibitor of the inducible nitric oxide synthase (iNOS),
was used to confirm that the enzyme was specifically induced during the de-
scribed activation process. For these experiments, cells were stimulated with
vesicles during 16 h in the presence of 3 M L-NIL and cultured with 1%
(vol/vol) Nutridoma-SP (Boehringer Mannheim) instead of FCS. To exclude the
possibility that cellular responses were affected by the presence of contaminant
LPS, control experiments were performed in the presence of purified polymyxin
B, an antibiotic that binds LPS and neutralizes its effects. Fungal vesicles and
LPS were incubated with macrophage-like cells in the presence of polymyxin B,
and nitric oxide production was evaluated. All experiments were performed in
triplicate sets and statistically analyzed by using one-way analysis of variance
(ANOVA) followed by a Bonferroni posttest.
Determination of internalization of C. neoformans by flow cytometry and
fluorescence microscopy. To determine internalization levels of C. neoformans
during interaction with RAW 264.7 cells, we adapted the method of Chaka et al.
(10). Briefly, yeast cells were incubated with fluorescein isothicyanate (FITC) at
0.5 mg/ml in PBS (25°C) for 10 min. After extensive washing with PBS, C.
1602 OLIVEIRA ET AL. INFECT.IMMUN.
by on March 18, 2010 iai.asm.orgDownloaded from
neoformans yeast cells were incubated with macrophage-like cells for3hata5:1
fungus/host cell ratio, followed by extensive washing with PBS for removal of
nonadherent fungi. The fluorescence intensity of the macrophage-like cells was
therefore a function of association with FITC-labeled C. neoformans. The in-
fected cells were then detached from tissue culture plates by pipetting or scrap-
ing, fixed with 4% paraformaldehyde, and analyzed in a FACSCalibur flow
cytometer (BD Biosciences). The data were analyzed using winMDI 2.9 software.
The data were statistically analyzed using the one-way ANOVA test followed by
the Bonferroni posttest. To visualize the overall aspect of the cells, RAW cells
infected with FITC-labeled yeast cells were washed with PBS, sealed under a
coverslip, and visualized by epifluorescence. Samples were observed with an
Axioplan 2 (Zeiss, Germany) fluorescence microscope. Images were acquired
using a Color View SX digital camera and processed with the analySIS software
system (Soft Image System).
Antifungal activity of RAW 264.7 stimulated with vesicles. Macrophage mono-
layers were stimulated for 16 h with vesicles at a final concentration correspond-
ing to 0.4 g/ml sterol. The supernatant was removed and the monolayer washed
with serum-free DMEM. Aliquots of fungal cells suspended in DMEM were
added to the macrophage monolayers at a 5:1 yeast/macrophage ratio. After
incubation for3hat37°C (7.5% CO
2
atmosphere), the samples were washed
three times with PBS to remove nonadherent fungi; fresh medium was added,
and incubation was continued for an additional 2 or 5 h (for a total of 5 or 8 h
of infection) under the conditions described above. After incubation, cells were
again washed and then lysed with sterile cold water. Macrophage lysates con-
taining fungal cells were immediately plated onto Sabouraud agar plates for CFU
determination. Controls consisted of supernatants of nonstimulated macro-
phages cultivated under the same conditions. All experiments were performed in
triplicate sets and statistically analyzed by one-way ANOVA followed by the
Bonferroni posttest.
RESULTS
Vesicles secreted by C. neoformans are internalized by mac-
rophages. Fungal vesicles are believed to be released to the
extracellular space during in vitro macrophage infection and
animal cryptococcosis (65). However, it is unknown whether
vesicular components are incorporated by host cells and
whether these cells respond to vesicles. In this context, we first
evaluated whether incubation of macrophages with vesicle
fractions isolated from cryptococcal supernatants would result
in their delivery into host cells. To follow internalization of
extracellular vesicles, we stained isolated vesicles with the red-
fluorescent lipophilic compound dialkylcarbocyanine iodide
(DiI). Coincubation of RAW 264.7 cells performed using Dil-
labeled vesicle preparations and confocal microscopy analysis
of RAW 264.7 revealed that they were internalized by the
macrophage-like cells (Fig. 1A). Vesicles that were smaller
than 200 nm were not visualized, a result which was possibly
related to the resolution limit of fluorescence microscopy. The
absence of colocalization with CtxB indicates that vesicles do
not fuse with plasma membranes. Three-dimensional recon-
struction of these cells is shown in movie S1 in the supplemen-
tal material.
We demonstrated previously that vesicles secreted by C.
neoformans also transport antigenic proteins (64). We there-
fore used serum samples from cryptococcosis patients to track
vesicle-associated antigenic components. In fact, components
showing serologic reactivity colocalized with vesicle compart-
ments labeled with DiI within macrophages (Fig. 1B). Serum
samples from healthy individuals did not react with vesicle-
treated RAW cells (data not shown).
Macrophage exposure to the vesicles for 16 h did not lead to
cell lysis (Fig. 1C), showing that, under the experimental con-
ditions used in this study, vesicular compounds do not cause
acute toxicity to these cells.
Secreted vesicles modulate NO and cytokine production by
macrophages. NO has a well-established role in the protective
responses that occur during C. neoformans infection (66). In
this context, we evaluated whether vesicles produced by encap-
sulated and acapsular C. neoformans strains would modulate
the production of NO by RAW macrophages. Vesicle fractions
from both wild-type parental and mutant strains induced
dose-dependent production of NO (Fig. 2A). However, larger
amounts of NO were produced upon stimulation with vesicles
from the acapsular mutant. In support of these data, NO pro-
duction was diminished when we treated macrophages with
vesicles produced by the acapsular mutant in the presence of
serotype A and D GXMs in comparison to macrophages sub-
jected to treatment with native vesicles from acapsular cells
(Fig. 2B). Together with the fact that vesicles from the two
encapsulated strains induced similar levels of NO (Fig. 2B),
these data suggest that different serotypes of GXM reduce
vesicle-induced NO production. The polysaccharide concentra-
tion used in this experiment was determined based on the GXM
concentration usually found in vesicles (30 g/ml; data not
shown). LPS was used as a positive control (data not shown).
Under our experimental conditions, NO production was de-
pendent on the activity of induced nitric oxide synthase
(iNOS), since NO production was drastically reduced by the
presence of L-NIL (Fig. 2B). High levels of NO were detected
after exposure to vesicles in the presence of polymyxin B. In
FIG. 1. Incorporation of vesicular components by macrophages.
DiI-stained vesicles (red fluorescence) were incubated with RAW
264.7 macrophages, followed by staining of the nucleus with DAPI
(blue fluorescence). (A) A plasma membrane was stained with FITC-
labeled cholera toxin subunit B (green fluorescence). (B) Macrophages
were incubated with serum samples from patients with cryptococcosis
followed by Alexa Fluor 488 anti-human IgG antibody. The merged
image at the right (third panel) demonstrates that DiI-stained com-
partments colocalize, at least partially, with antigenic vesicular com-
ponents (arrowheads). Scale bars, 10 m. (C) Macrophage viability is
apparently not affected by the vesicles, as demonstrated by the mea-
surement of LDH activity in supernatants of macrophages treated with
vesicles (at a 2 g/ml sterol concentration) or incubated in regular
medium. As a positive control for enzyme activity, the macrophages
were incubated with Triton X-100. (A
i
Ϫ A
f
)
340
nm indicates the
difference between the initial and final absorbance readings.
V
OL. 78, 2010 C. NEOFORMANS EXTRACELLULAR VESICLES 1603
by on March 18, 2010 iai.asm.orgDownloaded from
contrast, the effect of LPS was completely inhibited by the
presence of polymyxin B under these conditions (data not
shown).
The response of macrophage-like cells to stimulation with
vesicles was also determined by evaluation of their ability to
produce cytokines. The cytokines analyzed were TNF-, IL-10,
and TGF-. TNF- was analyzed because of its capacity to
induce the fungicidal activity of mouse peritoneal macrophages
against C. neoformans (31), while IL-10 and TGF- are posi-
tively modulated by the vesicular component GXM (42, 74).
To evaluate the influence of known modulatory effects of
GXM on macrophages (74), we compared the macrophage
responses to vesicles isolated from an encapsulated C. neofor-
mans isolate (HEC3393) with the responses to those isolated
from an acapsular mutant (CAP67). Vesicle fractions from
both strains induced a significant increase in TNF- produc-
tion compared to those from nonstimulated cells (Fig. 3). It is
noteworthy that CAP67 vesicles induced an even higher pro-
duction of TNF- in comparison to HEC3393 vesicles (Fig. 3),
possibly because these fractions lack GXM, which causes po-
tent depressive effects on immune function (44, 45, 70, 73).
IL-10 and TGF- levels were increased following exposure to
either vesicle fraction (Fig. 3). Although levels of TGF- pro-
duction by macrophages were similar in the two systems,
HEC3393 vesicles induced significantly higher production of
IL-10 than CAP67 vesicles. These data indicate that macro-
phages differentially respond to stimulation by vesicles of C.
neoformans depending on vesicle composition.
Phagocytic and microbicidal activity of macrophages is
stimulated by secreted vesicles. We evaluated the ability of
vesicle-primed macrophages to phagocytose nonopsonized en-
capsulated C. neoformans cells. Control and vesicle-stimulated
macrophages were challenged with FITC-labeled C. neofor-
mans. Association indices were then determined by flow cy-
tometry. We observed that 62% of macrophages activated with
CAP67 vesicles displayed fluorescence (Fig. 4A; M1 marker)
FIG. 2. Vesicle-stimulated macrophages produce nitric oxide. (A) Dose-dependent production of NO was observed for vesicle fractions from
both acapsular and encapsulated fungi. The asterisks indicate P Ͻ 0.05. Vesicle concentrations are expressed as a function of the sterol content
in their membranes. (B) Macrophages were incubated overnight with vesicles from different strains at a final concentration of 0.4 g/ml of sterol.
GXM and L-NIL were added in final concentrations of 30 g/ml and 3 M, respectively. The levels of nitric oxide production in the culture
medium (no vesicles) were below the detection limit of the technique. Bars represent averages of three measurements; brackets denote standard
deviations. Data shown are representative of the results of three independent experiments.
FIG. 3. Profile of cytokine production by macrophages in response to stimulation with vesicle fractions. Macrophages were stimulated with
vesicles isolated from culture supernatants from encapsulated (HEC3393) and acapsular (CAP67) C. neoformans cells. The sterol concentration
corresponded to 0.4 g/ml in all fractions. The basal production of each cytokine in the medium alone is also shown (first bars). TNF-, TGF-,
and IL-10 concentrations in the supernatant were determined by capture ELISA. Statistical significance values are highlighted; P Ͻ 0.05. Bars
represent averages of three measurements; brackets denote standard deviations. Data shown are representative of the results of three independent
experiments.
1604 OLIVEIRA ET AL. I
NFECT.IMMUN.
by on March 18, 2010 iai.asm.orgDownloaded from
as a consequence of infection with FITC-labeled C. neofor-
mans. Interestingly, 41% of the macrophages were positive for
FITC staining after exposure to HEC3393 vesicles. In contrast,
only 35% of control cells contained yeast cells. Although the
percentage of fluorescence in M1 tended to be higher in mac-
rophages treated with HEC3393 vesicles than in control mac-
rophages, there was no statistical significance to the difference.
Epifluorescence analysis suggested that yeast internalization
was maximized under the conditions used in our experiments.
The findings were supported by differential interference con-
trast (DIC) and fluorescence microscopy results (Fig. 4B).
Confirming the results of fluorescence-activated cell sorter
(FACS) analysis, macrophages stimulated with CAP67 vesicles
had more internalized fungi per macrophage than under any
other set of conditions (data not shown).
Since phagocytosis and production of antimicrobial com-
pounds increased after treatment with fungal vesicles, we eval-
uated the microbicidal activity of macrophages after exposing
them to vesicles from encapsulated and acapsular C. neofor-
mans strains. Control or vesicle-treated RAW cells were incu-
bated with nonopsonized C. neoformans. After 3 h, culture
fluids containing unattached fungi were removed and replaced
FIG. 4. Macrophages stimulated with vesicles exhibit enhanced phagocytosis. (A) Macrophages were treated overnight with vesicles derived
from CAP67 or HEC3393 at a sterol concentration of 1 M. Gray peaks represent uninfected macrophages; open histograms show the population
analysis after interaction with FITC-C. neoformans within the M1 region, and the corresponding macrophage fluorescence increase is indicated
numerically. These data are representative of the results of three individual experiments. The percentages of positive fluorescence (M1) are
indicated. Asterisks denote P Ͼ 0.05. (B) Analysis by fluorescence microscopy of the material prepared for flow cytometry. Infected cells were
visualized using differential interference contrast microscopy and fluorescence microscopy. Scale bars, 10 M.
V
OL. 78, 2010 C. NEOFORMANS EXTRACELLULAR VESICLES 1605
by on March 18, 2010 iai.asm.orgDownloaded from
by fresh media. This step was followed by an additional incu-
bation of 2 or 5 h. After these periods, supernatants were
removed and cell lysates of control and stimulated macro-
phages were assayed for the presence of viable cryptococci.
Macrophages treated with CAP67 vesicles manifested greater
killing activity than HEC3393 vesicle-activated macrophages
(Fig. 5). In fact, stimulation of macrophages with vesicles from
encapsulated yeast cells resulted in significant microbicidal ac-
tivity only after8hofincubation. Although we cannot ensure
that the only difference between the HEC3393 and CAP67
vesicles was the presence of GXM, it is conceivable that the
less efficient fungicidal activity observed of macrophages
treated with HEC3393 was related to the fact that GXM neg-
atively modulates the macrophage response against C. neofor-
mans (45, 73). Albeit a tendency of growth inhibition was
visualized on control macrophages after8hofincubation, it is
important that no statistical significance difference was ob-
served compared to the results obtained after5hofincuba-
tion.
DISCUSSION
The host response to microbial antigens is a complex process
that is influenced by a number of factors, including the con-
centration of immunogens, the efficacy of exposure of these
molecules to effector cells, and the ability of the infectious
agent to produce and secrete immunologically active com-
pounds. Consequently, the outcome of the interaction of in-
fectious agents with host cells includes multiple possibilities,
such as the persistence of the microbe within host tissues,
causing cell damage, and the limitation of microbial coloniza-
tion, resulting in disease control (8).
It was suggested that fungal cells secrete vesicles during
interaction with phagocytes in vivo (65). Therefore, one might
expect that the concentration of such a complex array of mol-
ecules in vesicular structures could directly influence the inter-
action of fungal pathogens with their hosts. Supporting this
hypothesis, Gram-negative bacteria have been shown to con-
stitutively secrete outer membrane vesicles composed of lipo-
polysaccharide, phospholipids, outer membrane proteins, and
soluble periplasmic proteins, most of which are important vir-
ulence determinants (5, 36). This secretion mechanism is
thought to be employed by bacteria to deliver virulence factors
in the host cell (29). Recently, Lee and colleagues demon-
strated that Gram-positive bacteria also secrete vesicles, sug-
gesting that this process could be involved with protein transfer
to other bacteria and is conserved among different organisms
(38). Although the origin of fungal extracellular vesicles stud-
ied by our group is very different from that of their bacterial
counterparts, we investigated whether cryptococcal vesicles
also affect host-pathogen interactions.
In the present work we demonstrate that extracellular se-
creted vesicles from pathogenic fungi directly modulate host-
pathogen interactions and propose a model for these interac-
tions. According to this model, differences in vesicle numbers
and composition, as well as in the presence of GXM, could
affect the outcome of the C. neoformans-macrophage interac-
tion by affecting the state of macrophage activation. Strongly
suggestive evidence that vesicles are made in tissue comes from
electron microscopy and the observation of a serum antibody
response to vesicle components in infected mice (64, 65). Al-
though our work indicates that comparable effects could occur
in vivo, such studies remain to be done, and currently there are
formidable technical hurdles interfering with the demonstra-
tion of in vivo effects. Nevertheless, our results are likely to
have anticipated the observation of extracellular vesicles as
important modulators of infection outcome and participation
in the activation of immune response, as recently described for
mammalian exosomes (69).
C. neoformans produces several exocellular antigens with
apparently conflicting functions with regard to pathogenesis.
For instance, GXM, the major cryptococcal virulence factor,
protects the fungus against phagocytosis and inhibits leukocyte
migration (17, 21, 22, 34). However, this polysaccharide, in its
capsular form, is also an efficient inducer of the activation of
the alternative pathway of the complement system, which re-
sults in the deposition of opsonins in the capsule and increased
phagocytosis (33, 35). Another example of apparently oppos-
ing functions of capsular components of C. neoformans in-
volves the soluble forms of GXM and GalXM. Both of these
well-known capsular components activate inducible nitric ox-
ide synthase and the consequent production of nitric oxide in
macrophages (74). However, a heavily encapsulated cryptococ-
cal strain suppressed the production of this antimicrobial com-
pound in CpG-oligodeoxynucleotide-stimulated macrophages
(78), which confirms previous studies showing that C. neofor-
mans cells fail to induce nitric oxide synthase in primed murine
macrophage-like cells (52). These results reveal the enormous
complexity of the events involved in the interaction of C. neo-
formans with phagocytes and the difficulty in predicting which
pathogenesis-related structure will have the greatest effect dur-
ing infection of host cells. The cryptococcal vesicles contain at
least 81 different fungal molecules with potentially different
functional impacts on immunity (64, 65). Therefore, the re-
sponse of host cells to vesicle preparations is virtually unpre-
dictable, which led us to experimentally address some of the
functions of macrophages after their exposure to vesicle frac-
tions of C. neoformans.
FIG. 5. Fungicidal activity of vesicle-stimulated macrophages.
Macrophages treated with vesicles isolated from encapsulated or acap-
sular C. neoformans were incubated with the encapsulated C. neofor-
mans strain for different periods for CFU determination of numbers of
intracellular C. neoformans. CFU values were multiplied using the
dilution factor to generate absolute cell numbers. Bars represent
averages of three measurements; brackets represent standard devi-
ations. Statistically significant differences are highlighted.
1606 OLIVEIRA ET AL. I
NFECT.IMMUN.
by on March 18, 2010 iai.asm.orgDownloaded from
As recently reported for bacterial vesicles (5), our results
indicate that macrophages are able to internalize vesicular
components. Exposure of macrophages to vesicle preparations
resulted in enhanced levels of IL-10 and TGF-, anti-inflam-
matory cytokines whose production is positively modulated by
the vesicular component GXM (11, 42). In agreement with the
role of GXM in the upregulation of IL-10, CAP67 vesicles
induced a significantly lower amount of this cytokine, although
the level was significantly greater than in untreated macro-
phages. The increased production of IL-10 was recently asso-
ciated with a protective immune response to cryptococcosis,
which was stimulated by a peptide mimotope of GXM (13).
This higher efficiency was inferred from (i) the lower concen-
tration of CAP67 vesicles required for induction of high levels
of NO, (ii) the enhanced ability to induce yeast internalization,
and (iii) the requirement of a shorter incubation period to
display potent microbicidal activity.
Transmission electron microscopy demonstrated that C.
neoformans vesicle fractions contain subpopulations that differ
in morphological characteristics, suggesting the existence of
different biosynthetic steps for vesicle biogenesis (62). More
importantly, the detection of different morphological types
suggests that vesicles of diverse molecular compositions occur.
In a recent report, we used gradient fractionation of vesicles to
demonstrate that GXM concentrations differ considerably in
fractions with different densities (57). The methods currently
used for vesicle purification do not discriminate between ves-
icles of different characteristics, resulting in heterogeneous
preparations. Here we showed that differences in molecular
composition of vesicles between two strains were translated
into functional differences in the modulation of host cell
functions. Although this vesicle heterogeneity complicates the
functional analysis of specific vesicle populations, it approxi-
mates the reality that C. neoformans simultaneously secretes
different vesicle types (57). Assuming that C. neoformans se-
cretes vesicles during interaction with host cells (65), the over-
all effects of the exposure of phagocytes to extracellular frac-
tions could be better reflected in experiments using “crude”
vesicle samples, although macrophage function could be dif-
ferentially affected by distinct vesicle fractions.
Although our results provide strong evidence that crypto-
coccal vesicle preparations stimulate macrophage-like cells to
alter their cytokine production and enhance their antifungal
function, there are some experimental limitations that pre-
clude more generalized conclusions at this time. Currently we
do not have a reliable method to accurately enumerate vesicles
in our preparations and consequently cannot estimate the
number of vesicles required for the activation effect. It is con-
ceivable, and probably likely, that the effect of the presence of
vesicles on macrophages depends on the number of vesicles
that interact with each phagocytic cell such that, at low vesicle/
macrophage ratios, the effect results in activation whereas, at
higher ratios, the effect is detrimental to the macrophage. The
in vivo milieu may also impact the interactions; for instance,
secreted host lipases could disrupt the vesicles, leading to ear-
lier release of vesicular contents. Furthermore, the impact of
vesicles produced by intracellular fungi may be different from
that of extracellular vesicle preparations. Notwithstanding
these uncertainties, the results further support our conclusion
that fungal vesicles are biologically active; this information
opens new venues of investigation in fungal-host cell interac-
tions.
In this report, we demonstrate that the recently described
extracellular fungal vesicles can modify the functions of mam-
malian cells, altering the fate of C. neoformans after phagocy-
tosis by macrophages. These results strongly suggest the po-
tential of these vesicular structures to modify the course of
cryptococcal infections through their effects on host phagocytic
cells. Fungal vesicles are apparently related to mammalian
exosomes (58, 64). In this regard, it is noteworthy that exosome
preparations from dendritic cells are being investigated as po-
tential anticancer vaccines (26). The current results and the
fact that vesicle-related proteins are immunogenic (1, 62, 64)
suggest that vesicle preparations and/or their components
could have utility as fungal vaccines, which will be a focus of
future studies in our laboratory.
ACKNOWLEDGMENTS
L.N. and M.L.R. are supported by grants from the Brazilian agencies
CNPq, FAPERJ, and FAPESP. L.N. was supported in part by an
Interhemispheric Research Training Grant in Infectious Diseases,
Fogarty International Center (NIH D43-TW007129). J.D.N. was sup-
ported in part by NIH RO1 AI052733. A.C. was supported by NIH
awards AI033774, HL059842, and 2R37AI033142. C.G.F.-D.-L. is sup-
ported by grants from CNPq, FAPERJ, and the National Institute of
Science and Technology in Vaccines (INCTV).
D.L.O. is a Ph.D. student at Instituto de Bioquímica Me´dica,
UFRJ.
We thank the AECOM Analytical Imaging Facility for help with
confocal experiments. We also thank Jorge Jose´Jo´ B. Ferreira for
helpful discussions.
REFERENCES
1. Albuquerque, P. C., E. S. Nakayasu, M. L. Rodrigues, S. Frases, A. Casade-
vall, R. M. Zancope-Oliveira, I. C. Almeida, and J. D. Nosanchuk. 2008.
Vesicular transport in Histoplasma capsulatum: an effective mechanism for
trans-cell wall transfer of proteins and lipids in ascomycetes. Cell Microbiol.
10:1695–1710.
2. Alvarez, M., and A. Casadevall. 2007. Cell-to-cell spread and massive vacu-
ole formation after Cryptococcus neoformans infection of murine macro-
phages. BMC Immunol. 8:16.
3. Alvarez, M., and A. Casadevall. 2006. Phagosome extrusion and host-cell
survival after Cryptococcus neoformans phagocytosis by macrophages. Curr.
Biol. 16:2161–2165.
4. Beauvais, A., M. Monod, J. Wyniger, J. P. Debeaupuis, E. Grouzmann, N.
Brakch, J. Svab, A. G. Hovanessian, and J. P. Latge. 1997. Dipeptidyl-
peptidase IV secreted by Aspergillus fumigatus, a fungus pathogenic to hu-
mans. Infect. Immun. 65:3042–3047.
5. Bomberger, J. M., D. P. Maceachran, B. A. Coutermarsh, S. Ye, G. A.
O’Toole, and B. A. Stanton. 2009. Long-distance delivery of bacterial viru-
lence factors by Pseudomonas aeruginosa outer membrane vesicles. PLoS
Pathog. 5:e1000382.
6. Bulmer, G. S., and M. D. Sans. 1968. Cryptococcus neoformans. III. Inhibi-
tion of phagocytosis. J. Bacteriol. 95:5–8.
7. Casadevall, A., J. D. Nosanchuk, P. Williamson, and M. L. Rodrigues. 2009.
Vesicular transport across the fungal cell wall. Trends Microbiol. 17:158–
162.
8. Casadevall, A., and L. Pirofski. 2001. Host-pathogen interactions: the at-
tributes of virulence. J. Infect. Dis. 184:337–344.
9. Chaffin, W. L., J. L. Lopez-Ribot, M. Casanova, D. Gozalbo, and J. P.
Martinez. 1998. Cell wall and secreted proteins of Candida albicans: identi-
fication, function, and expression. Microbiol. Mol. Biol. Rev. 62:130–180.
10. Chaka, W., J. Scharringa, A. F. Verheul, J. Verhoef, A. G. Van Strijp, and
I. M. Hoepelman. 1995. Quantitative analysis of phagocytosis and killing of
Cryptococcus neoformans by human peripheral blood mononuclear cells by
flow cytometry. Clin. Diagn. Lab Immunol. 2:753–759.
11. Chiapello, L. S., J. L. Baronetti, M. P. Aoki, S. Gea, H. Rubinstein, and D. T.
Masih. 2004. Immunosuppression, interleukin-10 synthesis and apoptosis are
induced in rats inoculated with Cryptococcus neoformans glucuronoxyloman-
nan. Immunology 113:392–400.
12. Cox, G. M., J. Mukherjee, G. T. Cole, A. Casadevall, and J. R. Perfect. 2000.
Urease as a virulence factor in experimental cryptococcosis. Infect. Immun.
68:443–448.
VOL. 78, 2010 C. NEOFORMANS EXTRACELLULAR VESICLES 1607
by on March 18, 2010 iai.asm.orgDownloaded from
13. Datta, K., A. Lees, and L. A. Pirofski. 2008. Therapeutic efficacy of a con-
jugate vaccine containing a peptide mimotope of cryptococcal capsular poly-
saccharide glucuronoxylomannan. Clin. Vaccine Immunol. 15:1176–1187.
14. De Jesus, M., A. M. Nicola, M. L. Rodrigues, G. Janbon, and A. Casadevall.
2009. Capsular localization of the Cryptococcus neoformans polysaccharide
component galactoxylomannan. Eukaryot. Cell 8:96–103.
15. Del Poeta, M. 2004. Role of phagocytosis in the virulence of Cryptococcus
neoformans. Eukaryot. Cell 3:1067–1075.
16. Djordjevic, J. T., M. Del Poeta, T. C. Sorrell, K. M. Turner, and L. C.
Wright. 2005. Secretion of cryptococcal phospholipase B1 (PLB1) is regu-
lated by a glycosylphosphatidylinositol (GPI) anchor. Biochem. J. 389:803–
812.
17. Dong, Z. M., and J. W. Murphy. 1997. Cryptococcal polysaccharides bind to
CD18 on human neutrophils. Infect. Immun. 65:557–563.
18. Douwes, J., P. Versloot, A. Hollander, D. Heederik, and G. Doekes. 1995.
Influence of various dust sampling and extraction methods on the measure-
ment of airborne endotoxin. Appl. Environ. Microbiol. 61:1763–1769.
19. Eigenheer, R. A., Y. Jin Lee, E. Blumwald, B. S. Phinney, and A. Gelli. 2007.
Extracellular glycosylphosphatidylinositol-anchored mannoproteins and pro-
teases of Cryptococcus neoformans. FEMS Yeast Res. 7:499–510.
20. Eisenman, H. C., S. Frases, A. M. Nicola, M. L. Rodrigues, and A. Casade-
vall. 2009. Vesicle-associated melanization in Cryptococcus neoformans. Mi-
crobiology 155:3860–3867.
21. Ellerbroek, P. M., D. J. Lefeber, R. van Veghel, J. Scharringa, E. Brouwer,
G. J. Gerwig, G. Janbon, A. I. Hoepelman, and F. E. Coenjaerts. 2004.
O-Acetylation of cryptococcal capsular glucuronoxylomannan is essential for
interference with neutrophil migration. J. Immunol. 173:7513–7520.
22. Ellerbroek, P. M., L. H. Ulfman, A. I. Hoepelman, and F. E. Coenjaerts.
2004. Cryptococcal glucuronoxylomannan interferes with neutrophil rolling
on the endothelium. Cell Microbiol. 6:581–592.
23. Feldmesser, M., S. Tucker, and A. Casadevall. 2001. Intracellular parasitism
of macrophages by Cryptococcus neoformans. Trends Microbiol. 9:273–278.
24. Ghosn, E. E., M. Russo, and S. R. Almeida. 2006. Nitric oxide-dependent
killing of Cryptococcus neoformans by B-1-derived mononuclear phagocyte.
J. Leukoc. Biol. 80:36–44.
25. Green, L. C., D. A. Wagner, J. Glogowski, P. L. Skipper, J. S. Wishnok, and
S. R. Tannenbaum. 1982. Analysis of nitrate, nitrite, and [15N]nitrate in
biological fluids. Anal. Biochem. 126:131–138.
26. Hao, S., T. Moyana, and J. Xiang. 2007. Cancer immunotherapy by exosome-
based vaccines. Cancer Biother. Radiopharm. 22:692–703.
27. Jacobson, E. S. 2000. Pathogenic roles for fungal melanins. Clin. Microbiol.
Rev. 13:708–717.
28. Jacobson, E. S., D. J. Ayers, A. C. Harrell, and C. C. Nicholas. 1982. Genetic
and phenotypic characterization of capsule mutants of Cryptococcus neofor-
mans. J. Bacteriol. 150:1292–1296.
29. Kadurugamuwa, J. L., and T. J. Beveridge. 1995. Virulence factors are
released from Pseudomonas aeruginosa in association with membrane vesi-
cles during normal growth and exposure to gentamicin: a novel mechanism
of enzyme secretion. J. Bacteriol. 177:3998–4008.
30. Kawakami, K., Y. Koguchi, M. H. Qureshi, S. Yara, Y. Kinjo, K. Uezu, and
A. Saito. 2000. NK cells eliminate Cryptococcus neoformans by potentiating
the fungicidal activity of macrophages rather than by directly killing them
upon stimulation with IL-12 and IL-18. Microbiol. Immunol. 44:1043–1050.
31. Kawakami, K., M. H. Qureshi, Y. Koguchi, T. Zhang, H. Okamura, M.
Kurimoto, and A. Saito. 1999. Role of TNF-alpha in the induction of fun-
gicidal activity of mouse peritoneal exudate cells against Cryptococcus neo-
formans by IL-12 and IL-18. Cell Immunol. 193:9–16.
32. Kozel, T. R., B. Highison, and C. J. Stratton. 1984. Localization on encap-
sulated Cryptococcus neoformans of serum components opsonic for phago-
cytosis by macrophages and neutrophils. Infect. Immun. 43:574–579.
33. Kozel, T. R., and G. S. Pfrommer. 1986. Activation of the complement
system by Cryptococcus neoformans leads to binding of iC3b to the yeast.
Infect. Immun. 52:1–5.
34. Kozel, T. R., G. S. Pfrommer, A. S. Guerlain, B. A. Highison, and G. J.
Highison. 1988. Role of the capsule in phagocytosis of Cryptococcus neofor-
mans. Rev. Infect. Dis. 10(Suppl. 2):S436–S439.
35. Kozel, T. R., M. A. Wilson, G. S. Pfrommer, and A. M. Schlageter. 1989.
Activation and binding of opsonic fragments of C3 on encapsulated Crypto-
coccus neoformans by using an alternative complement pathway reconsti-
tuted from six isolated proteins. Infect. Immun. 57:1922–1927.
36. Kuehn, M. J., and N. C. Kesty. 2005. Bacterial outer membrane vesicles and
the host-pathogen interaction. Genes Dev. 19:2645–2655.
37. Latge´, J. P., H. Kobayashi, J. P. Debeaupuis, M. Diaquin, J. Sarfati, J. M.
Wieruszeski, E. Parra, J. P. Bouchara, and B. Fournet. 1994. Chemical and
immunological characterization of the extracellular galactomannan of As-
pergillus fumigatus. Infect. Immun. 62:5424–5433.
38. Lee, E. Y., D. Y. Choi, D. K. Kim, J. W. Kim, J. O. Park, S. Kim, S. H. Kim,
D. M. Desiderio, Y. K. Kim, K. P. Kim, and Y. S. Gho. 2009. Gram-positive
bacteria produce membrane vesicles: proteomics-based characterization of
Staphylococcus aureus-derived membrane vesicles. Proteomics 9:5425–5436.
39. Liu, L., R. P. Tewari, and P. R. Williamson. 1999. Laccase protects Crypto-
coccus neoformans from antifungal activity of alveolar macrophages. Infect.
Immun. 67:6034–6039.
40. Ma, H., J. E. Croudace, D. A. Lammas, and R. C. May. 2007. Direct cell-
to-cell spread of a pathogenic yeast. BMC Immunol. 8:15.
41. Ma, H., J. E. Croudace, D. A. Lammas, and R. C. May. 2006. Expulsion of
live pathogenic yeast by macrophages. Curr. Biol. 16:2156–2160.
42. Mariano Andrade, R., G. Monteiro Almeida, G. Alexandre DosReis, and C.
Alves Melo Bento. 2003. Glucuronoxylomannan of Cryptococcus neoformans
exacerbates in vitro yeast cell growth by interleukin 10-dependent inhibition
of CD4
ϩ
T lymphocyte responses. Cell Immunol. 222:116–125.
43. Miller, M. F., and T. G. Mitchell. 1991. Killing of Cryptococcus neoformans
strains by human neutrophils and monocytes. Infect. Immun. 59:24–28.
44. Mirshafiey, A., D. T. Ngoc Dzung, J. W. Murphy, M. R. Khorramizadeh, F.
Saadat, F. Mehrabian, and P. Larsson. 2004. Tolerability and anti-inflam-
matory effects of glucuronoxylomannan in collagen-induced arthritis. Scand.
J. Immunol. 60:226–232.
45. Monari, C., F. Bistoni, A. Casadevall, E. Pericolini, D. Pietrella, T. R. Kozel,
and A. Vecchiarelli. 2005. Glucuronoxylomannan, a microbial compound,
regulates expression of costimulatory molecules and production of cytokines
in macrophages. J. Infect. Dis. 191:127–137.
46. Monari, C., A. Casadevall, C. Retini, F. Baldelli, F. Bistoni, and A. Vec-
chiarelli. 1999. Antibody to capsular polysaccharide enhances the function of
neutrophils from patients with AIDS against Cryptococcus neoformans.
AIDS 13:653–660.
47. Monari, C., T. R. Kozel, F. Bistoni, and A. Vecchiarelli. 2002. Modulation of
C5aR expression on human neutrophils by encapsulated and acapsular Cryp-
tococcus neoformans. Infect. Immun. 70:3363–3370.
48. Monari, C., E. Pericolini, G. Bistoni, A. Casadevall, T. R. Kozel, and A.
Vecchiarelli. 2005. Cryptococcus neoformans capsular glucuronoxylomannan
induces expression of Fas ligand in macrophages. J. Immunol. 174:3461–
3468.
49. Monod, M., K. Jaton-Ogay, and U. Reichard. 1999. Aspergillus fumigatus-
secreted proteases as antigenic molecules and virulence factors. Contrib.
Microbiol. 2:182–192.
50. Mukherjee, S., M. Feldmesser, and A. Casadevall. 1996. J774 murine mac-
rophage-like cell interactions with Cryptococcus neoformans in the presence
and absence of opsonins. J. Infect. Dis. 173:1222–1231.
51. Murphy, J. W., F. Schafer, A. Casadevall, and A. Adesina. 1998. Antigen-
induced protective and nonprotective cell-mediated immune components
against Cryptococcus neoformans. Infect. Immun. 66:2632–2639.
52. Naslund, P. K., W. C. Miller, and D. L. Granger. 1995. Cryptococcus neo-
formans fails to induce nitric oxide synthase in primed murine macrophage-
like cells. Infect. Immun. 63:1298–1304.
53. Nichols, K. L., S. K. Bauman, F. B. Schafer, and J. W. Murphy. 2002.
Differences in components at delayed-type hypersensitivity reaction sites in
mice immunized with either a protective or a nonprotective immunogen of
Cryptococcus neoformans. Infect. Immun. 70:591–600.
54. Nicola, A. M., A. Casadevall, and D. L. Goldman. 2008. Fungal killing by
mammalian phagocytic cells. Curr. Opin. Microbiol. 11:313–317.
55. Nicola, A. M., S. Frases, and A. Casadevall. 2009. Lipophilic dye staining of
Cryptococcus neoformans extracellular vesicles and capsule. Eukaryot. Cell
8:1373–1380.
56. Nosanchuk, J. D., L. Nimrichter, A. Casadevall, and M. L. Rodrigues. 2008.
A role for vesicular transport of macromolecules across cell walls in fungal
pathogenesis. Commun. Integr. Biol. 1:37–39.
57. Oliveira, D. L., L. Nimrichter, K. Miranda, S. Frases, K. F. Faull, A. Cas-
adevall, and M. L. Rodrigues. 2009. Cryptococcus neoformans cryoimmuno-
electronmicroscopy and vesicle fractionation reveals an intimate association
between membrane lipids and glucuronxylomannan. Fungal Genet. Biol.
46:956–963.
58. Panepinto, J., K. Komperda, S. Frases, Y. D. Park, J. T. Djordjevic, A.
Casadevall, and P. R. Williamson. 2009. Sec6-dependent sorting of fungal
extracellular exosomes and laccase of Cryptococcus neoformans. Mol. Micro-
biol. 71:1165–1176.
59. Pietrella, D., C. Monari, C. Retini, B. Palazzetti, T. R. Kozel, and A. Vec-
chiarelli. 1998. Cryptococcus neoformans and Candida albicans regulate CD4
expression on human monocytes. J. Infect. Dis. 178:1464–1471.
60. Puccia, R., S. Schenkman, P. A. Gorin, and L. R. Travassos. 1986. Exocel-
lular components of Paracoccidioides brasiliensis: identification of a specific
antigen. Infect. Immun. 53:199–206.
61. Reiss, E., M. Huppert, and R. Cherniak. 1985. Characterization of protein
and mannan polysaccharide antigens of yeasts, moulds, and actinomycetes.
Curr. Top. Med. Mycol. 1:172–207.
62. Rodrigues, M. L., M. Alvarez, F. L. Fonseca, and A. Casadevall. 2008.
Binding of the wheat germ lectin to Cryptococcus neoformans suggests an
association of chitinlike structures with yeast budding and capsular glucu-
ronoxylomannan. Eukaryot. Cell 7:602–609.
63. Rodrigues, M. L., C. S. Alviano, and L. R. Travassos. 1999. Pathogenicity of
Cryptococcus neoformans: virulence factors and immunological mechanisms.
Microbes Infect. 1:293–301.
64. Rodrigues, M. L., E. S. Nakayasu, D. L. Oliveira, L. Nimrichter, J. D.
Nosanchuk, I. C. Almeida, and A. Casadevall. 2008. Extracellular vesicles
1608 OLIVEIRA ET AL. INFECT.IMMUN.
by on March 18, 2010 iai.asm.orgDownloaded from
produced by Cryptococcus neoformans contain protein components associ-
ated with virulence. Eukaryot. Cell 7:58–67.
65. Rodrigues, M. L., L. Nimrichter, D. L. Oliveira, S. Frases, K. Miranda, O.
Zaragoza, M. Alvarez, A. Nakouzi, M. Feldmesser, and A. Casadevall. 2007.
Vesicular polysaccharide export in Cryptococcus neoformans is a eukaryotic
solution to the problem of fungal trans-cell wall transport. Eukaryot. Cell
6:48–59.
66. Rossi, G. R., L. A. Cervi, M. M. Garcia, L. S. Chiapello, D. A. Sastre, and
D. T. Masih. 1999. Involvement of nitric oxide in protecting mechanism
during experimental cryptococcosis. Clin. Immunol. 90:256–265.
67. Schaller, M., M. Bein, H. C. Korting, S. Baur, G. Hamm, M. Monod, S.
Beinhauer, and B. Hube. 2003. The secreted aspartyl proteinases Sap1 and
Sap2 cause tissue damage in an in vitro model of vaginal candidiasis based on
reconstituted human vaginal epithelium. Infect. Immun. 71:3227–3234.
68. Syme, R. M., J. C. Spurrell, E. K. Amankwah, F. H. Green, and C. H. Mody.
2002. Primary dendritic cells phagocytose Cryptococcus neoformans via man-
nose receptors and Fcgamma receptor II for presentation to T lymphocytes.
Infect. Immun. 70:5972–5981.
69. The´ry, C., M. Ostrowski, and E. Segura. 2009. Membrane vesicles as con-
veyors of immune responses. Nat. Rev. Immunol. 9:581–593.
70. Tissi, L., M. Puliti, F. Bistoni, P. Mosci, T. R. Kozel, and A. Vecchiarelli.
2004. Glucuronoxylomannan, the major capsular polysaccharide of Crypto-
coccus neoformans, inhibits the progression of group B streptococcal arthri-
tis. Infect. Immun. 72:6367–6372.
71. Tucker, S. C., and A. Casadevall. 2002. Replication of Cryptococcus neofor-
mans in macrophages is accompanied by phagosomal permeabilization and
accumulation of vesicles containing polysaccharide in the cytoplasm. Proc.
Natl. Acad. Sci. USA 99:3165–3170.
72. Vecchiarelli, A., D. Pietrella, P. Lupo, F. Bistoni, D. C. McFadden, and A.
Casadevall. 2003. The polysaccharide capsule of Cryptococcus neoformans
interferes with human dendritic cell maturation and activation. J. Leukoc.
Biol. 74:370–378.
73. Vecchiarelli, A., C. Retini, D. Pietrella, C. Monari, C. Tascini, T. Beccari,
and T. R. Kozel. 1995. Downregulation by cryptococcal polysaccharide of
tumor necrosis factor alpha and interleukin-1 beta secretion from human
monocytes. Infect. Immun. 63:2919–2923.
74. Villena, S. N., R. O. Pinheiro, C. S. Pinheiro, M. P. Nunes, C. M. Takiya,
G. A. DosReis, J. O. Previato, L. Mendonca-Previato, and C. G. Freire-de-
Lima. 2008. Capsular polysaccharides galactoxylomannan and glucuronoxy-
lomannan from Cryptococcus neoformans induce macrophage apoptosis me-
diated by Fas ligand. Cell Microbiol. 10:1274–1285.
75. Vyas, K. A., H. V. Patel, A. A. Vyas, and R. L. Schnaar. 2001. Segregation of
gangliosides GM1 and GD3 on cell membranes, isolated membrane rafts,
and defined supported lipid monolayers. Biol. Chem. 382:241–250.
76. Wang, Y., P. Aisen, and A. Casadevall. 1995. Cryptococcus neoformans mel-
anin and virulence: mechanism of action. Infect. Immun. 63:3131–3136.
77. Wozniak, K. L., and S. M. Levitz. 2008. Cryptococcus neoformans enters the
endolysosomal pathway of dendritic cells and is killed by lysosomal compo-
nents. Infect. Immun. 76:4764–4771.
78. Xiao, G., A. Miyazato, K. Inden, K. Nakamura, K. Shiratori, K. Nakagawa,
T. Miyazawa, K. Suzuki, M. Kaku, and K. Kawakami. 2008. Cryptococcus
neoformans inhibits nitric oxide synthesis caused by CpG-oligodeoxynucle-
otide-stimulated macrophages in a fashion independent of capsular polysac-
charides. Microbiol. Immunol. 52:171–179.
79. Yauch, L. E., J. S. Lam, and S. M. Levitz. 2006. Direct inhibition of T-cell
responses by the Cryptococcus capsular polysaccharide glucuronoxyloman-
nan. PLoS Pathog. 2:e120.
80. Zaragoza, O., C. J. Chrisman, M. V. Castelli, S. Frases, M. Cuenca-Estrella,
J. L. Rodriguez-Tudela, and A. Casadevall. 2008. Capsule enlargement in
Cryptococcus neoformans confers resistance to oxidative stress suggesting a
mechanism for intracellular survival. Cell Microbiol. 10:2043–2057.
81. Zaragoza, O., M. L. Rodrigues, M. De Jesus, S. Frases, E. Dadachova, and
A. Casadevall. 2009. The capsule of the fungal pathogen Cryptococcus neo-
formans. Adv. Appl. Microbiol. 68:133–216.
82. Zaragoza, O., C. P. Taborda, and A. Casadevall. 2003. The efficacy of
complement-mediated phagocytosis of Cryptococcus neoformans is depen-
dent on the location of C3 in the polysaccharide capsule and involves both
direct and indirect C3-mediated interactions. Eur. J. Immunol. 33:1957–
1967.
Editor: G. S. Deepe, Jr.
VOL. 78, 2010 C. NEOFORMANS EXTRACELLULAR VESICLES 1609
by on March 18, 2010 iai.asm.orgDownloaded from
89
Anexo 4:
Characterization of Yeast Extracellular Vesicles: Evidence
for the Participation of Different Pathways of Cellular
Traffic in Vesicle Biogenesis
De
´
bora L. Oliveira
1.
, Ernesto S. Nakayasu
2.¤
, Luna S. Joffe
1
, Allan J. Guimara
˜
es
3
, Tiago J. P. Sobreira
4
,
Joshua D. Nosanchuk
3,5
, Radames J. B. Cordero
5
, Susana Frases
6
, Arturo Casadevall
3,5
,IgorC.
Almeida
2"
, Leonardo Nimrichter
1"
, Marcio L. Rodrigues
1
*
1 Laborato
´
rio de Estudos Integrados em Bioq
´
mica Microbiana, Instituto de Microbiologia Professor Paulo de Go
´
es, Rio de Janeiro, Rio de Janeiro, Brazil, 2 Department of
Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, United States of America, 3 Department of Medicine, Albert
Einstein College of Medicine, Bronx, New York, United States of America, 4 Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sa
˜
o
Paulo, Sa
˜
o Paulo, Sa
˜
o Paulo, Brazil, 5 Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America,
6 Laborato
´
rio de Biotecnologia, Instituto Nacional de Metrologia, Normalizac¸a
˜
o e Qualidade Industrial, Rio de Janeiro, Rio de Janeiro, Brazil
Abstract
Background:
Extracellular vesicles in yeast cells are involved in the molecular traffic across the cell wall. In yeast pathogens,
these vesicles have been implicated in the transport of proteins, lipids, polysaccharide and pigments to the extracellular
space. Cellular pathways required for the biogenesis of yeast extracellular vesicles are largely unknown.
Methodology/Principal Findings:
We characterized extracellular vesicle production in wild type (WT) and mutant strains of
the model yeast Saccharomyces cerevisiae using transmission electron microscopy in combination with light scattering
analysis, lipid extraction and proteomics. WT cells and mutants with defective expression of Sec4p, a secretory vesicle-
associated Rab GTPase essential for Golgi-derived exocytosis, or Snf7p, which is involved in multivesicular body (MVB)
formation, were analyzed in parallel. Bilayered vesicles with diameters at the 100–300 nm range were found in extracellular
fractions from yeast cultures. Proteomic analysis of vesicular fractions from the cells aforementioned and additional mutants
with defects in conventional secretion pathways (sec1-1, fusion of Golgi-derived exocytic vesicles with the plasma
membrane; bos1-1, vesicle targeting to the Golgi complex) or MVB functionality (vps23, late endosomal trafficking) revealed
a complex and interrelated protein collection. Semi-quantitative analysis of protein abundance revealed that mutations in
both MVB- and Golgi-derived pathways affected the composition of yeast extracellular vesicles, but none abrogated vesicle
production. Lipid analysis revealed that mutants with defects in Golgi-related components of the secretory pathway had
slower vesicle release kinetics, as inferred from intracellular accumulation of sterols and reduced detection of these lipids in
vesicle fractions in comparison with WT cells.
Conclusions/Significance:
Our results suggest that both conventional and unconventional pathways of secretion are
required for biogenesis of extracellular vesicles, which demonstrate the complexity of this process in the biology of yeast
cells.
Citation: Oliveira DL, Nakayasu ES, Joffe LS, Guimara
˜
es AJ, Sobreira TJP, et al. (2010) Characterization of Yeast Extracellular Vesicles: Evidence for the Participation
of Different Pathways of Cellular Traffic in Vesicle Biogenesis. PLoS ONE 5(6): e11113. doi:10.1371/journal.pone.0011113
Editor: Michael Otto, National Institutes of Health, United States of America
Received March 24, 2010; Accepted May 21, 2010; Published June 14, 2010
Copyright: ß 2010 Oliveira et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: LN and MLR are supported by grants from the Brazilian agencies Conselho Nacional de Desenvolvimento Cientı
´
fico e Tecnolo
´
gico (CNPq), Fundac¸a
˜
ode
Amparo a
`
Pesquisa do Estado do Rio de Janeiro (FAPERJ) and Coordenac¸a
˜
o de Aperfeic¸oamento de Pessoal de
´
vel Superior (CAPES). AC is supported by NIH
awards HL059842, AI033774, AI052733, AI033142. AJG and LN were supported in part by an Interhemispheric Research Training Grant in Infectious Diseases,
Fogarty International Center (NIH D43-TW007129). JDN is supported in part by NIH AI52733-06A1 and AI056070-01A2, and a Hirschl/Weill-Caulier Career Scientist
Award. RJBC was supported by the Training Program in Cellular and Molecular 414 Biology and Genetics, T32 GM007491. ICA was partially supported by the NIH/
NCRR grant 5G12RR008124-16A1. ESN was partially supported by the George A. Krutilek memorial graduate scholarship from Graduate School, University of Texas
at El-Paso (UTEP). TJPS was supported by Fundac¸a
˜
o de Amparo a
`
Pesquisa do Estado de Sa
˜
o Paulo (FAPESP), Brazil. The authors thank the Biomolecule Analysis
Core Facility, Border Biomedical Research Center/Biology/University of Texas at El-Paso (NIH grants #5G12RR008124-16A1 and 5G12RR008124-16A1S1), for the
access to the LC-MS instrumentation. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: marcio@micro.ufrj.br
. These authors contributed equally to this work.
" These authors also contributed equally to this work.
¤ Current address: Pacific Northwest National Laboratory, Richland, Washington, United States of America
PLoS ONE | www.plosone.org 1 June 2010 | Volume 5 | Issue 6 | e11113
Introduction
Protein secretion is a complex process that involves many
organelles and accessory molecules. In eukaryotic cells, the most
well-studied pathway of protein secretion involves vesicular
migration from the endoplasmic reticulum to the trans face of
the Golgi and then loading into a complex network of vesicles, the
trans-Golgi reticulum [1]. The vesicular post-Golgi network is the
most prominent example of conventional mechanism of protein
secretion. These proteins are sorted in the trans-Golgi network into
transport vesicles that immediately move to and fuse with the
plasma membrane, releasing their contents by exocytosis [1].
There are multiple unconventional mechanisms of protein
secretion in eukaryotes [2]. One of these mechanisms requires
the formation of the exosomes, which are vesicles derived from
membrane invagination within endocytic compartments (endo-
somes). The formation of internal vesicles in the lumen of
endosomes generates the multivesicular bodies (MVBs), which can
fuse with the plasma membrane, resulting in the release of internal
vesicles to the extracellular milieu as exosomes [3]. Extracellular
vesicle formation could also require other cellular pathways, as
suggested for Dictyostelium discoideum. In this organism, it has been
hypothesized that the Golgi reassembly stacking protein (GRASP),
which is attached peripherally to the cytoplasmic surface of Golgi
membranes, is required for the vesicular release of acyl-CoA
binding protein [4].
Fungal cells secrete molecules of different chemical natures and
molecular masses. As typical eukaryotic organisms, fungal cells use
conventional pathways of secretion involving post-Golgi vesicles
that fuse with the plasma membrane to release their cargo [5,6]. In
fact, it is well known that yeast cells continuously secrete a number
of enzymes that remain localized in the periplasm [7] but, until
recently, trans-cell wall secretion in fungi is a relatively unknown
cellular event, which consequently, had not been extensively
studied. Over the past few years, the existence of trans-cell wall
transport of intact vesicles has been reported and partially
characterized in pathogenic and non-pathogenic species of fungi
[8,9,10,11,12,13,14,15]. These vesicles contain a number of
proteins, lipids, polysaccharides and pigments of a wide molecular
mass range [8,11,14,15]. Therefore, we have proposed that
extracellular vesicle secretion represents a eukaryotic solution to
the problem of trans-cell wall transport, especially for large
molecules [9,12].
Although fungal extracellular vesicles have been generally
termed ‘fungal exosomes’ [9,14,16], different studies suggest that
their release to the extracellular space requires elements of the
conventional post-Golgi secretory pathway [16,17]. Yoneda and
Doering demonstrated that a Cryptococcus neoformans strain defective
in the production of Sav1p, a homolog of the S. cerevisiae small
GTPase Sec4p, accumulates post-Golgi vesicles under restrictive
conditions [17], a morphological feature that was initially
described for S. cerevisiae [6,18,19]. More precisely, the sav1 mutant
of C. neoformans had defective protein secretion and accumulated
exocytic vesicles at the septum and the bud during cell division.
Remarkably, these vesicles also contained a polysaccharide
destined to the extracellular space, suggesting that post-Golgi
secretion is involved with the transfer of macromolecules through
the cell wall. These findings were further supported by an
independent study showing that exposure of yeast cells to brefeldin
A, which interferes with the retrograde protein transport from the
Golgi apparatus to the endoplasmic reticulum, results in the
inhibition of polysaccharide assembly at the outer layer of the C.
neoformans cell wall [20]. In agreement with these observations, a C.
neoformans RNAi mutant strain lacking expression of Sec6p, an
88 kDa subunit of the exocytic complex that mediates polarized
targeting of secretory vesicles to active sites of exocytosis, was
unable to produce extracellular vesicles [16].
Although the studies mentioned above suggested the involvement
of conventional secretory mechanisms in the vesicular export of
polysaccharides in C. neoformans, it remained unclear whether
extracellular polysaccharides were targeted to the cell surface
exclusively in post-Golgi vesicles or via recycling endosomes [17].
Hence, the possibility that that the post-Golgi polysaccharide-
containing vesicles are sorted to a compartment other than the
plasma membrane, such as the late endosomes and the MVBs cannot
be ruled out. In fact, endosomes and MVBs can be connected to the
trans-Golgi secretory pathway [3], thus both pathways could be
involved in vesicular polysaccharide export in fungi. The molecular
machinery implied in MVB formation and sorting is widely known in
S. cerevisiae [21], but studies have not evaluated extracellular vesicle
transport and the connection to exosomes.
In this study, we characterized extracellular vesicles produced by
the model yeast S. cerevisiae. Based on recent suggestions that fungal
extracellular vesicles resemble exosomes but originate from Golgi-
related pathways [9,14,16], we also characterized vesicle fractions
from culture supernatants of mutants with defects in MVB formation
and post-Golgi secretion. Our results indicate that mutations in both
pathways affect vesicle composition. Nevertheless, yeast cells
mutated in the SEC4 gene, previously shown to accumulate
cytoplasmic vesicles [19], were defective in the secretion of vesicles
to the extracellular space, suggesting a key role for the Golgi-derived
secretory pathway in the trans-cell wall traffic in yeast cells.
Materials and Methods
Strains
The S. cerevisiae strains used in this study included RSY255,
RSY113, SEY6210 and BY4741 wild type (WT) cells and several
yeast secretory mutants, as subsequently described in this section
and summarized in Table 1. Strains RSY782, SF2642-1D, and
RSY954 are respectively, temperature sensitive sec1-1, sec4-2 and
bos1-1 (also known as sec32-1) mutants. Strains EEY6-2 and EEY9
correspond to mutant cells lacking expression of Vps23p and
Snf7p (also known as Vps32p), respectively. Strain GRH1delta is
defective in the expression of GRASP. WT and mutant cells were
cultivated in Sabouraud dextrose broth for 24 h at 25uC with
shaking. Based on recent literature, strains SF2642-1D (sec4-2) and
EEY9 (snf7) were used as prototypes of yeast cells defective in post-
Golgi or endosome-dependent vesicular secretion, respectively
[17,22]. Therefore, except for proteomic analyses where all strains
listed above were used, experiments in this study were performed
using sec4 and snf7 mutants and related WT strains.
Vesicle isolation
Extracellular vesicles from strains SEY6210 and BY4741 and
corresponding vps and grasp mutants were isolated from culture
supernatants, using variations of methods previously described
[8,14,15]. For vesicle isolation from cultures of strains RSY255
and RSY113 and the corresponding sec mutants, supernatants
were removed from 24 h cultures, cells were washed three times,
and fresh medium was added for further incubation at 37uC for 1
to 18 h at which time supernatants were removed and processed
for vesicle isolation. Cell viability was similar in WT and mutant
cultures, as determined by colony forming unit counting (data not
shown). For all cultures, yeast cells and debris were removed by
sequential centrifugation at 4,000 and 15,000 g (15 min, 4uC)
[8,14,15]. Supernatants were collected and concentrated by
approximately 20-fold using an Amicon ultrafiltration system
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 2 June 2010 | Volume 5 | Issue 6 | e11113
(cutoff = 100 kDa). The concentrate was again centrifuged at
4,000 and 15,000 g (15 min, 4uC) and passed through filtering
membranes (0.8
mm pores). Filtered fractions were finally
centrifuged at 100,000 g for 1 h at 4uC. Pellets were then washed
by three sequential suspension and centrifugation steps, each
consisting of 100,000 g for 1 h at 4uC with 0.1 M Tris buffered
saline (TBS, pH 7.4). The resulting pellets were then suspended in
fixative solution for electron microscopy analysis or prepared for
lipid and protein determinations, as described below. To avoid the
possibility of artifactual isolation of intracellular organelles from
dead cells, similar protocols were used with suspensions of heat-
killed cells instead of living yeast as described previously [15].
Vesicle-like structures were not observed under these conditions.
Transmission electron microscopy (TEM)
Pellets obtained after centrifugation of cell-free supernatants at
100,000 g were fixed with 2% glutaraldehyde in 0.1 M cacodylate
at room temperature for 2 h, and then incubated overnight in 4%
formaldehyde, 1% glutaraldehyde, and 0.1% PBS. The samples
were incubated for 90 min in 2% osmium, serially dehydrated in
ethanol, and embedded in Spurrs epoxy resin. Thin sections were
obtained on a Reichart Ultracut UCT and stained with 0.5%
uranyl acetate and 0.5% lead citrate. Samples were observed in a
JEOL 1200EX transmission electron microscope operating at
80 kV.
Light scattering analysis of vesicles
Vesicles undergo Brownian motion that translates into light
scattering fluctuations in a liquid phase. This property can be
measured by dynamic light scattering (DLS) techniques providing
information on the size and heterogeneity of the sample [11].
Measurement of vesicle size by DLS was performed in a 90Plus/BI-
MAS Multi Angle Particle Sizing analyzer (Brookhaven Instruments)
as described recently by Einsenman and colleagues [11].
Sterol analysis
Sterols are structural components of fungal extracellular vesicles
and markers of vesicle membranes [15]. For this reason, these
molecules were used in our model as molecular markers of
vesicular secretion in indirect quantification of vesicle fractions.
Sterols were also analyzed in cellular fractions, for comparative
analyses in the different strains used in this study. Intact cells were
collected by centrifugation and washed with PBS. These pellets or
vesicle fractions were first suspended in methanol and then two
volumes of chloroform were added. The mixture was vigorously
vortexed and centrifuged to discard precipitates, dried by vacuum
centrifugation and partitioned according to [23]. The lower phase,
containing neutral lipids, was recovered for analysis by high
performance thin layer chromatography (HPTLC). The lipid
extract was loaded into HPTLC silica plates (Si 60F254s,
LiChrospher, Merck, Germany) and separated using a solvent
system containing hexane:ether:acetic acid (80:40:2, v/v/v). The
plate was sprayed with a solution of 50 mg ferric chloride (FeCl
3
)
in a mixture of 90 ml water, 5 ml acetic acid and 5 ml sulfuric
acid. After heating at 100uC for 3–5 min, the sterol spots were
identified by the appearance of a red-violet color. Stained HPTLC
plates were digitalized using Adobe Photoshop CS (version 8.0)
and densitometrically analyzed with the Scion Image software
(version Alpha 4.0.3.2). Sterol content was also evaluated using the
quantitative fluorimetric kit ‘‘Amplex Red Sterol Assay Kit’’
(Molecular Probes). Sensitivity of sterol detection in this test is
approximately 8 ng in intact membranes, with no requirement of
lipid extraction. Vesicle pellets were suspended in 500
ml PBS and
10% of the sample was evaluated in this assay according to
manufacturer’s instructions. In all analyses, sterol content in each
fraction was normalized to the number of viable cells in yeast
cultures.
Protein identification by liquid chromatography-tandem
mass spectrometry (LC-MS/MS)
Vesicle proteomics followed the methodology recently estab-
lished for the analysis of extracellular vesicle fractions from fungal
cells [8,14]. Briefly, purified vesicles were suspended in 40
ml
400 mM NH
4
HCO
3
, containing 8 M urea, and the disulfide
bonds were reduced by the addition of 10
ml 50 mM dithiotreitol,
followed by incubation for 15 min at 50uC. Cysteine residues were
alkylated by the addition of 100 mM iodoacetamide (10
ml),
followed by incubation for 15 min at room temperature under
protection from the light. The final concentration of urea was then
adjusted to 1 M and the mixture was supplemented with 4
mg
sequencing-grade trypsin (Promega) for overnight digestion at
37uC. Resulting tryptic peptides were desalted in C18 reverse-
phase in-house ziptip columns (POROS R2 50, Applied
Biosystems), as described by Jurado et al. [24]. Samples were
finally redissolved in 5% acetonitrile (ACN), containing 0.5%
formic acid (FA), and loaded onto a C18-trap column. The
separation was performed on a capillary reverse-phase column
connected to a nanoHPLC system (nanoLC 1D Plus, Eksigent).
Peptides were eluted in a linear gradient (5–40%) of solvent B
(solvent A: 5% ACN/0.1% FA; solvent B: 80% ACN/0.1% FA)
during 200 min and directly analyzed in an electrospray-linear ion
trap-mass spectrometer (LTQ XL/ETD, Thermo Fisher)
equipped at the front end with a Triversa NanoMate nanospray
source (Advion). The nanospray was set at 1.35 kV and 0.2 psi N
2
Table 1. Yeast strains.
WT strain Mutation (mutant strain) Cellular event affected by mutation Origin Reference
RSY255 sec1-1 (RSY782) Membrane fusion* Schekman laboratory [63]
RSY113 sec4-2 (SF2642-1D) Vesicle targeting to the cell surface* Schekman laboratory [32]
RSY255 bos1-1, also known as sec32.2
(RSY954)
Vesicle targeting to the Golgi complex* Schekman laboratory [47]
SEY6210 snf7, also known as vps32 (EEY9) Vesicle invagination within multivesicular bodies Emr laboratory [22,31]
SEY6210 vps23 (EEY6-2) Late endosomal trafficking Emr laboratory [46]
BY4741 grh1, also known as grasp
(GRH1delta)
Unconventional secretion of acyl coenzyme A–binding
protein
EUROSCARF/Malhotra laboratory [39]
(*) Phenotype observed under restrictive temperature.
doi:10.1371/journal.pone.0011113.t001
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 3 June 2010 | Volume 5 | Issue 6 | e11113
pressure using a chip A (Advion). MS spectra were collected in
centroid mode at the 400–1700 m/z range and the ten most
abundant ions were subjected twice to collision induced dissoci-
ation (CID) with 35% normalized collision energy, before being
dynamically excluded for 60 sec.
All MS/MS spectra from peptides with 800–3500 Da, more
than 10 counts, and at least 15 fragments were converted into
DTA files using Bioworks v.3.3.1 (Thermo Fisher). DTA files were
submitted to database search using TurboSequest [25] and the S.
cerevisiae protein database (downloaded on June 8
th
, 2008 from
www.yeastgenome.org) combined with human keratin and porcine
trypsin sequences (downloaded on June 8
th
, 2008 from www.ncbi.
nlm.nih.gov/protein). All the sequences were used in the correct
and reverse orientations, forming a database with 13,760 entries.
The database search parameters included: i) trypsin cleavage in
both peptide termini with one missed cleavage site allowed; ii)
carbamidomethylation of cysteine residues as a fixed modification;
iii) oxidation of methionine residues as a variable modification;
and iv) 2.0 Da and 1.0 Da for peptide and fragment mass
tolerance, respectively. TurboSequest outputs were filtered with
DCn $0.05, peptide probability #0.05, and Xcorr $1.5, 2.0, and
2.5 for singly-, doubly-, and triply-charged peptides, respectively.
After filtering, the files were exported into XML formats and the
peptides sequences were assembled into proteins using an in-house
written script (Nakayasu, Sobreira, and Almeida, unpublished
data). The protein hits were re-filtered with sum of peptide Xcorr
$3.5. The false-discovery rate (FDR) was estimated as described
previously [8,14]. Proteins with shared peptides were assembled
into groups to assess the redundancy issue. For semi-quantitative
calculations, another in-house script was elaborated to combine
respective peptides and spectral counts into their respective protein
groups (Nakayasu, Sobreira, and Almeida, unpublished data).
Semi-quantitative determinations of protein abundance in each
sample were based on the calculation of the
exponentially modified
protein abundance index (emPAI), according to the methodology
proposed by Ishihama et al. [26]. emPAI Data were further
validated by the methodology described by Liu et al. [27], using
the number of spectra acquired for each protein (spectral count).
The relevant differences in protein abundance were selected when
a two-fold incremental change variation (WT vs. mutant proteins)
after spectral count and emPAI calculation was observed. A total
of 400 proteins were identified; however, those (n = 273) with less
than 10 spectra after MS/MS analysis of peptides were excluded
from the spectral count analysis. Using this approach we could
semi-quantify 127 proteins. This group of proteins was used in the
analyses detailed in Tables S1 and S2 and related data. The whole
set of results is available in Tables S3, S4 and S5.
Bioinformatics
Venn diagrams were prepared using the Venny tool, available
at http://bioinfogp.cnb.csic.es/tools/venny/index.html. Func-
tional networks in vesicle proteins were prepared using the Osprey
Network Visualization software (version 1.2.0), with the S. cerevisiae
database (available from the software). Analysis of the putative
presence of glycosylphosphatidylinositol (GPI)-anchored sequences
in vesicle proteins was performed as described recently [28], using
the GPI-anchored protein prediction program FragAnchor
(http://navet.ics.hawaii.edu/,fraganchor/NNHMM/NNHMM.
html) [29]. Correlation graphs and R
2
calculations were performed
with GraphPad Prism version 5.00 for Windows, GraphPad
Software, San Diego California USA, www.graphpad.com. The
presence of signal peptide cleavage sites in amino acid sequences
from vesicle proteins was predicted with the SignalP 3.0 Server
(http://www.cbs.dtu.dk/services/SignalP/), as previously de-
scribed [30].
Results
Morphological features and diameter distribution of S.
cerevisiae vesicles
Vesicle morphology was analyzed in both WT and mutant S.
cerevisiae cells. The selection of mutant strains for this analysis was
based on the hypothesis that fungal extracellular vesicles derive
from MVB-related pathways for exosome formation or from post-
Golgi conventional secretion [9,14,16,17]. In this context, our
analyses included the snf7 mutant, which shows defective MVB
formation [31], and the sec4-2 mutant, due to the implication of
the SEC4 gene in the secretion of Golgi-derived vesicles in yeast
cells [17,32].
Morphological analysis of yeast extracellular vesicles by TEM
revealed bilayered structures with varying levels of electron density
(Figure 1A). The vesicles were generally round or ovoid and
sometimes resembled multivesicular structures. No morphologic
alterations were apparent in vesicles obtained from sec4-2 and snf7
mutants. Analysis of the effective diameter of vesicle fractions
obtained from WT cells revealed average values ranging from
133.964.2 (strain SEY6210, parental of snf7) to 183.9624.0
(strain RSY113, parental of sec4-2) nm (Figure 1B). In interpreting
these numbers, and the results below, it is noteworthy that
dynamic light scattering tends to overestimate vesicle diameter
relative to other techniques (for discussion on this effect see [11]).
However, for the purposes of this study we have used dimensions
obtained for dynamic light scattering for all comparisons thus
controlling for any systematic trends in vesicle size inherent to the
technique. Vesicles from strain SEY6210 were clearly distributed
in two different populations, ranging from 50 to 75 and 180 to
250 nm in effective diameter. Mutation in the SNF7 gene had little
or no influence in vesicle diameter, which shifted from
133.964.2 nm in the vesicle fraction from WT cells to
143.562.1 nm for vesicles from the mutant cells. The two-
population profile of distribution of vesicle diameter in fractions
from the snf7 mutant was very similar to that found in vesicles from
the parental strain SEY6210. On the other hand, mutation in the
SEC4 gene significantly affected the diameter of extracellular
vesicles. While most of the vesicles from WT cells (strain RSY113)
were in the range of 100 to 200 nm, sec4-2 vesicles were
distributed in two populations. The mutant vesicles were
distributed in either a population ranging from 80 to 120 nm or
in a population with very high diameters (400–550 nm). Average
values shifted from 183.9624.0 nm (RSY113 strain) to
294.56117.9 nm (sec4-2 mutant). Hence, a SEC4 mutation
resulted in a qualitative difference in the vesicles produced by S.
cerevisiae.
Proteomic analysis reveals a complex composition in S.
cerevisiae extracellular vesicles
Previous studies with C. neoformans and H. capsulatum revealed
that fungal extracellular vesicles carry proteins with highly diverse
functions [8,14]. Many of these proteins were previously reported
to also be components of mammalian exosomes [33,34,35]. In our
model, proteomic analysis was performed with vesicles obtained
from three different WT S. cerevisiae cells, including strains
RSY113, RSY225, and SEY6210. Four hundred proteins were
identified in our analysis, with a FDR of ,1.6% in protein level.
Within this group, those with more than 10 spectra after MS/MS
analysis of peptides were used for the analyses shown in Figures 2,
3, 4, and 5 and Tables S1 and S2.
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 4 June 2010 | Volume 5 | Issue 6 | e11113
A summary of the proteins found in these three strains is
summarized in Table S1 and their detailed characterization is
shown in Tables S3 and S4.
The S. cerevisiae vesicle proteome had many similarities to the
protein profiles observed for C. neoformans and H. capsulatum [8,14].
Proteins related to diverse metabolic processes consisted of the
most abundant functional class in the S. cerevisiae vesicles, as
previously described for similar models (Figure 2 and Table S1)
[8,14]. Other classes found in the vesicles included cell
organization and biogenesis, transporters, carbohydrate metabo-
lism, stress response, protein biosynthesis, protein degradation,
protein transport and sporulation. A few molecules with unknown
function were also identified (Figure 2A). Of note, twenty four
different proteins required for cell wall modeling were identified,
including seven different glucanases and three glucanosyl trans-
ferases. Eight different heat shock proteins and other stress-related
proteins were also found in the vesicles, as well as peptidases,
vacuolar and secretory proteins. Although the protein composition
of the S. cerevisiae extracellular vesicles was multifaceted in many
aspects, most of the proteins identified in these preparations (90%)
are putatively functionally connected with one or more molecules
within the vesicular proteome (Figure 2B). The proportion of
proteins showing a high probability of bearing GPI-anchored was
approximately 4% of the total vesicular proteome, a value 4-fold
higher than those predicted for the cellular S. cerevisiae proteome
[29]. In addition, prediction of the presence of signal peptide
cleavage sites revealed that vesicle fractions also concentrated
proteins targeted to the endoplasmic reticulum and the secretory
pathway (Figure 3).
Profiles of protein composition of the S. cerevisiae vesicles were
very similar in different WT cells, as inferred from the high
percentage of vesicular proteins shared by distinct strains
(Figure 4). The variation range (5–25%) in protein identification
observed between different runs of the same sample is within the
acceptable range previously reported [36,37]. Analysis of vesicles
obtained from strains RSY113 and RSY225, which were both
cultivated at 37uC, revealed that more than 90% of the proteins
identified were common to both preparations. These values were
also high even when these strains were compared with SEY6210
cells, which were cultivated at room temperature. Proteomic
Figure 1. Morphology and diameter of
S. cerevisiae
extracellular vesicles. A. TEM of vesicles isolated from WT (WT) and mutant cells. Each
individual panel exemplifies the typical vesicle morphology for the cell group specified in the top. WT fractions shown in left panels were obtained
from strain RSY113, parental of the sec4-2 mutant. Similar morphological features (not shown) were observed in vesicle fractions obtained from strain
SEY6210 (parental of the snf7 mutant). Scale bar, 100 nm. B. Light scattering analysis showing diameter distribution and average values of vesicles
obtained from WT (WT) or mutant (sec4-2 and snf7) cells.
doi:10.1371/journal.pone.0011113.g001
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 5 June 2010 | Volume 5 | Issue 6 | e11113
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 6 June 2010 | Volume 5 | Issue 6 | e11113
analysis was also performed in vesicle fractions from S. cerevisiae
secretion mutants (Table S3). Remarkably, most of the proteins
found in WT cells were detected in the mutant fractions. The
qualitative analysis shown in Figure 4 revealed that most of the
proteins found in WT cells were also present in both sec4-2 and
snf7 mutants, suggesting that the protein compositions were not
severely affected by mutations in the secretion genes studied here.
Aiming at a more reliable data interpretation, we included in this
analysis vesicle proteomics of additional secretion-related systems,
which comprised mutations in SEC1, BOS1 (post-Golgi secretion)
and VPS23 (MVB formation) genes. The results obtained were
very similar to those detailed in Figure 4 (data not shown).
Protein abundance in S. cerevisiae vesicles is affected by
mutations in SEC and MVB-related genes
Based on the fact that vesicle protein composition was barely
affected by mutations in SEC4, SEC1, BOS1, VPS23, and SNF7
genes, we evaluated whether the relative abundance of vesicle
proteins would be modified in cells bearing defects in expression of
the related proteins. These analyses were based on semi-
quantitative determinations of the abundance of each vesicle
protein [26,27], the same approach used before to describe the
proteomic composition of different compartments of the secretory
pathway [38]. These methods are based on the fact that the most
abundant proteins have a higher coverage by LC-MS/MS. For
these analyses we only considered the 127 proteins with at least 10
spectra on one of the samples (Table S5). Within this group,
seventy three proteins (57.5%) had changes in their abundance in
at least one of the yeast mutants relative to the wild type strain. In
the SEC mutant group, the abundance of 24 vesicular proteins
increased, whereas 11 decreased. Analysis of the VPS group of
mutants revealed that vesicular protein abundance increased in 14
proteins and decreased in 33. Notably, the collective analysis of
graphs correlating emPAI values in vesicle proteins from WT cells
with those from mutant strains (Figure 5) demonstrated that the
most expressive changes in protein abundance were observed in
the snf7 mutant, followed by sec4-2, sec1-1, bos1-1 and vps23 cells.
Individual analysis of protein abundance revealed that the greatest
changes (5-fold increase or decrease in comparison to protein
abundance in fractions from WT cells) were observed in the VPS
gene family (Table S5). This analysis revealed an increased
abundance of plasma membrane H+-ATPases and mannosyl-
transferases in the vps mutants, with a parallel decrease of
glucanases and cyclophilin. These results, which were essentially
confirmed by spectral count (data not shown), suggest that vesicle
formation is probably affected by multiple elements of the
secretory apparatus.
Mutations in genes involved in either conventional or
unconventional secretion affect S. cerevisiae extracellular
vesicles
The differences in protein composition led us to consider the
possibility that vesicle secretion was somehow altered in the S.
cerevisiae mutants. Due to the well known characteristic of
intracellular vesicle accumulation in the sec4-2 mutant of S.
cerevisiae and in the sav1 mutant of C. neoformans [17,19,32], we first
quantified vesicle release in these cells and in the snf7 mutant by
measuring the content of sterols in cellular and vesicle fractions by
different methods. Fluorimetric analysis (Figure 6A) revealed that
the sterol content in vesicle fractions from strains SEY6210 and
snf7 were very similar, whereas a large decrease in sterol detection
was observed in the mutant sec4-2, in comparison to its parental
strain (RSY113). These results were essentially confirmed by
chromatographic analysis (Figure 6B–C), which showed that, in
comparison to WT cells, the sec4-2 mutant showed intracellular
accumulation of sterols and reduced detection of extracellular
vesicular sterols. Similar results were observed for the sec1.1
mutant (data not shown). The distribution of vesicular and cellular
sterols was apparently not significantly affected by mutation in the
SNF7 gene.
The decreased vesicle production in the sec4-2 mutant led us to
analyze the kinetics of vesicle release by these cells, in comparison
to the WT strain. While the peak of vesicle secretion in the WT
strain occurred after 18 h, detection of sterols in vesicle fractions
from the mutant strain stagnated after 3 h (Figure 6D). This result
confirmed that release of vesicles to the extracellular space was
affected by mutation in the SEC4 gene.
Due to the clear involvement of elements of the post-Golgi
secretory pathway in the kinetics of vesicle release in yeast and the
potential involvement of MVB-related pathways with vesicle
composition, we asked whether other secretion pathways could
be related to vesicular secretion in S. cerevisiae. Extracellular vesicle
fractions from WT cells and a mutant lacking expression of
GRASP, which participates in secretory mechanisms connecting
Figure 2. Functional distribution of proteins in extracellular vesicle fractions obtained from
S. cerevisiae
WT cells. A. Proteins were
grouped by color as indicated according to their function in the cellular metabolism. B. Functional interrelationship in the collection of vesicular
proteins. For protein identification according with each individual code, see Table S1.
doi:10.1371/journal.pone.0011113.g002
Figure 3. Prediction of the presence of GPI-anchored sequenc-
es and signal peptide cleavage sites in
S. cerevisiae
cellular or
vesicle proteins. All values used in these analysis were obtained in
this study, except those related to GPI-anchored sequences in cellular
fractions (y) [29].
doi:10.1371/journal.pone.0011113.g003
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 7 June 2010 | Volume 5 | Issue 6 | e11113
early endosomal compartments and the MVB sorting pathway
[39], were analyzed. As observed for the sec4.2 mutant, yeast cells
lacking GRASP expression had significantly reduced sterol content
in vesicle fractions and increased intracellular sterol accumulation
(Figure 7). This result, which suggests the participation of an
unconventional secretion pathway in vesicle production, is in
agreement with the fact that approximately 75% of vesicular
proteins have no signal peptide sequences for ER translation
(Figure 3).
Discussion
The production of extracellular vesicles by yeast cells has now
been reported for at least 6 different fungal species
[8,10,11,13,14,15,16]. Fungal extracellular vesicles are believed
to function as carriers of distinct molecules to the extracellular
space which, in the case of pathogens, includes a wide range of
virulence factors, including polysaccharides, pigments, and lipids
[8,10,11,13,14]. As described for mammalian exosomes [40], the
vesicular transport in fungi may even include the release of nucleic
acids to the extracellular milieu [41]. The secretion of such a
complex array of different molecules is expected to impact the
interaction of fungal cells with their hosts. In fact, extracellular
vesicles isolated from the yeast pathogen C. neoformans were
recently reported to modulate macrophage functions [42].
Vesicular traffic in eukaryotes is a complex and multifunctional
cellular mechanism. Intracellular vesicles are required for the
traffic of proteins destined for secretion, in pathways that require
their movement from the endoplasmic reticulum to the Golgi
complex and then, via the trans-Golgi network, to the cell surface
[5,19,43]. These vesicles are expected to fuse with the plasma
membrane releasing their cargo. Vesicular secretion can also
involve exosomes, which originate from intracellular MVBs. These
organelles derive from endosomes, which form internal vesicles
that are released to the extracellular milieu as exosomes upon
plasma fusion with the plasma membrane [3]. Yeast extracellular
vesicles have been generally termed ‘fungal exosomes’ [9,14,16],
but different studies suggest that they are linked to elements of the
conventional post-Golgi secretory pathway [16,17,20]. It remains
unknown, nevertheless, whether the formation of MVBs is related
to the biogenesis of fungal extracellular vesicles.
C. neoformans represents the yeast cell model that extracellular
vesicles have been investigated in most detail. This species appears
to use trans-cell wall vesicular transport to release its major
capsular polysaccharide [15,16,17,20], a high molecular weight
polysaccharide known as glucuronoxylomannan (GXM). Although
it has been suggested that C. neoformans produces MVB- and
exosome-like structures [14,44,45], the vesicular traffic of GXM
apparently requires homologues of SEC4 and SEC6 genes [16,17],
suggesting that the export of polysaccharide-containing vesicles in
these cells requires events of the conventional post-Golgi secretory
apparatus. The events required for the biogenesis of extracellular
vesicles in these cells are unclear, but the complex and variable
morphology of extracellular cryptococcal vesicles analyzed by
TEM [14] strongly suggests that the fractions usually analyzed in
studies on fungal extracellular vesicles include mixed populations
of diverse cellular origin.
Although C. neoformans was the species that led to discovery of
extracellular vesicles, this fungus may not be the ideal system at
this time for genetic dissection of vesicular physiology because the
copious extracellular polysaccharide hinders several analytic
approaches such as mass spectrometry and genetic tools remain
more difficult to use relative to other model fungi. Consequently,
we turned our attention to S. cerevisiae, where we similarly detected
extracellular vesicles in culture supernatants by TEM [8].
Consequently we took advantage of the availability of S. cerevisiae
secretion mutants and characterized their extracellular vesicles,
aiming to identify key elements required for the generation of these
compartments in yeast cells. Two major prototypes were used in
our study, based on previous literature observations. Snf7,a
mutant strain with defective MVB formation [31], was selected as
a candidate to evaluate whether exosome formation was related to
extracellular vesicles in yeast cells. The sec4-2 mutant was selected
as the prototype mutant to evaluate whether events of the post-
Golgi conventional secretion were required for the release of
fungal extracellular vesicles, given a recent report that the
orthologue of SEC4 in C. neoformans is required for the export of
polysaccharide-containing vesicles [17]. Using the same rationale,
some of the experiments performed in this study included mutant
cells with related defects in post-Golgi secretion mechanisms (sec1-
1 and bos1-1) and MVB biogenesis (vps23) [19,46,47].
As determined in this work and in a previous study [11], the
diameter of fungal extracellular vesicles ranged from 50 to
500 nm. These dimensions contrast with the fact that extracellular
vesicles in other models are in a diameter range lower than
100 nm [3]. Different studies, however, have demonstrated that
larger membrane structures (300–500 nm in size) are the vehicles
Figure 4. Venn diagrams showing similarities of protein composition in vesicle fractions from WT (RSY113 and SEY6210 strains)
and mutant cells (
sec4-2
and
snf7
mutants).
doi:10.1371/journal.pone.0011113.g004
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 8 June 2010 | Volume 5 | Issue 6 | e11113
responsible for long distance, ER-to-Golgi and trans-Golgi to
plasma membrane transport of secretory molecules (reviewed in
[48]). Estimation of the diameter of cell wall pores in yeast cells
revealed values in the range of 200 to 400 nm [49], which would
theoretically permit the release of vesicles of different sizes to the
extracellular space. These observations and the high variability in
the morphology of fungal extracellular vesicles [14] support the
hypothesis that the vesicle populations originate from compart-
ments of distinct biogenesis, which could involve both Golgi- and
exosome-derived pathways.
Proteomic analysis of yeast extracellular vesicles revealed a
complex composition, as described for mammalian exosomes and
other fungal vesicles [14,33,34,35]. Several cytoplasmic proteins
with no apparent relation with secretory processes were observed
in the vesicle proteome, providing another parallel with mamma-
lian exosomes. Sorting of cytosolic proteins into exosomes is
normally explained by a random engulfment of small portions of
cytosol during the inward budding process of MVBs [3]. Of note,
many cell wall-degrading enzymes were found in the S. cerevisiae
vesicles, consistent with a prior notion that the passage of vesicles
through the cell wall could require hydrolysis of structural
components [8,9,12]. These enzymes were present in all fractions
analyzed in this study. Protein composition was consistently similar
in all vesicle fractions analyzed, providing confidence in the
Figure 5. Correlation analysis of the relative variation of protein abundance in vesicle fractions from WT (WT) or mutant
S. cerevisiae
cells. emPAI values for each parental strain were plotted against the values obtained for yeast mutant fractions analyzed by proteomics. SEC or MVB-
related mutants included snf7 (A), sec4-2 (B), vps23 (C), sec1-1 (D) and bos1-1 (E). Lower R
2
values suggest greater alterations in relative protein
distribution in vesicles from the mutants, in comparison to WT cells. Proteins whose abundance was increased in vesicle fractions from mutant cells
are represented by the red spots, whereas proteins that were more abundant in WT fractions are highlighted in blue.
doi:10.1371/journal.pone.0011113.g005
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 9 June 2010 | Volume 5 | Issue 6 | e11113
validity of the conclusion that vesicle proteins include many
different functional classes.
Most of the proteins found in the yeast vesicle proteome were
potentially associated other molecules identified in the vesicular
protein collection. More precisely, bioinformatics analyses sug-
gested that at least 219 protein-protein interactions were observed
within the vesicle proteome. Some of these interactions were in
fact expected. For example, YGR032W and YLR342W (glucan
synthases) were associated, and they are in the same functional
class. Apparently unrelated proteins, however, were also poten-
tially interacting. For instance, YER103W is a heat shock protein
that plays a role in protein-membrane targeting and translocation.
We found that the molecule interacted with other vesicular heat
shock proteins, but also with the metabolic enzyme glyceralde-
hyde-3-phosphate dehydrogenase [50,51,52]. Similarly,
YLR249W is a translational elongation factor that interacted with
four other elongation factors and two heat shock proteins, but also
with pyruvate decarboxylase, phosphoglucose isomerase and
alcohol dehydrogenase [53,54,55,56]. These observations illustrate
the complexity of the protein composition of fungal extracellular
vesicles, as well as the difficulties in unraveling their biosynthetic
steps.
Vesicular fractions had greater concentrations of secretory and
GPI-anchored proteins in comparison to intact S. cerevisiae cells.
Figure 6. Sterol analysis in vesicle and cellular fractions obtained from yeast cells. A–B. Indirect sterol-based vesicle quantification of
fractions obtained from cultures of WT (WT) or mutant cells. The sterol content was determined by fluorimetric methods (A) or by densitometric
analysis of bands obtained after HPTLC separation (B). Comparative analysis of the sterol content in vesicle fractions obtained from WT or mutant
cells suggested that the sec4-2 mutant has a defective release of extracellular vesicles. This supposition was supported by chromatographic analysis in
association with densitometry of cellular (C) or vesicle (D) fractions obtained from yeast cultures. The sec4-2 mutant, in contrast to the snf7 mutant,
showed intracellular accumulation of sterols (C) and a lower kinetics of release of sterol-containing vesicles (D). Arrows indicate the migration ofan
ergosterol standard in TLC plates. Results are representative of three independent analyses showing similar results.
doi:10.1371/journal.pone.0011113.g006
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 10 June 2010 | Volume 5 | Issue 6 | e11113
This observation suggests that the unconventional mechanism of
protein secretion by vesicle release includes elements of the
conventional secretory pathway. Furthermore, the fact that
sequences potentially containing GPI anchors and signals for ER
targeting are enriched in vesicle fractions confirms previous
observations that fungal extracellular vesicles are actively secreted
rather than released by dead cells, since they concentrate
secretion-related proteins.
Our semi-quantitative analysis of protein abundance in yeast
vesicles strongly suggests that fungal vesicles include compart-
ments related to the MVB-derived pathway of exosome
formation. Although vesicle release was similar in WT cells and
in the snf7 mutant, the abundance of 35 proteins was modified in
the mutant vesicles, suggesting that defects in MVB formation
also affect extracellular vesicles in fungi. In fact, the greatest
differences in protein abundance in WT/mutant systems
observed in this study involved vps mutants and, particularly,
snf7 cells. In vesicle fractions from these mutants, proteins with
the greatest levels of increase in relative abundance consisted of
two related plasma membrane proton ATPases (YGL008C and
YPL036W; Pma1p and Pma2p, respectively) and two Golgi
mannosyltransferases (YDR483W and YBR199W). The manno-
syltransferases YDR483W and YBR199W are known to interact
with other Golgi proteins, which include, respectively, members
of the exocyst complex and t-SNAREs required for vesicular
transport [57,58]. Similarly, Pma1p and its isoform Pma2p
interact with several elements of Golgi-associated pathways of
cellular traffic, including Ric1p, a protein involved in retrograde
transport to the cis-Golgi network, and Vps29p, which is essential
for endosome-to-Golgi retrograde transport [59,60]. Therefore,
we speculate that mutations in the VPS genes could led to the
activation of compensatory mechanisms of Golgi-associated
traffic, which could explain the increased abundance of Golgi-
related proteins in vesicles from snf7 and vps2 3 cells. On the other
hand, vesicles from the snf7 mutants had significantly decreased
levels of a protein of unknown function, two glucanases and
cyclophilin. The functional implication of each of these individual
changes in protein abundance is unknown, but the possibilities
are numerous. For instance, cyclophilin is supposed to interact
with 34 different proteins in S. cerevisiae, including elements of the
secretory apparatus [60] and cell wall architecture [61]. These
observations illustrate the fact that vesicular proteins with no
apparent connections with the secretory process may be directly
or indirectly linked to vesicle biogenesis.
Our results show that mutation of the SEC4 gene is associated
with a delay in vesicle release to the extracellular space. This result
is supportive and consistent with previous reports that a sec4-2
mutant of S. cerevisiae and a similar mutant in C. neoformans
accumulate intracellular vesicles [17,18]. This observation sup-
ports the view that the extracellular vesicles observed in fungal
cells may not be conventional exosomes, as suggested in
independent studies [14,16,17]. Nevertheless, it remains unknown
how post-Golgi vesicles, which are expected to fuse with the
plasma membrane to release their cargo, would leave the cell wall.
Different reports, however, suggest that not all secretory vesicles
fuse with the plasma membrane (reviewed in [48]), which supports
a prior study with a sec6 C. neoformans mutant [16] and our current
observations with S. cerevisiae strains showing that post-Golgi
secretion events are required for the release of extracellular
vesicles.
Remarkably, vesicle release was not completely abrogated in
any of the mutants analyzed in this study. This observation could
indicate that multiple cellular pathways are required for formation
of fungal extracellular vesicles, including elements of non-
conventional secretory mechanisms. In fact, in our study, mutant
cells lacking expression of GRASP, which is required for
unconventional secretion of an acyl coenzyme A–binding protein
in S. cerevisiae [39] and Dictyostelium discoideum [4], showed a
decreased content of extracellular vesicles, in comparison with WT
cells. This observation may be related to the fact that acyl
coenzyme A–binding protein plays an important role in the
cellular distribution of sphingolipids [62], which are important
structural components of fungal extracellular vesicles [15].
GRASP is involved in secretory mechanisms that require
autophagy genes, early endosomal compartments, and MVBs
[39]. The decreased vesicular release by the GRASP mutant
reinforces the importance of Golgi components in extracellular
vesicle formation and supports the notion that extracellular release
of vesicles in fungi is a multifactorial cellular event of high
complexity, and possibly involves considerable redundancy. We
cannot rule out the possibility, however, that the collection of
mutations analyzed in our study are affecting different types of
vesicles, since the methods currently used for vesicle purification
do not discriminate between vesicles of different origins, resulting
in heterogeneous preparations.
Our current results together with previous reports suggest
agreement with the fact that endosomes and MVBs can be
connected to the trans-Golgi secretory pathway [3], which could
directly affect the formation of fungal extracellular vesicles. In
summary, after analysis of eight different S. cerevisiae strains, our
results indicate that both MVB- and Golgi-derived cellular
pathways affect the formation and release of extracellular vesicles
Figure 7. Sterol analysis in vesicle (A) and cellular (B) fractions obtained from WT cells and a mutant strain lacking GRASP
expression. Comparative analysis of the sterol content in vesicle fractions obtained from WT or mutant cells suggested that GRASP is involved in the
release of extracellular vesicles. Chromatograms and related densitometric analyses are shown. Arrows indicate the chromatographic migration of
ergosterol standards. Results are representative of three independent analyses showing similar results.
doi:10.1371/journal.pone.0011113.g007
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 11 June 2010 | Volume 5 | Issue 6 | e11113
by fungal cells. We believe our observations with S. cerevisiae
extracellular vesicles will contribute to the understanding of a
complex event in the biology of yeast cells. Since yeast
extracellular vesicles in pathogens are presumably linked to fungal
virulence and the ability of fungal cells to modulate the host
immunity, these results could also be of use in the design of
pathogenic models aiming at the elucidation of the role of
secretion events in fungal virulence.
Supporting Information
Table S1 Proteomic analysis of S. cerevisiae extracellular
vesicles*.
Found at: doi:10.1371/journal.pone.0011113.s001 (0.16 MB
DOC)
Table S2 Changes in protein abundance in vesicle fractions
from secretion mutants of S. cerevisiae, in comparison to fractions
from WT cells.
Found at: doi:10.1371/journal.pone.0011113.s002 (0.09 MB
DOC)
Table S3 All identified proteins.
Found at: doi:10.1371/journal.pone.0011113.s003 (0.18 MB
XLS)
Table S4 Detailed information of peptide identification.
Found at: doi:10.1371/journal.pone.0011113.s004 (0.72 MB
XLS)
Table S5 emPAI calculation and spectral count for the 128
proteins used for the semi-quantitative analysis.
Found at: doi:10.1371/journal.pone.0011113.s005 (0.08 MB
XLS)
Acknowledgments
DLO is a PhD student at Instituto de Bioquı
´
mica Me´dica (Federal
University of Rio de Janeiro, Brazil). We thank the Biomolecule Analysis
Core Facility, Border Biomedical Research Center/Biology/University of
Texas at El-Paso for the access to the mass spectrometry instrumentation.
We are also grateful to Jorge J. Jo´ Bastos Ferreira for helpful comments and
Guylaine Poisson for help with use of Fraganchor. We are particularly
thankful to Drs. Randy Schekman, Scott Emr, Vivek Malhotra and Juan
Duran for donation of yeast mutants and helpful suggestions to this study.
We are particularly grateful to Dr. Vivek Malhotra for his initial suggestion
on the role of GRASP in the release of extracellular vesicles in yeast cells.
Author Contributions
Conceived and designed the experiments: DLO ESN JDN AC ICA LN
MLR. Performed the experiments: DLO ESN LSJ AJG RJBC SF LN.
Analyzed the data: DLO ESN TJPS RJBC ICA MLR. Contributed
reagents/materials/analysis tools: JDN AC ICA MLR. Wrote the paper:
DLO JDN AC ICA LN MLR.
References
1. Glick BS, Malhotra V (1998) The curious status of the Golgi apparatus. Cell 95:
883–889.
2. Nickel W, Rabouille C (2009) Mechanisms of regulated unconventional protein
secretion. Nat Rev Mol Cell Biol 10: 148–155.
3. Keller S, Sanderson MP, Stoeck A, Altevogt P (2006) Exosomes: from biogenesis
and secretion to biological function. Immunol Lett 107: 102–108.
4. Kinseth MA, Anjard C, Fuller D, Guizzunti G, Loomis WF, et al. (2007) The
Golgi-associated protein GRASP is required for unconventional protein
secretion during development. Cell 130: 524–534.
5. Schekman R (2002) Lasker Basic Medical Research Award. SEC mutants and
the secretory apparatus. Nat Med 8: 1055–1058.
6. Schekman R, Novick P (2004) 23 genes, 23 years later. Cell 116: S13–15,
11 p following S19.
7. Wickner W, Schekman R (2005) Protein translocation across biological
membranes. Science 310: 1452–1456.
8. Albuquerque PC, Nakayasu ES, Rodrigues ML, Frases S, Casadevall A, et al.
(2008) Vesicular transport in Histoplasma capsulatum: an effective mechanism
for trans-cell wall transfer of proteins and lipids in ascomycetes. Cell Microbiol
10: 1695–1710.
9. Casadevall A, Nosanchuk JD, Williamson P, Rodrigues ML (2009) Vesicular
transport across the fungal cell wall. Trends Microbiol 17: 158–162.
10. De Jesus M, Nicola AM, Rodrigues ML, Janbon G, Casadevall A (2009)
Capsular localization of the Cryptococcus neoformans polysaccharide compo-
nent galactoxylomannan. Eukaryot Cell 8: 96–103.
11. Eisenman HC, Frases S, Nicola AM, Rodrigues ML, Casadevall A (2009)
Vesicle-associated melanization in Cryptococcus neoformans. Microbiology 155:
3860–3867.
12. Nosanchuk JD, Nimrichter L, Casadevall A, Rodrigues ML (2008) A role for
vesicular transport of macromolecules across cell walls in fungal pathogenesis.
Commun Integr Biol 1: 37–39.
13. Oliveira DL, Nimrichter L, Miranda K, Frases S, Faull KF, et al. (2009)
Cryptococcus neoformans cryoultramicrotomy and vesicle fractionation reveals
an intimate association between membrane lipids and glucuronoxylomannan.
Fungal Genet Biol 46: 956–963.
14. Rodrigues ML, Nakayasu ES, Oliveira DL, Nimrichter L, Nosanchuk JD, et al.
(2008) Extracellular vesicles produced by Cryptococcus neoformans contain
protein components associated with virulence. Eukaryot Cell 7: 58–67.
15. Rodrigues ML, Nimrichter L, Oliveira DL, Frases S, Miranda K, et al. (2007)
Vesicular polysaccharide export in Cryptococcus neoformans is a eukaryotic
solution to the problem of fungal trans-cell wall transport. Eukaryot Cell 6:
48–59.
16. Panepinto J, Komperda K, Frases S, Park YD, Djordjevic JT, et al. (2009) Sec6-
dependent sorting of fungal extracellular exosomes and laccase of Cryptococcus
neoformans. Mol Microbiol 71: 1165–1176.
17. Yoneda A, Doering TL (2006) A eukaryotic capsular polysaccharide is
synthesized intracellularly and secreted via exocytosis. Mol Biol Cell 17:
5131–5140.
18. Walworth NC, Goud B, Kabcenell AK, Novick PJ (1989) Mutational analysis of
SEC4 suggests a cyclical mechanism for the regulation of vesicular traffic.
EMBO J 8: 1685–1693.
19. Schekman R, Esmon B, Ferro-Novick S, Field C, Novick P (1983) Yeast
secretory mutants: isolation and characterization. Methods Enzymol 96:
802–815.
20. Hu G, Steen BR, Lian T, Sham AP, Tam N, et al. (2007) Transcriptional
regulation by protein kinase A in Cryptococcus neoformans. PLoS Pathog 3:
e42.
21. Hurley JH, Emr SD (2006) The ESCRT complexes: structure and mechanism of
a membrane-trafficking network. Annu Rev Biophys Biomol Struct 35: 277–298.
22. Babst M, Katzmann DJ, Estepa-Sabal EJ, Meerloo T, Emr SD (2002) Escrt-III:
an endosome-associated heterooligomeric protein complex required for mvb
sorting. Dev Cell 3: 271–282.
23. Folch J, Lees M, Sloane Stanley GH (1957) A simple method for the isolation
and purification of total lipides from animal tissues. J Biol Chem 226: 497–509.
24. Jurado JD, Rael ED, Lieb CS, Nakayasu E, Hayes WK, et al. (2007)
Complement inactivating proteins and intraspecies venom variation in Crotalus
oreganus helleri. Toxicon 49: 339–350.
25. Eng JK, McCormack AL, Yates JR, 3rd (1994) An approach to correlate tandem
mass spectral data of peptides with amino acid sequences in a protein database.
J Am Soc Mass Spectrom 5: 976–989.
26. Ishihama Y, Oda Y, Tabata T, Sato T, Nagasu T, et al. (2005) Exponentially
modified protein abundance index (emPAI) for estimation of absolute protein
amount in proteomics by the number of sequenced peptides per protein. Mol
Cell Proteomics 4: 1265–1272.
27. Liu H, Sadygov RG, Yates JR, 3rd (2004) A model for random sampling and
estimation of relative protein abundance in shotgun proteomics. Anal Chem 76:
4193–4201.
28. Nakayasu ES, Yashunsky DV, Nohara LL, Torrecilhas AC, Nikolaev AV, et al.
(2009) GPIomics: global analysis of glycosylphosphatidylinositol-anchored
molecules of Trypanosoma cruzi. Mol Syst Biol 5: 261.
29. Poisson G, Chauve C, Chen X, Bergeron A (2007) FragAnchor: a large-scale
predictor of glycosylphosphatidylinositol anchors in eukaryote protein sequences
by qualitative scoring. Genomics Proteomics Bioinformatics 5: 121–130.
30. Bendtsen JD, Nielsen H, von Heijne G, Brunak S (2004) Improved prediction of
signal peptides: SignalP 3.0. J Mol Biol 340: 783–795.
31. Tu J, Vallier LG, Carlson M (1993) Molecular and genetic analysis of the SNF7
gene in Saccharomyces cerevisiae. Genetics 135: 17–23.
32. Salminen A, Novick PJ (1987) A ras-like protein is required for a post-Golgi
event in yeast secretion. Cell 49: 527–538.
33. Aoki N, Jin-no S, Nakagawa Y, Asai N, Arakawa E, et al. (2007) Identification
and characterization of microvesicles secreted by 3T3-L1 adipocytes: redox- and
hormone-dependent induction of milk fat globule-epidermal growth factor 8-
associated microvesicles. Endocrinology 148: 3850–3862.
34. Gatti JL, Metayer S, Belghazi M, Dacheux F, Dacheux JL (2005) Identification,
proteomic profiling, and origin of ram epididymal fluid exosome-like vesicles.
Biol Reprod 72: 1452–1465.
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 12 June 2010 | Volume 5 | Issue 6 | e11113
35. Mears R, Craven RA, Hanrahan S, Totty N, Upton C, et al. (2004) Proteomic
analysis of melanoma-derived exosomes by two-dimensional polyacrylamide gel
electrophoresis and mass spectrometry. Proteomics 4: 4019–4031.
36. Schirle M, Heurtier MA, Kuster B (2003) Profiling core proteomes of human cell
lines by one-dimensional PAGE and liquid chromatography-tandem mass
spectrometry. Mol Cell Proteomics 2: 1297–1305.
37. Faca V, Pitteri SJ, Newcomb L, Glukhova V, Phanstiel D, et al. (2007)
Contribution of protein fractionation to depth of analysis of the serum and
plasma proteomes. J Proteome Res 6: 3558–3565.
38. Gilchrist A, Au CE, Hiding J, Bell AW, Fernandez-Rodriguez J, et al. (2006)
Quantitative proteomics analysis of the secretory pathway. Cell 127: 1265–1281.
39. Duran JM, Anjard C, Stefan C, Loomis WF, Malhotra V () Unconventional
secretion of Acb1 is mediated by autophagosomes. J Cell Biol 188: 527–536.
40. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, et al. (2007) Exosome-
mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic
exchange between cells. Nat Cell Biol 9: 654–659.
41. Nicola AM, Frases S, Casadevall A (2009) Lipophilic dye staining of
Cryptococcus neoformans extracellular vesicles and capsule. Eukaryot Cell 8:
1373–1380.
42. Oliveira DL, Freire-de-Lima CG, Nosanchuk JD, Casadevall A, Rodrigues ML,
et al. Extracellular vesicles from Cryptococcus neoformans modulate macro-
phage functions. Infect Immun.
43. Novick P, Field C, Schekman R (1980) Identification of 23 complementation
groups required for post-translational events in the yeast secretory pathway. Cell
21: 205–215.
44. Takeo K, Uesaka I, Uehira K, Nishiura M (1973) Fine structure of Cryptococcus
neoformans grown in vivo as observed by freeze-etching. J Bacteriol 113:
1449–1454.
45. Takeo K, Uesaka I, Uehira K, Nishiura M (1973) Fine structure of Cryptococcus
neoformans grown in vitro as observed by freeze-etching. J Bacteriol 113:
1442–1448.
46. Babst M, Odorizzi G, Estepa EJ, Emr SD (2000) Mammalian tumor
susceptibility gene 101 (TSG101) and the yeast homologue, Vps23p, both
function in late endosomal trafficking. Traffic 1: 248–258.
47. Wuestehube LJ, Duden R, Eun A, Hamamoto S, Korn P, et al. (1996) New
mutants of Saccharomyces cerevisiae affected in the transport of proteins from
the endoplasmic reticulum to the Golgi complex. Genetics 142: 393–406.
48. Stephens DJ, Pepperkok R (2001) Illuminating the secretory pathway: when do
we need vesicles? J Cell Sci 114: 1053–1059.
49. de Souza Pereira R, Geibel J (1999) Direct observation of oxidative stress on the
cell wall of Saccharomyces cerevisiae strains with atomic force microscopy. Mol
Cell Biochem 201: 17–24.
50. Werner-Washburne M, Stone DE, Craig EA (1987) Complex interactions
among members of an essential subfamily of hsp70 genes in Saccharomyces
cerevisiae. Mol Cell Biol 7: 2568–2577.
51. Martineau CN, Beckerich JM, Kabani M (2007) Flo11p-independent control of
‘‘mat’’ formation by hsp70 molecular chaperones and nucleotide exchange
factors in yeast. Genetics 177: 1679–1689.
52. Gong Y, Kakihara Y, Krogan N, Greenblatt J, Emili A, et al. (2009) An atlas of
chaperone-protein interactions in Saccharomyces cerevisiae: implications to
protein folding pathways in the cell. Mol Syst Biol 5: 275.
53. Krogan NJ, Cagney G, Yu H, Zhong G, Guo X, et al. (2006) Global landscape
of protein complexes in the yeast Saccharomyces cerevisiae. Nature 440:
637–643.
54. Collins SR, Kemmeren P, Zhao XC, Greenblatt JF, Spencer F, et al. (2007)
Toward a comprehensive atlas of the physical interactome of Saccharomyces
cerevisiae. Mol Cell Proteomics 6: 439–450.
55. Chakraburtty K, Triana-Alonso FJ (1998) Yeast elongation factor 3: structure
and function. Biol Chem 379: 831–840.
56. Kovalchuke O, Kambampati R, Pladies E, Chakraburtty K (1998) Competition
and cooperation amongst yeast elongation factors. Eur J Biochem 258: 986–993.
57. Inadome H, Noda Y, Adachi H, Yoda K (2005) Immunoisolaton of the yeast
Golgi subcompartments and characterization of a novel membrane protein,
Svp26, discovered in the Sed5-containing compartments. Mol Cell Biol 25:
7696–7710.
58. De Craene JO, Coleman J, Estrada de Martin P, Pypaert M, Anderson S, et al.
(2006) Rtn1p is involved in structuring the cortical endoplasmic reticulum. Mol
Biol Cell 17: 3009–3020.
59. Luo W, Chang A (1997) Novel genes involved in endosomal traffic in yeast
revealed by suppression of a targeting-defective plasma membrane ATPase
mutant. J Cell Biol 138: 731–746.
60. Schuldiner M, Collins SR, Thompson NJ, Denic V, Bhamidipati A, et al. (2005)
Exploration of the function and organization of the yeast early secretory
pathway through an epistatic miniarray profile. Cell 123: 507–519.
61. Costanzo M, Baryshnikova A, Bellay J, Kim Y, Spear ED, et al. The genetic
landscape of a cell. Science 327: 425–431.
62. Gaigg B, Neergaard TB, Schneiter R, Hansen JK, Faergeman NJ, et al. (2001)
Depletion of acyl-coenzyme A-binding protein affects sphingolipid synthesis and
causes vesicle accumulation and membrane defects in Saccharomyces cerevisiae.
Mol Biol Cell 12: 1147–1160.
63. Novick P, Schekman R (1979) Secretion and cell-surface growth are blocked in a
temperature-sensitive mutant of Saccharomyces cerevisiae. Proc Natl Acad
Sci U S A 76: 1858–1862.
Yeast Extracellular Vesicles
PLoS ONE | www.plosone.org 13 June 2010 | Volume 5 | Issue 6 | e11113
www.landesbioscience.com Communicative & Integrative Biology 533
Communicative & Integrative Biology 3:6, 533-535; November/December 2010; ©2010 Landes Bioscience
AUTOPHAGIC PUNCTUM
ARTICLE ADDENDUM
Addendum to: Oliveira DL, Nakayasu ES, Joe LS,
Guimaraes AJ, Sobreira TJP, Nosanchuk JD, et al.
Characterization of yeast extracellular vesicles:
evidence for the participation of dierent path-
ways of cellular trac in vesicle biogenesis. PLoS
ONE 2010; 5:11113; PMID: 20559436; DOI: 0.1371/
jo ur nal .p on e.0 011113 .
Key words: secretion, extracellular
vesicles, exosome, trans-cell wall
transport, yeast box
Submitted: 06/21/10
Accepted: 06/21/10
Previously published online:
www.landesbioscience.com/journals/cib/
article/12756
DOI: 10.4161/cib.3.6.12756
*Correspondence to: Marcio L. Rodrigues;
T
he cellular events required for
unconventional protein secretion in
eukaryotic pathogens are beginning to be
revealed. In fungi, extracellular release
of proteins involves passage through
the cell wall by mechanisms that are
poorly understood. In recent years, sev-
eral studies demonstrated that yeast cells
produce vesicles that traverse the cell
wall to release a wide range of cellular
components into the extracellular space.
These studies suggested that extracellu-
lar vesicle release involves components of
both conventional and unconventional
secretory pathways, although the precise
mechanisms required for this process are
still unknown. We discuss here cellular
events that are candidates for regulating
this interesting but elusive event in the
biology of yeast cells.
Protein secretion is a widely studied cel-
lular phenomenon. To reach the extracel-
lular milieu, intracellularly synthesized
proteins are targeted to the cell surface for
release to the extracellular space.
1
In mam-
malian cells, the plasma membrane is the
final barrier to be crossed during secre-
tion. Such processes, which involve both
conventional and unconventional mecha-
nisms, have been studied in detail and a
number of excellent reviews are available
in the literature.
1-6
Secretory systems in microbes
and mammalian cells show points of
Biogenesis of extracellular vesicles in yeast
Many questions with few answers
Débora L. Oliveira,
1
Ernesto S. Nakayasu,
2,†
Luna S. Joffe,
1
Allan J. Guimarães,
3
Tiago J.P. Sobreira,
4
Joshua D. Nosanchuk,
3,5
Radames J.B. Cordero,
5
Susana Frases,
6
Arturo Casadevall,
3,5
Igor C. Almeida,
2
Leonardo Nimrichter
1
and Marcio L. Rodrigues
1,
*
1
Laboratório de Estudos Integrados em Bioquímica Microbiana; Instituto de Microbiologia Professor Paulo de Góes; Rio de Janeiro; Brazil;
2
The Border
Biomedical Research Center; Department of Biological Sciences; University of Texas at El Paso; El Paso, TX USA; Departments of
3
Medicine and
5
Microbiology
and Immunology; Albert Einstein College of Medicine; Bronx, New York USA;
4
Laboratory of Genetics and Molecular Cardiology; Heart Institute (InCor);
University of Sao Paulo; São Paulo, SP Brazil;
6
Laboratório de Biotecnologia; Instituto Nacional de Metrologia; Normalização e Qualidade Industrial; Rio de
Janeiro, Brazil
Current address: Pacific Northwest National Laboratory; Richland, WA USA
convergence and divergence.
7
Fungi and
prokaryotes are surrounded by thick cell
walls, a key difference in comparison with
mammalian and other eukaryotic cells
(e.g., protozoa) that adds significant com-
plexity to secretion systems in these organ-
isms. A number of mechanisms have been
proposed for the trans-cell wall molecular
transport in prokaryotes.
8
In fungi, how-
ever, the mechanisms required for passage
of molecules across the cell wall are poorly
understood. Recently, extracellular vesicle
release has been described as a mechanism
used by yeast cells to secrete many mol-
ecules across the cell wall.
9-12
Extracellular vesicles produced by
fungal cells share morphological and bio-
chemical similarities with mammalian
exosomes,
13,14
including an ability to mod-
ulate the function of immune cells.
15
Plant
cells also produce exosome-like vesicles,
16
supporting the notion that vesicular
release is a mechanism of trans-cell wall
passage shared by cell-wall containing
eukaryotes. In contrast to what is observed
for mammalian exosomes,
17
the pathways
required for extracellular vesicle biogenesis
and release in both plant and fungal cells
remain virtually unknown. One remark-
able feature of mammalian exosomes and
fungal extracellular vesicles is the abun-
dance of cytoplasmic proteins lacking a
signal peptide that directs proteins to the
endoplasmic reticulum in conventional
secretory processes.
13,14,18-20
534 Communicative & Integrative Biology Volume 3 Issue 6
were observed in extracellular vesicle sam-
ples obtained from S. cerevisiae cultures.
21
We hypothesize that these enzymes could
hydrolyze cell wall components to facilitate
vesicle passage through this cellular barrier.
The methods currently used for vesicle
purification do not discriminate between
vesicles of different origins. This implies
that heterogeneous preparations are
obtained during vesicle isolation. In this
context, the possibility that the collection
of mutations analyzed in our recent study
21
is affecting different types of vesicles can-
not be ruled out. The current knowledge on
how fungal extracellular vesicles are formed,
in fact, strongly suggests the involvement of
multipleand perhaps still unknown—
pathways of secretion.
9,11,13
As recently
described in independent studies, uncon-
ventional protein secretion can also involve
autophagosomes,
27,2 8
which are intracellular
structures whose functions were initially
attributed to many catabolic steps.
29
In autophagy, cytosolic material is
sequestered by an expanding membrane
compartment, the phagophore, resulting in
the formation of a double-membrane vesi-
cle, the autophagosome.
29
Autophagosomes
then fuse with the lysosome/vacuole where,
as initially supposed, the sequestered mate-
rial is degraded.
29
Independent studies,
however, have shown that yeast cells can
also direct the autophagic content for secre-
tion, in a process called exophagy.
27-29
In
fact, the autophagic machinery participates
in the packaging and delivery of the soluble
yeast protein acyl-Coenzyme A-binding
protein Acb1 to the cell surface. Therefore,
these studies suggest the existence of a
vesicular mechanism that utilizes the same
machinery for both secretion and degrada-
tion of cellular components. It is interesting
to note that secretion of Acb1 from yeast as
well as secretion of the Dictyostelium discoi-
deum Acb1 homologue, AcbA, depends on
the Golgi associated protein GRASP,
27,28,30
which is apparently required for extracel-
lular vesicle release in yeast cells.
21
These
observations add to an already long list of
candidates that can regulate vesicle forma-
tion in yeast cells.
After our initial description of fungal
extracellular vesicles in 2007,
12
eight differ-
ent studies showing their functions in fun-
gal physiology or pathogenesis have been
reported in the literature.
13-15,21,22,31-33
Vesicle
their cargo, so they are not expected
to interfere with formation of vesicular
structures outside the cell.
6
In our analy-
ses, however, yeast mutants lacking Sec4p,
a secretory vesicle-associated Rab GTPase
essential for Golgi-derived exocytosis,
6
had reduced kinetics of vesicle release to
the extracellular milieu.
21
The fact that
cells with defects in a post-Golgi event of
secretion, but not with disturbed MVB
formation, affected vesicle release raised
an obvious and still unanswered question:
how is a double layered vesicle secreted
from yeast cells?
The simplest and more tangible expla-
nation for the release of any extracellu-
lar vesicle is the fusion of MVB with the
plasma membrane. However, studies by
our group
25,26
clearly show that double-
layered vesicles can bud from the plasma
membrane of yeast cells (Fig. 1). Therefore,
one could speculate that proteins required
for post-Golgi conventional secretion could
be required for addressing vesicle compo-
nents to the plasma membrane. Vesicles
would then be formed by membrane bud-
ding and sequential transfer to the cell wall
and extracellular space. That would be con-
sistent with previous hypotheses raising the
possibility that formation of extracellular
vesicles can involve membrane budding.
4
Budding from the plasma membrane would
also be in line with the complex vesicle com-
position including cytoplasmic elements, as
observed in our analyses.
13,14,21
It remains
unknown how these vesicles traverse the cell
wall, but many cell wall degrading enzymes
In a recent study, we evaluated the
contribution of both conventional and
unconventional pathways of secretion in
the formation of extracellular vesicles in
the model yeast Saccharomyces cerevisiae.
21
Our approach was based on the study of
mutants with defects in two major secre-
tion pathways: conventional post-Golgi
secretion
2
and exosome formation, a mech-
anism of unconventional secretion.
17
The
use of this model was based on the facts
that: (1) genes required for conventional,
post-Golgi secretion were implicated in
the formation of extracellular vesicles in
fungi;
22
and (2) exosomes and fungal vesi-
cles share many similarities.
18-20,23,24
Defects in the formation of multive-
sicular bodies (MVB) are expected to
directly affect the formation of exosomes.
17
Surprisingly, yeast mutants with defects
in MVB formation produced similar
amounts of extracellular vesicles in
comparison to WT cells.
21
The protein
composition of vesicles from WT and
mutant cells was essentially equivalent,
but approximately 50% of these vesicu-
lar proteins had their abundance modi-
fied in mutant cells. Remarkably, most
of the proteins (75%) found in vesicular
fractions lacked signal peptides. These
puzzling results indicate that, although
MVB-related mutations apparently do
not affect vesicle release, MVB formation
is somehow related to the formation of
extracellular vesicles in yeast.
Post-Golgi secretory vesicles usually
fuse with the plasma membrane to release
Figure 1. Cryptococcus neoformans, a yeast pathogen, produce vesicle-like structures (arrows) that
apparently bud from the plasma membrane to be deposited at the cell wall, as evidenced from
transmission electron microscopy. Gold labeling represents reactivity of fungal glucosylceramide
with human antibodies. Scale bar, 0.1 µm. Asterisk denotes the cell wall. For experimental details,
see Rodrigues and colleagues.
26
Modied from Barreto-Bergter et al.
25
courtesy of Dr. Kildare
Miranda.
www.landesbioscience.com Communicative & Integrative Biology 535
17. van Niel G, Porto-Carreiro I, Simoes S, Raposo G.
Exosomes: a common pathway for a specialized func-
tion. J Biochem 2006; 140:13-21.
18. Aoki N, Jin-no S, Nakagawa Y, Asai N, Arakawa E,
Tamura N, et al. Identification and characterization
of microvesicles secreted by 3T3-L1 adipocytes:
redox- and hormone-dependent induction of milk
fat globule-epidermal growth factor 8-associated
microvesicles. Endocrinology 2007; 148:3850-62.
19. Gatti JL, Metayer S, Belghazi M, Dacheux F,
Dacheux JL. Identification, proteomic profiling and
origin of ram epididymal fluid exosome-like vesicles.
Biol Reprod 2005; 72:1452-65.
20. Mears R, Craven R A, Hanrahan S, Totty N, Upton
C, Young SL, et al. Proteomic analysis of melanoma-
derived exosomes by two-dimensional polyacryl-
amide gel electrophoresis and mass spectrometry.
Proteomics 2004; 4:4019-31.
21. Oliveira DL, Nakayasu ES, Joffe LS, Guimarães AJ,
Sobreira TJP, Nosanchuk JD, et al. Characterization
of yeast extracellular vesicles: evidence for the par-
ticipation of different pathways of cellular traffic in
vesicle biogenesis. PLoS ONE 2010; 5:11113.
22. Panepinto J, Komperda K, Frases S, Park YD,
Djordjevic JT, Casadevall A, et al. Sec6-dependent
sorting of fungal extracellular exosomes and lac-
case of Cryptococcus neoformans. Mol Microbiol 2009;
71:1165-76.
23. Valadi H, Ekstrom K, Bossios A, Sjostrand M,
Lee JJ, Lotvall JO. Exosome-mediated transfer of
mRNAs and microRNAs is a novel mechanism of
genetic exchange between cells. Nat Cell Biol 2007;
9:654-9.
24. Nicola AM, Frases S, Casadevall A. Lipophilic dye
staining of Cryptococcus neoformans extracellular vesi-
cles and capsule. Eukaryot Cell 2009; 8:1373-80.
25. Barreto-Bergter E, Pinto MR, Rodrigijes ML, Atta-
ur R. Structural and functional aspects of fun-
gal glycosphingolipids. Studies in Natural Products
Chemistry: Elsevier 2006; 1025-55.
26. Rodrigues ML, Travassos LR, Miranda KR, Franzen
AJ, Rozental S, de Souza W, et al. Human antibodies
against a purified glucosylceramide from Cryptococcus
neoformans inhibit cell budding and fungal growth.
Infect Immun 2000; 68:7049-60.
27. Duran JM, Anjard C, Stefan C, Loomis WF,
Malhotra V. Unconventional secretion of Acb1 is
mediated by autophagosomes. J Cell Biol 2010;
188:527-36.
28. Manjithaya R, Anjard C, Loomis WF, Subramani S.
Unconventional secretion of Pichia pastoris Acb1 is
dependent on GR ASP protein, peroxisomal functions
and autophagosome formation. J Cell Biol 2010;
188:537-46.
29. Abrahamsen H, Stenmark H. Protein secretion:
unconventional exit by exophagy. Curr Biol 2010;
20 : 415-8.
30. Kinseth MA, Anjard C, Fuller D, Guizzunti G,
Loomis WF, Malhotra V. The Golgi-associated protein
GRASP is required for unconventional protein secre-
tion during development. Cell 2007; 130:524-34.
31. De Jesus M, Nicola AM, Rodrigues ML, Janbon
G, Casadevall A. Capsular localization of the
Cryptococcus neoformans polysaccharide component
galactoxylomannan. Eukaryot Cell 2009; 8:96-103.
32. Oliveira DL, Nimrichter L, Miranda K, Frases S,
Faull KF, Casadevall A, et al. Cryptococcus neoformans
cryoultramicrotomy and vesicle fractionation reveals
an intimate association between membrane lipids
and glucuronoxylomannan. Fungal Genet Biol 2009;
46:956-63.
33. Eisenman HC, Frases S, Nicola AM, Rodrigues
ML, Casadevall A. Vesicle-associated melanization
in Cryptococcus neoformans. Microbiology 2009;
155 : 38 6 0 -7.
Jorge J. Jó Bastos Ferreira for his invaluable
suggestions on the role of vesicles in fun-
gal physiology. We are also grateful to the
Biomolecule Analysis Core Facility, Border
Biomedical Research Center, UTEP, funded
by NIH/NCRR grant 5G12RR008124-
16A1 and 5G12RR008124-16A1S1, for
continuous access to mass spectrometry
(LC-MS and GC-MS) instruments, which
have been fundamental for several of the
studies described here.
References
1. Jeremic A. Cell secretion: an update. J Cell Mol Med
2008; 12:1151-4.
2. Lee MC, Miller EA, Goldberg J, Orci L, Schekman
R. Bi-directional protein transport between the ER
and Golgi. Annu Rev Cell Dev Biol 2004; 20:87-
123.
3. Levi SK, Glick BS. GRASPing unconventional secre-
tion. Cell 2007; 130:407-9.
4. Nickel W, Rabouille C. Mechanisms of regulated
unconventional protein secretion. Nat Rev Mol Cell
Biol 2009; 10:148-55.
5. Herrmann JM, Malkus P, Schekman R. Out of the
ERoutfitters, escorts and guides. Trends Cell Biol
1999; 9:5-7.
6. Schekman RW. Regulation of membrane traffic in
the secretory pathway. Harvey Lect 1994; 90:41-57.
7. Rothblatt J, Schekman R. A hitchhiker’s guide to
analysis of the secretory pathway in yeast. Methods
Cell Biol 1989; 32:3-36.
8. Pugsley AP, Francetic O, Driessen AJ, de Lorenzo
V. Getting out: protein traffic in prokaryotes. Mol
Microbiol 2004; 52:3-11.
9. Nosanchuk JD, Nimrichter L, Casadevall A,
Rodrigues ML. A role for vesicular transport of mac-
romolecules across cell walls in fungal pathogenesis.
Commun Integr Biol 2008; 1:37-9.
10. Rodrigues ML, Nimrichter L, Oliveira DL,
Nosanchuk JD, Casadevall A. Vesicular trans-cell
wall transport in fungi: a mechanism for the deliv-
ery of virulence-associated macromolecules? Lipid
Insights 2008; 2008:27.
11. Casadevall A, Nosanchuk JD, Williamson P,
Rodrigues ML. Vesicular transport across the fungal
cell wall. Trends Microbiol 2009; 17:158-62.
12. Rodrigues ML, Nimrichter L, Oliveira DL, Frases
S, Miranda K, Zaragoza O, et al. Vesicular polysac-
charide export in Cryptococcus neoformans is a eukary-
otic solution to the problem of fungal trans-cell wall
transport. Eukaryot Cell 2007; 6:48-59.
13. Rodrigues ML, Nakayasu ES, Oliveira DL,
Nimrichter L, Nosanchuk JD, Almeida IC, et al.
Extracellular vesicles produced by Cryptococcus neo-
formans contain protein components associated with
virulence. Eukaryot Cell 2008; 7:58-67.
14. Albuquerque PC, Nakayasu ES, Rodrigues ML,
Frases S, Casadevall A, Zancope-Oliveira RM, et al.
Vesicular transport in Histoplasma capsulatum: an
effective mechanism for trans-cell wall transfer of
proteins and lipids in ascomycetes. Cell Microbiol
2008; 10:1695-710.
15. Oliveira DL, Freire-de-Lima CG, Nosanchuk
JD, Casadevall A, Rodrigues ML, Nimrichter L.
Extracellular vesicles from Cryptococcus neoformans
modulate macrophage functions. Infect Immun
2010; 78:1601-9.
16. Regente M, Corti-Monzon G, Maldonado AM,
Pinedo M, Jorrin J, de la Canal L. Vesicular fractions
of sunflower apoplastic fluids are associated with
potential exosome marker proteins. FEBS Lett 2009;
583:3363-6.
release has been associated to protein and
polysaccharide secretion,
12-14,22,32
surface
architecture,
31
virulence,
10,12,13
pigmenta-
tion
33
and modulation of macrophage
function.
15
Despite their apparent multiple
functions in yeast, the cellular components
controlling their biogenesis and release
remain elusive. We emphasize the sup-
position that the methods currently used
for preparation of extracellular fractions
containing vesicles may co-isolate vesicu-
lar compartments of different cellular ori-
gins, which limit the application of studies
based on the generation of punctual muta-
tions. Post-Golgi components required for
conventional secretion, proteins involved
in MVB formation, GRASP and even
autophagy-related events may be involved
in the formation of extracellular vesicles.
Although much progress has been made in
the last three years, the route to understand
how fungal extracellular vesicles are formed
still seems long and laborious.
Acknowledgements
D.L.O. is a Ph.D. student at Instituto de
Bioquímica Médica (Federal University
of Rio de Janeiro, Brazil). L.N. and
M.L.R. are supported by grants from the
Brazilian agencies Conselho Nacional de
Desenvolvimento Científico e Tecnológico
(CNPq), Fundão de Amparo à Pesquisa
do Estado do Rio de Janeiro (FAPERJ)
and Coordenação de Aperfeiçoamento de
Pessoal de Nível Superior (CAPES). A.C.
is supported by NIH awards HL059842,
AI033774, AI052733, AI033142. A.J.G.
and L.N. were supported in part by an
Interhemispheric Research Training Grant
in Infectious Diseases, Fogarty Inter-
national Center (NIH D43-TW007129).
J.D.N. is supported in part by NIH
AI52733-06A1 and AI056070-01A2 and
a Hirschl/Weill-Caulier Career Scientist
Award. R.J.B.C. was supported by the
Training Program in Cellular and Molecular
Biology and Genetics, T32 GM007491.
I.C.A. was partially supported by the NIH/
NCRR grant 5G12RR008124-16A1 and
5G12RR008124-16A1S1. E.S.N was par-
tially supported by the George A. Krutilek
memorial graduate scholarship from
Graduate School, University of Texas at
El-Paso (UTEP). T.J.P.S. was supported by
Fundação de Amparo à Pesquisa do Estado
de São Paulo (FAPESP), Brazil. We thank
103
Anexo 4 Supporting Information S1:
Protein
Description
Cellular distribution
metabolic process
YLR134W
Pdc5p, minor isoform of pyruvate decarboxylase
Nucleus, cytoplasm
YGR087C
Pdc6p, minor isoform of pyruvate decarboxylase
Cytoplasm
YKL152C
Gpm1p, tetrameric phosphoglycerate mutase
Cytosol, mitochondria
YML028W
Tsa1p, ubiquitous housekeeping thioredoxin peroxidase
Cytoplasm
YPR145W
Asn1p, asparagine synthetase
Cytoplasm
YLR303W
Met17p, methionine and cysteine synthase
Cytoplasm
YJL153C
Ino1p, inositol 1-phosphate synthase
Cytoplasm
YDR304C
Cpr5p, peptidyl-prolyl cis-trans isomerase (cyclophilin) of the
endoplasmic reticulum
Endoplasmic reticulum,
cytoplasm
YLR299W
Ecm38p, gamma-glutamyltranspeptidase
Cytoplasm, plasma
membrane
YDR483W
Kre2p, alpha1,2-mannosyltransferase of the Golgi involved in
protein mannosylation
Golgi apparatus, plasma
membrane, Golgi stack
YGR192C
Tdh3p, glyceraldehyde-3-phosphate dehydrogenase
Cytoplasm, mitochondria,
cell wall
YLR109W
Ahp1p, thiol-specific peroxiredoxin
Peroxisome, plasma
membrane, cytoplasm
YGR180C
Rnr4p, ribonucleotide-diphosphate reductase (RNR), small
subunit
Ribonucleoside-
diphosphate reductase,
nucleus, cytoplasm
YHR183W
Gnd1p, 6-phosphogluconate dehydrogenase
Mitochondria, cytoplasm
YHR179W
Oye2p, Widely conserved NADPH oxidoreductase containing
flavin mononucleotide (FMN), may be involved in sterol
metabolism
Nucleus, mitochondria,
cytoplasm
YGL234W
Ade5,7p, Bifunctional enzyme of the 'de novo' purine
nucleotide biosynthetic pathway
Cytoplasm
104
YOL011W
Plb3p, Phospholipase B (lysophospholipase); hydrolyzes
phosphatidylinositol and phosphatidylserine
Extracellular region,
plasma membrane
YLR245C
Cdd1p, Cytidine deaminase; catalyzes the modification of
cytidine to uridine
Nucleus, cytoplasm
YDL124W
Ydl124wp, NADPH-dependent alpha-keto amide reductase
Nucleus, cytoplasm
YML126C
Erg13p, 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) synthase
Endoplasmic reticulum,
mitochondria
YDR050C
Tpi1p, Triose phosphate isomerase, abundant glycolytic
enzyme.
Cytosol, mitochondria,
Cytoplasm
YLR044C
Pdc1p, Major of three pyruvate decarboxylase isozymes, key
enzyme in alcoholic fermentation.
Nucleus, cytoplasm
YGL202W
Aro8p, Aromatic aminotransferase I
Cytoplasm
YDR044W
Hem13p, Coproporphyrinogen III oxidase, catalyzes the sixth
step in the heme biosynthetic pathway
Mitochondrial inner
membrane
YEL047C
Yel047cp, Soluble fumarate reductase
Cytosol, ribosome,
mitochondria
YKL060C
Fba1p, Fructose 1,6-bisphosphate aldolase
Cytosol, mitochondria,
cytoplasm
YBR199W
Ktr4p, Putative mannosyltransferase involved in protein
glycosylation; member of the KRE2/MNT1
mannosyltransferase family
Golgi apparatus, integral to
membrane, membrane
YBR196C
Pgi1p, Glycolytic enzyme phosphoglucose isomerase
Cytosol, mitochondria
YCR012W
Pgk1p, 3-phosphoglycerate kinase; key enzyme in glycolysis
and gluconeogenesis
Cytosol, mitochondria,
cytoplasm
YJL052W
Tdh1p, Glyceraldehyde-3-phosphate dehydrogenase, isozyme
1, involved in glycolysis and gluconeogenesis
Lipid particle, Cell wall,
cytosol, mitochondria,
cytoplasm
YJR009C
Tdh2p, Glyceraldehyde-3-phosphate dehydrogenase, isozyme
2, involved in glycolysis and gluconeogenesis
Lipid particle, Cell wall,
cytosol, mitochondria,
cytoplasm
105
YAL038W
Cdc19p, Pyruvate kinase
Cytosol
YMR303C
Adh2p, Glucose-repressible alcohol dehydrogenase II
Cytoplasm
YGR254W
Eno1p, Enolase I, a phosphopyruvate hydratase
Mitochondria, vacuole,
cytoplasm
YOR133W
Eft1p, Elongation factor 2 (EF-2), catalyzes ribosomal
translocation during protein synthesis
Ribosome
YER091C
Met6p, Cobalamin-independent methionine synthase,
involved in amino acid biosynthesis
Cytoplasm
YCL043C
Pdi1p, Protein disulfide isomerase, multifunctional protein
resident in the endoplasmic reticulum lumen, essential for the
formation of disulfide bonds in secretory and cell-surface
proteins
Endoplasmic reticulum,
endoplasmic reticulum
lumen
YAL012W
Cys3p, Cystathionine gamma-lyase, catalyzes one of the two
reactions involved in the transsulfuration pathway that yields
cysteine from homocysteine
Cytoplasm
YDR533C
Hsp31p, Possible chaperone and cysteine protease
Soluble fraction
YDL045C
Fad1p, Flavin adenine dinucleotide (FAD) synthetase
Cytoplasm
YJR148W
Bat2p, Cytosolic branched-chain amino acid aminotransferase
Nucleus, cytoplasm
YHR057C
Cpr2p, Peptidyl-prolyl cis-trans isomerase (cyclophilin),
catalyzes the cis-trans isomerization of peptide bonds N-
terminal to proline residues; has a potential role in the
secretory pathway
Cellular component
unknown
YOR099W
Ktr1p, Alpha-1,2-mannosyltransferase involved in O- and N-
linked protein glycosylation
Golgi apparatus,
integral to membrane,
membrane, Golgi stack
YJR139C
Hom6p, Homoserine dehydrogenase (L-homoserine:NADP
oxidoreductase), dimeric enzyme that catalyzes the third step
in the common pathway for methionine and threonine
biosynthesis;
Nucleus, cytoplasm
YHR008C
Sod2p, Mitochondrial superoxide dismutase, protects cells
against oxygen toxicity
Mitochondrial matrix,
mitochondria
106
YER043C
Sah1p, S-adenosyl-L-homocysteine hydrolase
Cytoplasm
YNL117W
Mls1p, Malate synthase, enzyme of the glyoxylate cycle,
involved in utilization of non-fermentable carbon sources
Peroxisomal matrix,
glyoxysome, cytoplasm
YKL216W
Ura1p, Dihydroorotate dehydrogenase, catalyzes the fourth
enzymatic step in the de novo biosynthesis of pyrimidines
Extrinsic to membrane,
cytoplasm
YOR375C
Gdh1p, NADP(+)-dependent glutamate dehydrogenase
Nucleus, cytoplasm
YHR174W
Eno2p, Enolase II, a phosphopyruvate hydratase activity
Soluble fraction, vacuole,
mitochondria
YHR208W
Bat1p, Mitochondrial branched-chain amino acid
aminotransferase
Mitochondrial matrix,
mitochondria
YJL130C
Ura2p, Bifunctional carbamoylphosphate synthetase (CPSase)-
aspartate transcarbamylase (ATCase), catalyzes the first two
enzymatic steps in the de novo biosynthesis of pyrimidines
Mitochondria, cytoplasm,
integral to membrane
YAR071W
Pho11p, One of three repressible acid phosphatases, a
glycoprotein that is transported to the cell surface by the
secretory pathway
Extracellular region
YBR011C
Ipp1p, Cytoplasmic inorganic pyrophosphatase (PPase)
Cytosol
YJL026W
Rnr2p, Ribonucleotide-diphosphate reductase (RNR), small
subunit
Nucleus, cytoplasm
YGR256W
Gnd2p, 6-phosphogluconate dehydrogenase (decarboxylating),
catalyzes an NADPH regenerating reaction in the pentose
phosphate pathway
Cytosol
YOL086C
Adh1p, Alcohol dehydrogenase, fermentative isozyme
Cytosol
YKL157W
Ape2p, Zinc-dependent metallopeptidase yscII
Mitochondria, cell wall-
bounded periplasmic
space, cytoplasm
cell organization and biogenesis
YPL053C
Ktr6p, Probable mannosylphosphate transferase involved in
the synthesis of core oligosaccharides in protein glycosylation
pathway
Membrane fraction,
integral to membrane
107
YGR032W
Gsc2p, Catalytic subunit of 1,3-beta-glucan synthase, involved
in formation of the inner layer of the spore wall
Prospore membrane, 1,3-
beta-glucan synthase
complex, membrane
fraction, integral to
membrane, actin cap
YLR342W
Fks1p, Catalytic subunit of 1,3-beta-D-glucan synthase,
involved in cell wall synthesis and maintenance; localizes to
sites of cell wall remodeling
1,3-beta-glucan synthase
complex, integral to
membrane, actin cortical
patch, actin cap,
mitochondria
YLR355C
Ilv5p, Acetohydroxyacid reductoisomerase, mitochondrial
protein involved in amino acid biosynthesis, also required for
maintenance of mitochondrial DNA
Mitochondria,
mitochondrial nucleoid
YMR307W
Gas1p, Beta-1,3-glucanosyltransferase, required for cell wall
assembly; localizes to the cell surface via a
glycosylphosphatidylinositol (GPI) anchor
Cell wall, plasma
membrane, mitochondria
YGR282C
Bgl2p, Endo-beta-1,3-glucanase, major protein involved in cell
wall maintenance
Cell wall
YDR055W
Pst1p, Cell wall protein that contains a putative GPI-
attachment site; secreted by regenerating protoplasts; up-
regulated by cell wall damage
Cell wall, Plasma
membrane
YLR249W
Yef3p, Translational elongation factor 3, stimulates the binding
of aminoacyl-tRNA (AA-tRNA) to ribosomes
Ribosome
YDR261C
Exg2p, Exo-1,3-beta-glucanase, involved in cell wall beta-
glucan assembly; may be anchored to the plasma membrane
via a glycosylphosphatidylinositol (GPI) anchor
Cell wall, membrane
YBR078W
Ecm33p, GPI-anchored protein of unknown function, has a
possible role in apical bud growth
Membrane fraction, Cell
wall, mitochondria
YJR105W
Ado1p, Adenosine kinase, may be involved in recycling of
adenosine
Nucleus, cytoplasm
YLR300W
Exg1p, Major exo-1,3-beta-glucanase involved in cell wall beta-
glucan assembly
Cell wall
YLR121C
Yps3p, Aspartic protease, attached to the plasma membrane
Anchored to plasma
108
via a glycosylphosphatidylinositol (GPI) anchor
membrane
YMR186W
Hsc82p, Cytoplasmic chaperone of the Hsp90 family,
redundant in function and nearly identical with Hsp82p
Mitochondria, cytoplasm
YFL039C
Act1p, Actin, structural protein involved in cell polarization,
endocytosis, and other cytoskeletal functions
Cytoplasm
YLL026W
Hsp104p, Heat shock protein that cooperates to refold and
reactivate previously denatured, aggregated proteins;
responsive to stresses including: heat, ethanol, and sodium
arsenite
Nucleus, cytoplasm
YPL106C
Sse1p, ATPase that is a component of the heat shock protein
Hsp90 chaperone complex
Cytoplasm
Transporters
YPL036W
Pma2p, Plasma membrane H+-ATPase, isoform of Pma1p,
regulator of cytoplasmic pH and plasma membrane potential
Mitochondria, plasma
membrane, Integral to
membrane
YGL253W
Hxk2p, Hexokinase isoenzyme 2 that catalyzes
phosphorylation of glucose in the cytosol
Cytosol, mitochondria,
nucleus
YCR053W
Thr4p, Threonine synthase, conserved protein that catalyzes
formation of threonine
Nucleus, cytoplasm
YPR080W
Tef1p,Translational elongation factor EF-1 alpha; functions in
the binding reaction of aminoacyl-tRNA (AA-tRNA) to
ribosomes
Eukaryotic translation
elongation factor 1
complex, ribosome,
mitochondria
YJR048W
Cyc1p, Cytochrome c, isoform 1; electron carrier of the
mitochondrial intermembrane space
Mitochondrial
intermembrane space
Mitochondrial respiratory
chain
YGL008C
Pma1p, Plasma membrane H+-ATPase, major regulator of
cytoplasmic pH and plasma membrane potential
Lipid raft, mitochondria,
plasma membrane
YFR053C
Hxk1p, Hexokinase isoenzyme 1
Cytosol
YDL046W
Npc2p , Functional homolog of human NPC2/He1
Vacuole, cell cycle-
correlated morphology
109
YEL039C
Cyc7p, Cytochrome c isoform 2; electron carrier of the
mitochondrial intermembrane space
Mitochondrial respiratory
chain
carbohydrate metabolism
YGL028C
Scw11p , Cell wall protein with similarity to glucanases; may
play a role in conjugation during mating
Cell wall
YGR279C
Scw4p, Cell wall protein with similarity to glucanases
Cell wall
YNR067C
Dse4p, Daughter cell-specific secreted protein with similarity
to glucanases, degrades cell wall from the daughter side
causing daughter to separate from mother
Extracellular region, Cell
wall
YMR105C
Pgm2p, Phosphoglucomutase, catalyzes a key step in hexose
metabolism
Cytosol, cytoplasm
YLR354C
Tal1p, Transaldolase, enzyme in the non-oxidative pentose
phosphate pathway
Cytoplasm
YMR305C
Scw10p, Cell wall protein with similarity to glucanases; may
play a role in conjugation during mating
Endoplasmic reticulum,
Cell wall, cytoplasm
response to stress
YMR169C
Ald3p, Cytoplasmic aldehyde dehydrogenase, involved in beta-
alanine synthesis
Cytoplasm
YGR234W
Yhb1p, Nitric oxide oxidoreductase, flavohemoglobin involved
in nitric oxide detoxification; plays a role in the oxidative and
nitrosative stress responses
Mitochondrial matrix,
cytoplasm, cytosol
YDL229W
Ssb1p, Cytoplasmic ATPase that is a ribosome-associated
molecular chaperone
Soluble fraction, polysome
YNL160W
Ygp1p , Cell wall-related secretory glycoprotein; induced by
nutrient deprivation-associated growth arrest and upon entry
into stationary phase
Cell wall
YNL209W
Ssb2p, Cytoplasmic ATPase that is a ribosome-associated
molecular chaperone
Polysome, cytoplasm
YPL240C
Hsp82p, Hsp90 chaperone required for pheromone signaling
Cytoplasm
Protein biosynthesis
110
YLR075W
Rpl10p , Protein component of the large (60S) ribosomal
subunit
Cytosolic large ribosomal
subunit
YBR118W
Tef2p,Translational elongation factor EF-1 alpha; functions in
the binding reaction of aminoacyl-tRNA (AA-tRNA) to
ribosomes
Eukaryotic translation
elongation factor 1
complex, ribosome,
cytoplasm
YDR012W
Rpl4bp, Protein component of the large (60S) ribosomal
subunit
Cytosolic large ribosomal
subunit, cytoplasm
YDR385W
Eft2p, Elongation factor 2 (EF-2); catalyzes ribosomal
translocation during protein synthesis
Ribosome
YBR031W
Rpl4ap, N-terminally acetylated protein component of the
large (60S) ribosomal subunit
Cytosolic large ribosomal
subunit
YJR123W
Rps5p, Protein component of the small (40S) ribosomal
subunit
Cytosolic small ribosomal
subunit
protein degradation
YCL057W
Prd1, Zinc metalloendopeptidase, involved in degradation of
mitochondrial proteins and of presequence peptides cleaved
from imported proteins
Mitochondrial
intermembrane space,
Cytoplasm
YMR297W
Prc1p, Vacuolar carboxypeptidase Y (proteinase C), broad-
specificity C-terminal exopeptidase involved in non-specific
protein degradation in the vacuole; member of the serine
carboxypeptidase family; protein coding
Lumen of vacuole with cell
cycle-correlated
morphology, endoplasmic
reticulum, cytoplasm
YPL235W
Rvb2p, Essential protein involved in transcription regulation;
component of chromatin remodeling complexes
Nucleus, chromatin
remodeling complex,
SWR1 complex, INO80
complex
YFR044C
Dug1p, Probable di- and tri-peptidase; forms a complex to
degrade glutathione (GSH)
Ribosome, mitochondria,
cytoplasm
YGL203C
Kex1p, Protease involved in the processing of killer toxin and
alpha factor precursor; cleaves Lys and Arg residues from the
C-terminus of peptides and proteins; protein coding
Trans-Golgi network,
integral to membrane
protein transport
111
YJL034W
Kar2p , ATPase involved in protein import into the ER, also acts
as a chaperone to mediate protein folding in the ER and may
play a role in ER export of soluble proteins
Endoplasmic reticulum
lumen, endoplasmic
reticulum
YLL024C
Ssa2p , ATP binding protein involved in protein folding and
vacuolar import of proteins
Membrane of vacuole with
cell cycle-correlated
morphology, chaperonin-
containing T-complex, Cell
wall, mitochondria,
cytoplasm
YBL075C
Ssa3p, ATPase involved in protein folding and the response to
stress; plays a role in SRP-dependent cotranslational protein-
membrane targeting and translocation; member of the heat
shock protein 70 (HSP70) family
Cytosol
YAL005C
Ssa1p, ATPase involved in protein folding and nuclear
localization signal (NLS)-directed nuclear transport; member of
heat shock protein 70 (HSP70) family
Membrane of vacuole with
cell cycle-correlated
morphology, chaperonin-
containing T-complex, Cell
wall, nucleus, cytoplasm
YER103W
Ssa4p, Heat shock protein that is highly induced upon stress;
plays a role in SRP-dependent cotranslational protein-
membrane targeting and translocation
Nucleus, cytoplasm
Sporulation
YPL154C
Pep4p, Vacuolar aspartyl protease (proteinase A), required for
the posttranslational precursor maturation of vacuolar
proteinases; important for protein turnover after oxidative
damage
Lumen of vacuole with cell
cycle-correlated
morphology, mitochondria
YDR155C
Cpr1p, Cytoplasmic peptidyl-prolyl cis-trans isomerase
(cyclophilin), catalyzes the cis-trans isomerization of peptide
bonds N-terminal to proline residues
Histone deacetylase
complex, mitochondria,
nucleus
YER177W
Bmh1p, 14-3-3 protein, major isoform; controls proteome at
post-transcriptional level, binds proteins and DNA, involved in
regulation of many processes including exocytosis, vesicle
transport, Ras/MAPK signaling, and rapamycin-sensitive
signaling
Nucleus
YDR099W
Bmh2p, 14-3-3 protein, minor isoform; controls proteome at
Nucleus
112
post-transcriptional level, binds proteins and DNA, involved in
regulation of many processes including exocytosis, vesicle
transport, Ras/MAPK signaling, and rapamycin-sensitive
signaling
Unknown
YJL171C
Yjl171cp, GPI-anchored cell wall protein of unknown function;
induced in response to cell wall damaging agents and by
mutations in genes involved in cell wall biogenesis
Cell wall, mitochondria
YNL134C
Ynl134cp, Putative protein of unknown function with similarity
to dehydrogenases from other model organisms
Nucleus, cytoplasm
YLR178C
Tfs1p, Carboxypeptidase Y inhibitor,
phosphatidylethanolamine-binding protein involved in protein
kinase A signaling pathway
Membrane of vacuole with
cell cycle-correlated
morphology, soluble
fraction, lumen of vacuole
with cell cycle-correlated
morphology, cytoplasm
YLR179C
Ylr179cp, Protein of unknown function, transcription is
activated by paralogous proteins Yrm1p and Yrr1p along with
proteins involved in multidrug resistance
Nucleus, cytoplasm
YHR138C
Yhr138cp, Putative protein of unknown function; has similarity
to Pbi2p; double null mutant lacking Pbi2p and Yhr138p
exhibits highly fragmented vacuoles
Cellular component
unknown
YNL208W
Ynl208wp, Protein of unknown function; may interact with
ribosomes, based on co-purification experiments; authentic,
non-tagged protein is detected in purified mitochondria
Ribosome, mitochondria
YLR414C
Ylr414cp, Putative protein of unknown function;
transcriptional induced in response to cell wall damage;
YLR414C is not an essential gene
Cellular bud, cytoplasm
YDR262W
Ydr262wp, Putative protein of unknown function; induced in
response to the DNA-damaging agent MMS; gene expression
increases in response to Zymoliase treatment
Vacuole, cell cycle-
correlated morphology
YOL030W
Gas5p, 1,3-beta-glucanosyltransferase, has similarity to Gas1p;
localizes to the cell wall
Membrane fraction,
membrane, Cell wall
113
YMR215W
Gas3p, Putative 1,3-beta-glucanosyltransferase, has similarity
to Gas1p; localizes to the cell wall
Membrane fraction,
membrane, Cell wall
YPL225W
Ypl225wp, Protein of unknown function that may interact with
ribosomes
Ribosome, cytoplasm
YHR215W
Pho12p , One of three repressible acid phosphatases, a
glycoprotein that is transported to the cell surface by the
secretory pathway; nearly identical to Pho11p
Vacuole, cell cycle-
correlated morphology
* Proteins with less than 10 spectra after MS/MS analysis were not listed in this table.
118
V- Discussão:
Nesta tese e em estudos desenvolvidos por nosso grupo e outros autores (Rodrigues, et al.
2008; Panepinto, et al., 2009; Hu & Kronstad, et al. 2009) foi demonstrado que espécies de
fungos produzem vesículas extracelulares contendo bicamadas lipídicas morfologicamente
similares à exossomos de células mamíferas, com dimensões variando entre 60 e 300 nm. Este
novo mecanismo de secreção está implicado tanto em processos fisiológicos (como o
crescimento capsular) quanto na interação com célula hospedeira em C. neoformans,
comprovando que estas vesículas são biologicamente ativas. A caracterização de vesículas de
diversos mutantes em vias de tráfego sugere que a biogênese vesicular envolve componentes de
diferentes vias, tanto clássicas quanto não-convencionais, o que retrata a complexidade deste
processo secretório.
Fez-se necessário legitimar a existência das vesículas secretadas como um fenômeno
biológico, o que foi comprovado de três maneiras: (i) foi visto que a produção de vesículas é
dependente de viabilidade celular; (ii) experimentos de pulso e caça revelaram que componentes
vesiculares advêm do metabolismo celular e (iii) vesículas não incorporam componentes
exógenos presentes no meio de cultura e portanto não são formadas randomicamente pela
associação aleatória de lipídeos (Anexo 1). Posteriormente foi demonstrado que diversas
espécies de fungos são capazes de secretar vesículas, consolidando a existência de um
mecanismo de secreção vesicular comum aos fungos (Albuquerque, et al. 2008).
É sabido que o mutante de C. neoformans incapaz de expressar o produto do gene SAV1,
homólogo ao SEC4 de S. cerevisiae, apresenta acúmulo de vesículas citoplasmáticas contendo
119
GXM (Yoneda & Doering, 2006). Tais vesículas advêm do Aparato de Golgi e seguindo a via
secretória convencional, deveriam fundir-se à membrana plasmática liberando a GXM no
periplasma (Ponnambalam & Baldwin, 2003). Este modelo, entretanto, confronta-se com o fato
de vesículas intactas contendo GXM serem liberadas no ambiente extracelular (Anexo 1, Figura
4A). Mais do que isso, o modelo de secreção de GXM proposto por Yoneda e Doering não explica
de que forma o polissacarídeo de alto peso molecular atravessa a densa estrutura da parede
celular (Yoneda & Doering, 2006). É possível que após a síntese no Aparato de Golgi, vesículas
brotem da TGN carreando GXM e direcionem-se a uma via não- convencional de secreção ao
invés de se fundirem a membrana plasmática. De fato, compartimentos semelhantes à MVBs de
células mamíferas foram encontrados no citoplasma do C. neoformans (Rodrigues, et al. 2008
Takeo, et al. 1973). Além disso, experimentos de crioultramicrotomia seguidos de imuno-
marcação revelaram que GXM é transportada por diferentes compartimentos potencialmente
incluindo vacúolos, estruturas reticulares e vesículas citoplasmáticas (Anexo 2, Figura 3), o que
sugere que a GXM deve passar por vias de tráfego mais complexas do que a exocitose
convencional.
Os mecanismos que medeiam a passagem de vesículas pela parede celular de fungos são
ainda pouco conhecidos. Estudos de microscopia de força atômica revelaram a existência de
poros na parede celular da levedura S. cerevisiae com diâmetro variando entre 200-400 nm (de
Souza Pereira & Geibel, 1999), o que comportaria a passagem das vesículas. Ainda assim
permanece em aberto a questão de como as vesículas seriam transportadas através destes
poros. Especula-se que proteínas motoras possam estar envolvidas em tal evento. É sabido que
miosina e actina estão envolvidas em vários processos associados à parede celular, como
120
formação de septo, crescimento na hifa, germinação e esporulação (Oberholzer, et al. 2002; Bi,
2001; Esnault, et al. 1999). Entretanto, ainda não existem evidências da participação direta de
proteínas envolvidas em trânsito de moléculas e organelas citoplasmáticas agindo na matriz da
parede celular.
Análises feitas por microscopia eletrônica de transmissão revelaram a existência de grupos de
vesículas com elétron densidades distintas (Rodrigues, et al. 2008). Esta observação nos levou a
questionar se tal diferença morfológica seria reflexo de vias distintas de biogênese. O
fracionamento vesicular em gradiente de densidade evidenciou uma população de vesículas
enriquecida em GXM e outra população de vesículas contendo níveis mínimos do polissacarídeo
(Anexo 2, Figura 5). A existência de mais de uma população de vesículas que diferem em termos
de conteúdo, densidade e morfologia reforça, portanto, a hipótese de que mais de uma via de
biogênese deva estar implicada na produção desses compartimentos membranosos. Análises
genéticas necessárias para a identificação destas vias no C. neoformans estão em andamento,
sendo parte de estudos futuros dentro do nosso laboratório.
A caracterização proteômica de vesículas de C. neoformans revelou uma complexa
composição protéica incluindo chaperonas, proteínas de membrana, proteínas citoplasmáticas,
mitocondriais e até nucleares (Rodrigues, et al. 2008). Tal complexidade reflete uma
característica peculiar de fungos: proteínas localizam-se em mais de um compartimento celular
onde devem exercer funções distintas (Nimrichter, et al. 2005). Esse fenômeno, denominado
‘moonlighting’ pelo grupo de Jeffery (Jeffery, 1999), tem vários precedentes na área de Biologia
Celular de fungos. Em Histoplasma capsulatum, por exemplo, histonas são encontradas na
parede celular (Nosanchuk, et al. 2003). Em Paracoccidioides brasiliensis, a enzima glicolítica
121
gliceraldeído-3-fosfato desidrogenase também se encontra na parede celular, onde participa da
patogênese mediando adesão de leveduras a matriz extracelular (Barbosa, et al. 2006). A
proteína mitocondrial Mdj1 é também encontrada na parede celular em P. brasiliensis (Batista,
et al. 2006). É possível, portanto, que proteínas vesiculares de C. neoformans apresentem
funções no espaço extracelular, sobretudo em relação a patogênese, virulência e imunogênese.
De acordo com essa hipótese está o fato de que extratos de proteínas vesiculares reagem com
soro de pacientes infectados por C. neoformans (Rodrigues, et al. 2008), confirmando que as
mesmas são secretadas durante a infecção, funcionando como imunógenos ativadores da
resposta humoral.
Mecanismos de virulência e processos secretórios estão intimamente relacionados em
diversos modelos microbianos (Hauser, et al. 1998; Ghannoum, 1998; Hoegl, et al, 1996; Hube, et
al. 1996). Na presente tese foi demonstrado que vesículas de C. neoformans podem ser
internalizadas por macrófagos induzindo sua ativação, evento que leva a aumentos do índice de
fagocitose e de atividade microbicida (Anexo 3). Sabe-se que além de GXM, vesículas de C.
neoformans carreiam diversos outros moduladores de virulência como lacase, superóxido
dismutase, glucosilceramida e urease (Rodrigues, et al. 2008). No entanto estas estruturas não se
mostraram citotóxicas em modelo de linhagem de macrófagos (Anexo3, Figura 1C). O
empacotamento de diversas moléculas relacionadas à virulência dentro de uma mesma estrutura
poderia representar um mecanismo vantajoso de concentração de enzimas e moléculas
antioxidantes que agiriam na defesa da célula fúngica. Entretanto este coquetel de moléculas
fúngicas induz eficiente ativação celular e produção de NO e citocinas (Anexo 3). É possível que a
concentração de componentes do patógeno permita assimilação mais eficiente por parte do
122
fagócito do que se estas moléculas estivessem solúveis no meio, levando a uma resposta mais
eficiente na célula hospedeira. Preparações de TEM de pulmões de camundongos infectados
sugerem fortemente que vesículas são produzidas in vivo durante a infecção (Anexo 1, Figura 1).
É importante ressaltar que incubação de células com vesículas isoladas do sobrenadante de
cultivo não necessariamente mimetiza a produção de vesículas que ocorreria naturalmente
durante a infecção em termos de concentração vesicular, tempo de exposição e possivelmente
de composição.
É notório que a presença de GXM nas vesículas influencia diretamente o perfil de citocinas
secretadas e o resultado da interação entre C. neoformans e fagócitos. Macrófagos incubados
com vesículas isoladas do mutante acapsular produzem significativamente mais TNF-α do que os
incubados com vesículas de células capsuladas e ao mesmo tempo produzem significativamente
menos IL-10 (Anexo 3, Figura 3), o que corrobora com o conhecido papel antiinflamatório e
imunossupressor da GXM (Monari, et al. 2009; Lupo, et al. 2008; Monari, et al. 2006; Villena, et
al. 2008). Mais do que isso, células incubadas com vesículas do mutante acapsular apresentaram
uma maior capacidade fagocítica e um menor tempo de incubação necessário para visualização
da atividade microbicida quando comparadas com células ativadas com vesículas contendo GXM
(Anexo 3, Figuras 4 e 5). Tais resultados sugerem que vesículas isoladas do sobrenadante de
cultura de C. neoformans apresentam grande potencial para modular a resposta imune e o curso
da infecção, uma vez que células fagocíticas exercem papel crucial no controle da criptococose
(Miller & Mitchel, 1991). Estudos sobre exossomos derivados de células dendríticas revelaram
que estas estruturas são promissoras vacinas no tratamento antitumoral (Wolfers, et al. 2001).
Neste sentido, é razoável considerar o desenvolvimento de uma possível vacina a partir de
123
vesículas produzidas pelo C. neoformans como estratégia terapêutica, considerando o fato de
vesículas conterem várias proteínas imunogênicas (Rodrigues, et al. 2008).
Embora a secreção vesicular tenha sido primeiramente caracterizada em C. neoformans,
vários fatores tornam S. cerevisiae um modelo mais promissor para o estudo das vias de
biogênese vesicular em fungos. Sendo essa a primeira levedura a ter seu genoma seqüenciado,
além de ser essa a espécie eucariótica como maior volume de informações disponíveis com
relação a eventos da biologia celular em geral, várias vantagens técnicas se configuram no
modelo do S. cerevisiae. Encontra-se disponível, por exemplo, uma vasta coleção de mutantes
com defeitos em vias de tráfego celular com fenótipos caracterizados (Schekman, et al. 1983;
Winzeler, et al. 1999; Giaever, et al. 2002). Em conseqüência disso optamos por utilizar S.
cerevisiae com o intuito de definir genes cujos produtos poderiam estar relacionados com
produção e/ou liberação de vesículas extracelulares. Utilizando os protocolos estabelecidos por
nosso grupo para C. neoformans, vesículas extracelulares foram isoladas do sobrenadante de
cultivo de S. cerevisiae, conforme visualizado por TEM (Anexo 4, Figura 1). Dois mutantes
principais foram utilizados como protótipos neste estudo: o mutante sec4-2 ts, o qual é afetado
na via secretora clássica e o mutante snf7
, o qual apresenta defeitos na formação de
exossomos. O gene SEC4 foi selecionado por ser homólogo do gene SAV1 de C. neoformans, cujo
silenciamento leva ao acúmulo de vesículas citoplasmáticas contendo GXM (Yoneda & Doering,
2006). O gene SNF7 foi escolhido com base no fato de que seu produto, a Snf7p, atua
diretamente na formação de vesículas internas durante a maturação do endossoma em MVB,
afetando portanto a formação de exossomos (Tu, et al. 1993). Outros mutantes usados incluíram
o gene BOS1, envolvido no transporte a partir do RE, SEC1, relacionado à fusão de vesículas
124
oriundas do Aparato de Golgi com a membrana plasmática, e VPS23, que codifica um dos
componentes do complexo ESCRT-I, responsável pelo endereçamento de proteínas ao
compartimento MVB (Schekman, et al. 1983; Babst, et al. 2000; Wuestehube, et al. 1996). Um
mutante incapaz de expressar o produto do gene GRH1 também foi analisado, uma vez que seu
envolvimento em um mecanismo de secreção não- convencional foi recentemente descrito em S.
cerevisiae (Duran, et al. 2010).
A secreção vesicular não foi completamente abolida em nenhum dos mutantes analisados, o
que pode indicar que múltiplas vias estejam implicadas neste processo. A secreção vesicular pelo
mutante sec 4-2 ts, entretanto, foi expressivamente diminuída (Anexo 4, Figura 6), o que indica a
participação de um componente da via de secreção clássica na geração de vesículas. Contudo, o
mecanismo pelo qual vesículas extracelulares surgem a partir de vesículas citoplasmáticas
oriundas do Aparato de Golgi permanece desconhecido. É possível que eventos posteriores
viabilizem topologicamente a formação de tais vesículas utilizando maquinarias de secreção não-
convencional. De fato, foi demonstrado neste estudo que células mutantes deficientes na
expressão do produto do gene GRH1, as quais são deficientes no funcionamento de uma via de
secreção não-convencional dependente de mecanismos de autofagia (Duran, et al. 2010),
também apresentam diminuição acentuada da secreção de vesículas (Anexo 4, Figura 7). A
influência de mecanismos autofágicos na secreção vesicular será avaliada futuramente pelo
nosso grupo através de estudos de secreção em mutantes ATG, já em processo de preparação.
A caracterização proteômica das vesículas de S. cerevisiae revelou uma composição bastante
complexa, incluindo ao menos 127 proteínas (Anexo 4, Figura 2 e Tabela suplementar S1). Esse
perfil composicional foi consistentemente similar em todas as cepas analisadas (3 diferentes
125
cepas selvagens e 5 diferentes mutantes) (Anexo 4, Figura 4). Muitas destas proteínas são
citoplasmáticas e não se correlacionam com processos secretórios. A presença dessas proteínas,
entretanto, (por exemplo: piruvato quinase e glucose-6- fosfato isomerase) é freqüentemente
descrita em exossomos de mamíferos (Schorey & Bhatnagar, 2008). Especula-se que essa
observação estaria relacionada ao fato de que as invaginações na membrana do compartimento
endocítico levam à incorporação aleatória de porções do citosol (Schorey & Bhatnagar, 2008). A
complexidade da composição proteômica das vesículas de S. cerevisiae está aparentemente
relacionada a interações biológicas múltiplas. Em nosso estudo, estimamos que ocorram 219
diferentes interações dentre as 127 proteínas descritas (Anexo 4, Figura 2B). Diversas enzimas
que hidrolisam componentes estruturais da parede celular também foram encontradas nas
vesículas, sugerindo que o processo de passagem de vesículas através da parede possa envolver
remodelamento dessa estrutura. A análise proteômica revelou ainda um enriquecimento
importante de proteínas GPI-ancoradas bem como de proteínas sintetizadas no RE em frações
vesiculares. É sabido que proteínas GPI ancoradas são enriquecidas em microdomínios lipídicos
de membrana, o que poderia sugerir pistas da origem intracelular dessas membranas. Sabe-se
que exossomos de origem endossomal, bem como sítios de saída do RE, são enriquecidos em tais
microdomínios (Subra, et al. 2007; Mayor & Riezman, 2004).
Mutações relacionadas à formação de MVB não alteraram a quantidade de vesículas
produzidas por S. cerevisiae, além de não afetar seu perfil protéico qualitativo. Análises semi-
quantitativas do proteoma das vesículas, entretanto, mostraram alterações drásticas na
abundância relativa de proteínas (Anexo 4, Figura 5), sugerindo que defeitos na via de formação
de MVBs afetam a composição de vesículas extracelulares. A via de secreção não-convencional
126
dependente da proteína Grh1 também parece convergir para via de secreção de exossomos
(Duran, et al. 2010). Os dados semi-quantitativos de proteínas juntamente com análises
quantitativas de lipídeos sugerem que componentes de ltiplas vias devem estar relacionados
à produção de vesículas extracelulares. A secreção de vesículas parece ser um processo
multifatorial complexo que possivelmente envolve vias redundantes.
Esta tese propõe a existência de um novo veículo de transporte de macromoléculas para o
ambiente extracelular em fungos. Tal achado gera diversos questionamentos sobre o impacto
deste processo na fisiologia celular e na patôgenese de fungos. Outros aspectos funcionais das
vesículas, como por exemplo em processos de comunicação célula-célula ou transferência lateral
de material genético, devem ser investigados. Sobretudo, os mecanismos subjacentes a secreção
vesicular e transporte através da parede celular ainda precisam ser esclarecidos. Em suma, o
presente trabalho abre potenciais novas áreas de investigação sobre estrutura da parede celular
de fungos, mecanismos virulência e rotas não convencionais de secreção de proteínas.
127
VI- Referências Bibliográficas:
Alam, S.L., and Sundquist, W.I. (2007). Structural biology: ESCRT service. Nature 447, 921-922.
Albuquerque, P.C., Nakayasu, E.S., Rodrigues, M.L., Frases, S., Casadevall, A., Zancope-Oliveira,
R.M., Almeida, I.C., and Nosanchuk, J.D. (2008). Vesicular transport in Histoplasma capsulatum:
an effective mechanism for trans-cell wall transfer of proteins and lipids in ascomycetes. Cell
Microbiol 10, 1695-1710.
Altfeld, M., Addo, M.M., Kreuzer, K.A., Rockstroh, J.K., Dumoulin, F.L., Schliefer, K., Leifeld, L.,
Sauerbruch, T., and Spengler, U. (2000). T(H)1 to T(H)2 shift of cytokines in peripheral blood of
HIV-infected patients is detectable by reverse transcriptase polymerase chain reaction but not by
enzyme-linked immunosorbent assay under nonstimulated conditions. J Acquir Immune Defic
Syndr 23, 287-294.
Alvarez, M., and Casadevall, A. (2006). Phagosome extrusion and host-cell survival after
Cryptococcus neoformans phagocytosis by macrophages. Curr Biol 16, 2161-2165.
Andrei, C., Dazzi, C., Lotti, L., Torrisi, M.R., Chimini, G., and Rubartelli, A. (1999). The secretory
route of the leaderless protein interleukin 1beta involves exocytosis of endolysosome-related
vesicles. Mol Biol Cell 10, 1463-1475.
Andrei, C., Margiocco, P., Poggi, A., Lotti, L.V., Torrisi, M.R., and Rubartelli, A. (2004).
Phospholipases C and A2 control lysosome-mediated IL-1 beta secretion: Implications for
inflammatory processes. Proc Natl Acad Sci U S A 101, 9745-9750.
Anjard, C., and Loomis, W.F. (2005). Peptide signaling during terminal differentiation of
Dictyostelium. Proc Natl Acad Sci U S A 102, 7607-7611.
Arora, S., Hernandez, Y., Erb-Downward, J.R., McDonald, R.A., Toews, G.B., and Huffnagle, G.B.
(2005). Role of IFN-gamma in regulating T2 immunity and the development of alternatively
activated macrophages during allergic bronchopulmonary mycosis. J Immunol 174, 6346-6356.
128
Babst, M., Odorizzi, G., Estepa, E.J., and Emr, S.D. (2000). Mammalian tumor susceptibility gene
101 (TSG101) and the yeast homologue, Vps23p, both function in late endosomal trafficking.
Traffic 1, 248-258.
Barbosa, F.M., Fonseca, F.L., Figueiredo, R.T., Bozza, M.T., Casadevall, A., Nimrichter, L., and
Rodrigues, M.L. (2007). Binding of glucuronoxylomannan to the CD14 receptor in human A549
alveolar cells induces interleukin-8 production. Clin Vaccine Immunol 14, 94-98.
Barbosa, M.S., Bao, S.N., Andreotti, P.F., de Faria, F.P., Felipe, M.S., dos Santos Feitosa, L.,
Mendes-Giannini, M.J., and Soares, C.M. (2006). Glyceraldehyde-3-phosphate dehydrogenase of
Paracoccidioides brasiliensis is a cell surface protein involved in fungal adhesion to extracellular
matrix proteins and interaction with cells. Infect Immun 74, 382-389.
Barlowe, C., Orci, L., Yeung, T., Hosobuchi, M., Hamamoto, S., Salama, N., Rexach, M.F.,
Ravazzola, M., Amherdt, M., and Schekman, R. (1994). COPII: a membrane coat formed by Sec
proteins that drive vesicle budding from the endoplasmic reticulum. Cell 77, 895-907.
Barr, F.A., Puype, M., Vandekerckhove, J., and Warren, G. (1997). GRASP65, a protein involved in
the stacking of Golgi cisternae. Cell 91, 253-262.
Batista, W.L., Matsuo, A.L., Ganiko, L., Barros, T.F., Veiga, T.R., Freymuller, E., and Puccia, R.
(2006). The PbMDJ1 gene belongs to a conserved MDJ1/LON locus in thermodimorphic
pathogenic fungi and encodes a heat shock protein that localizes to both the mitochondria and
cell wall of Paracoccidioides brasiliensis. Eukaryot Cell 5, 379-390.
Bauman, S.K., Nichols, K.L., and Murphy, J.W. (2000). Dendritic cells in the induction of protective
and nonprotective anticryptococcal cell-mediated immune responses. J Immunol 165, 158-167.
Beatty, W.L., and Russell, D.G. (2000). Identification of mycobacterial surface proteins released
into subcellular compartments of infected macrophages. Infect Immun 68, 6997-7002.
Beenhouwer, D.O., Shapiro, S., Feldmesser, M., Casadevall, A., and Scharff, M.D. (2001). Both Th1
and Th2 cytokines affect the ability of monoclonal antibodies to protect mice against
Cryptococcus neoformans. Infect Immun 69, 6445-6455.
129
Bendtsen, J.D., Nielsen, H., von Heijne, G., and Brunak, S. (2004). Improved prediction of signal
peptides: SignalP 3.0. J Mol Biol 340, 783-795.
Bhatnagar, S., and Schorey, J.S. (2007). Exosomes released from infected macrophages contain
Mycobacterium avium glycopeptidolipids and are proinflammatory. J Biol Chem 282, 25779-
25789.
Bhatnagar, S., Shinagawa, K., Castellino, F.J., and Schorey, J.S. (2007). Exosomes released from
macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro
and in vivo. Blood 110, 3234-3244.
Bi, E. (2001). Cytokinesis in budding yeast: the relationship between actomyosin ring function
and septum formation. Cell Struct Funct 26, 529-537.
Bingle, L.E., Bailey, C.M., and Pallen, M.J. (2008). Type VI secretion: a beginner's guide. Curr Opin
Microbiol 11, 3-8.
Blackstock, R., Buchanan, K.L., Adesina, A.M., and Murphy, J.W. (1999). Differential regulation of
immune responses by highly and weakly virulent Cryptococcus neoformans isolates. Infect
Immun 67, 3601-3609.
Blackstock, R., and Murphy, J.W. (1997). Secretion of the C3 component of complement by
peritoneal cells cultured with encapsulated Cryptococcus neoformans. Infect Immun 65, 4114-
4121.
Blackstock, R., and Murphy, J.W. (2004). Role of interleukin-4 in resistance to Cryptococcus
neoformans infection. Am J Respir Cell Mol Biol 30, 109-117.
Bock, J.B., Matern, H.T., Peden, A.A., and Scheller, R.H. (2001). A genomic perspective on
membrane compartment organization. Nature 409, 839-841.
Bonifacino, J.S., and Glick, B.S. (2004). The mechanisms of vesicle budding and fusion. Cell 116,
153-166.
130
Bonifacino, J.S., and Lippincott-Schwartz, J. (2003). Coat proteins: shaping membrane transport.
Nat Rev Mol Cell Biol 4, 409-414.
Bose, I., Reese, A.J., Ory, J.J., Janbon, G., and Doering, T.L. (2003). A yeast under cover: the
capsule of Cryptococcus neoformans. Eukaryot Cell 2, 655-663.
Bowers, K., and Stevens, T.H. (2005). Protein transport from the late Golgi to the vacuole in the
yeast Saccharomyces cerevisiae. Biochim Biophys Acta 1744, 438-454.
Cantin, R., Diou, J., Belanger, D., Tremblay, A.M., and Gilbert, C. (2008). Discrimination between
exosomes and HIV-1: purification of both vesicles from cell-free supernatants. J Immunol
Methods 338, 21-30.
Casadevall, A.,Perfect, J.R. (1998). Cryptococcus neoformans, American Society for Microbiology,
Washington.
Chaffin, W.L., Lopez-Ribot, J.L., Casanova, M., Gozalbo, D., and Martinez, J.P. (1998). Cell wall and
secreted proteins of Candida albicans: identification, function, and expression. Microbiol Mol Biol
Rev 62, 130-180.
Chaka, W., Scharringa, J., Verheul, A.F., Verhoef, J., Van Strijp, A.G., and Hoepelman, I.M. (1995).
Quantitative analysis of phagocytosis and killing of Cryptococcus neoformans by human
peripheral blood mononuclear cells by flow cytometry. Clin Diagn Lab Immunol 2, 753-759.
Chang, Y.C., and Kwon-Chung, K.J. (1994). Complementation of a capsule-deficient mutation of
Cryptococcus neoformans restores its virulence. Mol Cell Biol 14, 4912-4919.
Chang, Y.C., and Kwon-Chung, K.J. (1998). Isolation of the third capsule-associated gene, CAP60,
required for virulence in Cryptococcus neoformans. Infect Immun 66, 2230-2236.
Chang, Y.C., and Kwon-Chung, K.J. (1999). Isolation, characterization, and localization of a
capsule-associated gene, CAP10, of Cryptococcus neoformans. J Bacteriol 181, 5636-5643.
131
Chang, Y.C., Penoyer, L.A., and Kwon-Chung, K.J. (1996). The second capsule gene of
cryptococcus neoformans, CAP64, is essential for virulence. Infect Immun 64, 1977-1983.
Charlier, C., Nielsen, K., Daou, S., Brigitte, M., Chretien, F., and Dromer, F. (2009). Evidence of a
role for monocytes in dissemination and brain invasion by Cryptococcus neoformans. Infect
Immun 77, 120-127.
Chayakulkeeree, M., and Perfect, J.R. (2006). Cryptococcosis. Infect Dis Clin North Am 20, 507-
544, v-vi.
Chen, L.C., Goldman, D.L., Doering, T.L., Pirofski, L., and Casadevall, A. (1999). Antibody response
to Cryptococcus neoformans proteins in rodents and humans. Infect Immun 67, 2218-2224.
Cherniak, R., Jones, R.G., and Slodki, M.E. (1988). Type-specific polysaccharides of Cryptococcus
neoformans. n.m.r.-spectral study of a glucuronomannan chemically derived from a Tremella
mesenterica exopolysaccharide. Carbohydr Res 182, 227-239.
Cherniak, R., and Sundstrom, J.B. (1994). Polysaccharide antigens of the capsule of Cryptococcus
neoformans. Infect Immun 62, 1507-1512.
Cianciotto, N.P. (2005). Type II secretion: a protein secretion system for all seasons. Trends
Microbiol 13, 581-588.
Cox, G.M., McDade, H.C., Chen, S.C., Tucker, S.C., Gottfredsson, M., Wright, L.C., Sorrell, T.C.,
Leidich, S.D., Casadevall, A., Ghannoum, M.A., et al. (2001). Extracellular phospholipase activity is
a virulence factor for Cryptococcus neoformans. Mol Microbiol 39, 166-175.
Craig, L., and Li, J. (2008). Type IV pili: paradoxes in form and function. Curr Opin Struct Biol 18,
267-277.
de Gassart, A., Geminard, C., Fevrier, B., Raposo, G., and Vidal, M. (2003). Lipid raft-associated
protein sorting in exosomes. Blood 102, 4336-4344.
132
de Souza Pereira, R., and Geibel, J. (1999). Direct observation of oxidative stress on the cell wall
of Saccharomyces cerevisiae strains with atomic force microscopy. Mol Cell Biochem 201, 17-24.
Del Poeta, M. (2004). Role of phagocytosis in the virulence of Cryptococcus neoformans.
Eukaryot Cell 3, 1067-1075.
Dessauer, A., Rother, U., and Rother, K. (1984). Freeze-thaw activation of the complement attack
phase: I. Separation of two steps in the formation of the active C--56 complex. Acta Pathol
Microbiol Immunol Scand Suppl 284, 75-81.
Diamond, R.D., and Bennett, J.E. (1973). Growth of Cryptococcus neoformans within human
macrophages in vitro. Infect Immun 7, 231-236.
Diamond, R.D., May, J.E., Kane, M.A., Frank, M.M., and Bennett, J.E. (1974). The role of the
classical and alternate complement pathways in host defenses against Cryptococcus neoformans
infection. J Immunol 112, 2260-2270.
Doering, T.L., Nosanchuk, J.D., Roberts, W.K., and Casadevall, A. (1999). Melanin as a potential
cryptococcal defence against microbicidal proteins. Med Mycol 37, 175-181.
Dong, Z.M., and Murphy, J.W. (1997). Cryptococcal polysaccharides bind to CD18 on human
neutrophils. Infect Immun 65, 557-563.
Driessen, A.J., Fekkes, P., and van der Wolk, J.P. (1998). The Sec system. Curr Opin Microbiol 1,
216-222.
Drubin, D.G., and Nelson, W.J. (1996). Origins of cell polarity. Cell 84, 335-344.
Duran, J.M., Anjard, C., Stefan, C., Loomis, W.F., and Malhotra, V. Unconventional secretion of
Acb1 is mediated by autophagosomes. J Cell Biol 188, 527-536.
Eisenman, H.C., Frases, S., Nicola, A.M., Rodrigues, M.L., and Casadevall, A. (2009). Vesicle-
associated melanization in Cryptococcus neoformans. Microbiology 155, 3860-3867.
133
Ellen, A.F., Albers, S.V., Huibers, W., Pitcher, A., Hobel, C.F., Schwarz, H., Folea, M., Schouten, S.,
Boekema, E.J., Poolman, B., et al. (2009). Proteomic analysis of secreted membrane vesicles of
archaeal Sulfolobus species reveals the presence of endosome sorting complex components.
Extremophiles 13, 67-79.
Eng, J.K., McCormack, A.L., Yates, J.R., 3rd (1994). An approach to correlate tandem mass spectral
data of peptides with amino acid sequences in a protein database. J Am Soc Mass Spectrom 5:
976989.
Esnault, K., el Moudni, B., Bouchara, J.P., Chabasse, D., and Tronchin, G. (1999). Association of a
myosin immunoanalogue with cell envelopes of Aspergillus fumigatus conidia and its
participation in swelling and germination. Infect Immun 67, 1238-1244.
Falkow, S. (1988). Molecular Koch's postulates applied to microbial pathogenicity. Rev Infect Dis
10 Suppl 2, S274-276.
Feldmesser, M., Kress, Y., and Casadevall, A. (2001). Dynamic changes in the morphology of
Cryptococcus neoformans during murine pulmonary infection. Microbiology 147, 2355-2365.
Feldmesser, M., Kress, Y., Novikoff, P., and Casadevall, A. (2000). Cryptococcus neoformans is a
facultative intracellular pathogen in murine pulmonary infection. Infect Immun 68, 4225-4237.
Feldmesser, M., Tucker, S., and Casadevall, A. (2001). Intracellular parasitism of macrophages by
Cryptococcus neoformans. Trends Microbiol 9, 273-278.
Flesch, I.E., Schwamberger, G., and Kaufmann, S.H. (1989). Fungicidal activity of IFN-gamma-
activated macrophages. Extracellular killing of Cryptococcus neoformans. J Immunol 142, 3219-
3224.
Fonseca, F.L., Nohara, L.L., Cordero, R.J., Frases, S., Casadevall, A., Almeida, I.C., Nimrichter, L.,
and Rodrigues, M.L. Immunomodulatory effects of serotype B glucuronoxylomannan from
Cryptococcus gattii correlate with polysaccharide diameter. Infect Immun 78, 3861-3870.
134
Franzot, S.P., Fries, B.C., Cleare, W., and Casadevall, A. (1998). Genetic relationship between
Cryptococcus neoformans var. neoformans strains of serotypes A and D. J Clin Microbiol 36,
2200-2204.
Frases, S., Nimrichter, L., Viana, N.B., Nakouzi, A., and Casadevall, A. (2008). Cryptococcus
neoformans capsular polysaccharide and exopolysaccharide fractions manifest physical,
chemical, and antigenic differences. Eukaryot Cell 7, 319-327.
French, N., Gray, K., Watera, C., Nakiyingi, J., Lugada, E., Moore, M., Lalloo, D., Whitworth, J.A.,
and Gilks, C.F. (2002). Cryptococcal infection in a cohort of HIV-1-infected Ugandan adults. AIDS
16, 1031-1038.
Garcia-Hermoso, D., Janbon, G., and Dromer, F. (1999). Epidemiological evidence for dormant
Cryptococcus neoformans infection. J Clin Microbiol 37, 3204-3209.
Garcia-Rivera, J., Chang, Y.C., Kwon-Chung, K.J., and Casadevall, A. (2004). Cryptococcus
neoformans CAP59 (or Cap59p) is involved in the extracellular trafficking of capsular
glucuronoxylomannan. Eukaryot Cell 3, 385-392.
Ghannoum, M.A. (1998). Extracellular phospholipases as universal virulence factor in pathogenic
fungi. Nippon Ishinkin Gakkai Zasshi 39, 55-59.
Ghannoum, M.A. (2000). Potential role of phospholipases in virulence and fungal pathogenesis.
Clin Microbiol Rev 13, 122-143, table of contents.
Giaever, G., Chu, A.M., Ni, L., Connelly, C., Riles, L., Veronneau, S., Dow, S., Lucau-Danila, A.,
Anderson, K., Andre, B., et al. (2002). Functional profiling of the Saccharomyces cerevisiae
genome. Nature 418, 387-391.
Goldman, D.L., Khine, H., Abadi, J., Lindenberg, D.J., Pirofski, L., Niang, R., and Casadevall, A.
(2001). Serologic evidence for Cryptococcus neoformans infection in early childhood. Pediatrics
107, E66.
Gomez, B.L., and Nosanchuk, J.D. (2003). Melanin and fungi. Curr Opin Infect Dis 16, 91-96.
135
Green, L.C., Wagner, D.A., Glogowski, J., Skipper, P.L., Wishnok, J.S., and Tannenbaum, S.R.
(1982). Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 126, 131-
138.
Hauser, A.R., Kang, P.J., and Engel, J.N. (1998). PepA, a secreted protein of Pseudomonas
aeruginosa, is necessary for cytotoxicity and virulence. Mol Microbiol 27, 807-818.
Hoag, K.A., Lipscomb, M.F., Izzo, A.A., and Street, N.E. (1997). IL-12 and IFN-gamma are required
for initiating the protective Th1 response to pulmonary cryptococcosis in resistant C.B-17 mice.
Am J Respir Cell Mol Biol 17, 733-739.
Hoegl, L., Ollert, M., and Korting, H.C. (1996). The role of Candida albicans secreted aspartic
proteinase in the development of candidoses. J Mol Med 74, 135-142.
Horwitz, M.A. (1983). The Legionnaires' disease bacterium (Legionella pneumophila) inhibits
phagosome-lysosome fusion in human monocytes. J Exp Med 158, 2108-2126.
Hu, G., and Kronstad, J.W. A putative P-type ATPase, Apt1, is involved in stress tolerance and
virulence in Cryptococcus neoformans. Eukaryot Cell 9, 74-83.
Hu, G., Steen, B.R., Lian, T., Sham, A.P., Tam, N., Tangen, K.L., and Kronstad, J.W. (2007).
Transcriptional regulation by protein kinase A in Cryptococcus neoformans. PLoS Pathog 3, e42.
Hube, B. (1996). Candida albicans secreted aspartyl proteinases. Curr Top Med Mycol 7, 55-69.
Hurley, J.H., and Emr, S.D. (2006). The ESCRT complexes: structure and mechanism of a
membrane-trafficking network. Annu Rev Biophys Biomol Struct 35, 277-298.
Hyun, T.K., Uddin, M.N., Rim, Y., and Kim, J.Y. Cell-to-cell trafficking of RNA and RNA silencing
through plasmodesmata. Protoplasma.
Ishihama, Y., Oda, Y., Tabata, T., Sato, T., Nagasu, T., Rappsilber, J., and Mann, M. (2005).
Exponentially modified protein abundance index (emPAI) for estimation of absolute protein
amount in proteomics by the number of sequenced peptides per protein. Mol Cell Proteomics 4,
1265-1272.
136
Jacobson, E.S., and Ikeda, R. (2005). Effect of melanization upon porosity of the cryptococcal cell
wall. Med Mycol 43, 327-333.
Jahn, R., Lang, T., and Sudhof, T.C. (2003). Membrane fusion. Cell 112, 519-533.
Jeffery, C.J. (1999). Moonlighting proteins. Trends Biochem Sci 24, 8-11.
Johnstone, R.M., Adam, M., and Pan, B.T. (1984). The fate of the transferrin receptor during
maturation of sheep reticulocytes in vitro. Can J Biochem Cell Biol 62, 1246-1254.
Kapteyn, J.C., Van Den Ende, H., and Klis, F.M. (1999). The contribution of cell wall proteins to the
organization of the yeast cell wall. Biochim Biophys Acta 1426, 373-383.
Katzmann, D.J., Sarkar, S., Chu, T., Audhya, A., and Emr, S.D. (2004). Multivesicular body sorting:
ubiquitin ligase Rsp5 is required for the modification and sorting of carboxypeptidase S. Mol Biol
Cell 15, 468-480.
Kawakami, K., Hossain Qureshi, M., Zhang, T., Koguchi, Y., Xie, Q., Kurimoto, M., and Saito, A.
(1999). Interleukin-4 weakens host resistance to pulmonary and disseminated cryptococcal
infection caused by combined treatment with interferon-gamma-inducing cytokines. Cell
Immunol 197, 55-61.
Kawakami, K., Kohno, S., Kadota, J., Tohyama, M., Teruya, K., Kudeken, N., Saito, A., and Hara, K.
(1995). T cell-dependent activation of macrophages and enhancement of their phagocytic activity
in the lungs of mice inoculated with heat-killed Cryptococcus neoformans: involvement of IFN-
gamma and its protective effect against cryptococcal infection. Microbiol Immunol 39, 135-143.
Kinseth, M.A., Anjard, C., Fuller, D., Guizzunti, G., Loomis, W.F., and Malhotra, V. (2007). The
Golgi-associated protein GRASP is required for unconventional protein secretion during
development. Cell 130, 524-534.
Kirchhausen, T. (2000). Three ways to make a vesicle. Nat Rev Mol Cell Biol 1, 187-198.
137
Kleinschek, M.A., Muller, U., Brodie, S.J., Stenzel, W., Kohler, G., Blumenschein, W.M.,
Straubinger, R.K., McClanahan, T., Kastelein, R.A., and Alber, G. (2006). IL-23 enhances the
inflammatory cell response in Cryptococcus neoformans infection and induces a cytokine pattern
distinct from IL-12. J Immunol 176, 1098-1106.
Klis, F.M., Mol, P., Hellingwerf, K., and Brul, S. (2002). Dynamics of cell wall structure in
Saccharomyces cerevisiae. FEMS Microbiol Rev 26, 239-256.
Klutts, J.S., Levery, S.B., and Doering, T.L. (2007). A beta-1,2-xylosyltransferase from Cryptococcus
neoformans defines a new family of glycosyltransferases. J Biol Chem 282, 17890-17899.
Kornfeld, S., and Mellman, I. (1989). The biogenesis of lysosomes. Annu Rev Cell Biol 5, 483-525.
Kozel, T.R., deJong, B.C., Grinsell, M.M., MacGill, R.S., and Wall, K.K. (1998). Characterization of
anticapsular monoclonal antibodies that regulate activation of the complement system by the
Cryptococcus neoformans capsule. Infect Immun 66, 1538-1546.
Kozel, T.R., MacGill, R.S., and Wall, K.K. (1998). Bivalency is required for anticapsular monoclonal
antibodies to optimally suppress activation of the alternative complement pathway by the
Cryptococcus neoformans capsule. Infect Immun 66, 1547-1553.
Kozel, T.R., and Pfrommer, G.S. (1986). Activation of the complement system by Cryptococcus
neoformans leads to binding of iC3b to the yeast. Infect Immun 52, 1-5.
Kuehn, M.J., and Kesty, N.C. (2005). Bacterial outer membrane vesicles and the host-pathogen
interaction. Genes Dev 19, 2645-2655.
Kwon-Chung, K. (1998). Gene disruption to evaluate the role of fungal candidate virulence genes.
Curr Opin Microbiol 1, 381-389.
Leal-Morales, C.A., Bracker, C.E., and Bartnicki-Garcia, S. (1994). Subcellular localization,
abundance and stability of chitin synthetases 1 and 2 from Saccharomyces cerevisiae.
Microbiology 140 ( Pt 9), 2207-2216.
138
Lee, E.Y., Choi, D.Y., Kim, D.K., Kim, J.W., Park, J.O., Kim, S., Kim, S.H., Desiderio, D.M., Kim, Y.K.,
Kim, K.P., et al. (2009). Gram-positive bacteria produce membrane vesicles: proteomics-based
characterization of Staphylococcus aureus-derived membrane vesicles. Proteomics 9, 5425-5436.
Letourneur, F., Gaynor, E.C., Hennecke, S., Demolliere, C., Duden, R., Emr, S.D., Riezman, H., and
Cosson, P. (1994). Coatomer is essential for retrieval of dilysine-tagged proteins to the
endoplasmic reticulum. Cell 79, 1199-1207.
Levin, D.E. (2005). Cell wall integrity signaling in Saccharomyces cerevisiae. Microbiol Mol Biol
Rev 69, 262-291.
Levitz, S.M., Dupont, M.P., and Smail, E.H. (1994). Direct activity of human T lymphocytes and
natural killer cells against Cryptococcus neoformans. Infect Immun 62, 194-202.
Levitz, S.M., Nong, S.H., Seetoo, K.F., Harrison, T.S., Speizer, R.A., and Simons, E.R. (1999).
Cryptococcus neoformans resides in an acidic phagolysosome of human macrophages. Infect
Immun 67, 885-890.
Levitz, S.M., Tabuni, A., Kozel, T.R., MacGill, R.S., Ingalls, R.R., and Golenbock, D.T. (1997). Binding
of Cryptococcus neoformans to heterologously expressed human complement receptors. Infect
Immun 65, 931-935.
Littman, M.L. (1958). Capsule synthesis by Cryptococcus neoformans. Trans N Y Acad Sci 20, 623-
648.
Liu, H., Sadygov, R.G., and Yates, J.R., 3rd (2004). A model for random sampling and estimation of
relative protein abundance in shotgun proteomics. Anal Chem 76, 4193-4201.
Lupo, P., Chang, Y.C., Kelsall, B.L., Farber, J.M., Pietrella, D., Vecchiarelli, A., Leon, F., and Kwon-
Chung, K.J. (2008). The presence of capsule in Cryptococcus neoformans influences the gene
expression profile in dendritic cells during interaction with the fungus. Infect Immun 76, 1581-
1589.
Ma, H., Croudace, J.E., Lammas, D.A., and May, R.C. (2006). Expulsion of live pathogenic yeast by
macrophages. Curr Biol 16, 2156-2160.
139
Makarova, K.S., Yutin, N., Bell, S.D., and Koonin, E.V. Evolution of diverse cell division and vesicle
formation systems in Archaea. Nat Rev Microbiol 8, 731-741.
Manjithaya, R., Anjard, C., Loomis, W.F., and Subramani, S. Unconventional secretion of Pichia
pastoris Acb1 is dependent on GRASP protein, peroxisomal functions, and autophagosome
formation. J Cell Biol 188, 537-546.
Marzesco, A.M., Janich, P., Wilsch-Brauninger, M., Dubreuil, V., Langenfeld, K., Corbeil, D., and
Huttner, W.B. (2005). Release of extracellular membrane particles carrying the stem cell marker
prominin-1 (CD133) from neural progenitors and other epithelial cells. J Cell Sci 118, 2849-2858.
Mayor, S., and Riezman, H. (2004). Sorting GPI-anchored proteins. Nat Rev Mol Cell Biol 5, 110-
120.
McFadden, D.C., De Jesus, M., and Casadevall, A. (2006). The physical properties of the capsular
polysaccharides from Cryptococcus neoformans suggest features for capsule construction. J Biol
Chem 281, 1868-1875.
McGrath, J.P., and Varshavsky, A. (1989). The yeast STE6 gene encodes a homologue of the
mammalian multidrug resistance P-glycoprotein. Nature 340, 400-404.
Mednick, A.J., Feldmesser, M., Rivera, J., and Casadevall, A. (2003). Neutropenia alters lung
cytokine production in mice and reduces their susceptibility to pulmonary cryptococcosis. Eur J
Immunol 33, 1744-1753.
Milam, J.E., Herring-Palmer, A.C., Pandrangi, R., McDonald, R.A., Huffnagle, G.B., and Toews, G.B.
(2007). Modulation of the pulmonary type 2 T-cell response to Cryptococcus neoformans by
intratracheal delivery of a tumor necrosis factor alpha-expressing adenoviral vector. Infect
Immun 75, 4951-4958.
Miller, M.F., and Mitchell, T.G. (1991). Killing of Cryptococcus neoformans strains by human
neutrophils and monocytes. Infect Immun 59, 24-28.
Monari, C., Bevilacqua, S., Piccioni, M., Pericolini, E., Perito, S., Calvitti, M., Bistoni, F., Kozel, T.R.,
and Vecchiarelli, A. (2009). A microbial polysaccharide reduces the severity of rheumatoid
140
arthritis by influencing Th17 differentiation and proinflammatory cytokines production. J
Immunol 183, 191-200.
Monari, C., Bistoni, F., and Vecchiarelli, A. (2006). Glucuronoxylomannan exhibits potent
immunosuppressive properties. FEMS Yeast Res 6, 537-542.
Mukherjee, J., Nussbaum, G., Scharff, M.D., and Casadevall, A. (1995). Protective and
nonprotective monoclonal antibodies to Cryptococcus neoformans originating from one B cell. J
Exp Med 181, 405-409.
Muller, U., Stenzel, W., Kohler, G., Werner, C., Polte, T., Hansen, G., Schutze, N., Straubinger,
R.K., Blessing, M., McKenzie, A.N., et al. (2007). IL-13 induces disease-promoting type 2
cytokines, alternatively activated macrophages and allergic inflammation during pulmonary
infection of mice with Cryptococcus neoformans. J Immunol 179, 5367-5377.
Nickel, W., and Rabouille, C. (2009). Mechanisms of regulated unconventional protein secretion.
Nat Rev Mol Cell Biol 10, 148-155.
Nimrichter, L., Frases, S., Cinelli, L.P., Viana, N.B., Nakouzi, A., Travassos, L.R., Casadevall, A., and
Rodrigues, M.L. (2007). Self-aggregation of Cryptococcus neoformans capsular
glucuronoxylomannan is dependent on divalent cations. Eukaryot Cell 6, 1400-1410.
Nimrichter, L., Rodrigues, M.L., Rodrigues, E.G., and Travassos, L.R. (2005). The multitude of
targets for the immune system and drug therapy in the fungal cell wall. Microbes Infect 7, 789-
798.
Nosanchuk, J.D., Steenbergen, J.N., Shi, L., Deepe, G.S., Jr., and Casadevall, A. (2003). Antibodies
to a cell surface histone-like protein protect against Histoplasma capsulatum. J Clin Invest 112,
1164-1175.
Novick, P., Field, C., and Schekman, R. (1980). Identification of 23 complementation groups
required for post-translational events in the yeast secretory pathway. Cell 21, 205-215.
141
Novick, P., and Schekman, R. (1979). Secretion and cell-surface growth are blocked in a
temperature-sensitive mutant of Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 76, 1858-
1862.
Novick, P., and Schekman, R. (1983). Export of major cell surface proteins is blocked in yeast
secretory mutants. J Cell Biol 96, 541-547.
Oberholzer, U., Marcil, A., Leberer, E., Thomas, D.Y., and Whiteway, M. (2002). Myosin I is
required for hypha formation in Candida albicans. Eukaryot Cell 1, 213-228.
Ortiz, D., and Novick, P.J. (2006). Ypt32p regulates the translocation of Chs3p from an internal
pool to the plasma membrane. Eur J Cell Biol 85, 107-116.
Osterholzer, J.J., Milam, J.E., Chen, G.H., Toews, G.B., Huffnagle, G.B., and Olszewski, M.A. (2009).
Role of dendritic cells and alveolar macrophages in regulating early host defense against
pulmonary infection with Cryptococcus neoformans. Infect Immun 77, 3749-3758.
Palade, G. (1975). Intracellular aspects of the process of protein synthesis. Science 189, 347-358.
Pan, B.T., Teng, K., Wu, C., Adam, M., and Johnstone, R.M. (1985). Electron microscopic evidence
for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol
101, 942-948.
Panepinto, J., Komperda, K., Frases, S., Park, Y.D., Djordjevic, J.T., Casadevall, A., and Williamson,
P.R. (2009). Sec6-dependent sorting of fungal extracellular exosomes and laccase of
Cryptococcus neoformans. Mol Microbiol 71, 1165-1176.
Park, B.J., Wannemuehler, K.A., Marston, B.J., Govender, N., Pappas, P.G., and Chiller, T.M.
(2009). Estimation of the current global burden of cryptococcal meningitis among persons living
with HIV/AIDS. AIDS 23, 525-530.
Pierini, L.M., and Doering, T.L. (2001). Spatial and temporal sequence of capsule construction in
Cryptococcus neoformans. Mol Microbiol 41, 105-115.
142
Pietrella, D., Corbucci, C., Perito, S., Bistoni, G., and Vecchiarelli, A. (2005). Mannoproteins from
Cryptococcus neoformans promote dendritic cell maturation and activation. Infect Immun 73,
820-827.
Poisson, G., Chauve, C., Chen, X., and Bergeron, A. (2007). FragAnchor: a large-scale predictor of
glycosylphosphatidylinositol anchors in eukaryote protein sequences by qualitative scoring.
Genomics Proteomics Bioinformatics 5, 121-130.
Polacheck, I. (1991). The discovery of melanin production in Cryptococcus neoformans and its
impact on diagnosis and the study of virulence. Zentralbl Bakteriol 276, 120-123.
Ponnambalam, S., and Baldwin, S.A. (2003). Constitutive protein secretion from the trans-Golgi
network to the plasma membrane. Mol Membr Biol 20, 129-139.
Qu, Y., Franchi, L., Nunez, G., and Dubyak, G.R. (2007). Nonclassical IL-1 beta secretion stimulated
by P2X7 receptors is dependent on inflammasome activation and correlated with exosome
release in murine macrophages. J Immunol 179, 1913-1925.
Raposo, G., Nijman, H.W., Stoorvogel, W., Liejendekker, R., Harding, C.V., Melief, C.J., and Geuze,
H.J. (1996). B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183, 1161-1172.
Rawat, D.S., Upreti, H.B., and Das, S.K. (1984). Lipid composition of Cryptococcus neoformans.
Microbiologica 7, 299-307.
Regente, M., Corti-Monzon, G., Maldonado, A.M., Pinedo, M., Jorrin, J., and de la Canal, L.
(2009). Vesicular fractions of sunflower apoplastic fluids are associated with potential exosome
marker proteins. FEBS Lett 583, 3363-3366.
Rhodes, J.C. (1985). Contribution of complement component C5 to the pathogenesis of
experimental murine cryptococcosis. Sabouraudia 23, 225-234.
Rhodes, J.C., Wicker, L.S., and Urba, W.J. (1980). Genetic control of susceptibility to Cryptococcus
neoformans in mice. Infect Immun 29, 494-499.
Rice, L.M., and Brunger, A.T. (1999). Crystal structure of the vesicular transport protein Sec17:
implications for SNAP function in SNARE complex disassembly. Mol Cell 4, 85-95.
143
Rittershaus, P.C., Kechichian, T.B., Allegood, J.C., Merrill, A.H., Jr., Hennig, M., Luberto, C., and
Del Poeta, M. (2006). Glucosylceramide synthase is an essential regulator of pathogenicity of
Cryptococcus neoformans. J Clin Invest 116, 1651-1659.
Rivera, J., Cordero, R.J., Nakouzi, A.S., Frases, S., Nicola, A., and Casadevall, A. Bacillus anthracis
produces membrane-derived vesicles containing biologically active toxins. Proc Natl Acad Sci U S
A 107, 19002-19007.
Rivera, J., Feldmesser, M., Cammer, M., and Casadevall, A. (1998). Organ-dependent variation of
capsule thickness in Cryptococcus neoformans during experimental murine infection. Infect
Immun 66, 5027-5030.
Rodrigues, M.L., Nakayasu, E.S., Oliveira, D.L., Nimrichter, L., Nosanchuk, J.D., Almeida, I.C., and
Casadevall, A. (2008). Extracellular vesicles produced by Cryptococcus neoformans contain
protein components associated with virulence. Eukaryot Cell 7, 58-67.
Rodrigues, M.L., Shi, L., Barreto-Bergter, E., Nimrichter, L., Farias, S.E., Rodrigues, E.G., Travassos,
L.R., and Nosanchuk, J.D. (2007). Monoclonal antibody to fungal glucosylceramide protects mice
against lethal Cryptococcus neoformans infection. Clin Vaccine Immunol 14, 1372-1376.
Rodrigues, M.L., Travassos, L.R., Miranda, K.R., Franzen, A.J., Rozental, S., de Souza, W., Alviano,
C.S., and Barreto-Bergter, E. (2000). Human antibodies against a purified glucosylceramide from
Cryptococcus neoformans inhibit cell budding and fungal growth. Infect Immun 68, 7049-7060.
Rothman, J.E. (1994). Mechanisms of intracellular protein transport. Nature 372, 55-63.
Sakaguchi, N., Baba, T., Fukuzawa, M., and Ohno, S. (1993). Ultrastructural study of Cryptococcus
neoformans by quick-freezing and deep-etching method. Mycopathologia 121, 133-141.
Schekman, R. (2002). Lasker Basic Medical Research Award. SEC mutants and the secretory
apparatus. Nat Med 8, 1055-1058.
144
Schekman, R., Esmon, B., Ferro-Novick, S., Field, C., and Novick, P. (1983). Yeast secretory
mutants: isolation and characterization. Methods Enzymol 96, 802-815.
Schorey, J.S., and Bhatnagar, S. (2008). Exosome function: from tumor immunology to pathogen
biology. Traffic 9, 871-881.
Segev, N. (2001). Ypt/rab gtpases: regulators of protein trafficking. Sci STKE 2001, re11.
Shapiro, S., Beenhouwer, D.O., Feldmesser, M., Taborda, C., Carroll, M.C., Casadevall, A., and
Scharff, M.D. (2002). Immunoglobulin G monoclonal antibodies to Cryptococcus neoformans
protect mice deficient in complement component C3. Infect Immun 70, 2598-2604.
Shoham, S., Huang, C., Chen, J.M., Golenbock, D.T., and Levitz, S.M. (2001). Toll-like receptor 4
mediates intracellular signaling without TNF-alpha release in response to Cryptococcus
neoformans polysaccharide capsule. J Immunol 166, 4620-4626.
Shorter, J., Watson, R., Giannakou, M.E., Clarke, M., Warren, G., and Barr, F.A. (1999). GRASP55,
a second mammalian GRASP protein involved in the stacking of Golgi cisternae in a cell-free
system. EMBO J 18, 4949-4960.
Simons, M., and Raposo, G. (2009). Exosomes--vesicular carriers for intercellular communication.
Curr Opin Cell Biol 21, 575-581.
Singh, N., Alexander, B.D., Lortholary, O., Dromer, F., Gupta, K.L., John, G.T., del Busto, R.,
Klintmalm, G.B., Somani, J., Lyon, G.M., et al. (2008). Pulmonary cryptococcosis in solid organ
transplant recipients: clinical relevance of serum cryptococcal antigen. Clin Infect Dis 46, e12-18.
Soler, N., Marguet, E., Verbavatz, J.M., and Forterre, P. (2008). Virus-like vesicles and
extracellular DNA produced by hyperthermophilic archaea of the order Thermococcales. Res
Microbiol 159, 390-399.
Sommer, U., Liu, H., and Doering, T.L. (2003). An alpha-1,3-mannosyltransferase of Cryptococcus
neoformans. J Biol Chem 278, 47724-47730.
Southwick, F.S., and Purich, D.L. (1996). Intracellular pathogenesis of listeriosis. N Engl J Med 334,
770-776.
145
Steenbergen, J.N., and Casadevall, A. (2003). The origin and maintenance of virulence for the
human pathogenic fungus Cryptococcus neoformans. Microbes Infect 5, 667-675.
Stephens, D.J., and Pepperkok, R. (2001). Illuminating the secretory pathway: when do we need
vesicles? J Cell Sci 114, 1053-1059.
Strasser, J.E., Newman, S.L., Ciraolo, G.M., Morris, R.E., Howell, M.L., and Dean, G.E. (1999).
Regulation of the macrophage vacuolar ATPase and phagosome-lysosome fusion by Histoplasma
capsulatum. J Immunol 162, 6148-6154.
Sturgill-Koszycki, S., Schlesinger, P.H., Chakraborty, P., Haddix, P.L., Collins, H.L., Fok, A.K., Allen,
R.D., Gluck, S.L., Heuser, J., and Russell, D.G. (1994). Lack of acidification in Mycobacterium
phagosomes produced by exclusion of the vesicular proton-ATPase. Science 263, 678-681.
Subra, C., Laulagnier, K., Perret, B., and Record, M. (2007). Exosome lipidomics unravels lipid
sorting at the level of multivesicular bodies. Biochimie 89, 205-212.
Sutton, R.B., Fasshauer, D., Jahn, R., and Brunger, A.T. (1998). Crystal structure of a SNARE
complex involved in synaptic exocytosis at 2.4 A resolution. Nature 395, 347-353.
Tokuyasu, K.T. (1973). A technique for ultracryotomy of cell suspensions and tissues. J Cell Biol
57, 551-565.
Tu, J., Vallier, L.G., and Carlson, M. (1993). Molecular and genetic analysis of the SNF7 gene in
Saccharomyces cerevisiae. Genetics 135, 17-23.
Tucker, S.C., and Casadevall, A. (2002). Replication of Cryptococcus neoformans in macrophages
is accompanied by phagosomal permeabilization and accumulation of vesicles containing
polysaccharide in the cytoplasm. Proc Natl Acad Sci U S A 99, 3165-3170.
Vaishnav, V.V., Bacon, B.E., O'Neill, M., and Cherniak, R. (1998). Structural characterization of the
galactoxylomannan of Cryptococcus neoformans Cap67. Carbohydr Res 306, 315-330.
146
Valadi, H., Ekstrom, K., Bossios, A., Sjostrand, M., Lee, J.J., and Lotvall, J.O. (2007). Exosome-
mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between
cells. Nat Cell Biol 9, 654-659.
Valdivia, R.H., and Schekman, R. (2003). The yeasts Rho1p and Pkc1p regulate the transport of
chitin synthase III (Chs3p) from internal stores to the plasma membrane. Proc Natl Acad Sci U S A
100, 10287-10292.
Vecchiarelli, A. (2000). Immunoregulation by capsular components of Cryptococcus neoformans.
Med Mycol 38, 407-417.
Villena, S.N., Pinheiro, R.O., Pinheiro, C.S., Nunes, M.P., Takiya, C.M., DosReis, G.A., Previato, J.O.,
Mendonca-Previato, L., and Freire-de-Lima, C.G. (2008). Capsular polysaccharides
galactoxylomannan and glucuronoxylomannan from Cryptococcus neoformans induce
macrophage apoptosis mediated by Fas ligand. Cell Microbiol 10, 1274-1285.
Voelz, K., and May, R.C. Cryptococcal interactions with the host immune system. Eukaryot Cell 9,
835-846.
Waterman, S.R., Hacham, M., Panepinto, J., Hu, G., Shin, S., and Williamson, P.R. (2007). Cell wall
targeting of laccase of Cryptococcus neoformans during infection of mice. Infect Immun 75, 714-
722.
Waters, M.G., Serafini, T., and Rothman, J.E. (1991). 'Coatomer': a cytosolic protein complex
containing subunits of non-clathrin-coated Golgi transport vesicles. Nature 349, 248-251.
Weber, T., Zemelman, B.V., McNew, J.A., Westermann, B., Gmachl, M., Parlati, F., Sollner, T.H.,
and Rothman, J.E. (1998). SNAREpins: minimal machinery for membrane fusion. Cell 92, 759-772.
Williamson, P.R. (1997). Laccase and melanin in the pathogenesis of Cryptococcus neoformans.
Front Biosci 2, e99-107.
Winzeler, E.A., Shoemaker, D.D., Astromoff, A., Liang, H., Anderson, K., Andre, B., Bangham, R.,
Benito, R., Boeke, J.D., Bussey, H., et al. (1999). Functional characterization of the S. cerevisiae
genome by gene deletion and parallel analysis. Science 285, 901-906.
147
Wolfers, J., Lozier, A., Raposo, G., Regnault, A., Thery, C., Masurier, C., Flament, C., Pouzieux, S.,
Faure, F., Tursz, T., et al. (2001). Tumor-derived exosomes are a source of shared tumor rejection
antigens for CTL cross-priming. Nat Med 7, 297-303.
Wuestehube, L.J., Duden, R., Eun, A., Hamamoto, S., Korn, P., Ram, R., and Schekman, R. (1996).
New mutants of Saccharomyces cerevisiae affected in the transport of proteins from the
endoplasmic reticulum to the Golgi complex. Genetics 142, 393-406.
Yamaguchi, M., Ohkusu, M., Sameshima, M., and Kawamoto, S. (2005). Safe specimen
preparation for electron microscopy of pathogenic fungi by freeze-substitution after
glutaraldehyde fixation. Nippon Ishinkin Gakkai Zasshi 46, 187-192.
Yeo, S.C., Xu, L., Ren, J., Boulton, V.J., Wagle, M.D., Liu, C., Ren, G., Wong, P., Zahn, R., Sasajala,
P., et al. (2003). Vps20p and Vta1p interact with Vps4p and function in multivesicular body
sorting and endosomal transport in Saccharomyces cerevisiae. J Cell Sci 116, 3957-3970.
Yoneda, A., and Doering, T.L. (2006). A eukaryotic capsular polysaccharide is synthesized
intracellularly and secreted via exocytosis. Mol Biol Cell 17, 5131-5140.
Yonezawa, H., Osaki, T., Kurata, S., Fukuda, M., Kawakami, H., Ochiai, K., Hanawa, T., and Kamiya,
S. (2009). Outer membrane vesicles of Helicobacter pylori TK1402 are involved in biofilm
formation. BMC Microbiol 9, 197.
Zaragoza, O., and Casadevall, A. (2004). Experimental modulation of capsule size in Cryptococcus
neoformans. Biol Proced Online 6, 10-15.
Zaragoza, O., Chrisman, C.J., Castelli, M.V., Frases, S., Cuenca-Estrella, M., Rodriguez-Tudela, J.L.,
and Casadevall, A. (2008). Capsule enlargement in Cryptococcus neoformans confers resistance
to oxidative stress suggesting a mechanism for intracellular survival. Cell Microbiol 10, 2043-
2057.
Zaragoza, O., Rodrigues, M.L., De Jesus, M., Frases, S., Dadachova, E., and Casadevall, A. (2009).
The capsule of the fungal pathogen Cryptococcus neoformans. Adv Appl Microbiol 68, 133-216.
148
Zaragoza, O., Telzak, A., Bryan, R.A., Dadachova, E., and Casadevall, A. (2006). The polysaccharide
capsule of the pathogenic fungus Cryptococcus neoformans enlarges by distal growth and is
rearranged during budding. Mol Microbiol 59, 67-83.
Zitvogel, L., Regnault, A., Lozier, A., Wolfers, J., Flament, C., Tenza, D., Ricciardi-Castagnoli, P.,
Raposo, G., and Amigorena, S. (1998). Eradication of established murine tumors using a novel
cell-free vaccine: dendritic cell-derived exosomes. Nat Med 4, 594-600.
149
Anexo 5: Esta seção reune os demais artigos nos quais participei como autora
no decorrer do meu doutorado.
EUKARYOTIC CELL, Jan. 2008, p. 58–67 Vol. 7, No. 1
1535-9778/08/$08.00ϩ0 doi:10.1128/EC.00370-07
Copyright © 2008, American Society for Microbiology. All Rights Reserved.
Extracellular Vesicles Produced by Cryptococcus neoformans Contain
Protein Components Associated with Virulence
Marcio L. Rodrigues,
1
* Ernesto S. Nakayasu,
2
Debora L. Oliveira,
1
Leonardo Nimrichter,
1
Joshua D. Nosanchuk,
3,4
Igor C. Almeida,
2
and Arturo Casadevall
3,4
*
Laborato´rio de Estudos Integrados em Bioquı´mica Microbiana, Departamento de Microbiologia Geral, Instituto de
Microbiologia Professor Paulo de Go´es, Universidade Federal do Rio de Janeiro, CEP 21941590 Rio de Janeiro, Brazil
1
;
Department of Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso,
El Paso, Texas 79968-0519
2
; and Department of Microbiology and Immunology
3
and Division of
Infectious Diseases,
4
Department of Medicine, Albert Einstein College of Medicine,
1300 Morris Park Ave., Bronx, New York 10461
Received 8 October 2007/Accepted 6 November 2007
Cryptococcus neoformans produces vesicles containing its major virulence factor, the capsular polysaccharide
glucuronoxylomannan (GXM). These vesicles cross the cell wall to reach the extracellular space, where the
polysaccharide is supposedly used for capsule growth or delivered into host tissues. In the present study, we
characterized vesicle morphology and protein composition by a combination of techniques including electron
microscopy, proteomics, enzymatic activity, and serological reactivity. Secretory vesicles in C. neoformans
appear to be correlated with exosome-like compartments derived from multivesicular bodies. Extracellular
vesicles manifested various sizes and morphologies, including electron-lucid membrane bodies and electron-
dense vesicles. Seventy-six proteins were identified by proteomic analysis, including several related to virulence
and protection against oxidative stress. Biochemical tests indicated laccase and urease activities in vesicles. In
addition, different vesicle proteins were recognized by sera from patients with cryptococcosis. These results
reveal an efficient and general mechanism of secretion of pathogenesis-related molecules in C. neoformans,
suggesting that extracellular vesicles function as “virulence bags” that deliver a concentrated payload of fungal
products to host effector cells and tissues.
Cryptococcus neoformans is the causative agent of cryptococ-
cosis and a major pathogen for immunosuppressed individuals
(39). Cryptococcosis is rampant in Africa and Asia in associa-
tion with human immunodeficiency virus infection, where it is
associated with high morbidity and mortality (5). C. neofor-
mans provides a unique model in cell biology studies because
it is the only eukaryotic pathogen with a polysaccharide cap-
sule, a structure that is essential for virulence (26, 32, 39). The
major capsular polysaccharide, glucuronoxylomannan (GXM),
is released extracellularly during infection and induces a num-
ber of deleterious effects to the host, including interference
with phagocytosis, inhibition of leukocyte migration to infected
tissues, and modulation of cytokine production (reviewed in
reference 32). It also represents a potential component for
vaccines (13) and is the target of potentially therapeutic anti-
bodies (7).
The mechanisms of secretion of macromolecules by fungal
cells have not been well elucidated. Fungal cells are encased in
a rigid, pore-containing cell wall consisting of polysaccharides,
proteins, and pigments (14, 35). Different studies demonstrate
that structures with molecular masses higher than 1,000 kDa
can cross the cell wall and reach the extracellular milieu (26,
27, 47). GXM, for instance, has an average molecular mass
ranging from 1.7 ϫ 10
6
to 7 ϫ 10
6
Da (27). Several studies by
our group and others indicate that C. neoformans synthesizes
GXM intracellularly and then transports the polysaccharide to
the extracellular space for assembly into a capsule (17, 20, 45,
54, 55). The mechanism by which capsular polysaccharide is
synthesized inside the cell and exported to the extracellular
environment for capsule assembly and release is a central ques-
tion in cryptococcal cell biology.
We have recently described how GXM-containing vesicles
accumulate in supernatants of C. neoformans cultures (45).
These extracellular vesicles contain bilayered membranes en-
riched with key fungal lipids, such as glucosylceramide and
sterols. This observation led to the proposal that extracellular
export of GXM is accomplished by vesicular transport (45).
The existence of a vesicular transport mechanism raises the
possibility that other fungal molecules could be released using
the same cellular apparatus. Indeed, a secretion mutant of C.
neoformans that accumulates secretory vesicles in the cyto-
plasm has severe defects in protein secretion (54). We there-
fore hypothesized that vesicular extracellular secretion in C.
neoformans is a general mechanism used for the trans-cell-wall
transport of protein, lipid, and carbohydrate components to
the extracellular environment.
In the present work, we used microscopic, serological, bio-
chemical, and proteomic approaches to analyze the extracel-
lular vesicles of C. neoformans. Electron microscopy suggests
* Corresponding author. Mailing address for M. L. Rodrigues: In-
stituto de Microbiologia Professor Paulo de Go´es, Universidade Fed-
eral do Rio de Janeiro, 21941590 Rio de Janeiro, Brazil. Phone: 55 21
2562 6740. Fax: 55 21 25608344. E-mail: [email protected].
Mailing address for A. Casadevall: Department of Microbiology and
Immunology, Albert Einstein College of Medicine, 1300 Morris Park
Avenue, Bronx, NY 10461. Phone: (718) 430-2215. Fax: (718) 430-8968.
† Supplemental material for this article may be found at http://ec
.asm.org/.
Published ahead of print on 26 November 2007.
58
that vesicle secretion derives from the traffic of multivesicle-
like compartments to the cell surface. The results indicate that
C. neoformans extracellular vesicles represent a heterogeneous
population of “virulence bags” containing numerous molecules
associated with fungal survival and host pathogenicity.
MATERIALS AND METHODS
Strains. The C. neoformans isolates used in this study included strains H99
(serotype A, wild type), Cap 67 (derived from serotype D and lacking a GXM
capsule), and 2E-TU and 2E-TUC (serotype D, LAC1 gene deletion and recon-
stituted laccase mutants) (46). Yeast cells were cultivated in a minimal medium
composed of dextrose (15 mM), MgSO
4
(10 mM), KH
2
PO
4
(29.4 mM), glycine
(13 mM), and thiamine-HCl (3 M). Fungal cells were cultivated for 48 h at
30°C, with continuous shaking. Strain Cap 67 was used for electron microscopy.
Strains 2E-TU and 2E-TUC were used for laccase assays. Strain H99 was used
for all other experiments. Proteomic analyses were performed using strains Cap
67, H99, and 2E-TUC.
Vesicle purification. Isolation of extracellular vesicles was done using the
protocol described by Rodrigues et al. (45). Briefly, cell-free culture supernatants
were obtained by sequential centrifugation at 4,000 and 15,000 ϫ g (15 min, 4°C).
These supernatants contained vesicles and were concentrated by approximately
20-fold using an Amicon ultrafiltration system (cutoff of 100 kDa). The concen-
trate was again centrifuged at 4,000 and 15,000 ϫ g (15 min, 4°C) and then at
100,000 ϫ g for1hat4°C. The supernatants were discarded, and pellets were
washed by five sequential suspension and centrifugation steps, each consisting of
100,000 ϫ g for1hat4°Cwith 0.1 M Tris-buffered saline. To remove extrave-
sicular GXM contamination, vesicles were subjected to passage through a col-
umn packed with cyanogen bromide-activated Sepharose coupled to a monoclo-
nal antibody to GXM, as described previously (45). Fractions that were not
bound to the monclonal antibody-containing column were again centrifuged at
100,000 ϫ g. The resulting pellets were then suspended in fixative solution for
electron microscopy analysis or prepared for biochemical, proteomic, and West-
ern blotting analyses, as described below.
TEM. For transmission electron microscopy (TEM), C. neoformans cells were
grown in minimal medium, serially washed in phosphate-buffered saline (PBS),
and fixed for1hin0.1Msodium cacodylate buffer (pH 7.2) containing 4%
paraformaldehyde and 2% glutaraldehyde. Cells were then infiltrated for 2 h in
a solution containing 25% polyvinylpyrrolidone and 2.1 M sucrose and then
rapidly frozen by immersion in liquid nitrogen. They were transferred to a
cryo-ultramicrotome (Ultracut; Reichert), and cryosections were obtained in a
temperature range of Ϫ70 to Ϫ90°C. The material was collected with a sucrose
loop and transferred to Formvar-carbon-coated grids. Specimens were observed
in a Zeiss 900 transmission electron microscope operating at 80 kV.
Pellets obtained after centrifugation of cell-free supernatants at 100,000 ϫ g
were fixed with 2% glutaraldehyde in 0.1 M cacodylate at room temperature for
2 h and then incubated overnight in 4% formaldehyde, 1% glutaraldehyde, and
0.1% PBS. The samples were incubated for 90 min in 2% osmium, serially
dehydrated in ethanol, and embedded in Spurrs epoxy resin. Thin sections were
obtained on a Reichart Ultracut UCT and stained with 0.5% uranyl acetate and
0.5% lead citrate. Samples were observed in a JEOL 1200EX transmission
electron microscope operating at 80 kV.
Biochemical detection of enzymatic activities. Laccase and urease activity in
vesicle preparations was assayed spectrophotometrically. Acid phosphatase, a
cryptococcal protein that mediates adhesion of cryptococci to epithelial cells and
whose secretion has been associated with vesicle production (8, 54), was also
assayed. Pellets obtained after centrifugation at 100,000 ϫ g were suspended in
PBS and serially diluted in media appropriate for the reactions catalyzed by
laccase, urease, or acid phosphatase. The laccase reaction medium corresponded
to 0.2% (10 mM)
L-DOPA in PBS, while the medium for urease activity con-
tained 4% urea, 0.02% yeast extract, 0.002% phenol red, 0.273% KH
2
PO
4
, and
0.285% Na
2
HPO-
4
. For phosphatase determination, the reaction medium con
-
sisted of acetate buffer (pH 5.0) supplemented with 5 mg/ml p-nitrophenyl
phosphate. Vesicle suspensions were incubated overnight at room temperature
and protected from the light. Reactions were quantified by reading at 450
(laccase), 405 (phosphatase), or 540 (urease) nm with a Multiscan mass spec-
trometer (MS) (Labsystem, Helsinki, Finland). The amount of vesicles in each
system was assumed to be related to the protein concentration in each vesicular
suspension. The protein concentration at the starting dilution in each system
corresponded to 0.3 g/ml. Enzymatic assays were repeated at least three times,
producing similar results.
Western blot analysis. Vesicles were suspended in loading buffer (1% so-
dium dodecyl sulfate [SDS], 10% glycerol, 10 mM Tris-Cl [pH 6.8], 1 mM
2-mercaptoethanol, and 0.05 mg/ml bromophenol blue) and a final amount of
proteins corresponding to 8 g was loaded onto 12% SDS-polyacrylamide gel
electrophoresis (PAGE) gel. Separated proteins were transferred to nitrocel-
lulose membranes, which were sequentially blocked in PBS containing 1%
bovine serum albumin and incubated for1hatroom temperature with pooled
sera from 10 individuals diagnosed with cryptococcosis based on positive
serum reactivity in latex agglutination tests for cryptococcal GXM. Alterna-
tively, the membranes were incubated with pooled normal human sera from
healthy volunteers with no previous diagnosis of any systemic mycosis. Pooled
sera were used at a 1:100 dilution in PBS-bovine serum albumin. After
extensive washing, the membranes were incubated in the presence of a per-
oxidase-labeled anti-human immunoglobulin antibody followed by immuno-
detection by chemiluminescence (Pierce). To exclude the possibility of non-
specific recognition of samples by secondary antibodies, samples were
incubated directly with the peroxidase-labeled antihuman antibody, produc-
ing negative results (not shown). Serological analyses were repeated twice,
with similar results.
Protein identification by liquid chromatography-tandem mass spectrometry.
Purified vesicles were suspended in 400 mM NH
4
HCO
3
(40 l) containing 8 M
urea, and 50 mM dithiothreitol (10 l) was then added for reduction of disulfide
bonds. After incubation at 50°C for 15 min, the cysteine residues were alkyl-
ated through the addition of 100 mM iodoacetamide (10 l), followed by
incubation for 15 min at room temperature under protection from the light.
The final concentration of urea was then adjusted to1Mbytheaddition of
high-performance liquid chromatography (HPLC)-grade water (Sigma). The
mixture was supplemented with 4 g sequencing-grade trypsin (Promega) and
digested overnight at 37°C. The resulting sample was purified in reverse-
phase ZipTip columns (POROS R2 50; Applied Biosystems) as described by
Jurado et al. (24). Released peptides were then fractionated on a strong
cation-exchange ZipTip column (POROS HS 50 resin; Applied Biosystems),
preequilibrated with 25% acetonitrile (ACN)–0.5% formic acid (FA). After
loading the peptide mixture, the column was washed with 25% ACN–0.5%
FA and the peptides were eluted with the same solution supplemented with
NaCl concentrations ranging from 0 to 500 mM. Each fraction was dried in a
vacuum centrifuge (Eppendorf) and again purified by reverse-phase chroma-
tography in POROS R2 50 ZipTip columns. Fractions were finally suspended
in 0.05% trifluoracetic acid (30 l). An 8-l aliquot of each fraction was
loaded into a C
18
trap column (1 lC
18
; OPTI-PAK). The separation was
performed on a capillary reverse-phase column (Acclaim; LC Packings [3 m
C
18
,75m by 25 cm]) connected to a nanoHPLC system (nanoLC 1D Plus;
Eksigent). Peptides were eluted with increasing concentrations of ACN (0 to
40%) in 0.1% FA during 100 min and directly analyzed in an electrospray-
linear ion trap MS equipped with a nanospray source (LTQ XL; Thermo
Fisher). MS spectra were collected in centroid mode at the 400 to 1,700 m/z
range, and the five most abundant ions were subjected twice to collision-
induced dissociation with 35% normalized collision energy, before being
dynamically excluded for 120 s.
All tandem MS spectra from peptides with 600 to 4,000 Da, more than 100 counts,
and at least 15 fragments were converted into DTA files using Bioworks v.3.3.1
(Thermo). The DTA files were submitted to a database search using TurboSequest
(15), available in Bioworks, and the C. neoformans protein database, available at
www.broad.mit.edu/annotation/fungi/cryptococcus_neoformans. Common contami-
nant sequences (retrieved from GenBank at http://www.ncbi.nlm.nih.gov/ and the
International Protein Index at http://www.ebi.ac.uk/IPI) and 100,000 randomly gen-
erated sequences were used to supplement the C. neoformans database. The data-
base search parameters included (i) trypsin cleavage in both peptide termini with
one missed cleavage site allowed; (ii) carbamidomethylation of cysteine residues as
a fixed modification; (iii) oxidation of methionine residues as a variable modification;
and (iv) 2.0 and 1.0 Da for peptide and fragment mass tolerance, respectively. To
ensure the quality of protein identification, the false-positive rate (FPR) was esti-
mated using the TurboSequest output and the following formula: FPR ϭ number of
proteins matching random sequences/total number of proteins.
The FPR was calculated after applying the following filters in Bioworks:
distinct peptides (for exclusion of redundant hits); DCn, Ն0.1; protein proba-
bility, Յ1 ϫ 10
Ϫ3
; and Xcorr, Ն1.5, 2.2, and 2.7 for singly, doubly, and triply
charged peptides, respectively. When necessary, protein consensus scores were
also applied to limit the number of false-positive hits. All data sets showed an
FPR lower than 3.2%.
VOL. 7, 2008 C. NEOFORMANS VESICLES AND VIRULENCE PROTEINS 59
RESULTS
The extracellular vesicles produced by C. neoformans consist
of a complex population. The individual analysis of micro-
graphs of 419 secreted vesicles produced by C. neoformans
revealed considerable morphological variation. Using morpho-
logical criteria, we identified four main vesicle groups (Fig. 1).
The groups included electron-dense and electron-lucid vesi-
cles, vesicular structures with membrane-associated electron-
dense regions, and vesicles containing hyper-dense structures
resembling a dark pigment.
Pathogenesis-related molecules are present in the extracel-
lular vesicles. The microscopic analysis illustrated in Fig. 1
suggested that pigment synthesis could occur in vesicular struc-
tures. We therefore evaluated whether laccase, a cryptococcal
enzyme characterized as a virulence factor responsible for mel-
anin synthesis (46), was present in vesicular preparations. Lac-
case activity was detected in vesicle preparations from wild-
type cells (Fig. 2A). Enzyme activity was absent in vesicles from
a C. neoformans mutant lacking the ability to produce LAC1,
while vesicular structures from a laccase-reconstituted strain
had enzyme activity similar to wild-type cells. These results,
combined with a recent report by our group (45), indicated
that extracellular vesicles in C. neoformans contain at least
three virulence determinants: GXM (reviewed in reference
32), glucosylceramides (44), and laccase (46). We therefore
evaluated whether other molecules related to pathogenesis
were present in the C. neoformans vesicular structures.
We examined the activities of urease, a well-described ex-
tracellular virulence factor of C. neoformans (11), and acid
phosphatase, an adhesion-related molecule (8) suggested to be
released to the extracellular space in secretory vesicles (54).
Dose-dependent activities of both enzymes were observed
when the extracellular vesicles were incubated with urease and
phosphatase substrates (Fig. 2B and C).
Vesicle proteins are recognized by sera from cryptococcosis
patients. Vesicle proteins were separated by SDS-PAGE and
analyzed by reactivity with human sera by immunoblotting
(Fig. 3). No significant reactivity was observed using sera from
normal individuals. However, when the vesicle sample was
probed with a pool of sera from cryptococcosis patients, seven
major bands were observed, with relative molecular masses
corresponding to 131, 101, 67, 48, 38, 27, and 19 kDa. Diffuse
FIG. 1. Electron microscopic appearance (left panels) and preva-
lence (right panels) of the four major vesicle morphological groups
observed in preparations of extracellular vesicles from C. neoformans.
The total population analyzed consisted of 419 different vesicles. Scale
bars, 200 nm.
FIG. 2. Laccase (A), urease (B), and phosphatase (C) activities are associated with extracellular vesicles in C. neoformans. (A). Vesicles purified
from culture supernatants of strains H99 and the serotype D laccase mutants 2E-TUC (complemented strain) and 2E-TU (LAC1 deletion strain)
were incubated in the presence of L-DOPA and analyzed spectrophotometrically. Vesicles purified from the supernatants of strain H99 were also
incubated in urease (B) and phosphatase (C) reaction media, followed by spectrophotometric determination of enzyme activity.
FIG. 3. Vesicle-associated proteins are recognized by sera from
cryptococcosis patients. Vesicle-associated proteins (a and b) were
separated by SDS-PAGE and incubated with pooled sera from healthy
individuals (a) or cryptococcosis patients (b). Molecular masses for
standard (left values) or vesicle (right values) proteins are indicated.
60 RODRIGUES ET AL. E
UKARYOT.CELL
TABLE 1. Protein components of C. neoformans extracellular vesicles
a
Hit no. Accession no./identification Function (reference) Strain(s)
Pathogenesis/immune
response
1 CNAG_01727.1/heat shock 70-kDa protein 2*
(644 aa, 69.53 kDa)
Chaperone Cap67, 2E-TUC, H99
2 CNAG_03891.1/60-kDa chaperonin* (582 aa,
61.36 kDa)
Chaperone Cap67
3 CNAG_06150.1/heat shock protein 90* (700 aa,
79.15 kDa)
Chaperone Cap67
4 CNAG_00334.1/heat shock protein 70* (615 aa,
67.09 kDa)
Chaperone, C. neoformans
immunogen (25)
Cap67, 2E-TUC, H99
5 CNAG_02292.1/superoxide dismutase copper
chaperone* (288 aa, 30.32 kDa)
Antioxidant defense, C. neoformans
virulence (9, 34)
Cap67
6 CNAG_02944.1/acid phosphatase (552 aa, 59.32
kDa)
Adhesion of C. neoformans to
epithelial cells (8)
Cap67, 2E-TUC
7 CNAG_02801.1/thioredoxin (105 aa, 11.44 kDa)* Protection against oxidative stress,
survival in the oxidative
environment of macrophages, C.
neoformans virulence (29)
Cap67, H99
8 CNAG_05847.1/thioredoxin reductase (372 aa,
39.84 kDa)
C. neoformans viability (30) Cap67
9 CNAG_06917.1/thiol-specific antioxidant protein
(234 aa, 25.90 kDa)
C. neoformans virulence, resistance
to nitric oxide and peroxide (31)
H99, 2E-TUC, Cap67
10 CNAG_00575.1/catalase A (683 aa, 76.42 kDa) C. neoformans antioxidant defense
(22)
2E-TUC
11 CNAG_03322.1/UDP-glucuronic acid
decarboxylase Uxs1p (411 aa, 46.47 kDa)
Converts UDP-glucuronic acid to
UDP-xylose, capsule synthesis in
C. neoformans (3)
Cap67
12 CNAG_04969.1/UDP-glucose dehydrogenase
(469 aa, 51.36 kDa)
C. neoformans growth at 37°C and
capsule biosynthesis (33)
Cap67, H99
Signal transduction
13 CNAG_04762.1/Ras2 (364 aa, 39.47 kDa) Mating and high-temp growth in C.
neoformans (52)
Cap67
14 CNAG_03315.1/Rho1* (198 aa, 21.68 kDa) GTP-binding protein, cell wall
integrity in C. neoformans (19)
Cap67
15 CNAG_05235.1/14-3-3 protein* (257 aa, 28.98
kDa)
Cell cycle regulation in C.
neoformans (53)
Cap67, H99, 2E-TUC
16 CNAG_04577.1/nucleoside-diphosphate kinase
(216 aa, 23.36 kDa)
Exchange of phosphate groups
between different nucleoside
diphosphates
Cap67
17 CNAG_03052.1/protein phosphatase type 2C
(560 aa, 59.40 kDa)
Stress response and cell cycle
regulation
Cap67
Ribosomal proteins*
18 CNAG_06447.1/60S ribosomal protein l17 (183
aa, 20.49 kDa)
Ribosomal protein Cap67, H99
19 CNAG_03283.1/60S ribosomal protein L24 (L30)
(151 aa, 16.89 kDa)
Ribosomal protein Cap67
20 CNAG_00232.1/60S ribosomal protein l30-1 (l32)
(114 aa, 12.33 kDa)
Ribosomal protein 2E-TUC
21 CNAG_05762.1/ribosomal protein P2 (112 aa,
11.07 kDa)
Ribosomal protein Cap67
22 CNAG_00771.1/ribosomal protein L35 (128 aa,
14.67 kDa)
Ribosomal protein Cap67
23 CNAG_00779.1/ribosomal protein L27 (137 aa,
15.76 kDa)
Ribosomal protein Cap67
24 CNAG_06605.1/ribosomal protein S2 (257 aa,
27.62 kDa)
Ribosomal protein Cap67
25 CNAG_02144.1/60S ribosomal protein l1-a (l10a)
(225 aa, 25.50 kDa)
Ribosomal protein Cap67, 2E-TUC
26 CNAG_04114.1/40S ribosomal protein S0 (293
aa, 31.46 kDa)
Ribosomal protein Cap67
27 CNAG_01480.1/ribosomal protein L12 (166 aa,
17.55 kDa)
Ribosomal protein Cap67
28 CNAG_06633.1/40S ribosomal protein S15 (151
aa, 17.12 kDa)
Ribosomal protein Cap67
29 CNAG_00656.1/60S ribosomal protein l7 (251
aa, 28.25 kDa)
Ribosomal protein Cap67
Continued on following page
V
OL. 7, 2008 C. NEOFORMANS VESICLES AND VIRULENCE PROTEINS 61
TABLE 1—Continued
Hit no. Accession no./identification Function (reference) Strain(s)
30 CNAG_00034.1/60S ribosomal protein l9 (192
aa, 21.17 kDa)
Ribosomal protein Cap67
31 CNAG_06095.1/ribosomal protein L13 (206 aa,
23.31 kDa)
Ribosomal protein Cap67
32 CNAG_04011.1/60S ribosomal protein l37a (93
aa, 10.07 kDa)
Ribosomal protein Cap67
33 CNAG_02818.1/ribosomal protein S11 (410 aa,
44.31 kDa)
Ribosomal protein Cap67
Sugar and lipid
metabolism
34 CNAG_06699.1/glyceraldehyde-3-phosphate
dehydrogenase* (340 aa, 36.28 kDa)
Glucose metabolism, secreted
enzyme
Cap67, 2E-TUC
35 CNAG_00061.1/citrate synthase* (465 aa, 51.02
kDa)
Energy metabolism Cap67, 2E-TUC
36 CNAG_05653.1/malate synthase* (539 aa, 59.96
kDa)
Glucose metabolism 2E-TUC
37 CNAG_03072.1/phosphopyruvate hydratase
(enolase)* (434 aa, 46.44 kDa)
Cell wall constituent in Candida
(40)
Cap67, H99, 2E-TUC
38 CNAG_04659.1/pyruvate decarboxylase* (624
aa,)
Decarboxylation of pyruvic acid to
acetaldehyde and carbon dioxide
Cap67, H99
39 CNAG_03225.1/malate dehydrogenase* (339 aa,
35.58 kDa)
Conversion of malate into
oxaloacetate, major antigen in P.
brasiliensis (12)
Cap67
40 CNAG_02620.1/phosphogluconate
dehydrogenase* (decarboxylating) (492 aa,
53.85 kDa)
Pentose phosphate pathway Cap67, 2E-TUC
41 CNAG_03920.1/isocitrate dehydrogenase* (453
aa, 50.60 kDa)
Citric acid cycle Cap67
42 CNAG_02100.1/fatty-acid synthase complex
protein* (1,439 aa, 157.47 kDa)
Fatty acid biosynthetic process Cap67
43 CNAG_06140.1/long-chain fatty acid transporter
(111 aa, 11.91 kDa)
Fatty acid biosynthetic process Cap67
44 CNAG_06572.1/pyruvate dehydrogenase e1
component alpha subunit, mitochondrial
precursor (414 aa, 45.66 kDa)
Conversion of pyruvate to acetyl
coenzyme A and CO
2
Cap67
Nuclear proteins
45 CNAG_01648.1/histone H4* (104 aa, 11.40 kDa) DNA assembly H99, Cap67, 2E-TUC
46 CNAG_05221.1/histone H2A* variant (139 aa,
14.70 kDa)
DNA assembly Cap67, 2E-TUC
Protein/amino acid
metabolism
47 CNAG_00370.1/ubiquitin-carboxy extension
protein fusion* (130 aa, 14.64 kDa)
Posttranslational protein
modification
H99, 2E-TUC
48 CNAG_02500.1/endoplasmic reticulum-
associated protein, catabolism-related protein
(552 aa, 60.97 kDa), similar to fungal calnexin
Retention of unfolded or
unassembled N-linked
glycoproteins in the endoplasmic
reticulum
Cap67, H99
49 CNAG_00441.1/inosine 5-monophosphate
dehydrogenase (545 aa, 57.85 kDa)
Oxidation of inosine 5Ј-
monophosphate to xanthosine
5Ј-monophosphate
Cap67
50 CNAG_01890.1/5-
methyltetrahydropteroyltriglutamate-
homocysteine S-methyltransferase (764 aa,
85.32 kDa)
Amino acid metabolism,
cobalamin-independent
methionine synthase
Cap67, H99
51 CNAG_05725.1/acetohydroxy acid
reductoisomerase (402 aa, 44.31 kDa)
Amino acid metabolism Cap67
52 CNAG_00457.1/glutamine synthetase (359 aa,
39.49 kDa)
Amino acid metabolism Cap67
53 CNAG_00785.1/translation initiation factor* (402
aa, 45.13 kDa)
Protein synthesis Cap67
54 CNAG_00930.1/argininosuccinate synthase (430
aa, 47.34 kDa)
Amino acid metabolism Cap67
55 CNAG_04601.1/glycine hydroxymethyltransferase
(500 aa, 54.39 kDa)
Amino acid metabolism Cap67, H99
Continued on following page
62 RODRIGUES ET AL. E
UKARYOT.CELL
areas of serological reaction were also observed in the molec-
ular mass ranges of 50 to 65 and 70 to 100 kDa. These results
suggest that vesicle-related cryptococcal proteins are produced
during human infection and also that potentially effective pro-
tein immunogens are present in extracellular vesicles. Unfor-
tunately, the identification of the proteins recognized by the
sera of cryptococcosis patients has been hampered by the lack
of sufficient amount of vesicular material needed for liquid
chromatography-tandem MS sequencing from SDS-PAGE gel
bands.
Proteomic analysis of the C. neoformans extracellular vesi-
cles. To further characterize the protein components of the
secreted vesicles, the material purified by centrifugation was
enzymatically digested, fractionated by chromatographic meth-
ods, and analyzed by liquid chromatography-tandem MS.
Overall, 76 different proteins were identified (Table 1). The
TABLE 1—Continued
Hit no. Accession no./identification Function (reference) Strain(s)
56 CNAG_00450.1/3-isopropylmalate dehydrogenase
(374 aa, 39.85 kDa)
Amino acid metabolism Cap67
57 CNAG_00992.1/homocitrate synthase (490 aa,
53.51 kDa)
Amino acid metabolism Cap67
58 CNAG_06125.1/translation elongation factor 1*
(461 aa, 50.32 kDa)
Protein synthesis Cap67, H99, 2E-TUC
59 CNAG_06908.1/aminomethyltransferase,
mitochondrial precursor (338 aa, 34.92 kDa)
Amino acid metabolism Cap67
Plasma membrane
proteins
60 CNAG_06101.1/ADP/ATP carrier (314 aa, 33.77
kDa)
ATP/ADP translocation, multipass
membrane protein
Cap67, 2E-TUC
61 CNAG_06400.1/plasma membrane H(ϩ)-ATPase
(999 aa, 108.47 kDa)
Proton pump Cap67
62 CNAG_03058.1/Hmp1 protein (115 aa, 11.74
kDa)
Similar to -catenins, proteins
found in complexes with
cadherin cell adhesion
2E-TUC
63 CNAG_04758.1/ammonium transporter (497 aa,
53.04 kDa)
Multipass membrane protein Cap67
Cytoskeleton
proteins
64 CNAG_00483.1/actin* (378 aa, 42.02 kDa) Cytoskeleton protein Cap67, 2E-TUC, H99
65 CNAG_03787.1/-tubulin* (449 aa, 49.67 kDa) Cytoskeleton protein Cap67
66 CNAG_01840.1/tubulin chain* (452 aa, 49.99
kDa)
Cytoskeleton protein Cap67
Miscellaneous
67 CNAG_01577.1/glutamate dehydrogenase (452
aa, 49.16 kDa)
Urea synthesis Cap67, 2E-TUC
68 CNAG_05750.1/ATP synthase chain,
mitochondrial precursor (541 aa, 58.02 kDa)
ATP synthesis Cap67, H99, 2E-TUC
69 CNAG_05918.1/ATP synthase chain* (548 aa,
58.63 kDa)
ATP synthesis Cap67, 2E-TUC
70 CNAG_02974.1/voltage-dependent ion-selective
channel (293 aa, 30.61 kDa)
Membrane transport Cap67, 2E-TUC, H99
71 CNAG_01539.1/inositol-3-phosphate synthase
(559 aa, 61.34 kDa)
Metabolism of inositol-containing
molecules
Cap67
72 CNAG_05909.1/electron transporter, transferring
electrons within CoQH2-cytochrome c
reductase complex (320 aa, 34.97 kDa)
Mitochondrial protein, electron
transport
2E-TUC
73 CNAG_00799.1/hypothetical protein similar to
glycosyl hydrolase family 5 protein from
Stigmatella aurantiaca (790 aa, 85.25 kDa)
Cell wall assembly (putative) 2E-TUC
74 CNAG_03537.1/FA and derivative metabolism-
related protein (1022 aa, 108.90 kDa)
FA metabolism Cap67, H99
75 CNAG_00965.1/probable transketolase (688 aa,
74.28 kDa)
NADPH metabolism Cap67, H99
76 CNAG_05759.1/acetyl coenzyme A carboxylase
(2238 aa, 247.95 kDa)
Synthesis of malonyl coenzyme A Cap67
a
Biological functions of the proteins detected by proteomics are referenced when an association of the molecule with the biology or pathogenesis of C. neoformans
or other fungal pathogens was previously described. General functions in other models were obtained from the ExPASy Proteomics Server (http://ca.expasy.org/). The
number of amino acids (aa) and molecular mass values are presented for each protein. Twenty-five proteins with unknown functions were characterized but not
presented in this table. For detailed information, see the supplemental material. Proteins that were already characterized in extracellular vesicles produced by
mammalian cells are marked with an asterisk. For details, see references 1, 21, 28, 42, 48, and 49.
VOL. 7, 2008 C. NEOFORMANS VESICLES AND VIRULENCE PROTEINS 63
yield of protein detection varied considerably, depending on
the presence of capsular structures, suggesting that the poly-
saccharides in extracellular fractions might interfere with pro-
tein digestion, as previously suggested for Aspergillus oryzae
(38). Accordingly, no proteins were detected by proteomic
approaches in preliminary analysis of vesicles from the highly
encapsulated strain 24067 (data not shown). Twenty different
proteins were identified using strain H99, which presents
smaller capsules than isolate 24067. A higher number of pro-
teins (n ϭ 26) was identified using the encapsulated strain
2E-TUC, but protein characterization was maximum using the
acapsular mutant Cap 67 (70 proteins). The results obtained
with each strain were combined and are summarized in Table
1. A complete, detailed list of proteins analyzed by mass spec-
trometry is available (see Table S1 in the supplemental mate-
rial). Chaperones, including heat shock proteins (Hsp70 and
Hsp90) and superoxide dismutase, signal transduction regula-
tors, antioxidant and cytosolic proteins, and enzymes were
identified. Of note, nuclear proteins such as histones and ri-
bosomal proteins were also identified, as previously described
in vesicles secreted by mammalian cells (1, 42). Overall, 27 of
the 76 proteins identified in the C. neoformans vesicles were
already reported as vesicular proteins in mammalian exosomes
(1, 21, 28, 42, 48, 49), including a ubiquitin-related protein.
Figure 4 shows identified C. neoformans vesicle proteins dis-
tributed according to their functions in fungal cells.
Intracellular distribution of vesicles in C. neoformans. Mam-
malian exosomes consist of secreted vesicles that originate
from key compartments of the endocytic pathway, namely mul-
tivesicular bodies (MVBs) (51). Since the profile of protein
detection in the C. neoformans vesicles resembled those found
in mammalian exosomes, ultrathin sections of yeast cells were
searched for the presence of MVB-like structures. Vesicular
structures with dimensions similar to those found at the extra-
cellular fractions were observed inside vacuole-like compart-
ments (Fig. 5), resembling endosome-derived MVBs (51).
These vesicle-containing vacuoles were detected in all the sec-
tions analyzed, independently of the technique used for TEM
(data not shown). Bilayered membranes were present at the
vacuole-like structures, which were sometimes found in fusion
with the plasma membrane. These results suggest that vesicle
secretion in C. neoformans may derive from MVBs or vesicle-
containing vacuoles.
DISCUSSION
The secretion of virulence factors is a mechanism used by
different pathogens to cause damage to host cells (6). In C.
neoformans, secreted virulence factors include superoxide dis-
mutase (9), phospholipase B (10), urease (11), and GXM, a
high-molecular-weight polysaccharide (reviewed in references
26 and 32). GXM is synthesized in intracellular compartments
(17, 20, 54) and transported through the cell wall in secretory
vesicles (45). The virulence regulator glucosylceramide (44) is
also present in the cryptococcal extracellular vesicles (45), but
the presence of other molecules in these membrane compart-
ments is unknown. In this context, we hypothesized that vesic-
ular secretion in C. neoformans could be a general secretory
mechanism, possibly used for the delivery of different mole-
cules related to pathogenic mechanisms to the extracellular
space.
Polysaccharide-containing vesicles in C. neoformans ap-
peared to be associated with the Golgi apparatus-derived se-
cretory pathway (54). Golgi apparatus-derived secretory vesi-
cles, however, are expected to fuse with the plasma membrane
and release their internal content to the extracellular space
(41). In C. neoformans, this model would satisfactorily explain
how GXM reaches the periplasmic space, although it does not
explain how the polysaccharide would reach the outer layer of
the cell wall to be incorporated into the growing capsule. Fur-
thermore, such a mechanism could not account for the origin
of the GXM-containing extracellular vesicles that we recently
described. In fact, the existence of GXM-containing extracel-
lular vesicles implies the existence of a vesicle secretion mech-
anism whereby there is no fusion of secretory vesicles and the
plasma membrane. In this context, we examined whether the
secretory vesicles of C. neoformans could have a relationship
with exosomes, which are defined as non-plasma-membrane-
derived vesicles (18).
Exosomes are the only type of bioactive vesicles originating
from an intracellular compartment, named MVBs on the basis
of their morphology (18, 51). These intracellular compart-
ments are derived from endosomes and have well-known func-
tions as intermediates in the degradation of proteins internal-
ized from the cell surface or sorted from the trans-Golgi
network (18, 51). Internal vesicles of MVBs are generated by
budding from the limiting membrane into the lumen of endo-
somes (51). In the degradation pathway, MVBs fuse with ly-
sosomes. However, in several hematopoietic and nonhemato-
poietic cells, MVBs fuse with the plasma membrane, resulting
in the release of internal vesicles to the extracellular milieu as
exosomes (18, 51). In this context, we evaluated whether MVB-
like compartments were present in C. neoformans cells. In fact,
several vacuole-like compartments containing vesicles with di-
mensions similar to those of exosomes and the C. neoformans
extracellular vesicles were observed. Some of these vesicle-
containing vacuoles were in close association with the plasma
membrane and the cell wall, suggesting that the release of
extracellular vesicles to the extracellular space in C. neofor-
mans involves MVB-like compartments. Since endosomes and
MVBs can be connected to the trans-Golgi secretory pathway
(51), we speculate that the previously described Golgi appara-
tus-derived vesicles containing GXM (54) are linked to MVBs
in C. neoformans, which would result in the release of polysac-
FIG. 4. Functional classification of the C. neoformans vesicle proteins.
The number of proteins found for each class is shown. Unidentified
proteins are not shown. For details, see Table S1 in the supplemental
material.
64 RODRIGUES ET AL. E
UKARYOT.CELL
charide-containing vesicles into the periplasmic space. In ac-
cordance with this supposition, treatment of C. neoformans
with brefeldin A, an inhibitor of the Golgi apparatus-derived
transport of molecules, results in a significant inhibition of
capsule expression (23).
Fungal proteins frequently have more than a single function
and are found in different cellular locations (2, 4, 16, 37). For
example, histones have been described as being present at the
cell wall of Histoplasma capsulatum, where they are targeted by
antifungal antibodies (37). Glyceraldehyde-3-phosphate dehy-
drogenase, a major protein component of the glycolytic path-
way, is present in the cell wall of Paracoccidioides brasiliensis,
where it participates in the pathogenic processes mediating the
adhesion of yeast cells to host cells and the extracellular matrix
(2). In the same model, the mitochondrial protein Mdj1 was
detected not only in the mitochondria, where it is apparently
sorted, but also in the cell wall (4). Proteomic analysis of the C.
neoformans vesicles revealed a complex protein composition
that included chaperone and membrane, cytoplasmic, and even
nuclear and mitochondrial proteins. Several of these proteins
were similar to those described in mammalian exosomes, which
usually contain cytoplasmic proteins such as elongation factors,
tubulin, actin, actin-binding proteins, annexins, and Rab pro-
tein, molecules responsible for signal transduction, and heat-
shock proteins such as Hsp70 and Hsp90 (1, 21, 28, 42, 48, 49).
Sorting of cytosolic proteins into exosomes is normally ex-
plained by a random engulfment of small portions of cytosol
during the inward budding process of MVBs (51). These ob-
servations, together with morphological data, could support
the supposition that the extracellular vesicles produced by C.
neoformans are exosome-like structures.
Protein composition and morphological analyses indicate
that the C. neoformans extracellular vesicles are not a uniform
population. Vesicles with clearly different electron densities
were observed, and some of them were observed to carry
pigment-like structures. The observation of electron-dense
spots in the inner vesicle compartments suggests the presence
of the molecular machinery necessary for the synthesis of mel-
anin, a pigment that has been concretely associated with the
virulence of C. neoformans (46). Melanin is autopolymerized
from the oxidation of diphenolic compounds by the enzyme
laccase (36). In this context, we incubated the vesicular sus-
pension with the laccase substrate
L-DOPA, which demon-
strated laccase activity in C. neoformans vesicles. The finding of
FIG. 5. TEM of C. neoformans suggesting the presence of cytoplasmic vacuole-containing vesicles reminiscent of exosome-like structures. (A) Over-
view of a C. neoformans cell with different cytoplasmic vacuoles containing vesicles (black asterisks). The white asterisk indicates the cell wall. Scale bar,
500 nm. A magnified view of the vesicle-containing vacuoles is shown. Panel B demonstrates that these structures are surrounded by a bilayered
membrane, which sometimes invaginates (arrow). A close association with the cell wall (white asterisk) was observed, suggesting fusion with the plasma
membrane. Scale bar, 200 nm. (C) Intracellular and extracellular vesicles (black arrows) have similar dimensions. Scale bar, 200 nm.
V
OL. 7, 2008 C. NEOFORMANS VESICLES AND VIRULENCE PROTEINS 65
laccase in vesicles that are possibly derived from MVB has an
intriguing parallel in mammalian systems where tyrosinase-
containing melanosomes are shed from melanocytes after syn-
thesis from early endosomal vesicles (43). However, laccase
was not detected by the proteomic approach. This observation
is probably a false-negative result related to a low protein
concentration, since pigmented vesicles are the less-abundant
fraction in the vesicle population (15%). A relatively low pro-
tein concentration could also explain why urease is also detect-
able by a sensitive enzymatic colorimetric assays but not by
proteomic approaches. Hence, we postulate that the proteins
identified here may be only a subset of the total proteins found
in vesicles.
We identified several virulence-related molecules in C. neo-
formans vesicles. This group of molecules includes well-known
virulence factors such as GXM and glucosylceramide, which
were characterized as vesicle components in a previous study
(45). In the present study, we demonstrate the presence of
several other components associated with virulence in vesicular
fractions, such as enzymes related to capsule synthesis (3),
urease (11), laccase (46), acid phosphatase (8), heat shock
proteins (25), and several antioxidant proteins such as super-
oxide dismutase (9, 34), thioredoxin (29), thioredoxin reduc-
tase (30), thiol-specific antioxidant protein (31), and catalase A
(22). Some of the vesicle proteins were recognized by sera from
cryptococcosis patients, suggesting that these proteins are pro-
duced during human infection. The combined presence of lip-
ids, pigments, polysaccharides, and virulence-related and im-
munogenic proteins suggests that C. neoformans uses vesicular
secretion as a single mechanism to deliver virulence factors
into the fungal extracellular space. Since vesicle production has
been observed in vivo and during macrophage infection (45),
we suggest that the C. neoformans extracellular vesicles func-
tion as “virulence factor delivery bags” that could expressively
influence the interaction of fungal cells with the host. Different
types of vesicles may carry different types of toxic payloads.
Clearly, the presence of numerous virulence-associated com-
ponents in vesicular preparations would allow C. neoformans to
deliver a toxic concentrated payload to target cells such as
predatory amoebae and macrophages. Vesicle secretion could
also presumably occur in phagosomal spaces and allow delivery
of toxic payloads to cells that have ingested cryptococcal cells.
Vesicular delivery of concentrated virulence-associated com-
ponents could be significantly more effective in damaging toxic
cells than if such components were secreted separately and had
to reach target cells through diffusion. In this context, it has
been recently suggested by our group that vesicle secretion
occurs during infection of host macrophages (45). This obser-
vation could be related to the known ability of C. neoformans
to secrete GXM during intracellular infection of phagocytes
(50), which results in host cell toxicity and release of fungal
cells to extracellular host sites.
In summary, we report the identification of numerous
virulence-associated components in C. neoformans vesicle
preparations. The similarities in the protein content of C.
neoformans vesicles and mammalian exosome-like struc-
tures combined with electron microscopic morphological evi-
dence of exosome-like structures in cryptococcal cells led us to
propose that the extracellular vesicles originate from fungal
exosomes. The complexity of vesicle populations with respect
to their morphology and cargo suggest numerous new avenues
for the investigation of their role in virulence and cryptococcal
cell biology.
ACKNOWLEDGMENTS
M.L.R. and L.N. are supported by grants from the Brazilian agencies
CNPq, CAPES, FAPESP, and FAPERJ. A.C. is supported by NIH
grants AI033142, AI033774, AI052733, and HL059842. I.C.A. is sup-
ported by NIH grant 5G12RR008124 (to the Border Biomedical Re-
search Center [BBRC]/University of Texas at El Paso [UTEP]). J.D.N.
is supported by NIH AI056070-01A2 and AI52733. We are thankful to
the Biomolecule Analysis Core Facility/BBRC/UTEP, supported by
NIH/NCRR grant 5G12RR008124. D.L.O. is a Ph.D. student at the
Instituto de Bioquimica Medica, UFRJ.
We thank Kildare Miranda and the Albert Einstein College of
Medicine Analytical Imaging Facility staff for help with the electron
microscopy. We are also indebted to Fabio Gozzo (Laboratorio Na-
cional de Luz Sincrontron, Campinas, Brazil) for the 100,000 random
sequences used for statistics in the proteomic analysis.
REFERENCES
1. Aoki, N., S. Jin-no, Y. Nakagawa, N. Asai, E. Arakawa, N. Tamura, T.
Tamura, and T. Matsuda. 2007. Identification and characterization of mi-
crovesicles secreted by 3T3-L1 adipocytes: redox- and hormone-dependent
induction of milk fat globule-epidermal growth factor 8-associated mi-
crovesicles. Endocrinology 148:3850–3862.
2. Barbosa, M. S., S. N. Bao´, P. F. Andreotti, F. P. de Faria, M. S. S. Felipe, L.
dos Santos Feitosa, M. J. S. Mendes-Giannini, and C. M. d. A. Soares. 2006.
Glyceraldehyde-3-phosphate dehydrogenase of Paracoccidioides brasiliensis
is a cell surface protein involved in fungal adhesion to extracellular matrix
proteins and interaction with cells. Infect. Immun. 74:382–389.
3. Bar-Peled, M., C. L. Griffith, and T. L. Doering. 2001. Functional cloning
and characterization of a UDP-glucuronic acid decarboxylase: the patho-
genic fungus Cryptococcus neoformans elucidates UDP-xylose synthesis.
Proc. Natl. Acad. Sci. USA 98:12003–12008.
4. Batista, W. L., A. L. Matsuo, L. Ganiko, T. F. Barros, T. R. Veiga, E.
Freymu¨ller, and R. Puccia. 2006. The PbMDJ1 gene belongs to a conserved
MDJ1/LON locus in thermodimorphic pathogenic fungi and encodes a heat
shock protein that localizes to both the mitochondria and cell wall of Para-
coccidioides brasiliensis. Eukaryot. Cell 5:379–390.
5. Bicanic, T., and T. S. Harrison. 2004. Cryptococcal meningitis. Br. Med.
Bull. 72:99–118.
6. Casadevall, A., and L. A. Pirofski. 2003. The damage-response framework of
microbial pathogenesis. Nat. Rev. Microbiol. 1:17–24.
7. Casadevall, A., W. Cleare, M. Feldmesser, A. Glatman-Freedman, D. L.
Goldman, T. R. Kozel, N. Lendvai, J. Mukherjee, L.-A. Pirofski, J. Rivera,
A. L. Rosas, M. D. Scharff, P. Valadon, K. Westin, and Z. Zhong. 1998.
Characterization of a murine monoclonal antibody to Cryptococcus neofor-
mans polysaccharide that is a candidate for human therapeutic studies.
Antimicrob. Agents Chemother. 42:1437–1446.
8. Collopy-Junior, I., F. F. Esteves, L. Nimrichter, M. L. Rodrigues, C. S.
Alviano, and J. R. Meyer-Fernandes. 2006. An ectophosphatase activity in
Cryptococcus neoformans. FEMS Yeast Res. 6:1010–1017.
9. Cox, G. M., T. S. Harrison, H. C. McDade, C. P. Taborda, G. Heinrich, A.
Casadevall, and J. R. Perfect. 2003. Superoxide dismutase influences the
virulence of Cryptococcus neoformans by affecting growth within macro-
phages. Infect. Immun. 71:173–180.
10. Cox, G. M., H. C. McDade, S. C. Chen, S. C. Tucker, M. Gottfredsson, L. C.
Wright, T. C. Sorrell, S. D. Leidich, A. Casadevall, M. A. Ghannoum, and
J. R. Perfect. 2001. Extracellular phospholipase activity is a virulence factor
for Cryptococcus neoformans. Mol. Microbiol. 39:166–175.
11. Cox, G. M., J. Mukherjee, G. T. Cole, A. Casadevall, and J. R. Perfect. 2000.
Urease as a virulence factor in experimental cryptococcosis. Infect. Immun.
68:443–448.
12. Da Fonseca, C. A., R. S. Jesuino, M. S. Felipe, D. A. Cunha, W. A. Brito, and
C. M. Soares. 2001. Two-dimensional electrophoresis and characterization
of antigens from Paracoccidioides brasiliensis. Microbes Infect. 3:535–542.
13. Datta, K., and L. A. Pirofski. 2006. Towards a vaccine for Cryptococcus
neoformans: principles and caveats. FEMS Yeast Res. 6:525–536.
14. De Souza Pereira, R., and J. Geibel. 1999. Direct observation of oxidative
stress on the cell wall of Saccharomyces cerevisiae strains with atomic force
microscopy. Mol. Cell. Biochem. 201:17–24.
15. Eng, J. K., A. L. McCormack, and J. R. Yates III. 1994. An approach to
correlate tandem mass spectral data of peptides with amino acid sequences
in a protein database. J. Am. Soc. Mass Spectrom. 5:976–989.
16. Esnault, K., B. el Moudni, J.-P. Bouchara, D. Chabasse, and G. Tronchin.
1999. Association of a myosin immunoanalogue with cell envelopes of As-
66 RODRIGUES ET AL. EUKARYOT.CELL
pergillus fumigatus conidia and its participation in swelling and germination.
Infect. Immun. 67:1238–1244.
17. Feldmesser, M., Y. Kress, and A. Casadevall. 2001. Dynamic changes in the
morphology of Cryptococcus neoformans during murine pulmonary infection.
Microbiology 147:2355–2365.
18. Fevrier, B., and G. Raposo. 2004. Exosomes: endosomal-derived vesicles
shipping extracellular messages. Curr. Opin. Cell Biol. 16:415–421.
19. Fuchs, B. B., G. P. Tegos, M. R. Hamblin, and E. Mylonakis. 2007. Suscep-
tibility of Cryptococcus neoformans to photodynamic inactivation is associ-
ated with cell wall integrity. Antimicrob. Agents Chemother. 51:2929–2936.
20. Garcia-Rivera, J., Y. C. Chang, K. J. Kwon-Chung, and A. Casadevall. 2004.
Cryptococcus neoformans CAP59 (or Cap59p) is involved in the extracellular
trafficking of capsular glucuronoxylomannan. Eukaryot. Cell 3:385–392.
21. Gatti, J. L., S. Metayer, M. Belghazi, F. Dacheux, and J. L. Dacheux. 2005.
Identification, proteomic profiling, and origin of ram epididymal fluid exo-
some-like vesicles. Biol. Reprod. 72:1452–1465.
22. Giles, S. S., J. E. Stajich, C. Nichols, Q. D. Gerrald, J. A. Alspaugh, F.
Dietrich, and J. R. Perfect. 2006. The Cryptococcus neoformans catalase gene
family and its role in antioxidant defense. Eukaryot. Cell 5:1447–1459.
23. Hu, G., B. R. Steen, T. Lian, A. P. Sham, N. Tam, K. L. Tangen, and J. W.
Kronstad. 2007. Transcriptional regulation by protein kinase A in Crypto-
coccus neoformans. PLoS Pathog. 3:e42.
24. Jurado, J. D., E. D. Rael, C. S. Lieb, E. Nakayasu, W. K. Hayes, S. P. Bush,
and J. A. Ross. 2007. Complement inactivating proteins and intraspecies
venom variation in Crotalus oreganus helleri. Toxicon 49:339–350.
25. Kakeya, H., H. Udono, S. Maesaki, E. Sasaki, S. Kawamura, M. A. Hossain,
Y. Yamamoto, T. Sawai, M. Fukuda, K. Mitsutake, Y. Miyazaki, K. Tomono,
T. Tashiro, E. Nakayama, and S. Kohno. 1999. Heat shock protein 70
(hsp70) as a major target of the antibody response in patients with pulmo-
nary cryptococcosis. Clin. Exp. Immunol. 115:485–490.
26. McFadden, D., O. Zaragoza, and A. Casadevall. 2006. The capsular dynam-
ics of Cryptococcus neoformans. Trends Microbiol. 14:497–505.
27. McFadden, D. C., M. De Jesus, and A. Casadevall. 2006. The physical
properties of the capsular polysaccharides from Cryptococcus neoformans
suggest features for capsule construction. J. Biol. Chem. 281:1868–1875.
28. Mears, R., R. A. Craven, S. Hanrahan, N. Totty, C. Upton, S. L. Young, P.
Patel, P. J. Selby, and R. E. Banks. 2004. Proteomic analysis of melanoma-
derived exosomes by two-dimensional polyacrylamide gel electrophoresis
and mass spectrometry. Proteomics 4:4019–4031.
29. Missall, T. A., and J. K. Lodge. 2005. Function of the thioredoxin proteins in
Cryptococcus neoformans during stress or virulence and regulation by puta-
tive transcriptional modulators. Mol. Microbiol. 57:847–858.
30. Missall, T. A., and J. K. Lodge. 2005. Thioredoxin reductase is essential for
viability in the fungal pathogen Cryptococcus neoformans. Eukaryot. Cell
4:487–489.
31. Missall, T. A., J. M. Moran, J. A. Corbett, and J. K. Lodge. 2005. Distinct
stress responses of two functional laccases in Cryptococcus neoformans are
revealed in the absence of the thiol-specific antioxidant Tsa1. Eukaryot. Cell
4:202–208.
32. Monari, C., F. Bistoni, and A. Vecchiarelli. 2006. Glucuronoxylomannan
exhibits potent immunosuppressive properties. FEMS Yeast Res. 6:537–542.
33. Moyrand, F., and G. Janbon. 2004. UGD1, encoding the Cryptococcus neo-
formans UDP-glucose dehydrogenase, is essential for growth at 37°C and for
capsule biosynthesis. Eukaryot. Cell 3:1601–1608.
34. Narasipura, S. D., J. G. Ault, M. J. Behr, V. Chaturvedi, and S. Chaturvedi.
2003. Characterization of Cu, Zn superoxide dismutase (SOD1) gene knock-
out mutant of Cryptococcus neoformans var. gattii: role in biology and viru-
lence. Mol. Microbiol. 47:1681–1694.
35. Nimrichter, L., M. L. Rodrigues, E. G. Rodrigues, and L. R. Travassos. 2005.
The multitude of targets for the immune system and drug therapy in the
fungal cell wall. Microbes Infect. 7:789–798.
36. Nosanchuk, J. D., and A. Casadevall. 2006. Impact of melanin on microbial
virulence and clinical resistance to antimicrobial compounds. Antimicrob.
Agents Chemother. 50:3519–3528.
37. Nosanchuk, J. D., J. N. Steenbergen, L. Shi, G. S. Deepe, Jr., and A.
Casadevall. 2003. Antibodies to a cell surface histone-like protein protect
against Histoplasma capsulatum. J. Clin. Investig. 112:1164–1175.
38. Oda, K., D. Kakizono, O. Yamada, H. Iefuji, O. Akita, and K. Iwashita. 2006.
Proteomic analysis of extracellular proteins from Aspergillus oryzae grown
under submerged and solid-state culture conditions. Appl. Environ. Micro-
biol. 72:3448–3457.
39. Perfect, J. R., and A. Casadevall. 2002. Cryptococcosis. Infect. Dis. Clin. N.
Am. 16:837–874, v–vi.
40. Pitarch, A., A. Jimenez, C. Nombela, and C. Gil. 2006. Decoding serological
response to Candida cell wall immunome into novel diagnostic, prognostic,
and therapeutic candidates for systemic candidiasis by proteomic and bioin-
formatic analyses. Mol. Cell Proteomics 5:79–96.
41. Ponnambalam, S., and S. A. Baldwin. 2003. Constitutive protein secretion
from the trans-Golgi network to the plasma membrane. Mol. Membr. Biol.
20:129–139.
42. Potolicchio, I., G. J. Carven, X. Xu, C. Stipp, R. J. Riese, L. J. Stern, and L.
Santambrogio. 2005. Proteomic analysis of microglia-derived exosomes:
metabolic role of the aminopeptidase CD13 in neuropeptide catabolism.
J. Immunol. 175:2237–2243.
43. Raposo, G., and M. S. Marks. 2007. Melanosomes—dark organelles en-
lighten endosomal membrane transport. Nat. Rev. Mol. Cell Biol. 8:786–797.
44. Rittershaus, P. C., T. B. Kechichian, J. C. Allegood, A. H. Merrill, Jr., M.
Hennig, C. Luberto, and M. Del Poeta. 2006. Glucosylceramide synthase is
an essential regulator of pathogenicity of Cryptococcus neoformans. J. Clin.
Investig. 116:1651–1659.
45. Rodrigues, M. L., L. Nimrichter, D. O. Oliveira, S. Frases, K. Miranda, O.
Zaragoza, M. Alvarez, A. Nakouzi, M. Feldmesser, and A. Casadevall. 2007.
Vesicular polysaccharide export in Cryptococcus neoformans is a eukaryotic
solution to the problem of fungal trans-cell wall transport. Eukaryot. Cell
6:48–59.
46. Salas, S. D., J. E. Bennett, K. J. Kwon-Chung, J. R. Perfect, and P. R.
Williamson. 1996. Effect of the laccase gene CNLAC1, on virulence of
Cryptococcus neoformans. J. Exp. Med. 184:377–386.
47. Schmid, F., F. Separovic, B. M. McDougall, B. A. Stone, R. T. Brownlee, and
R. J. Seviour. 2007. Characterisation of the extracellular polysaccharides
produced by isolates of the fungus Acremonium. Carbohydr. Res. [Epub
ahead of print.]
48. Segura, E., S. Amigorena, and C. Thery. 2005. Mature dendritic cells secrete
exosomes with strong ability to induce antigen-specific effector immune
responses. Blood Cells Mol. Dis. 35:89–93.
49. Thery, C., M. Boussac, P. Veron, P. Ricciardi-Castagnoli, G. Raposo, J.
Garin, and S. Amigorena. 2001. Proteomic analysis of dendritic cell-derived
exosomes: a secreted subcellular compartment distinct from apoptotic vesi-
cles. J. Immunol. 166:7309–7318.
50. Tucker, S. C., and A. Casadevall. 2002. Replication of Cryptococcus neofor-
mans in macrophages is accompanied by phagosomal permeabilization and
accumulation of vesicles containing polysaccharide in the cytoplasm. Proc.
Natl. Acad. Sci. USA 99:3165–3170.
51. van Niel, G., I. Porto-Carreiro, S. Simoes, and G. Raposo. 2006. Exosomes:
a common pathway for a specialized function. J. Biochem. (Tokyo) 140:
13–21.
52. Waugh, M. S., C. B. Nichols, C. M. DeCesare, G. M. Cox, J. Heitman, and
J. A. Alspaugh. 2002. Ras1 and Ras2 contribute shared and unique roles in
physiology and virulence of Cryptococcus neoformans. Microbiology 148:191–
201.
53. Woyke, T., M. E. Berens, D. B. Hoelzinger, G. R. Pettit, G. Winkelmann, and
R. K. Pettit. 2004. Differential gene expression in auristatin PHE-treated
Cryptococcus neoformans. Antimicrob. Agents Chemother. 48:561–567.
54. Yoneda, A., and T. L. Doering. 2006. A eukaryotic capsular polysaccharide is
synthesized intracellularly and secreted via exocytosis. Mol. Biol. Cell 17:
5131–5140.
55. Zaragoza, O., A. Telzak, R. A. Bryan, E. Dadachova, and A. Casadevall.
2006. The polysaccharide capsule of the pathogenic fungus Cryptococcus
neoformans enlarges by distal growth and is rearranged during budding. Mol.
Microbiol. 59:67–83.
VOL. 7, 2008 C. NEOFORMANS VESICLES AND VIRULENCE PROTEINS 67
27
REVIEW – SPECIAL ISSUE
Correspondence: Marcio L. Rodrigues, Universidade Federal do Rio de Janeiro, Instituto de Microbiologia
Professor Paulo de Góes. Avenida Carlos Chagas Filho, 373, Cidade Universitária CCS, Bloco I. Rio de
Janeiro—RJ, 21941-902, Brazil. Tel: 55 21 25626740; Fax: 55 21 25608344. Email:[email protected]
Copyright in this article, its metadata, and any supplementary data is held by its author or authors. It is published under the
Creative Commons Attribution By licence. For further information go to: http://creativecommons.org/licenses/by/3.0/.
Vesicular Trans-Cell Wall Transport in Fungi: A Mechanism
for the Delivery of Virulence-Associated Macromolecules?
Marcio L. Rodrigues
1
, Leonardo Nimrichter
1
, Debora L. Oliveira
1
, Joshua D.
Nosanchuk
2,3
and Arturo Casadevall
2,3
1
Laboratório de Estudos Integrados em Bioquímica Microbiana, Instituto de Microbiologia
Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941590, Brazil.
2
Department
of Microbiology and Immunology and the
3
Division of Infectious Diseases of the Department
of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, U.S.A. 10461.
Abstract: Fungal cells are encaged in rigid, complex cell walls. Until recently, there was remarkably little information
regarding the trans-fungal cell wall transfer of intracellular macromolecules to the extracellular space. Recently, several
studies have begun to elucidate the mechanisms that fungal cells utilize to secrete a wide variety of macromolecules through
the cell wall. The combined use of transmission electron microscopy, serology, biochemistry, proteomics and lipidomics
have revealed that the fungal pathogens Cryptococcus neoformans, Histoplasma capsulatum, Candida albicans, Candida
parapsilosis and Sporothrix schenckii, as well as the model yeast Saccharomyces cerevisiae, each produces extracellular
vesicles that carry lipids, proteins, polysaccharides and pigment-like structures of unquestionable biological signi cance.
Compositional analysis of the C. neoformans and H. capsulatum extracellular vesicles suggests that they may function as
‘virulence bags’, with the potential to modulate the host-pathogen interaction in favor of the fungus. The cellular origin of
the extracellular vesicles remains unknown, but morphological and biochemical features indicate that they are similar to
the well-described mammalian exosomes.
Keywords: trans-cell wall transport, extracellular vesicles, secretion
Insights into Secretory Mechanisms and Vesicular Transport Systems
Pathogenic mechanisms and secretory processes in microbes are closely associated. Secreted virulence
factors and potent extracellular immunogens have been described for a number of prokaryotic and
eukaryotic pathogens (Engel et al. 1998; Ghannoum, 1998; Hoegl et al, 1996; Hube, 1996; Liu and
Nizet, 2004; McFadden et al. 2006a; Mitchell, 2006; Rodrigues et al. 2008; Rodrigues et al. 2007).
Therefore, the understanding of secretory mechanisms and their regulation in microbial pathogens may
represent a promising strategy for the design of new drugs and prophylactic agents.
There are numerous secretory pathways in eukaryotic and prokaryotic organisms that utilize diverse
mechanisms for secretion (Gorelick and Shugrue, 2001; Lee et al. 2004; Mazar and Cotter, 2007; Rigel
and Braunstein, 2008; Salama and Schekman, 1995; Wickner and Schekman, 2005). In eukaryotes, the
most well-studied pathway of protein secretion involves vesicular migration from the endoplasmic
reticulum to the trans face of the Golgi and then loading into a complex network of vesicles, the trans-
Golgi reticulum (Glick and Malhotra, 1998). These proteins are sorted in the trans-Golgi network into
transport vesicles that immediately move to and fuse with the plasma membrane, releasing their contents
by exocytosis (Glick and Malhotra, 1998). In this regard, the generation of a collection of temperature-
sensitive mutant Saccharomyces cerevisiae yeast strains (sec mutants in which secretion and cell surface
assembly of proteins were blocked at different steps of the secretory pathway) was extremely important
for the elucidation of the sequential events required for secretion (Novick et al. 1980; Novick and
Schekman, 1979; Schekman, 2002; Schekman et al. 1983; Schekman and Novick, 2004). In these cells,
inhibition of protein secretion at high (non-permissive) temperature is usually accompanied by morpho-
logical and biochemical changes, as well as intracellular vesicle accumulation. Other so-called ‘conven-
tional’ mechanisms of secretion involve, for instance, ATP binding cassette type transporters, which are
common to both eukaryotes and prokaryotes (Davidson and Maloney, 2007; Niimi et al. 2005).
Lipid Insights 2008:2 27–40
28
Rodrigues et al
Lipid Insights 2008:2
Proteins that do not use the classical ER-Golgi
pathway or membrane transporters can be secreted
through various nonclassical pathways, as
recently reviewed by (Nickel and Seedorf, 2008).
Non-classical protein secretion may require vesicle
release to the extracellular space, in a process that
involves the formation of the so-called exosomes.
During exosome biogenesis, small vesicles are
formed by membrane invagination within endo-
cytic compartments (endosomes). The formation
of internal vesicles in the lumen of endosomes
generates the so-called multivesicular bodies,
which usually fuse with lysosomes in degradation
pathways. However, multivesicular bodies can also
fuse with the plasma membrane, resulting in the
release of internal vesicles to the extracellular
milieu as exosomes (Keller et al. 2006).
In contrast to most eukaryotic cells, fungi and
bacteria are cell wall-containing organisms, making
secretion topologically more complex. The presence
of the cell wall, at the very least, implies the exis-
tence of trans-cell wall mechanisms for the release
of molecules to the extracellular space. In prokary-
otes, the mechanisms of transport of proteins across
the cell wall are multiple. A general protein secre-
tion pathway involving multiple genes (SECA,
SECY, SECE, and SECG) and a number of accessory
proteins has been rmly documented (Mori and Ito,
2001; Veenendaal et al. 2004). In Gram-negative
bacteria, secretion systems have been widely
reported (reviewed in (Saier, 2006)). This secretion
machinery operates to release proteins extracellu-
larly and, in the case of some pathogens, to inject
toxins within host cells. At least six types of bacte-
rial secretion systems have been de ned, i.e. type I
to type VI. Comprehensive reviews on this topic
are available in the literature (Bingle et al. 2008;
Cianciotto, 2005; Craig and Li, 2008; Henderson
et al. 2004). Other protein secretion pathways in
bacteria include two-partner secretion systems
(Mazar and Cotter, 2007), which export large
exoproteins across the outer membranes using
channel-forming β-barrel proteins, twin-arginine
transporters (Sargent, 2007), a membrane-bound
transport apparatus that translocate proteins in a
fully folded conformation, and secretion of extracel-
lular vesicles (Mashburn-Warren et al. 2008).
Vesicle-mediated toxin delivery is a potent virulence
mechanism exhibited by diverse Gram-negative
pathogens.
Trans-cell wall secretion in fungi has remained
poorly explored for many years. Although it is well
known that fungal cells secrete molecules of
different chemical natures and molecular masses,
the mechanisms by which extracellular structures
leave intracellular compartments and cross the cell
wall are virtually unknown. As typical eukaryotic
organisms, fungal cells use conventional pathways
of secretion involving post-Golgi vesicles that fuse
with the plasma membrane to release their cargo
(Schekman, 2002; Schekman and Novick, 2004).
In fact, it is well known that yeast cells continu-
ously secrete a number of enzymes that remain
localized in the periplasm (Wickner and Schekman,
2005). The discovery that the capsular polysac-
charide of C. neoformans had a molecular weight
that could exceed 1 million Daltons (McFadden
et al. 2006b), however, revealed the need for con-
sidering new mechanisms of trans-cell wall trans-
port mechanism that could deliver macromolecules
from the periplasmic space outside of the cell.
Recent studies reported the characterization
of extracellular vesicles in pathogenic and
non-pathogenic species of fungi. C. neoformans,
Histoplasma capsulatum, Candida albicans,
Candida parapsilosis, Sporothrix schenckii and
S. cerevisiae were demonstrated to produce extra-
cellular vesicles containing lipid, polysaccharide
and protein components (Albuquerque et al. 2008;
Rodrigues et al. 2008; Rodrigues et al. 2007).
Therefore, extracellular vesicle secretion may
represent a eukaryotic solution to the problem of
trans-cell wall transport. Remarkably, the vesicles
produced by C. neoformans and H. capsulatum
contain key virulence determinants (Albuquerque
et al. 2008; Rodrigues et al. 2008; Rodrigues et al.
2007), suggesting that, as described for bacteria
(Mashburn-Warren et al. 2008), extracellular
vesicles in fungi may represent an ef cient mech-
anism of virulence factor delivery that may be
crucial for the success of the infection. In this
review, we discuss different models of extracel-
lular vesicle secretion, as well as putative pathways
of biogenesis and the impact of vesicle excretion
on fungal pathogenesis.
Extracellular Vesicles and Trans-Cell
Wall Transport: The Cryptococcus
Neoformans Model of Polysaccharide
and Protein Export
The most distinctive characteristic of the yeast
pathogen C. neoformans is the expression of a
polysaccharide capsule, a common feature of
29
Trans-cell wall secretion in fungi
Lipid Insights 2008:2
prokaryotic pathogens which is usually not
observed in eukaryotic microbes. Another particu-
larity of C. neoformans is the fact that the synthe-
sis of capsular polysaccharides occurs in the
cytoplasm (Feldmesser et al. 2001; Garcia-Rivera
et al. 2004; Yoneda and Doering, 2006). In
prokaryotes, capsule synthesis usually occurs at
surface and extracellular sites. In Escherichia coli,
for instance, capsular polysaccharides are initially
assembled by enzymes associated with the
plasma membrane (Whit eld, 2006; Whit eld
et al. 2003).
The capsule of C. neoformans is primarily
composed of two polysaccharides, namely gluc-
uronoxylomannan (GXM) and galactoxylomannan
(GalXM) (McFadden et al. 2006a). GXM, the best
studied capsular component of C. neoformans, is
the main virulence factor in this pathogen
(McClelland et al. 2005). GXM is not only surface
associated, but also continuously secreted to the
extracellular space where it accumulates in tissues
(McFadden et al. 2006a). Interestingly, GXM has
a molecular mass in the range of 1 to 7 million
Daltons (McFadden et al. 2006b), suggesting the
existence of mechanisms of transport of the poly-
saccharide from intracellular sites to the extracel-
lular environment. Early and recent studies
suggested that the transfer of intracellular polysac-
charides to the extracellular space in fungi could
require vesicular transport.
Vesicular transport in fungi was hinted at in
several earlier reports. Heath and colleagues
showed almost 40 years ago that fungal vesicles
contained cell wall precursors, suggesting that
intracellular vesicles can migrate to the cell surface
to release substrates for wall synthesis (Heath et al.
1971). More recently, the cell wall of C. neoformans
was described as the major cellular site of the
glycosphingolipid glucosylceramide (Rodrigues
et al. 2000), which is a membrane component of
vesicles that migrate from the plasma membrane
to the cell wall (Nimrichter et al. 2005b; Rodrigues
et al. 2007; Rodrigues et al. 2000). Early studies
by Takeo and co-workers also demonstrated that
C. neoformans secrete vesicles outside the cell
membrane (Takeo et al. 1973a, b). Interestingly,
despite the fact that no molecular evidence was
provided at that time, the authors proposed that
“capsular material is synthesized in and released
via the vesicles”. There the story rested for almost
20 years, until Feldmesser et al. used immunogold
electron microscopy and noticed clumped labeling
of the cell wall consistent with vesicular transport
of GXM (Feldmesser et al. 2001). Those observa-
tions were further extended by that of Garcia-
Rivera and colleagues who showed that antibodies
to GXM recognized cytoplasmic and cell wall
clusters resembling vesicles (Garcia-Rivera et al.
2004). In essence, these studies provide support
for the suggestion that the capsular polysaccharide
was synthesized in the cytoplasm and exported to
the exterior of the cell in secretory vesicles that
traversed the cell wall.
Based on the classical studies of Schekman and
Novick on yeast mutants defective in protein
synthesis (Novick et al. 1980; Novick and
Schekman, 1979; Schekman, 2002; Schekman
et al. 1983; Schekman and Novick, 2004), Yoneda
and Doering (Yoneda and Doering, 2006) used site-
directed mutagenesis to generate a C. neoformans
strain defective in the production of Sav1p, a
homolog of the S. cerevisiae small GTPase Sec4p.
In S. cerevisiae, sec4 mutants accumulate post-
Golgi vesicles under restrictive conditions
(Walworth et al. 1989), a morphological feature
that was also observed in the C. neoformans mutant
(Yoneda and Doering, 2006). More precisely, the
sav1 mutant of C. neoformans had defective
protein secretion and accumulated exocytic vesi-
cles at the septum and the bud during cell division.
Strikingly, these vesicles were strongly recognized
by an antibody to GXM (Yoneda and Doering,
2006), confirming and extending previous
suppositions that the polysaccharide was synthe-
sized intracellularly and secreted in vesicles.
Since the capsule of C. neoformans enlarges by
apical growth (Zaragoza et al. 2006), one might
deduce that capsular components are secreted to
the extracellular space and incorporated into the
external layers of the growing capsule. Therefore,
assuming that secretion of intracellularly synthe-
sized GXM is an essential element of capsular
growth, one might hypothesize that extracellular
vesicles lled up with capsular components may
exist in C. neoformans culture supernatants.
Indeed, cell wall-bound vesicles, as well as extra-
cellular vesicles in association with surface struc-
tures, were observed in vitro and during animal
infection by C. neoformans (Rodrigues et al. 2007).
In addition, the fractionation of culture superna-
tants using centrifugation procedures previously
designed for the puri cation of secreted vesicles
in mammalian systems allowed the isolation of
extracellular lipid-containing fractions. The analysis
30
Rodrigues et al
Lipid Insights 2008:2
of these fractions by transmission electron
microscopy revealed the presence of round vesi-
cles, in the range of 20 to 400 nm, containing
bilayered membranes (Rodrigues et al. 2008;
Rodrigues et al. 2007). Vesicle morphology
included usually round compartments with differ-
ent levels of electron-density. Vesicular structures
with hyper-dense structures resembling a dark
pigment were also observed. Importantly, some of
the isolated vesicles were recognized by an anti-
body to GXM (Rodrigues et al. 2007), con rming
that these compartments were involved in polysac-
charide trans-cell wall secretion. In addition, GXM
detection in vesicles was correlated with capsule
enlargement, suggesting that polysaccharide deliv-
ery in extracellular vesicles is necessary for assem-
bly of the capsule (Rodrigues et al. 2007).
The production of extracellular vesicles requires
cell viability (Rodrigues et al. 2007), con rming
that these compartments are actively secreted
rather than passively released by dead cells. Trans-
mission electron microscopy of intact C. neofor-
mans cells also revealed that the secretory vesicles
were recognized by an antibody to glucosylce-
ramide (Rodrigues et al. 2007). Since glucosylce-
ramide is an important regulator of growth and
virulence in C. neoformans (Rittershaus et al. 2006;
Rodrigues et al. 2000), the possibility that the
extracellular vesicles carry bioactive lipids was
raised. The analysis of lipid components of isolated
extracellular vesicles by chromatographic and
spectrometric methods revealed the presence of
glucosylceramide (GlcCer) and other structures
(Rodrigues et al. 2007). By thin-layer chromatog-
raphy, bands with migration rates corresponding
to GlcCer and ergosterol were detected in lipid
extracts of vesicles produced by both acapsular
and encapsulated C. neoformans cells. Vesicle
lipids from encapsulated cells were also examined
by electrospray ionization mass spectrome-
try, revealing a complex lipid composition.
GlcCer analysis in vesicle fractions revealed the
presence of N-2’-hydroxyhexadecanoyl- and N-2’-
hydroxyoctadecanoyl-1-beta-
D-glucopyranosyl-9-
methyl-4,8-sphingadienine. Analysis of sterols
showed that ergosterol and 4,14-dimethylergosta-
24(24
1
)-en-3β-ol, an obtusifoliol-like molecule,
are also vesicle components in C. neoformans
(Rodrigues et al. 2007).
In addition to polysaccharides and lipids,
the extracellular vesicles of C. neoformans also
carried a complex array of proteins to the
extracellular milieu. Employing a proteomic
approach, 76 different proteins were identi ed as
vesicle components (Rodrigues et al. 2008).
Chaperones, including heat shock proteins and
superoxide dismutase, signal transduction regula-
tors, anti-oxidant and cytosolic proteins and
enzymes were identified. It is intriguing that
approximately one third of proteins identi ed in
the C. neoformans vesicles corresponded to
molecules that were previously described to
compose mammalian exosomes (Rodrigues
et al. 2008).
Vesicular Trans-Cell Wall Transport
in Histoplasma Capsulatum
and Other Fungal Species
Following the identi cation of extracellular vesi-
cles in the basidiomycetes C. neoformans, fungi
in the phylum Ascomycota were examined for
vesicle production. The ascomycetes H. capsula-
tum, C. albicans, C. parapsilosis, S. schenckii, and
S. cerevisiae each were found to generate hetero-
geneous extracellular vesicles (Albuquerque et al.
2008). Transmission electron microscopy studies
of supernatants subjected to fractional centrifuga-
tion revealed that each fungus produced extracel-
lular spherical, bilayered vesicles that varied in
their electron density and ranged in diameter from
10 to 350 nm. Vesicles were not present in super-
natants from media inoculated with dead fungal
cells. Notably, the percentage of large versus small
vesicles varied signi cantly between the different
ascomycetes. For example, only 4% of S. cerevi-
siae vesicles were larger than 50 nm compared to
38 and 54% of vesicles from H. capsulatum and
C. parapsilosis, respectively.
The movement of membrane-bound vesicles
across C. albicans cell walls was previously
proposed by Anderson and co-workers (Anderson
et al. 1990) who demonstrated the presence of
“pimples” in the cell wall of opaque switch variants
of strain WO-1 where vesicles were identi ed
within some channels or appeared to emerge from
the “pimples” with their double membrane intact.
The analysis of intact H. capsulatum yeast cells by
transmission electron microscopy similarly
demonstrated vesicular structures engaging the
internal aspects of the cell wall, within the cell
wall, and emerging from the cell wall consis-
tent with trans-cell wall transport of vesicles
(Albuquerque et al. 2008).
31
Trans-cell wall secretion in fungi
Lipid Insights 2008:2
To further define the constituents of the
ascomycetes vesicles, purified fractions from
H. capsulatum were subjected to lipidomic and
proteomic analyses (Albuquerque et al. 2008).
Using electrospray ionization time-of- ight mass
spectrometry, phosphatidylethanolamine, phospha-
tidylserine (PS), and phosphatidylcholine (PC)
were identi ed as the major phospholipid species
comprising the vesicles, which is similar to the
typical distribution of lipids in pathogenic
yeast (Rattray et al. 1975). The protein compo-
nents of the vesicles were analyzed by cation
exchange chromatography and analyzed by liquid
chromatography-tandem mass spectrometry and
283 proteins were validated, with 206 proteins
identi ed by sequence analysis. Notably, several
proteins involved in H. capsulatum pathogenesis
and host immune responses were detected,
including chaperones (Hsp30, Hsp70, and Hsp60),
superoxide dismutase, and catalase B. Further-
more, proteins involved in signal transduction,
vesicle formation, cell wall and cytoskeleton
regulation, cell growth, and sugar, lipid and amino
acid metabolism were identi ed.
The fact that proteins associated with H. capsu-
latum virulence were identi ed in the puri ed
extracellular vesicles raised the question of whether
the vesicle proteins were recognized by the host
immune system. In this regard, hyperimmune
human serum reacted strongly with diverse vesic-
ular proteins, including Hsp60 and histone 2B.
Notably, H. capsulatum Hsp60 has been associated
with virulence (Allendoerfer et al. 1996; Deepe and
Gibbons, 2001; Deepe and Gibbons, 2002; Gomez
et al. 1995b; Scheckelhoff and Deepe, 2002) and
antibody to histone 2B can modify experimental
histoplasmosis (Nosanchuk et al. 2003). Hence, it
is likely that proteins transported via vesicles are
involved in host-pathogen interactions.
Putative Mechanisms of Vesicle
Diversity, Biogenesis and Passage
Through the Cell Wall
The ndings of Yoneda and Doering (Yoneda and
Doering, 2006) suggesting that GXM-containing
vesicles in C. neoformans were derived from the
Golgi apparatus was further supported by Hu and
co-workers (Hu et al. 2007). These authors
reported that exposure of C. neoformans to
brefeldin A resulted in a massive reduction of
capsule size. The target of brefeldin A in mammalian
cells is a subset of Sec7-type GTP-exchange
factors that catalyze the activation of a small
GTPase called Arf1p (Nebenfuhr et al. 2002).
Arf1, in turn, is responsible for the recruitment
of coat proteins to membranes, resulting in the
formation of transport vesicles (Scales et al.
2000). Arf1p and brefeldin A-sensitive GTP-
exchange factors are localized to the Golgi
apparatus of mammalian and yeast cells (Spang
et al. 2001). Therefore, it seems clear that GXM
assembly at the cell surface requires the secretion
of post-Golgi vesicles lled up with the polysac-
charide. These vesicles would be targeted to the
plasma membrane for exocytosis (Yoneda and
Doering, 2006), which would result in the release
of the polysaccharide to the periplasmic space.
Therefore, for capsule assembly, additional
mechanisms of trans-cell wall transport of GXM
would be required. However, a model of exocy-
tosis with discharge into the periplasmic space
did not account for transport of GXM across the
cell wall for assembly into a capsule in the
extracellular space.
Mutation of the SEC4 homolog that resulted in
the cytoplasmic accumulation of post-Golgi vesi-
cles did not lead to any apparent alteration in the
capsular expression phenotype (Yoneda and
Doering, 2006). This observation could be a
consequence of continued polysaccharide secretion
before the temperature shift, which blocks secre-
tion, or incomplete inhibition of the secretion
caused by the mutation. It was impossible to
distinguish in C. neoformans whether GXM was
targeted to the cell surface exclusively in post-
Golgi vesicles or via recycling endosomes. In fact,
endosomes and related multivesicular bodies
(MVBs) can be connected to the trans-Golgi secre-
tory pathway, so both pathways could be involved
in polysaccharide secretion in C. neoformans.
It is therefore possible that the post-golgi
GXM-enriched vesicles are sorted to another
compartment than the plasma membrane such as
the late endosomes and the MVBs.
The existence of MVB-like structures in
C. neoformans was suggested in early and recent
studies (Rodrigues et al. 2008; Takeo et al.
1973a, b) (Fig. 1). Some of these compartments
were found to be clearly merging with the plasma
membrane, indicating that the extracellular vesicles
of C. neoformans could be related to mammalian
exosomes. Exosomes are extracellular vesicles
derived from the fusion of MVBs with the plasma
32
Rodrigues et al
Lipid Insights 2008:2
membrane. Their isolation was extensively
described in several animal cell models such as
reticulocytes, dendritic cells, B lymphocytes,
mastocytes and epithelial cells (Johnstone et al.
1987; Lamparski et al. 2002; Raposo et al. 1996;
Skokos et al. 2003; van Niel et al. 2001).
The molecular machinery implied in MVB
formation and sorting is widely known in
S. cerevisiae (Hurley and Emr, 2006), but these
studies had never shown extracellular exosomes
in fungal cells. Early observations by Takeo and
colleagues (Takeo et al. 1973a) suggested the
occurrence of what the authors de ned as an “early
stage of formation of spherical invaginations which
secrete the vesicles outside the cell membrane”,
suggesting the occurrence of exosomes in
fungi. That supposition was strengthened by the
highly heterogeneous protein composition of
C. neoformans and H. capsulatum vesicles
(Albuquerque et al. 2008; Rodrigues et al. 2008).
This heterogeneous protein pro le is also seen in
mammalian exosomes, indicating a possible
common origin for both extracellular vesicles.
Moreover, detergent-resistant lipid microdomains
are known to compose exosome membranes at
least in some mammalian cell types (de Gassart
et al. 2003). Lipid microdomains—or lipid
rafts – are mainly characterized by high sterol
content and presence of glycosphingolipids, as
described in fungal vesicles (Rodrigues et al.
2007). They are seemingly involved on the
sorting of exosomal proteins as well as in
vesicle morphology and formation (de Gassart
et al. 2003).
Laccase, an enzyme involved in melanin
synthesis, is another important component of
C. neoformans vesicles (Rodrigues et al. 2008).
Interestingly, it was recently shown that a
C. neoformans mutant defective in the vps34 gene
has a considerable decrease on melanin expression
(Hu et al. 2008). Vps34 (vacuolar protein sorting 34)
is involved in vesicular sorting of vacuolar hydro-
lases from the trans-Golgi-network to the late
endosomes and in the internal vesicle formation
within MVBs (Futter et al. 2001; Kihara et al.
2001). These data strongly support the idea that
laccase-containing vesicles originate from the
endocytic pathway.
CW PM
Cy
100 nm
MVB (?)
Figure 1. Typical MVB-like structures in fusion with the plasma membrane in C. neoformans. MVB internal vesicles (arrowheads) are released
outside the external layer of the plasma membrane, representing a potential mechanism of exosome biogenesis in fungi. This gure was
originally published by Takeo and colleagues (Takeo et al. 1973b). Reproduced with permission from the American Society for Microbiology.
CW, cell wall; PM, plasma membrane; Cy, cytoplasm; MVB, multivesicular body.
33
Trans-cell wall secretion in fungi
Lipid Insights 2008:2
Although different lines of evidence point to a
relationship between fungal extracellular vesicles
and exosomes, other possibilities can not be ruled
out. The methods currently used for vesicle puri-
cation do not discriminate between vesicles of
different origins, resulting in heterogeneous
preparations. Indeed, electron microscopy com-
bined to other approaches demonstrated that
vesicle fractions contain different subpopulations
which differ in morphological characteristics
(Rodrigues et al. 2008), suggesting the existence
of different intracellular compartments involved
in vesicle biogenesis (Fig. 2). Cellular vesicle
secretion strategies other than exosome formation
(Fig. 2B) could consist of simple membrane budding
(Fig. 2C) and other unconventional mechanisms
of vesicle release (Fig. 2D). The existence of
different mechanisms of vesicle biogenesis could,
in turn, result in the simultaneous generation of
extracellular membrane compartments of different
cellular origins.
Further investigations at the molecular and ultra-
structural levels will need to be done for the devel-
opment of reliable concepts of vesicle biogenesis.
In this context, the discovery of biochemical
markers for different vesicle sub-populations is
likely to be fundamental criterion for vesicular
identi cation and characterization. In this scenario,
genetic approaches will be powerful tools to under-
stand where the vesicles are assembled and how
they reach the extracellular space.
It is unknown how vesicles cross the cell wall to
reach the extracellular space. Transmission
electron microscopy revealed that the cell wall
of C. neoformans and H. capsulatum contain
numerous vesicle-like structures (Rodrigues et al.
2008; Albuquerque et al. 2008). The cell wall is now
described as a compact although malleable structure
AB
C
D
Periplasm
cell wall
cytoplasm
Figure 2. Mechanisms for vesicle biogenesis in fungal cells. (A) Vesicles sorted to the cell surface by conventional secretion fuse with the
plasma membrane, releasing their cargo into the periplasm. MVB formation followed by fusion with the plasma membrane (B) would result
in the extracellular release of exosomes. Membrane budding (C) could also result in the periplasmic release of vesicles, as also proposed
for other unconventional secretion mechanisms (D). For conceptual clari cation, see (Kinseth et al. 2007) and (Keller et al. 2006). Of note,
vesicle content could vary according with biogenesis pathways. The possibility that different mechanisms result in the simultaneous generation
of extracellular vesicles cannot be ruled out.
34
Rodrigues et al
Lipid Insights 2008:2
(Nimrichter et al. 2005a), likely to suffer rearrangements,
that could allow vesicle passage. Indeed, it was
demonstrated by atomic force microscopy that the
cell wall of fungi contain pores that would allow
vesicle passage (de Souza Pereira and Geibel, 1999;
Eisenman et al. 2005). Trans-cell wall vesicle secre-
tion could also involve vesicle-mediated cell wall
remodeling. For example, H. capsulatum extracel-
lular vesicles contain enzymes regulating synthesis
and hydrolysis of cell wall components (Albuquerque
et al. 2008). In yeast cells, wall remodeling for
vesicle passage could be facilitated during budding,
when the cell wall is thinner in the bud than in non-
dividing areas of the cell surface (Linnemans et al.
1977). Finally, a myosin analog was described as a
cell wall component of Aspergillus fumigatus
(Esnault et al. 1999). In the cytosolic space, motor
proteins like myosin and dynein contribute with the
movement of transport vesicles (Schliwa and Woe-
hlke, 2003). However, it is completely unknown if
such proteins would be functional at the cell wall
microenvironment.
‘Virulence Bags’: Fungal Extracellular
Vesicles Concentrate Fungal
Molecules Involved in Pathogenesis
It is clear that fungal vesicles are released to the
extracellular milieu carrying a complex panel of
proteins and lipids and, in the case of C. neoformans,
polysaccharides (Rodrigues et al. 2007; Rodrigues
et al. 2008). Current data strongly suggest that vesicle
secretion occurs not only in vitro, but also in vivo
during animal infections by fungal pathogens.
Secreted vesicles were detected when murine mac-
rophages were incubated with C. neoformans yeasts
and also in sections of lung excised from mice infected
with this pathogen (Rodrigues et al. 2007). Vesicles
secreted during infection could directly mediate host
cell damage and/or modulation of immune response.
Indeed, similar functions were proposed for vesicles
released from the outer membrane of Gram negative
bacteria (Mashburn-Warren et al. 2008).
Lipid components of fungal vesicles have the
potential to modulate interactions of fungi with their
hosts. GlcCer is required for fungal growth in
alkaline pH and high CO
2
levels, as well as for the
virulence of C. neoformans (Rittershaus et al. 2006;
Saito et al. 2006). The fact that the vesicles secreted
by S. cerevisiae, which does not synthesize
GlcCer, are in a smaller size range (Albuquerque
et al. 2008) suggests an involvement of GlcCer in
vesicle assembly. As a vesicle-associated compound,
GlcCer could be constantly delivered to host cells,
facilitating the production of antimicrobial antibodies
(Rodrigues et al. 2000). In addition, lipid turnover
is known to be a well regulated phenomenon that
occurs in mammalian cells. Glycosphingolipids are
also involved with sorting of lipids and proteins in
mammalian cells (Degroote et al. 2004). Although
fungal and mammalian GlcCer are structurally dif-
ferent (Barreto-Bergter et al. 2004), secreted fungal
GlcCer could potentially interfere with recycling
and sorting of lipids and proteins in host cells. Other
lipids, including phosphatidylcholine, phosphatidyl-
ethanolamine and phosphatidylserine, were also
characterized in fungal vesicles (Albuquerque et al.
2008). Although these are common components of
biological membranes, their involvement in immune
responses has been already reported. Liposomes
carrying phosphatidylserine, for instance, can
modulate cytokine production, decrease microbial
killing and inhibit nitric oxide production by
macrophages (Aramaki, 2000; Gilbreath et al. 1986;
Hoffmann et al. 2005).
In bacteria, adhesins, toxins, and immunomodula-
tory compounds are the components of extracellular
vesicles, which directly mediate bacterial binding
and invasion, cause cytotoxicity, and modulate the
host immune response (Kuehn and Kesty, 2005).
In fungal vesicles, several proteins involved with
fungal pathogenesis were identi ed (Albuquerque
et al. 2008; Rodrigues et al. 2008). Concentration of
pathogenic determinants in the vesicles could provide
an ef cient mechanism of release of virulence factors
into host tissues. Vesicle membranes could also
protect fungal structures against hydrolysis by host
extracellular enzymes. In addition, membrane fusion
involving vesicle elements and the surface of host
cells could result in the direct delivery of fungal
structures into host tissues.
Different immunogenic compounds are released
in fungal vesicles. Chaperones, including heat
shock proteins 60 and 70 from H. capsulatum and
C. neoformans, respectively, were detected in
extracellular vesicles by proteomics and serology
(Albuquerque et al. 2008; Rodrigues et al. 2008).
Hsp60 is responsible for binding of H. capsulatum
to the CD11/C18 receptor in human macrophages
(Long et al. 2003). This protein, which was initially
described as a glycoprotein present at membrane
and cell wall of H. capsulatum, confers protec-
tion against lethal intravenous challenge with
this pathogen (Gomez et al. 1995a). Besides
35
Trans-cell wall secretion in fungi
Lipid Insights 2008:2
representing an ef cient vehicle for delivery of
immunogens during infection, vesicles carrying
Hsp60 could sequestrate anti-Hsp60 antibodies
produced by the host. In C. neoformans, the role
of Hsp70 is still unknown, but this protein is
considered to be a major immunogen in cryptococ-
cosis (Kakeya et al. 1997; Kakeya et al. 1999).
Pathogenic-related enzymes have also been
described as components of fungal extracellular
vesicles (Table 1) (Albuquerque et al. 2008;
Rodrigues et al. 2008). A number of proteins with
general anti-oxidant functions are also contained in
the extracellular vesicles, suggesting that vesicle
secretion could be bene cial for intracellular parasit-
ism and fungal persistence within host tissues.
Virulence factors of C. neoformans such as urease
and laccase were also vesicle associated (Rodrigues
et al. 2008). Interestingly, some of the vesicles had
electron dense structures consistent with pigments.
Laccase is responsible for melanin synthesis in
many fungal species, but it also interferes with
prostaglandin metabolism (Erb-Downward et al.
2008). Prostaglandins are lipid mediators that
regulate components of the immune response,
including defense against infection. For instance,
PGE
2
inhibits B and T lymphocyte proliferation, as
well as macrophage functions (Chouaib et al. 1985;
Simkin et al. 1987; Taffet et al. 1981; Taffet and
Russell, 1981; Xu et al, 2008). Prostaglandin produc-
tion by laccase present in vesicles has the potential
to negatively modulate the immune response.
As discussed in previous sections, cryptococcal
GXM is also secreted within vesicles that reach
the extracellular space. GXM is recognized by
different cell types and involved with a series of
immunomodulatory effects (Vecchiarelli, 2005).
Polysaccharide vesicular export, therefore, could
be not only a physiological solution for capsule
assembly, but also a fungal strategy to cause tissue
damage through the delivery of virulence factors
and defense proteins into host tissues. Interest-
ingly, two different enzymes with well-de ned
roles in capsule synthesis and virulence were also
found in cryptococcal vesicles (Rodrigues
et al. 2008).
Unsolved Problems and Future
Perspectives
The discovery of a trans-cell wall vesicular trans-
port system in C. neoformans, and its subsequent
extension to ascomycetous fungi, provides a
Table 1. Pathogenic determinants found in fungal extracellular vesicles.
Virulence
determinant
Function Organism
Catalases A and B Anti-oxidant defense C. neoformans (CatA)
and H. capsulatum (CatB)
Glucosylceramide Fungal growth C. neoformans
GXM Immunomodulation C. neoformans
Heat shock proteins Immunogen; adhesion
to host cells
C. neoformans
and H. capsulatum
Laccase Pigment synthesis;
prostaglandin metabolism
C. neoformans
Superoxide dismutase Anti-oxidant defense C. neoformans
and H. capsulatum
Thiol-speci c
antioxidant protein
Anti-oxidant defense C. neoformans
and H. capsulatum
Thioredoxin Anti-oxidant defense C. neoformans
Thioredoxin reductase Anti-oxidant defense C. neoformans
UDP-glucose
dehydrogenase
Conversion of UDP-glucose
into UDP-glucuronic acid
(capsule synthesis)
C. neoformans
UDP-glucuronic acid
decarboxylase
Converts UDP-glucuronic
acid to UDP-xylose
(capsule synthesis)
C. neoformans
36
Rodrigues et al
Lipid Insights 2008:2
solution to the problem of exporting macromole-
cules to the cellular exterior. The nding of similar
vesicles in ascomycetes and basidiomycetes
suggest that the vesicular transport system is
ancient, such that its existence predated the diver-
gence of these fungal branches 0.5–1.0 billion
years ago. For C. neoformans the existence of such
an export mechanism would appear to be necessary
for capsular synthesis given this structure is
composed of macromolecules with mass that can
exceed 1 million Daltons (McFadden et al. 2006b).
The nding that such vesicles contain numerous
fungal products associated with virulence suggests
that they also have a concentrative function that
could allow these compounds to function more
ef ciently, and possibly synergistically. However,
the discovery of this trans-cell wall vesicular trans-
port system also poses a new set of questions for
future studies.
How many types of vesicles
are there?
Electron microscopy reveals that vesicles differ
in shape, size, and inter-vesicular contents.
EM surveys suggest the existence of at least four
types of vesicles in C. neoformans supernatants
(Rodrigues et al. 2008). Classifying vesicular
types is important because each type would sug-
gest the existence of a separate cellular synthetic
mechanism. Currently, vesicle types have been
discriminated based on morphology and size.
However, morphological criteria may not be
adequate to ascertain the various types of vesicles
since similar appearance does not necessarily
imply similar content. Consequently, continued
progress in this area will require the develop-
ment of methods for vesicular fractionation and
chemical characterization. Furthermore, a full
description of the vesicular transport system will
require a correlation of vesicular content with
morphological characteristics, which is likely to
involve combined proteomic analysis, serological
reactivity, enzymatic assays, etc.
Where do vesicles originate from?
Analysis of protein components and the nding of
vesicles within vacuolar structures led to the
proposal that extracellular vesicles originate in
exosome-like structures (Rodrigues et al. 2008).
However, such structures have yet to be described
in fungi. De nition of the various vesicle types
combined with mutational analysis could be used to
ascertain the number of vesicular synthetic pathways
in fungal cells. Understanding the sites of vesicular
synthesis and the relationship of extracellular vesi-
cles to cytoplasmic vesicles will undoubtedly be a
major area of investigation in future years.
How are vesicles transported across
the cell wall?
There is no information on the mechanism used to
shuttle vesicles across the cell wall. The existence
of pores on the cell wall with dimensions that
approximate those of vesicles suggest that these
could be used for vesicular transport. Vesicular
transport across pores raises a variety of other
questions including the energy dynamics of such
processes and the types of motors and gears that
would be needed to shuttle such large structures
across rigid cell walls. This is made even more
complex when one considers that certain fungi,
such as C. neoformans, H. capsulatum and
S. schenckii can form dense melanin layers within
their cell walls (for review, (Nosanchuk and
Casadevall, 2006)).
What is the fate of released vesicles?
In the case of C. neoformans, vesicular transport
has been proposed as the mechanism to deliver
macromolecules for capsular assembly to the exte-
rior of the cell (Rodrigues et al. 2007). If that is
the case, it raises the question of how vesicles are
directed to the capsule surface and their cargo
unloaded. Extracellular phospholipases may
degrade phospholipids membranes and allow cargo
discharge. C. neoformans and most pathogenic
fungi have extracellular phospholipase activity
(Chayakulkeeree et al. 2008; Ghannoum, 1998;
Hruskova-Heidingsfeldova, 2008; Simockova
et al. 2008). A related issue is the relative paucity
of vesicles relative to the number of cells in solu-
tion. This could be interpreted as indicating that
vesicles are relatively short lived structures that
release their contents in the extracellular space. It
is possible that vesicles recovered from superna-
tants re ect a very small minority of the secreted
vesicles and that isolation of the overwhelming
majority of vesicles produced by fungal cells would
require the development of more sophisticated
recovery techniques. Hence, understanding the
37
Trans-cell wall secretion in fungi
Lipid Insights 2008:2
dynamics of vesicular transport will require studies
on the fate of released vesicles.
What is the relationship between
vesicles and host cell toxicity?
C. neoformans intracellular pathogenesis is
characterized by the appearance of numerous
vesicles in the cytoplasm of phagocytic host cells
that contain cryptococcal capsular polysaccharide
(Feldmesser et al. 2000). These vesicles are
believed to be toxic to the host cells since their
accumulation is temporally correlated with cyto-
toxic changes that include membrane blebs and
phagosomal membrane leakiness (Feldmesser et al.
2000; Tucker and Casadevall, 2002). The discovery
of a vesicular transport system for C. neoformans
raises the possibility that these cytoplasmic vesicles
originated as fungal vesicles that were released from
yeast-containing phagosomes. Hence, knowing
about the effect of shed vesicles on host cells and
their relationship to the polysaccharide-containing
vesicles that accumulate in host cells would seem
to be an important question for understanding the
pathogenesis of cryptococcal infections. Further-
more, it is possible that shed vesicles are also
involved in the pathogenesis of other facultative
intracellular fungi such as H. capsulatum.
What is the internal organization
of vesicular components?
Given that vesicles contain multiple fungal compo-
nents that include lipids, polysaccharides, carbohy-
drates and proteins, it is possible that these are
packaged inside vesicles in an organized manner that
could reflect their addition following or during
vesicular synthesis. For example, it is conceivable
that vesicles destined for capsular synthesis contain
capsular components arranged in a manner that
facilitates their function. Understanding the packaging
characteristics of vesicles is likely to be an essential
aspect of understanding their function and mecha-
nisms of action. Such studies will almost certainly
require the application of new microscopy techniques
in combination with speci c reagents, such as mono-
clonal antibodies, that delineate the position of spe-
ci c components within the vesicular structure.
Finally, there is the notion that knowledge of
vesicular transport systems may be exploited in
the design of new types of antifungal drugs and
vaccines. The complex choreography of vesicular
transport implies the existence of numerous steps
that could be targets of drug development. One could
imagine drugs that reduce the virulence of fungi by
interfering with vesicular synthesis and transport.
Identi cation of common pathways used by differ-
ent pathogenic fungi for vesicular transport could
lead to the design of broad-spectrum antifungal
drugs. Since trans-cell wall vesicular transport
mechanisms have no counterpart on mammalian
cells such drugs could potentially have high speci-
city for fungal cells while concomitantly having
limited host cells toxicity. Furthermore, it is possible
that vesicular preparations can be developed into
vaccine formulations. The nding of many fungal
components associated with virulence in vesicles
and the fact that vesicle-related proteins are immu-
nogenic (Albuquerque et al. 2008; Rodrigues et al.
2008) suggest that vesicle preparations and/or their
components could have utility as fungal vaccines.
In this regard, it is noteworthy that exosome prepa-
rations from dendritic cells are been investigated as
potential anti-cancer vaccines (Hao et al. 2007).
It is now apparent that the discovery of vesicles in
cryptococcal culture supernatants has opened a new
window into fungal cell biology that promises to
revolutionize our thinking on fungal product export
and its impact on pathogenesis. It is also clearly appar-
ent that this area of investigation is extremely complex
and will require the development of new experimen-
tal tools and reagents for continued progress.
Acknowledgements
MLR and LN are supported by grants from the
Brazilian agencies FAPERJ and CNPq. AC is
supported by NIH grants AI033142, AI033774,
AI052733, and HL059842. JDN is supported by
NIH AI52733 and AI056070-01A2. We are
thankful to Igor C. Almeida, Rosana Puccia and
Luiz R. Travassos for helpful discussions.
Disclosure
The authors report no con icts of interest.
References
Albuquerque, P.C., Nakayasu, E.S., Rodrigues, M.L., Frases, S.,
Casadevall, A., Zancope-Oliveira, R.M., Almeida, I.C. and
Nosanchuk, J.D. 2008. Vesicular transport in Histoplasma capsulatum:
an effective mechanism for trans-cell wall transfer of proteins and
lipids in ascomycetes. Cell. Microbiol.
Allendoerfer, R., Maresca, B. and Deepe, G.S., Jr., 1996. Cellular immune
responses to recombinant heat shock protein 70 from Histoplasma
capsulatum. Infect. Immun., 64:4123–8.
38
Rodrigues et al
Lipid Insights 2008:2
Anderson, J., Mihalik, R. and Soll, D.R. 1990. Ultrastructure and antigenicity
of the unique cell wall pimple of the Candida opaque phenotype.
J. Bacteriol., 172:224–35.
Aramaki, Y. 2000. Liposomes as immunomodulator—inhibitory effect of
liposomes on NO production from macrophages. Biol. Pharm. Bull.,
23:1267–74.
Barreto-Bergter, E., Pinto, M.R. and Rodrigues, M.L. 2004. Structure and
biological functions of fungal cerebrosides. An Acad. Bras. Cienc.,
76:67–84.
Bingle, L.E., Bailey, C.M. and Pallen, M.J. 2008. Type VI secretion: a
beginners guide. Curr. Opin. Microbiol., 11:3–8.
Chayakulkeeree, M., Sorrell, T.C., Siafakas, A.R., Wilson, C.F., Pantarat, N.,
Gerik, K.J., Boadle, R. and Djordjevic, J.T. 2008. Role and
Mechanism of Phosphatidylinositol-Speci c Phospholipase C in
Survival and Virulence of Cryptococcus neoformans. Mol.
Microbiol.
Chouaib, S., Welte, K., Mertelsmann, R. and Dupont, B. 1985. Prostaglandin
E2 acts at two distinct pathways of T lymphocyte activation: inhibi-
tion of interleukin 2 production and down-regulation of transferrin
receptor expression. J. Immunol., 135:1172–9.
Cianciotto, N.P. 2005. Type II secretion: a protein secretion system for all
seasons. Trends Microbiol., 13:581–8.
Craig, L. and Li, J. 2008. Type IV pili: paradoxes in form and function.
Curr. Opin. Struct. Biol., 18:267–77.
Davidson, A.L. and Maloney, P.C. 2007. ABC transporters: how small
machines do a big job. Trends Microbiol., 15:448–55.
de Gassart, A., Geminard, C., Fevrier, B., Raposo, G. and Vidal, M. 2003.
Lipid raft-associated protein sorting in exosomes. Blood,
102:4336–44.
de Souza Pereira, R. and Geibel, J. 1999. Direct observation of oxidative
stress on the cell wall of Saccharomyces cerevisiae strains with atomic
force microscopy. Mol. Cell Biochem., 201:17–24.
Deepe, G.S., Jr., and Gibbons, R. 2001. V beta 6+ T cells are obligatory for
vaccine-induced immunity to Histoplasma capsulatum. J. Immunol.,
167:2219–26.
Deepe, G.S., Jr., and Gibbons, R.S. 2002. Cellular and molecular regulation
of vaccination with heat shock protein 60 from Histoplasma
capsulatum. Infect. Immun., 70:3759–67.
Degroote, S., Wolthoorn, J. and van Meer, G. 2004. The cell biology of
glycosphingolipids. Semin. Cell. Dev. Biol., 15:375–87.
Eisenman, H.C., Nosanchuk, J.D., Webber, J.B., Emerson, R.J., Camesano,
T.A. and Casadevall, A. 2005. Microstructure of cell wall-associated
melanin in the human pathogenic fungus Cryptococcus neoformans.
Biochemistry, 44:3683–93.
Engel, L.S., Hill, J.M., Caballero, A.R., Green, L.C. and O’Callaghan, R.J.
1998. Protease IV, a unique extracellular protease and virulence fac-
tor from Pseudomonas aeruginosa. J. Biol. Chem., 273:16792–7.
Erb-Downward, J.R., Noggle, R.M., Williamson, P.R. and Huffnagle, G.B.
2008. The role of laccase in prostaglandin production by Cryptococcus
neoformans. Mol. Microbiol., 68:1428–37.
Esnault, K., el Moudni, B., Bouchara, J.P., Chabasse, D. and Tronchin, G.
1999. Association of a myosin immunoanalogue with cell envelopes
of Aspergillus fumigatus conidia and its participation in swelling and
germination. Infect. Immun., 67:1238–44.
Feldmesser, M., Kress, Y. and Casadevall, A. 2001. Dynamic changes in
the morphology of Cryptococcus neoformans during murine
pulmonary infection. Microbiology, 147:2355–65.
Feldmesser, M., Kress, Y., Novikoff, P. and Casadevall, A. 2000.
Cryptococcus neoformans is a facultative intracellular pathogen in
murine pulmonary infection. Infect. Immun., 68:4225–37.
Futter, C.E., Collinson, L.M., Backer, J.M. and Hopkins, C.R. 2001. Human
VPS34 is required for internal vesicle formation within multivesicular
endosomes. J. Cell Biol., 155:1251–64.
Garcia-Rivera, J., Chang, Y.C., Kwon-Chung, K.J. and Casadevall, A. 2004.
Cryptococcus neoformans CAP59 (or Cap59p) is involved in the
extracellular traf cking of capsular glucuronoxylomannan.
Eukaryot.
Cell, 3:385–92.
Ghannoum, M.A. 1998. Extracellular phospholipases as universal virulence
factor in pathogenic fungi. Nippon Ishinkin Gakkai Zasshi,
39:55–9.
Gilbreath, M.J., Hoover, D.L., Alving, C.R., Swartz, G.M., Jr., and
Meltzer, M.S. 1986. Inhibition of lymphokine-induced macrophage
microbicidal activity against Leishmania major by liposomes:
characterization of the physicochemical requirements for liposome
inhibition. J. Immunol., 137:1681–7.
Glick, B.S. and Malhotra, V. 1998. The curious status of the Golgi apparatus.
Cell., 95:883–9.
Gomez, F.J., Allendoerfer, R. and Deepe, G.S., Jr., 1995a. Vaccination with
recombinant heat shock protein 60 from Histoplasma capsulatum
protects mice against pulmonary histoplasmosis. Infect. Immun.,
63:2587–95.
Gomez, F.J., Allendoerfer, R. and Deepe, G.S., Jr., 1995b. Vaccination with
recombinant heat shock protein 60 from Histoplasma capsulatum
protects mice against pulmonary histoplasmosis. Infect. Immun.,
63:2587–95.
Gorelick, F.S. and Shugrue, C. 2001. Exiting the endoplasmic reticulum.
Mol. Cell Endocrinol., 177:13–8.
Hao, S., Moyana, T. and Xiang, J. 2007. Review: cancer immunotherapy
by exosome-based vaccines. Cancer Biother. Radiopharm.,
22:692–703.
Heath, I.B., Gay, J.L. and Greenwood, A.D. 1971. Cell wall formation in
the Saprolegiales: cytoplasmicvesicles underlying developing walls.
Henderson, I.R., Navarro-Garcia, F., Desvaux, M., Fernandez, R.C. and
Ala’Aldeen, D. 2004. Type V protein secretion pathway: the
autotransporter story. Microbiol. Mol. Biol. Rev., 68:692–744.
Hoegl, L., Ollert, M. and Korting, H.C. 1996. The role of Candida albicans
secreted aspartic proteinase in the development of candidoses. J. Mol.
Med., 74:135–42.
Hoffmann, P.R., Kench, J.A., Vondracek, A., Kruk, E., Daleke, D.L.,
Jordan, M., Marrack, P., Henson, P.M. and Fadok, V.A. 2005. Interaction
between phosphatidylserine and the phosphatidylserine receptor
inhibits immune responses in vivo. J. Immunol., 174:1393–404.
Hruskova-Heidingsfeldova, O. 2008. Secreted proteins of Candida albicans.
Front Biosci., 13:7227–42.
Hu, G., Hacham, M., Waterman, S.R., Panepinto, J., Shin, S., Liu, X.,
Gibbons, J., Valyi-Nagy, T., Obara, K., Jaffe, H.A., Ohsumi, Y. and
Williamson, P.R. 2008. PI3K signaling of autophagy is required for
starvation tolerance and virulenceof Cryptococcus neoformans.
J. Clin. Invest., 118:1186–97.
Hu, G., Steen, B.R., Lian, T., Sham, A.P., Tam, N., Tangen, K.L. and
Kronstad, J.W. 2007. Transcriptional regulation by protein kinase
A in Cryptococcus neoformans. PLoS Pathog., 3:e42.
Hube, B. 1996. Candida albicans secreted aspartyl proteinases. Curr. Top
Med. Mycol., 7:55–69.
Hurley, J.H. and Emr, S.D. 2006. The ESCRT complexes: structure and
mechanism of a membrane-traf cking network. Annu. Rev. Biophys.
Biomol. Struct., 35:277–98.
Johnstone, R.M., Adam, M., Hammond, J.R., Orr, L. and Turbide, C. 1987.
Vesicle formation during reticulocyte maturation. Association of
plasma membrane activities with released vesicles (exosomes).
J. Biol. Chem., 262:9412–20.
Kakeya, H., Udono, H., Ikuno, N., Yamamoto, Y., Mitsutake, K., Miyazaki,
T., Tomono, K., Koga, H., Tashiro, T., Nakayama, E. and Kohno, S.
1997. A 77-kilodalton protein of Cryptococcus neoformans, a member
of the heat shock protein 70 family, is a major antigen detected in the
sera of mice with pulmonary cryptococcosis. Infect. Immun.,
65:1653–8.
Kakeya, H., Udono, H., Maesaki, S., Sasaki, E., Kawamura, S., Hossain, M.A.,
Yamamoto, Y., Sawai, T., Fukuda, M., Mitsutake, K., Miyazaki, Y.,
Tomono, K., Tashiro, T., Nakayama, E. and Kohno, S. 1999. Heat
shock protein 70 (hsp70) as a major target of the antibody response
in patients with pulmonary cryptococcosis. Clin. Exp. Immunol.,
115:485–90.
J. Gen. Microbiol., 65:225–232.
39
Trans-cell wall secretion in fungi
Lipid Insights 2008:2
Keller, S., Sanderson, M.P., Stoeck, A. and Altevogt, P. 2006. Exosomes:
from biogenesis and secretion to biological function. Immunol. Lett.,
107:102–8.
Kihara, A., Noda, T., Ishihara, N. and Ohsumi, Y. 2001. Two distinct Vps34
phosphatidylinositol 3-kinase complexes function in autophagy and
carboxypeptidase Y. sorting in Saccharomyces cerevisiae. J. Cell
Biol., 152:519–30.
Kinseth, M.A., Anjard, C., Fuller, D., Guizzunti, G., Loomis, W.F. and
Malhotra, V. 2007. The Golgi-associated protein GRASP is required
for unconventional protein secretion during development. Cell,
130:524–34.
Kuehn, M.J. and Kesty, N.C. 2005. Bacterial outer membrane vesicles and
the host-pathogen interaction. Genes Dev., 19:2645–55.
Lamparski, H.G., Metha-Damani, A., Yao, J.Y., Patel, S., Hsu, D.H.,
Ruegg, C. and Le Pecq, J.B. 2002. Production and characterization
of clinical grade exosomes derived from dendritic cells. J. Immunol.
Methods, 270:211–26.
Lee, M.C., Miller, E.A., Goldberg, J., Orci, L. and Schekman, R. 2004.
Bi-directional protein transport between the ER. and Golgi. Annu.
Rev. Cell Dev. Biol., 20:87–123.
Linnemans, W.A., Boer, P. and Elbers, P.F. 1977. Localization of acid
phosphatase in Saccharomyces cerevisiae: a clue to cell wall
formation. J. Bacteriol., 131:638–44.
Liu, G.Y. and Nizet, V. 2004. Extracellular virulence factors of group B.
Streptococci. Front Biosci., 9:1794–802.
Long, K.H., Gomez, F.J., Morris, R.E. and Newman, S.L. 2003. Identi ca-
tion of heat shock protein 60 as the ligand on Histoplasma capsulatum
that mediates binding to CD18 receptors on human macrophages.
J. Immunol., 170:487–94.
Mashburn-Warren, L., McLean, R.J. and Whiteley, M. 2008. Gram-negative
outer membrane vesicles: beyond the cell surface. Geobiology,
6:214–9.
Mazar, J. and Cotter, P.A. 2007. New insight into the molecular mechanisms
of two-partner secretion. Trends Microbiol., 15:508–15.
McClelland, E.E., Bernhardt, P. and Casadevall, A. 2005. Coping with multiple
virulence factors: which is most important? PLoS Pathog., 1:e40.
McFadden, D., Zaragoza, O. and Casadevall, A. 2006a. The capsular dynamics
of Cryptococcus neoformans. Trends Microbiol., 14:497–505.
McFadden, D.C., De Jesus, M. and Casadevall, A. 2006b. The physical
properties of the capsular polysaccharides from Cryptococcus
neoformans suggest features for capsule construction. J. Biol. Chem.,
281:1868–75.
Mitchell, A.P. 2006. Cryptococcal virulence: beyond the usual suspects.
J. Clin. Invest, 116:1481–3.
Mori, H. and Ito, K. 2001. The Sec protein-translocation pathway. Trends
Microbiol., 9:494–500.
Nebenfuhr, A., Ritzenthaler, C. and Robinson, D.G. 2002. Brefeldin A:
deciphering an enigmatic inhibitor of secretion. Plant Physiol.,
130:1102–8.
Nickel, W. and Seedorf, M. 2008. Unconventional Mechanisms of Protein
Transport to the Cell. Surface of Eukaryotic Cells. Annu. Rev. Cell
Dev. Biol.,
Niimi, M., Tanabe, K., Wada, S., Yamazaki, A., Uehara, Y., Niimi, K.,
Lamping, E., Holmes, A.R., Monk, B.C. and Cannon, R.D. 2005.
[ABC transporters of pathogenic fungi: recent advances in functional
analyses]. Nippon Ishinkin Gakkai Zasshi, 46:249–60.
Nimrichter, L., Cerqueira, M.D., Leitao, E.A., Miranda, K., Nakayasu, E.
S., Almeida, S.R., Almeida, I.C., Alviano, C.S., Barreto-Bergter, E.
and Rodrigues, M.L. 2005a. Structure, cellular distribution,
antigenicity, and biological functions of Fonsecaea pedrosoi ceramide
monohexosides. Infect. Immun., 73:7860–8.
Nimrichter, L., Rodrigues, M.L., Rodrigues, E.G. and Travassos, L.R. 2005b.
The multitude of targets for the immune system and drug therapy in
the fungal cell wall. Microbes. Infect., 7:789–98.
Nosanchuk, J.D. and Casadevall, A. 2006. Impact of melanin on microbial
virulence and clinical resistance to antimicrobial compounds.
Antimicrob. Agents Chemother., 50:3519–28.
Nosanchuk, J.D., Steenbergen, J.N., Shi, L., Deepe, G.S., Jr., and Casadevall,
A. 2003. Antibodies to a cell surface histone-like protein protect
against Histoplasma capsulatum. J. Clin. Invest., 112:1164–75.
Novick, P., Field, C. and Schekman, R. 1980. Identification of
23 complementation groups required for post-translational events in
the yeast secretory pathway. Cell., 21:205–15.
Novick, P. and Schekman, R. 1979. Secretion and cell-surface growth are
blocked in a temperature-sensitive mutant of Saccharomyces
cerevisiae. Proc. Natl. Acad. Sci. U.S.A., 76:1858–62.
Raposo, G., Nijman, H.W., Stoorvogel, W., Liejendekker, R., Harding, C.V.,
Melief, C.J. and Geuze, H.J. 1996. B. lymphocytes secrete antigen-
presenting vesicles. J. Exp. Med., 183:1161–72.
Rattray, J.B., Schibeci, A. and Kidby, D.K. 1975. Lipids of yeasts. Bacteriol
Rev., 39:197–231.
Rigel, N.W. and Braunstein, M. 2008. A new twist on an old pathway—accessory
secretion systems. Mol. Microbiol.,
Rittershaus, P.C., Kechichian, T.B., Allegood, J.C., Merrill, A.H., Jr.,
Hennig, M., Luberto, C. and Del Poeta, M. 2006. Glucosylceramide
synthase is an essential regulator of pathogenicity of Cryptococcus
neoformans. J. Clin. Invest., 116:1651–9.
Rodrigues, M.L., Nakayasu, E.S., Oliveira, D.L., Nimrichter, L.,
Nosanchuk, J.D., Almeida, I.C. and Casadevall, A. 2008. Extracellular
vesicles produced by Cryptococcus neoformans contain protein
components associated with virulence. Eukaryot. Cell., 7:58–67.
Rodrigues, M.L., Nimrichter, L., Oliveira, D.L., Frases, S., Miranda, K.,
Zaragoza, O., Alvarez, M., Nakouzi, A., Feldmesser, M. and
Casadevall, A. 2007. Vesicular polysaccharide export in Cryptococcus
neoformans is a eukaryotic solution to the problem of fungal trans-cell
wall transport. Eukaryot. Cell., 6:48–59.
Rodrigues, M.L., Travassos, L.R., Miranda, K.R., Franzen, A.J., Rozental,
S., de Souza, W., Alviano, C.S. and Barreto-Bergter, E. 2000. Human
antibodies against a puri ed glucosylceramide from Cryptococcus
neoformans inhibit cell budding and fungal growth. Infect. Immun.,
68:7049–60.
Saier, M.H., Jr., 2006. Protein secretion and membrane insertion systems
in gram-negative bacteria. J. Membr. Biol., 214:75–90.
Saito, K., Takakuwa, N., Ohnishi, M. and Oda, Y. 2006. Presence of gluco-
sylceramide in yeast and its relation to alkali tolerance of yeast. Appl.
Microbiol. Biotechnol., 71:515–21.
Salama, N.R. and Schekman, R.W. 1995. The role of coat proteins in the
biosynthesis of secretory proteins. Curr. Opin. Cell Biol., 7:536–43.
Sargent, F. 2007. The twin-arginine transport system: moving folded proteins
across membranes. Biochem. Soc. Trans., 35:835–47.
Scales, S.J., Gomez, M. and Kreis, T.E. 2000. Coat proteins regulating
membrane traf c. Int. Rev. Cytol., 195:67–144.
Scheckelhoff, M. and Deepe, G.S., Jr., 2002. The protective immune response
to heat shock protein 60 of Histoplasma capsulatum is mediated by
a subset of V beta 8.1/8.2+ T cells. J. Immunol., 169:5818–26.
Schekman, R. 2002. Lasker Basic Medical Research Award. SEC mutants
and the secretory apparatus. Nat. Med., 8:1055–8.
Schekman, R., Esmon, B., Ferro-Novick, S., Field, C. and Novick, P. 1983.
Yeast secretory mutants: isolation and characterization. Methods
Enzymol., 96:802–15.
Schekman, R. and Novick, P. 2004. 23 genes, 23 years later. Cell., 116:
S13–5,1pfollowingS9.
Schliwa, M. and Woehlke, G. 2003. Molecular motors. Nature,
422:759–65.
Simkin, N.J., Jelinek, D.F. and Lipsky, P.E. 1987. Inhibition of human B. cell
responsiveness by prostaglandin E2. J. Immunol., 138:1074–81.
Simockova, M., Holic, R., Tahotna, D., Patton-Vogt, J. and Griac, P. 2008.
Yeast Pgc1p (YPL206c) controls the amount of phosphatidylglycerol
via a phospholipase C-type degradation mechanism. J. Biol. Chem.,
283:17107–15.
Skokos, D., Botros, H.G., Demeure, C., Morin, J., Peronet, R., Birkenmeier,
G., Boudaly, S. and Mecheri, S. 2003. Mast cell-derived exosomes
induce phenotypic and functional maturation of dendritic cells and elicit
speci c immune responses in vivo. J. Immunol., 170:3037–45.
40
Rodrigues et al
Lipid Insights 2008:2
Spang, A., Herrmann, J.M., Hamamoto, S. and Schekman, R. 2001. The ADP
ribosylation factor-nucleotide exchange factors Gea1p and Gea2p have
overlapping, but not redundant functions in retrograde transport from
the Golgi to the endoplasmic reticulum. Mol. Biol. Cell., 12:1035–45.
Taffet, S.M., Pace, J.L. and Russell, S.W. 1981. Lymphokine maintains
macrophage activation for tumor cell killing by interfering with the
negative regulatory effect of prostaglandin E2. J. Immunol., 127:121–4.
Taffet, S.M. and Russell, S.W. 1981. Macrophage-mediated tumor cell
killing: regulation of expression of cytolytic activity by prostaglandin
E. J. Immunol., 126:424–7.
Takeo, K., Uesaka, I., Uehira, K. and Nishiura, M. 1973a. Fine structure
of Cryptococcus neoformans grown in vitro as observed by
freeze-etching. J. Bacteriol., 113:1442–8.
Takeo, K., Uesaka, I., Uehira, K. and Nishiura, M. 1973b. Fine structure
of Cryptococcus neoformans grown in vivo as observed by
freeze-etching. J. Bacteriol., 113:1449–54.
Tucker, S.C. and Casadevall, A. 2002. Replication of Cryptococcus neoformans
in macrophages is accompanied by phagosomal permeabilization and
accumulation of vesicles containing polysaccharide in the cytoplasm.
Proc. Natl. Acad. Sci. U.S.A., 99:3165–70.
van Niel, G., Raposo, G., Candalh, C., Boussac, M., Hershberg, R.,
Cerf-Bensussan, N. and Heyman, M. 2001. Intestinal epithelial cells
secrete exosome-like vesicles. Gastroenterology, 121:337–49.
Vecchiarelli, A. 2005. The cellular responses induced by the capsular poly-
saccharide of Cryptococcus neoformans differ depending on the
presence or absence of speci c protective antibodies. Curr. Mol.
Med., 5:413–20.
Veenendaal, A.K., van der Does, C. and Driessen, A.J. 2004.
The protein-conducting channel SecYEG. Biochim. Biophys. Acta.,
1694:81–95.
Walworth, N.C., Goud, B., Kabcenell, A.K. and Novick, P.J. 1989.
Mutational analysis of SEC4 suggests a cyclical mechanism for the
regulation of vesicular traf c. EMBO J., 8:1685–93.
Whit eld, C. 2006. Biosynthesis and assembly of capsular polysaccharides
in Escherichia coli. Annu. Rev. Biochem., 75:39–68.
Whit eld, C., Kaniuk, N. and Frirdich, E. 2003. Molecular insights into the
assembly and diversity of the outer core oligosaccharide in lipopoly-
saccharides from Escherichia coli and Salmonella. J. Endotoxin Res.,
9:244–9.
Wickner, W. and Schekman, R. 2005. Protein translocation across biological
membranes. Science, 310:1452–6.
Xu, X.J., Reichner, J.S., Mastrofrancesco, B., Henry, W.L., Jr., and Albina,
J.E. 2008. Prostaglandin E2 suppresses lipopolysaccharide-stimulated
IFN-beta production. J. Immunol., 180:2125–31.
Yoneda, A. and Doering, T.L. 2006. A eukaryotic capsular polysaccharide
is synthesized intracellularly and secreted via exocytosis. Mol. Biol.
Cell., 17:5131–40.
Zaragoza, O., Telzak, A., Bryan, R.A., Dadachova, E. and Casadevall, A.
2006. The polysaccharide capsule of the pathogenic fungus
Cryptococcus neoformans enlarges by distal growth and is rearranged
during budding. Mol. Microbiol., 59:67–83.
The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and
metabolism in the human pathogen Cryptococcus neoformans
Lívia Kmetzsch
a
, Charley Christian Staats
a,b
, Elisa Simon
a
, Fernanda L. Fonseca
c
, Débora L. Oliveira
c
,
Luna S. Joffe
c
, Jéssica Rodrigues
c
, Rogério F. Lourenço
d
, Suely L. Gomes
d
, Leonardo Nimrichter
c
,
Marcio L. Rodrigues
c
, Augusto Schrank
a,b
, Marilene Henning Vainstein
a,b,
*
a
Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Av. Bento Gonçalves 9500, 43421, Caixa Postal 15005, Porto Alegre, RS 91501-970, Brazil
b
Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Brazil
c
Laboratório de Estudos Integrados em Bioquímica Microbiana, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas
Filho 373, CCS, Bloco I. Rio de Janeiro, RJ, 21941-902, Brazil
d
Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Professor Lineu Prestes, 748, 05508-900, São Paulo, SP, Brazil
a r t i c l e i n f o
Article history:
Received 7 June 2010
Accepted 22 July 2010
Available online xxxx
Keywords:
Nitrogen metabolism
Nitrogen Catabolite Repression
Cryptococcus neoformans
a b s t r a c t
Nitrogen uptake and metabolism are essential to microbial growth. Gat1 belongs to a conserved family of
zinc finger containing transcriptional regulators known as GATA-factors. These factors activate the tran-
scription of Nitrogen Catabolite Repression (NCR) sensitive genes when preferred nitrogen sources are
absent or limiting. Cryptococcus neoformans GAT1 is an ortholog to the Aspergillus nidulans AreA and Can-
dida albicans GAT1 genes. In an attempt to define the function of this transcriptional regulator in C. neo-
formans, we generated null mutants (gat1
D
) of this gene. The gat1 mutant exhibited impaired growth on
all amino acids tested as sole nitrogen sources, with the exception of arginine and proline. Furthermore,
the gat1 mutant did not display resistance to rapamycin, an immunosuppressant drug that transiently
mimics a low-quality nitrogen source. Gat1 is not required for C. neoformans survival during macrophage
infection or for virulence in a mouse model of cryptococcosis. Microarray analysis allowed the identifica-
tion of target genes that are regulated by Gat1 in the presence of proline, a poor and non-repressing nitro-
gen source. Genes involved in ergosterol biosynthesis, iron uptake, cell wall organization and capsule
biosynthesis, in addition to NCR-sensitive genes, are Gat1-regulated in C. neoformans.
Ó 2010 Elsevier Inc. All rights reserved.
1. Introduction
Pathogenic fungi have to adapt and survive in distinct nutritive
environments and, in this context, nitrogen uptake and its metab-
olism are critical to fungal growth. Nitrogen Catabolite Repression
(NCR) is a mechanism that controls the fungi selective utilization
of optimal nitrogen sources in preference to poor ones (
Marzluf,
1997). NCR-sensitive genes are repressed when preferred nitrogen
sources are available. Under limitation of these sources, the
expression of genes encoding permeases and catabolic enzymes,
required to utilize poor nitrogen sources, is activated by specific
GATA-factor family of transcription factors. Members of this family
contain a zinc-finger domain and are conserved in fungi (
Coffman
and Cooper, 1997; Coffman et al., 1996; Magasanik and Kaiser,
2002; Marzluf, 1997
). In Saccharomyces cerevisiae, two GATA-fac-
tors involved in activation of NCR-sensitive genes (Gat1 and
Gln3) were described (
Stanbrough et al., 1995). The GATA-type
transcriptional activator Gat1 and Gln3 orthologs of Candida albi-
cans regulate nitrogen metabolism and virulence during host-path-
ogen interactions (Liao et al., 2008; Limjindaporn et al., 2003). The
knockout of GAT1 in C. albicans results in reduced capacity to
metabolize some secondary nitrogen sources, but dimorphism is
not impaired (
Limjindaporn et al., 2003). The ortholog of GAT1 in
Aspergillus fumigatus (AreA) also participates in nitrogen regulation
and virulence, since null mutants for AreA show attenuated viru-
lence in a murine model of pulmonary aspergillosis (
Hensel
et al., 1998
).
Exposure of S. cerevisiae cells to rapamycin mimics a low-qual-
ity nitrogen source, which results in activation of NCR-sensitive
genes by Gat1 and Gln3 transcription factors (
Hardwick et al.,
1999; Scherens et al., 2006
). This immunosuppressant drug inhib-
its a conserved signaling cascade for cell proliferation regulated by
target of rapamycin (TOR) in response to nutrient availability
(
Jiang and Broach, 1999; Thomas and Hall, 1997). S. cerevisiae
Gln3 and Gat1 are phosphorylated in a Tor-dependent mechanism,
resulting in interaction with the cytoplasmic protein Ure2. Upon
rapamycin treatment, Gln3 and Gat1 are dephosphorylated,
1087-1845/$ - see front matter Ó 2010 Elsevier Inc. All rights reserved.
doi:
10.1016/j.fgb.2010.07.011
* Corresponding author at: Centro de Biotecnologia, Universidade Federal do Rio
Grande do Sul, Avenida Bento Gonçalves 9500, 43421, Setor 4, Porto Alegre, RS
91501-970, Brazil. Fax: +55 51 3308 7309.
E-mail address:
[email protected] (M.H. Vainstein).
Fungal Genetics and Biology xxx (2010) xxx–xxx
Contents lists available at ScienceDirect
Fungal Genetics and Biology
jo u rn a l h om ep a ge : w w w. e ls ev i er . co m /l o ca te / yf g bi
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
released and directed to the nucleus, leading to transcription of
NCR-sensitive genes (
Beck and Hall, 1999; Bertram et al., 2000).
Null mutants of GLN3 or GAT1 in C. albicans were resistant to rap-
amycin, suggesting that the TOR signaling pathway acts through
Gln3 and Gat1 in this pathogen (
Liao et al., 2008).
The life cycle of the human pathogen Cryptococcus neoformans is
influenced by nitrogen availability, since in response to nitrogen
limitation this organism initiates monokaryotic fruiting or mating
(
Wickes et al., 1996). These two processes lead to the production of
spores, potential infectious propagules that are pathogenic in mice
(
Giles et al., 2009; Lengeler et al., 2000; Wickes et al., 1996). Fur-
thermore, the C. neoformans ammonium permease Amt2, which
is activated in response to nitrogen starvation, is required to induce
ammonium-responsive invasive growth and mating, indicating the
relevance of nitrogen metabolism in important aspects of C. neofor-
mans biology (
Rutherford et al., 2008). Here we report the identifi-
cation of the GATA-type transcriptional activator Gat1 in C.
neoformans, which influences nitrogen uptake and controls the
transcription of genes involved in NCR, ergosterol biosynthesis,
iron uptake, cell wall organization and capsule biosynthesis.
2. Material and methods
2.1. Fungal strains, plasmids and media
C. neoformans H99 strain was the recipient for target gene dele-
tion. C. neoformans strains were maintained on YPD medium (1%
yeast extract, 2% peptone, 2% dextrose, and 1.5% agar). YPD plates
amended with hygromycin (200
l
g/ml) were used to select C. neo-
formans gat1 mutant strains. YPD plates amended with nourseo-
thricin (100
l
g/ml) were used to select C. neoformans gat1::GAT1
complemented strains. Plasmid pJAF15 (
Fraser et al., 2003) was
the source of a hygromycin resistance cassette and plasmid pAI4
(
Idnurm et al., 2004) was the source of a nourseothricin resistance
cassette.
2.2. In silico analysis of the C. neoformans GATA-type transcription
factor GAT1 ortholog
The putative C. neoformans GAT1 gene sequence was identified
by a BLAST search of the C. neoformans strain H99 genomic data-
base at the Broad Institute using GAT1 sequence of S. cerevisiae
[GenBank: NP_116632.1]. The amino acid sequences of Gat1 ortho-
logs from S. cerevisiae, C. albicans, A. nidulans, Ustilago maydis, Neu-
rospora crassa and C. neoformans were aligned using ClustalX2
(Larkin et al., 2007). Mega4 was utilized for phylogenetic analysis
applying the Neighbor-Joining method and the tree architecture
was inferred from 1000 bootstraps (
Tamura et al., 2007). Pfam
database (
http://pfam.sanger.ac.uk/) was used to search for con-
served domains in the Gat1 ortholog proteins.
2.3. Disruption and complementation of C. neoformans GAT1
The Delsgate methodology (
Garcia-Pedrajas et al., 2008) was
employed for disruption of GAT1. The hygromycin resistance cas-
sette from pJAF15 was subcloned into the EcoRV site of pDONR201
(Gateway donor vector, Invitrogen) to construct pDONRHYG plas-
mid. The 5
0
and 3
0
GAT1 flanks (827 and 857 bp, respectively) were
PCR amplified, and gel purified using Illustra GFX PCR DNA and Gel
Band Purification kit (GE Healthcare). Approximately 300 ng of
pDONRHYG vector and 30 ng of each PCR product were utilized
in the BP clonase reaction, according to manufacturer’s instruc-
tions (Invitrogen). This reaction was transformed into Escherichia
coli OmniMAX 2-T1. After confirmation of the correct deletion
construct, the plasmid was linearized by I-SceI digestion prior to
C. neoformans biolistic transformation (
Toffaletti et al., 1993).
Transformants were screened by colony PCR, and the deletion
was confirmed by Southern blot analysis and semi-quantitative
RT-PCR. For complementation, a 6.2 Kb genomic PCR fragment con-
taining the wild-type GAT1 gene was cloned into the SmaI site of
the pAI4 plasmid. The resulting plasmid was used for transforma-
tion of the gat1 mutant strain. Random genomic insertion of the
complemented gene was confirmed by Southern blot analysis
and semi-quantitative RT-PCR. The primers utilized in these con-
structions are listed in
Table S1.
2.4. Phenotypic characterization assays
Nitrogen source utilization was assessed in YCB medium (Yeast
Carbon Base, Difco). Wild type (WT), gat1 mutant and comple-
mented strains were pre-cultured in YPD medium at 30 °C for
18 h. The cells were collected by centrifugation, washed three
times with sterile dH
2
O, suspended in YCB at a cell density of
10
8
cells/ml and incubated at 30 °C for 12 h to deplete nitrogen.
Cells suspensions (10
7
cells/ml) were serially 10-fold diluted and
3
l
l from each dilution was spotted onto YCB agar supplemented
with 2 mM of each amino acid, 100 mM of urea, or 37 mM of
ammonium sulfate. Sensitivity to rapamycin was assessed on
YPD agar medium supplemented with 100 ng/ml and 200 ng/ml
of rapamycin. WT, gat1 mutant and complemented cells were cul-
tured overnight in YPD, washed and suspended to a density of
10
7
cells/ml. The cells were diluted and spotted as described above.
As control, cells were grown in YPD agar only. The plates were
incubated for 2 days at 30 °C and photographed. Capsule formation
was examined by microscopy after incubation for 24 h at 30 °C in a
minimal medium and prepared with India ink. Relative capsule
sizes were defined as the distance between the cell wall and the
capsule outer border by cell diameter. ImageJ software was utilized
to determine capsule measurements of one hundred cells of each
strain. The content of extracellular glucuronoxylomannan (GXM)
in culture supernatants was determined by ELISA (
Fonseca et al.,
2009
).
2.5. Thin Layer Chromatography
Analyses of amino acids in culture supernatants of WT and gat1
mutant strains were assessed by Thin Layer Chromatography (TLC).
Briefly, starter cultures of WT and gat1 mutant cells were grown
overnight in YPD at 37 °C with shaking. Cells were washed three
times, suspended in YNB (with no ammonia nor amino acids and
amended with 2% of glucose) and incubated at 37 °C for 12 h to de-
plete nitrogen. Then, 10
8
cells of each strain were inoculated in
YNB supplemented with 2 mM proline or aspartic acid. After 20 h
incubation at 37 °C, the cells were removed by centrifugation for
10 min at 10,000g. The supernatants were passed through a
0.45
l
m pore filter, followed by an ultra-filtration in membrane
disks (1 kDa pore size). Silica gel TLC plates were spotted with
5
l
l of each supernatant, and the mobile phase utilized for chroma-
tography was butanol: acetic acid: water (40:10:10). For amino
acids visualization, the plate was dipped in a ninhydrin solution
(0.2% in ethanol) for 10 s. After 30 min incubation at 60 °C, the
TLC plates were photographed. These analyses were performed in
biological and technical duplicates.
2.6. Macrophage infection assay
The susceptibility of fungal cells to the antifungal action of
phagocytes was determined by counting colony forming units
(CFU) after interaction of WT, gat1 mutant and gat1::GAT1 comple-
mented strains with the murine macrophage-like cell line RAW
264.7. Prior to interaction, fungal cells were opsonized with mono-
2 L. Kmetzsch et al. / Fungal Genetics and Biology xxx (2010) xxx–xxx
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
clonal antibody 18B7 (1
l
g/ml), a gift from Dr. Arturo Casadevall
(Albert Einstein College of Medicine, USA). Macrophages were
seeded at a concentration of 10
5
cells/well in a 96-well cell culture
plate, and incubated overnight at 37 °C in 5% CO
2
in Dulbecco’s
modified Eagle’s medium (DMEM) supplemented with 5% heat-
inactivated fetal bovine serum (FBS). Fungal cells (10
6
) were inoc-
ulated in each well, and after 1 h the wells were washed to remove
unattached, extracellular fungal cells. After 20 h of incubation, in-
fected cultures were again washed and sterile ice-cold dH
2
O was
added to each well to promote macrophage lysis. Fungal viability
was measured by plating the lysates on YPD for CFU determination
after cultivation of the plates for 48 h at 30 °C. The assay was per-
formed in triplicate sets for each strain. Student’s t test was used to
determine the statistical significance of differences in fungal
survival.
2.7. Virulence assay
Virulence studies were conducted according to a previously de-
scribed intranasal inhalation infection model in mice (
Cox et al.,
2000). Fungal cells were cultured in 50 ml of YPD medium at
30 °C overnight with shaking, washed twice and re-suspended in
PBS. Groups of eight female BALB/c mice (approximately 5 weeks
old) were infected with 10
7
yeast cells suspended in 50
l
l PBS
and monitored daily. Kaplan–Meier analysis of survival was per-
formed using GraphPad Prism Software. Animal studies were ap-
proved by the Federal University of Rio Grande do Sul Ethics
Committee.
2.8. Microarray analysis
For RNA extraction, starter cultures of WT and gat1 mutant cells
were grown overnight in YPD at 37 °C with shaking. Cells were
washed three times and suspended in YNB with no ammonia nor
amino acids and amended with 2% of glucose. This medium was
supplemented with 10 mM proline as a nitrogen source and incu-
bated for 3 h at 37 °C. Three independent sets of RNA samples from
independent experiments were prepared using Trizol reagent
(Invitrogen) according to the manufacturer’s protocol. After DNase
treatment, RNA preparations were purified using RNAeasy mini
columns (Qiagen). The CyScribe First-Strand cDNA Labeling Kit
(GE Life Sciences) was utilized for preparation and purification of
Cy3-dUTP and Cy5-dUTP labeled cDNA probes during first-strand
cDNA synthesis reactions, according to the manufacture’s protocol.
These fluorescent labeled cDNAs were synthesized from 25
l
g to-
tal RNA from each strain tested. Cy5-labeled cDNA from WT was
mixed with Cy3-labeled cDNA from the gat1 mutant strain and
the mixture was hybridized to C. neoformans microarray slides
for 18 h at 42 °C in a humid hybridization chamber. Arrays were
washed with SSC buffer (1Â) with 0.2% (w/v) SDS for 10 min at
room temperature, followed by two washes in SSC (0.1Â) with
0.2% (w/v) SDS for 5 min and scanned on a GenePix 4000B Scanner
(Molecular Devices). Image preprocessing and data quantification
was done using GenePix Pro Software and the raw expression data
was obtained. For further data analysis, the TIGR microarray soft-
ware suite (
http://www.tm4.org/), which includes Midas and
MeV software, was used. Data sets were first processed by Midas
using total-intensity and LOWESS normalizations, and standard-
deviation regulation, so that for each gene a normalized expression
value was attributed. Following, MeV Software was used to per-
form one sample Student’s t test in order to identify genes with
statistically significant changes. Fold changes (WT/gat1
D
) were de-
rived from the mean expression levels from three independent
experiments. The complete list of differentially expressed genes
(2-fold up or down regulated) with P values of <0.05 is presented
in
Table S2. The C. neoformans arrays (version 2) were purchased
from an academic consortium of Genome Sequencing Center at
School of Medicine in Washington University in St. Louis (
http://
genomeold.wustl.edu/activity/ma/cneoformans/
).
2.9. Quantitative real time RT-PCR analysis
Real-time PCR reactions were performed in an Applied Biosys-
tems 7500 Real-Time PCR System. PCR thermal cycling conditions
were an initial step at 95 °C for 5 min followed by 40 cycles at 95 °C
for 15 s, 60 °C for 20 s and 72 °C for 20 s. Platinum SYBR green qPCR
Supermix (Invitrogen) was used as reaction mix, supplemented
with 5 pmol of each primer and 2
l
l of the cDNA template in a final
volume of 25
l
l. All experiments were done in three independent
cultures and each cDNA sample was analyzed in duplicate with
each primer pair. Melting curve analysis was performed at the
end of the reaction to confirm a single PCR product. Data was nor-
malized to actin cDNAs amplified in each set of PCR experiments.
Relative expression was determined by the 2
À
D
CT
method (Livak
and Schmittgen, 2001
). The primers utilized in these experiments
are listed in
Table S1.
3. Results
3.1. Identification of a GATA-type transcription factor Gat1 ortholog in
C. neoformans
The GAT1 gene [Broad Institute: CNAG_00193.2] was identified
in the C. neoformans var. grubii H99 genomic database available at
the Broad Institute (
http://www.broadinstitute.org/annotation/
genome/cryptococcus_neoformans/MultiHome.html
), based on its
similarity to GAT1 from S. cerevisiae. The C. neoformans GAT1 coding
region is 4239 bp long, contains three introns and encodes a puta-
tive 1277-amino-acid protein. Members of the GATA-factor family
of transcription factors have a GATA zinc-finger domain [Pfam:
PF00320] (
Marzluf, 1997) that is also present in the C. neoformans
Gat1 ortholog. Furthermore, a phylogenetic analysis including Gat1
sequences from different fungi species was performed (
Fig. 1A) and
showed that C. neoformans Gat1 is most similar to Gat1 from U.
maydis and least similar to Gat1 from S. cerevisiae. The domain
architecture of the C. neoformans Gat1 was compared to the ortho-
logs herein analyzed showing that all orthologs have the GATA
zinc-finger domain [Pfam: PF00320] plus a domain of unknown
function DUF1752 [Pfam: 08550] (
Fig. 1B).
3.2. The gat1 mutant exhibited normal growth only when arginine and
proline were used as sole nitrogen sources
In order to perform a functional analysis of Gat1 in C. neofor-
mans, knockout and complemented strains were constructed. Dele-
tion and complementation of GAT1 were confirmed by Southern
blot analysis and semi-quantitative RT-PCR, as shown in
Fig. 2.
To assess the role of C. neoformans GAT1 in nitrogen uptake and
metabolism, the ability of WT, gat1 mutant and gat1::GAT1 com-
plemented strains to grow in distinct nitrogen sources was evalu-
ated. WT and complemented strains grew well on all nitrogen
sources tested, but slightly growth differences between these
strains were observed. However, the gat1 mutant had strongly im-
paired growth on isoleucine, leucine, alanine, asparagine, lysine,
aspartic acid, methionine, cysteine, phenylalanine, glutamic acid,
threonine, glutamine, tryptophan, glycine, valine, serine, tyrosine,
histidine, urea or ammonium sulfate as nitrogen sources. The
gat1 mutant exhibited normal growth only when the nitrogen
sources utilized were arginine and proline (
Fig. 3A). These gat1 mu-
tant phenotypes can be due to a nitrogen metabolism defect or to
an amino acid transport defect. To address this point, TLC analyses
L. Kmetzsch et al. / Fungal Genetics and Biology xxx (2010) xxx–xxx
3
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
of the supernatants of WT and gat1 mutant cultures in the presence
of proline or aspartic acid were performed. As seen in
Fig. 3B, after
20 h of incubation, proline was completely utilized by both WT and
gat1 mutant strains, in contrast with aspartic acid, which was con-
sumed only by WT strain. Since the aspartic acid concentration was
not altered in gat1 mutant supernatants in comparison to the con-
centration observed in the fresh medium, we can conclude that
gat1 mutant has a transport defect of some amino acids.
To test whether the TOR signaling pathway acts through the
activity of GAT1 in C. neoformans, as demonstrated for the orthologs
in C. albicans and S. cerevisiae (
Beck and Hall, 1999; Cardenas et al.,
1999; Liao et al., 2008
), we conducted rapamycin sensitivity as-
says. Unlike C. albicans and S. cerevisiae, the C. neoformans gat1 mu-
tant strain had almost identical sensibility to rapamycin in
comparison to WT and complemented strains (
Fig. 3C), indicating
that the loss of GAT1 does not influence the TOR signaling pathway
in C. neoformans.
3.3. Disruption of GAT1 does not influence capsule size, but decreases
extracellular GXM secretion
The gat1 mutant strain was tested for its ability to grow at 37 °C,
for capsule size and for melanin production since these traits are
considered key virulence factors in C. neoformans (
McClelland
et al., 2005). The loss of GAT1 apparently did not interfere with
any of these traits (
Fig. 4A and data not shown). The disruption of
GAT1, however, decreased the levels of extracellular GXM, since
lower polysaccharide contents were observed in culture superna-
tants from the gat1 mutant strain, in comparison to WT (P < 0.05)
(
Fig. 4B). This finding suggests a role for C. neoformans Gat1 on
the release of capsular polysaccharides to the extracellular
environment.
3.4. Gat1 is neither required for C. neoformans survival during
macrophage infection nor for virulence in a mouse intranasal model of
cryptococcosis
The GATA-type transcriptional regulators Gat1 of C. albicans and
AreA of A. fumigatus act in virulence of these human pathogens
(
Hensel et al., 1998; Limjindaporn et al., 2003). Therefore we tested
whether C. neoformans Gat1 is essential during macrophage infec-
tion in vitro and in mice. Unlike as occurs in C. albicans and
A.
fumigatus, Gat1 does not influence C. neoformans virulence in mice
(
Fig. 4C). Furthermore, no differences were observed in survival of
WT, gat1 mutant and complement cells during an in vitro macro-
phage infection, as seen by CFU count showed in
Fig. 4D.
3.5. The GATA-type transcriptional activator Gat1 regulates genes
involved in NCR, ergosterol biosynthesis, iron uptake, cell wall
organization and capsule biosynthesis in C. neoformans
Microarray analyses were conducted to identify target genes
that are regulated by Gat1. We examined changes in global gene
expression due to deletion of GAT1 in the presence of proline, a
Fig. 1. Identification of a GATA-type transcriptional activator Gat1 ortholog in C. neoformans A. Phylogenetic analysis applying the Neighbor-Joining method including Gat1
sequences from distinct fungi, as follows: S. cerevisiae [Genbank: NP_116632.1], C. albicans [Genbank: AAP50501.1], A. nidulans [Genbank: XP_681936.1], U. maydis [Broad
Institute: UM_04252], N. crassa [Genbank: AAB03891.1] and C. neoformans [Broad Institute: CNAG_00193.2]. The bar marker indicates the genetic distance, which is
proportional to the number of amino acid substitutions. B. Domain architecture in the Gat1 orthologs of different fungi. The GATA zinc-finger domain [Pfam: PF00320] is
represented by white ovals, and the domain of unknown function DUF1752 [Pfam: 08550] is represented by black bars. The length of each protein sequence (in amino acids)
is indicated in the right.
Fig. 2. Construction of C. neoformans GAT1 knockout and complemented strains. A. Gat1 deletion scheme. TV represents the targeting vector constructed by Delsgate
methodology. 5 GAT and 3 GAT represent the 5
0
and 3
0
gene flanks of GAT1 gene, respectively. 5F and 5R: primers utilized to amplify 5
0
flank of GAT1 gene. 3F and 3R: primers
utilized to amplify 3
0
flank of GAT1 gene. Hyg
R
: cassette that confers hygromicin resistance. WT represents the wild type locus of the GAT1 gene in H99 strain.
D
represents the
GAT1 locus in the gat1 mutant strain. The cleavage sites of HindIII restriction enzyme are indicated. B. Confirmation by Southern blot. Genomic DNA (10
l
g) from WT (lane 1),
gat1 mutant (lane 2) and gat1::GAT1 complemented (lane 3) strains were digested with HindIII restriction enzyme. The 3
0
gene flank was used as probe in Southern
hybridization. Left numbers (in base pairs) indicate the hybridization signal sizes based upon the position of molecular size marker. C. Semi-quantitative RT-PCR using cDNA
from WT (lane 1), gat1 mutant (lane 2) and gat1::GAT1 complemented (lane 3) strains as template. Right numbers (in bp) indicate the length of the transcript amplification for
GAT1 (upper panel) and ACT1 (lower panel) genes. Lane 4: positive control using genomic DNA as template. NC: negative control of the PCR reaction.
4 L. Kmetzsch et al. / Fungal Genetics and Biology xxx (2010) xxx–xxx
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
poor and non-repressing nitrogen source. We found that 127 genes
were differentially expressed at least 2-fold in the WT strain.
Among these, 54 genes were upregulated in WT in comparison to
gat1 mutant, including genes related to nitrogen metabolism and
NCR mechanism (proline dehydrogenase, glutamate dehydroge-
nase, and amino acid transporter), iron uptake (high-affinity iron
permease FTR1), and ergosterol biosynthesis (squalene monooxy-
genase). Seventy-three genes were found to be downregulated in
the WT strain, including genes associated with capsule biosynthe-
sis (capsular associated proteins), carbohydrate metabolism
(alpha-
L
-rhamnosidase, UDP-glucose dehydrogenase, UDP-glucu-
ronic acid decarboxylase, alpha 1–3 mannosyltransferase), cell wall
integrity (chitin synthase 2), oxidative metabolism (copper zinc
superoxide dismutase), and signal transduction (protein kinase
C).
Table 1 summarizes the microarray results. Some genes with
less than 2-fold change were included in
Table 1 due to their rele-
vance to possibly explain the distinct phenotypes observed in the
gat1 mutant strain as that involved in capsule biosynthesis (capsu-
lar associated protein and alpha-1,3-mannosyltransferase) and
nitrogen metabolism (glutamate-rich WD repeat containing one
protein and amino acid transporter). The complete list of differen-
tially expressed genes (2-fold up and down regulated) with
P values of <0.05 is in Table S2. To validate the microarray results,
the differential expression of seven selected genes was confirmed
by quantitative real time RT-PCR (
Fig. 5).
4. Discussion
Fungi can utilize a wide range of nitrogen sources and the key
regulators that control nitrogen acquisition are well conserved in
these organisms (
Marzluf, 1997). In the present study, the GATA-
type transcriptional activator Gat1 of C. neoformans was described.
GATA-factors are responsible for activation of NCR-sensitive genes,
as seen for Gat1 orthologs from S. cerevisiae, C. albicans and A.
fumigatus (
Hensel et al., 1998; Limjindaporn et al., 2003; Stanb-
rough et al., 1995
). Members of the GATA-factor family of tran-
scription factors are characterized by the presence of a GATA
zinc-finger domain [Pfam: PF00320] (
Marzluf, 1997). As expected,
C. neoformans Gat1 is required for optimal growth on a variety of
nitrogen sources, indicating that this GATA-factor is involved in
nitrogen uptake and metabolism. A similar response was observed
for C. albicans Gat1 and for Gln3, another GATA-type transcription
factor that acts in parallel to Gat1 to control nitrogen uptake in this
pathogen (
Liao et al., 2008; Limjindaporn et al., 2003). Further-
more, N. crassa nit-2 mutants had diminished growth on a diversity
of amino acids sources including branched chain amino acids, tryp-
tophan and phenylalanine, but not tyrosine (
Facklam and Marzluf,
1978).
The TOR signaling pathway, that regulates cell proliferation in
response to nutrient availability, is the target of inhibition by the
drug rapamycin in C. neoformans and C. albicans (
Cruz et al.,
2001). Rapamycin binds to FKBP12 protein, and this complex inter-
acts and inhibits the Tor1 kinase (
Cruz et al., 2001). The sensitivity
of C. albicans to rapamycin was greatly decreased by deletion of
either GLN3 or GAT1 (
Liao et al., 2008). However, C. neoformans
gat1 mutant strain had almost identical sensitivity to rapamycin
in comparison to WT and complemented strains, indicating that
the TOR signaling pathway does not act through Gat1 in C. neofor-
mans. This phenotype may indicate the presence of redundant
pathways or overlapping functions with other proteins that could
Fig. 3. Gat1 is involved in nitrogen uptake in C. neoformans. A. Nitrogen source utilization assay. Ten-fold serial dilutions of wild-type H99 (WT), gat1 mutant (gat1
D
) and
gat1::GAT1 complemented (gat1
D
::GAT1) cells were plated in YCB medium supplemented with different nitrogen sources, as indicated. B. Thin Layer Chromatography.
Culture supernatants of wild-type H99 (WT) and gat1 mutant strains were analyzed for amino acids (proline or aspartic acid) visualization. As controls, 2 mM of each amino
acid (C1) and fresh media supplemented with 2 mM of each amino acid (C2) were also spotted in the TLC plates. C. Rapamycin sensitivity assay. Ten-fold serial dilutions of
wild-type H99 (WT), gat1 mutant (gat1
D
) and gat1 ::GAT1 complemented (gat1
D
::GAT1) cells were plated in YPD medium supplemented with 200 ng/ml of rapamycin. The
plates were incubated for 2 days at 30 °C. As control, cells were grown in YPD agar only.
L. Kmetzsch et al. / Fungal Genetics and Biology xxx (2010) xxx–xxx
5
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
compensate the loss of GAT1 in C. neoformans during rapamycin
treatment.
The deletion of C. neoformans GAT1 did not affect capsule size,
but decreased the levels of extracellular GXM in culture superna-
Fig. 4. Evaluation of virulence-related phenotypes of the gat1 mutant strain. A. Relative capsule size of WT and gat1 mutant cells. B. Content of extracellular GXM in culture
supernatants of WT and gat1 mutant cells determined by ELISA.
Ã
P < 0.001. C. Virulence assay of WT, gat1 mutant and gat1::GAT1 complemented strains in an intranasal
inhalation infection model using BALB/c mice. D. Macrophage infection assay. CFU counting after macrophage infection with WT, gat1 mutant and gat1::GAT1 complemented
strains.
Table 1
List of GAT1-regulated genes in C. neoformans.
Category Description
a
/accession number at Broad Institute
Ã
or Genbank
ÃÃ
databases Fold change
b
(WT/gat1
D
)
Capsule biosynthesis Capsular associated protein|CNK01140
ÃÃ
0.54
Capsular associated protein|CNAG_00721
Ã
0.63
Carbohydrate metabolism Alpha-
L
-rhamnosidase|CNAG_02587
Ã
0.50
UDP-glucose dehydrogenase|CNAG_04969
Ã
0.22
UDP-glucuronic acid decarboxylase|CNG02560
Ã
0.35
Alpha-1,3-mannosyltransferase|CNAG_05142
Ã
1.54
Cell wall integrity Chitin synthase 2|CNAG_03326
Ã
0.56
Ergosterol biosynthesis Squalene monooxygenase|CNAG_06829
Ã
2.02
Squalene monooxygenase, putative|CND06110
Ã
2.01
Iron metabolism High-affinity iron permease CaFTR1|CNAG_02959
Ã
2.45
Nitrogen metabolism Proline dehydrogenase|CNAG_02049
Ã
5.10
Glutamate dehydrogenase (NADP+)|CNC00920
ÃÃ
11.37
Glutamate-rich WD repeat containing 1|CNAG_01600
Ã
1.55
Amino acid transporter|CNAG_02539
Ã
1.42
Oxidative metabolism Copper zinc superoxide dismutase|CND01490
ÃÃ
0.22
Signal transduction Protein kinase C, putative|CNC03300
ÃÃ
0.42
Transporters Potassium transport protein|CNAG_02856
Ã
0.33
ATP-binding cassette (ABC) transporter|CND00300
ÃÃ
0.39
Low-affinity zinc ion transporter, putative|CND00350
ÃÃ
0.45
a
Descriptions were obtained from NCBI database (http://www.ncbi.nlm.nih.gov/) and Broad Institute Database (http://www.broadinstitute.org/annotation/genome/
cryptococcus_neoformans/MultiHome.html
).
b
Data are presented as the average changes in expression of genes in WT cells compared with their expression in gat1 mutant cells during growth in YNB supplemented
with 10 mM proline as secondary nitrogen source. List contains a set of statistically significant genes identified by Student’s t test that have average changes of greater than 2-
fold. Genes with less than 2-fold change (italicized) are included in
Table 1 due to their relevance to explain the distinct phenotypes observed in the gat1 mutant strain.
6 L. Kmetzsch et al. / Fungal Genetics and Biology xxx (2010) xxx–xxx
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
tants. Although the concentration of extracellular GXM is fre-
quently associated with capsule enlargement, this same phenotype
was observed for a sec6 mutant of C. neoformans (Panepinto et al.,
2009). We speculate that the basal levels of GXM secretion mani-
fested by the gat1 mutant are sufficient to warrant capsule assem-
bly, although they are below the regular levels of polysaccharide
secretion by C. neoformans. The facts that Gat1 orthologs of C. albi-
cans and A. fumigatus are involved in virulence (
Hensel et al., 1998;
Limjindaporn et al., 2003
), together with our observation of de-
creased GXM levels in C. neoformans, led us to test the role of
Gat1 during macrophage infection in vitro and in a mice model of
cryptococcosis. No differences between WT, gat1 mutant and com-
plemented strains were observed in these assays, indicating that C.
neoformans Gat1 is not necessary for survival during host infection.
We hypothesize that this observation may be related to the fact
that arginine and proline are probably available in host tissues
and fluids in their free form or obtained from the peptidase-med-
iated hydrolysis of host proteins (
Pinti et al., 2007). For example,
the utilization of serum amino acids by C. albicans gat1 mutant
and WT strains was examined. After 24 h of incubation, approxi-
mately 200
l
M of proline (corresponding to 100%) had been con-
sumed by both WT and gat1 mutant strains, which indicate the
availability and utilization of this amino acid by C. albicans in ser-
um (Limjindaporn et al., 2003). Another possibility is that nitrogen
transport in the C. neoformans gat1 mutant in the in vivo environ-
ment differs from that in the in vitro environment, which could
be sufficient for virulence and survival in macrophages. However,
it remains unknown whether the observed phenotype for the
gat1 mutant in vitro also occurs during infection. Although the
expression of GAT1 was not required for fungal pathogenesis, we
cannot rule out the possibility that its product and related proteins
are required for infection, since nitrogen metabolism is relevant in
essential aspects of C. neoformans biology (
Kingsbury and McCus-
ker, 2008; Kingsbury et al., 2004; Wickes et al., 1996
).
C. neoformans transcription factor Gat1 activates the expression
of known NCR-sensitive genes, as occurs with GATA-factors ortho-
logs in other fungi. We found that genes encoding proline dehydro-
genase, glutamate dehydrogenase, and amino acid transporter, the
corresponding orthologs of S. cerevisiae PUT1, GDH2 and GAP1,
respectively, are upregulated in the WT strain in comparison with
gat1 mutant during growth with proline as nitrogen source. The
upregulation of such genes is in agreement with the observed ef-
fect on S. cerevisiae NCR-sensitive genes (
Hofman-Bang, 1999;
Scherens et al., 2006
). C. albicans Gat1 also regulates the expression
of the general amino acid transporter GAP1 ortholog (
Limjindaporn
et al., 2003
).
The expression of genes related to capsule biosynthesis and car-
bohydrate metabolism are also controlled by C. neoformans Gat1
(Table 1). One of the genes encoding capsular associated protein
is downregulated in the WT strain. Interestingly, UDP-glucose
dehydrogenase and UDP-glucuronic acid decarboxylase genes are
also downregulated. These enzymes directly participate in GXM
biosynthesis, which may explain the reduced levels of extracellular
GXM produced by the gat1 mutant. The basic building units of
GXM are UDP-glucuronic acid, UDP-xylose and GDP-mannose,
and the connections between these units are done by glycosyl-
transferases (reviewed in
Zaragoza et al. (2009)). Mannose is the
most abundant sugar unit in GXM followed by glucuronic acid.
The addition of glucuronic acid to the polysaccharide chain re-
quires UDP-glucuronic acid synthesis, which occurs via oxidation
of UDP-glucose by UDP-glucose dehydrogenase in the cytoplasm.
Xylose is the third component of GXM. UDP-xylose, the target of
fungal glycosyltransferases, is synthesized from the decarboxyl-
ation of UDP-glucuronic acid by UDP-glucuronic acid decarboxyl-
ase (UGD1) (reviewed in
Zaragoza et al. (2009)). C. neoformans
UGD1 is essential for growth at 37 °C and for capsule biosynthesis
(
Moyrand and Janbon, 2004). The gene encoding a
a
-1,3-manno-
syltransferase, which catalyzes the transfer of mannose from
GDP-mannose to
a
-1,3-linked mannose disaccharides (Sommer
et al., 2003), is upregulated in the WT in comparison to gat1 mu-
tant strain. Cryptococcal mannosyltransferase-1 gene (CMT1) was
shown to be homologous to C. neoformans CAP59, a gene involved
in capsule synthesis and GXM export (
Garcia-Rivera et al., 2004;
Zaragoza et al., 2009
). The loss of C. neoformans GAT1 negatively
influences the contents of released GXM in culture supernatants.
This phenotype, found in gat1 mutant strain, can be related to
the remodeled pattern of gene expression of key genes that act
in the GXM biosynthesis. The expression of squalene monooxygen-
ase, a rate-limiting step in sterol biosynthesis, is also reduced in
the gat1 mutant. A decrease in sterol production could potentially
impair vesicle biogenesis and secretion through unconventional
pathways, a mechanism used by C. neoformans to transport GXM
to extracellular environments (
Rodrigues et al., 2007).
The Sre1 transcription factor of C. neoformans regulates oxygen
sensing, sterol homeostasis and virulence (
Chang et al., 2007).
Gene expression analysis of WT and sre1 mutant strains in the
presence of cobalt chloride (a hypoxia mimicking agent) revealed
that genes involved in ergosterol biosynthesis and in iron/copper
transport are Sre1-regulated (
Lee et al., 2007). Surprisingly, at least
23 genes related to nitrogen metabolism were also regulated by
Sre1 during growth with 0.6 mM CoCl
2
, including C. neoformans
transcription factor GAT1 (transcriptional activator) [GenBank:
CNA01820] (Lee et al., 2007). As seen for Sre1, Gat1 also regulates
genes involved in ergosterol biosynthesis, cell wall integrity and
iron uptake. These findings suggest that these two transcription
factors are somehow linked and can act in parallel regulating
important pathways in C. neoformans.
5. Conclusions
In conclusion, we have shown that Gat1, a GATA-type transcrip-
tion factor in C. neoformans, is directly involved in nitrogen uptake
Fig. 5. Validation of the microarray results by quantitative real time RT-PCR.
Relative expression of amino acid transporter, proline dehydrogenase, high-affinity
iron permease FTR1, alpha-1,3-mannosyltransferase, squalene monooxygenase,
chitin synthase 2, and capsular associated protein transcripts during growth of WT
and gat1 mutant cells in YNB amended with 10 mM of proline as nitrogen source.
The measured quantity of the mRNA in each of the samples was normalized using
the Ct values obtained for the actin gene. Data are shown as mean ± SD.
Ã
P < 0.05.
ÃÃ
P < 0.01.
L. Kmetzsch et al. / Fungal Genetics and Biology xxx (2010) xxx–xxx
7
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
and metabolism. Rapamycin sensitivity assays revealed that TOR
signaling pathway does not act through Gat1 in C. neoformans.
Additionally, Gat1 is neither required for C. neoformans survival
during macrophage infection nor for virulence in a mouse intrana-
sal model of cryptococcosis. As revealed by microarray analysis,
Gat1 controls the transcription of NCR-sensitive genes, and genes
involved in ergosterol biosynthesis, iron uptake, cell wall organiza-
tion and capsule biosynthesis in C. neoformans.
Acknowledgments
This work was supported by grants from the Brazilian agencies
Conselho Nacional de Desenvolvimento Científico e Tecnológico
(CNPq, Brazil), Coordenação de Aperfeiçoamento de Pessoal de
Nível Superior (CAPES, Brazil), Fundação de Amparo a Pesquisa
no Estado do Rio de Janeiro (FAPERJ, Brazil) and Financiadora de
Estudos e Projetos (FINEP, Brazil). The authors thank Dr. Joseph
Heitman and Dr. Alex Idnurm for providing pJAF15, pAI4 plasmids
and Dr. Arturo Casadevall for providing the monoclonal antibody
anti-GXM (18B7). Automated DNA sequencing was performed at
the facilities of the Brazilian Genome Network at the Center of Bio-
technology, CBiot-UFRGS-RS.
Appendix A. Supplementary material
Supplementary data associated with this article can be found, in
the online version, at
doi:10.1016/j.fgb.2010.07.011.
References
Beck, T., Hall, M.N., 1999. The TOR signalling pathway controls nuclear localization
of nutrient-regulated transcription factors. Nature 402, 689–692.
Bertram, P.G., Choi, J.H., Carvalho, J., Ai, W., Zeng, C., Chan, T.F., Zheng, X.F., 2000.
Tripartite regulation of Gln3p by TOR, Ure2p, and phosphatases. J. Biol. Chem.
275, 35727–35733.
Cardenas, M.E., Cutler, N.S., Lorenz, M.C., Di Como, C.J., Heitman, J., 1999. The TOR
signaling cascade regulates gene expression in response to nutrients. Genes
Dev. 13, 3271–3279.
Chang, Y.C., Bien, C.M., Lee, H., Espenshade, P.J., Kwon-Chung, K.J., 2007. Sre1p, a
regulator of oxygen sensing and sterol homeostasis, is required for virulence in
Cryptococcus neoformans. Mol. Microbiol. 64, 614–629.
Coffman, J.A., Cooper, T.G., 1997. Nitrogen GATA factors participate in
transcriptional regulation of vacuolar protease genes in Saccharomyces
cerevisiae. J. Bacteriol. 179, 5609–5613.
Coffman, J.A., Rai, R., Cunningham, T., Svetlov, V., Cooper, T.G., 1996. Gat1p, a GATA
family protein whose production is sensitive to nitrogen catabolite repression,
participates in transcriptional activation of nitrogen-catabolic genes in
Saccharomyces cerevisiae. Mol. Cell Biol. 16, 847–858.
Cox, G.M., Mukherjee, J., Cole, G.T., Casadevall, A., Perfect, J.R., 2000. Urease as a
virulence factor in experimental cryptococcosis. Infect. Immun. 68, 443–448.
Cruz, M.C., Goldstein, A.L., Blankenship, J., Del Poeta, M., Perfect, J.R., McCusker, J.H.,
Bennani, Y.L., Cardenas, M.E., Heitman, J., 2001. Rapamycin and less
immunosuppressive analogs are toxic to Candida albicans and Cryptococcus
neoformans via FKBP12-dependent inhibition of TOR. Antimicrob. Agents
Chemother. 45, 3162–3170.
Facklam, T.J., Marzluf, G.A., 1978. Nitrogen regulation of amino acid catabolism in
Neurospora crassa. Biochem. Genet. 16, 343–354.
Fonseca, F.L., Frases, S., Casadevall, A., Fischman-Gompertz, O., Nimrichter, L.,
Rodrigues, M.L., 2009. Structural and functional properties of the Trichosporon
asahii glucuronoxylomannan. Fungal Genet. Biol. 46, 496–505.
Fraser, J.A., Subaran, R.L., Nichols, C.B., Heitman, J., 2003. Recapitulation of the
sexual cycle of the primary fungal pathogen Cryptococcus neoformans var. Gattii:
implications for an outbreak on Vancouver Island, Canada. Eukaryot. Cell. 2,
1036–1045.
Garcia-Pedrajas, M.D., Nadal, M., Kapa, L.B., Perlin, M.H., Andrews, D.L., Gold, S.E.,
2008. DelsGate, a robust and rapid gene deletion construction method. Fungal
Genet. Biol. 45, 379–388.
Garcia-Rivera, J., Chang, Y.C., Kwon-Chung, K.J., Casadevall, A., 2004. Cryptococcus
neoformans CAP59 (or Cap59p) is involved in the extracellular trafficking of
capsular glucuronoxylomannan. Eukaryot. Cell. 3, 385–392.
Giles, S.S., Dagenais, T.R., Botts, M.R., Keller, N.P., Hull, C.M., 2009. Elucidating the
pathogenesis of spores from the human fungal pathogen Cryptococcus
neoformans. Infect. Immun. 77, 3491–3500.
Hardwick, J.S., Kuruvilla, F.G., Tong, J.K., Shamji, A.F., Schreiber, S.L., 1999.
Rapamycin-modulated transcription defines the subset of nutrient-sensitive
signaling pathways directly controlled by the Tor proteins. Proc. Natl. Acad. Sci.
USA 96, 14866–14870.
Hensel, M., Arst Jr., H.N., Aufauvre-Brown, A., Holden, D.W., 1998. The role of the
Aspergillus fumigatus areA gene in invasive pulmonary aspergillosis. Mol. Gen.
Genet. 258, 553–557.
Hofman-Bang, J., 1999. Nitrogen catabolite repression in Saccharomyces cerevisiae.
Mol. Biotechnol. 12, 35–73.
Idnurm, A., Reedy, J.L., Nussbaum, J.C., Heitman, J., 2004. Cryptococcus neoformans
virulence gene discovery through insertional mutagenesis. Eukaryot. Cell. 3,
420–429.
Jiang, Y., Broach, J.R., 1999. Tor proteins and protein phosphatase 2A reciprocally
regulate Tap42 in controlling cell growth in yeast. EMBO J. 18, 2782–2792.
Kingsbury, J.M., McCusker, J.H., 2008. Threonine biosynthetic genes are essential in
Cryptococcus neoformans. Microbiology 154, 2767–2775.
Kingsbury, J.M., Yang, Z., Ganous, T.M., Cox, G.M., McCusker, J.H., 2004. Cryptococcus
neoformans Ilv2p confers resistance to sulfometuron methyl and is required for
survival at 37 °C and in vivo. Microbiology 150, 1547–1558.
Larkin, M.A., Blackshields, G., Brown, N.P., Chenna, R., McGettigan, P.A., McWilliam,
H., Valentin, F., Wallace, I.M., Wilm, A., Lopez, R., Thompson, J.D., Gibson, T.J.,
Higgins, D.G., 2007. Clustal W and Clustal X version 2.0. Bioinformatics 23,
2947–2948.
Lee, H., Bien, C.M., Hughes, A.L., Espenshade, P.J., Kwon-Chung, K.J., Chang, Y.C.,
2007. Cobalt chloride, a hypoxia-mimicking agent, targets sterol synthesis in
the pathogenic fungus Cryptococcus neoformans. Mol. Microbiol. 65, 1018–1033.
Lengeler, K.B., Wang, P., Cox, G.M., Perfect, J.R., Heitman, J., 2000. Identification of
the MATa mating-type locus of Cryptococcus neoformans
reveals a serotype A
MATa strain thought to have been extinct. Proc. Natl. Acad. Sci. USA 97, 14455–
14460.
Liao, W.L., Ramon, A.M., Fonzi, W.A., 2008. GLN3 encodes a global regulator of
nitrogen metabolism and virulence of C. Albicans. Fungal Genet. Biol. 45, 514–
526.
Limjindaporn, T., Khalaf, R.A., Fonzi, W.A., 2003. Nitrogen metabolism and virulence
of Candida albicans require the GATA-type transcriptional activator encoded by
GAT1. Mol. Microbiol. 50, 993–1004.
Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using
real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25,
402–408.
Magasanik, B., Kaiser, C.A., 2002. Nitrogen regulation in Saccharomyces cerevisiae.
Gene 290, 1–18.
Marzluf, G.A., 1997. Genetic regulation of nitrogen metabolism in the fungi.
Microbiol. Mol. Biol. Rev. 61, 17–32.
McClelland, E.E., Bernhardt, P., Casadevall, A., 2005. Coping with multiple virulence
factors: which is most important? PLoS Pathog. 1, e40.
Moyrand, F., Janbon, G., 2004. UGD1, encoding the Cryptococcus neoformans UDP-
glucose dehydrogenase, is essential for growth at 37 °C and for capsule
biosynthesis. Eukaryot. Cell. 3, 1601–1608.
Panepinto, J., Komperda, K., Frases, S., Park, Y.D., Djordjevic, J.T., Casadevall, A.,
Williamson, P.R., 2009. Sec6-dependent sorting of fungal extracellular
exosomes and laccase of Cryptococcus neoformans. Mol. Microbiol. 71, 1165–
1176.
Pinti, M., Orsi, C.F., Gibellini, L., Esposito, R., Cossarizza, A., Blasi, E., Peppoloni, S.,
Mussini, C., 2007. Identification and characterization of an aspartyl protease
from Cryptococcus neoformans. FEBS Lett. 581, 3882–3886.
Rodrigues, M.L., Nimrichter, L., Oliveira, D.L., Frases, S., Miranda, K., Zaragoza, O.,
Alvarez, M., Nakouzi, A., Feldmesser, M., Casadevall, A., 2007. Vesicular
polysaccharide export in Cryptococcus neoformans is a eukaryotic solution to
the problem of fungal trans-cell wall transport. Eukaryot. Cell. 6, 48–59.
Rutherford, J.C., Lin, X., Nielsen, K., Heitman, J., 2008. Amt2 permease is required to
induce ammonium-responsive invasive growth and mating in Cryptococcus
neoformans. Eukaryot. Cell. 7, 237–246.
Scherens, B., Feller, A., Vierendeels, F., Messenguy, F., Dubois, E., 2006. Identification
of direct and indirect targets of the Gln3 and Gat1 activators by transcriptional
profiling in response to nitrogen availability in the short and long term. FEMS
Yeast Res. 6, 777–791.
Sommer, U., Liu, H., Doering, T.L., 2003. An alpha-1,3-mannosyltransferase of
Cryptococcus neoformans. J. Biol. Chem. 278, 47724–47730.
Stanbrough, M., Rowen, D.W., Magasanik, B., 1995. Role of the GATA factors Gln3p
and Nil1p of Saccharomyces cerevisiae in the expression of nitrogen-regulated
genes. Proc. Natl. Acad. Sci. USA 92, 9450–9454.
Tamura, K., Dudley, J., Nei, M., Kumar, S., 2007. MEGA4: molecular evolutionary
genetics analysis (MEGA) software version 4.0. Mol. Biol. Evol. 24, 1596–1599.
Thomas, G., Hall, M.N., 1997. TOR signalling and control of cell growth. Curr. Opin.
Cell Biol. 9, 782–787.
Toffaletti, D.L., Rude, T.H., Johnston, S.A., Durack, D.T., Perfect, J.R., 1993. Gene
transfer in Cryptococcus neoformans by use of biolistic delivery of DNA. J.
Bacteriol. 175, 1405–1411.
Wickes, B.L., Mayorga, M.E., Edman, U., Edman, J.C., 1996. Dimorphism and haploid
fruiting in Cryptococcus neoformans: association with the alpha-mating type.
Proc. Natl. Acad. Sci. USA 93, 7327–7331.
Zaragoza, O., Rodrigues, M.L., De Jesus, M., Frases, S., Dadachova, E., Casadevall, A.,
2009. The capsule of the fungal pathogen Cryptococcus neoformans. Adv. Appl.
Microbiol. 68, 133–216.
8 L. Kmetzsch et al. / Fungal Genetics and Biology xxx (2010) xxx–xxx
Please cite this article in press as: Kmetzsch, L., et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake and metabolism in the hu-
man pathogen Cryptococcus neoformans. Fungal Genet. Biol. (2010), doi:
10.1016/j.fgb.2010.07.011
CURRICULUM VITAE
Nome: Débora Leite de Oliveira
Nascimento: 03/10/1983
Naturalidade: Rio de Janeiro
Formação Acadêmica
Bacharel em Microbiologia e Imunologia pela Universidade Federal do Rio de Janeiro, de
março de 2002 à dezembro de 2005.
Doutorado em Química Biológica no Instituto de Bioquímica Médica Universidade
Federal do Rio de Janeiro.
Orientação de Monografia
O papel das proteínas Sec4 e Grasp na formação de vesículas extracelulares em
Saccharomyces cerevisiae
Luna Sobrino Joffe, Monografia apresentada ao Instituto de Microbiologia
Universidade Federal do Rio de Janeiro
Atividade imunobiológica de vesículas secretadas pelo patógeno fúngico Candida
albicans
Gabriele Vargas Cesar, Monografia apresentada ao Instituto de Microbiologia
Universidade Federal do Rio de Janeiro
Comunicações em congress
13 comunicações em congressos nacionais
Publicações
1. Oliveira, Débora L. et al. Biogenesis of extracellular vesicles in yeast Many questions with few
answers, v. 3, p. 1-4, 2010.
2. OLIVEIRA, D. L. ; et al. Extracellular Vesicles from Cryptococcus neoformans Modulate Macrophage
Functions. Infection and Immunity, v. 78, p. 1601-1609, 2010
3. Oliveira, Débora L. et al. Characterization of Yeast Extracellular Vesicles: Evidence for the
Participation of Different Pathways of Cellular Traffic in Vesicle Biogenesis. Plos One , v. 5, p.
e11113, 2010.
4. Kmetzsch, Lívia ; et al. The GATA-type transcriptional activator Gat1 regulates nitrogen uptake
and metabolism in the human pathogen Cryptococcus neoformans. Fungal Genetics and Biology
(Print) , p. 1-2, 2010.
5. Kmetzsch, Lívia ; et al. The Vacuolar Ca2+ Exchanger Vcx1 is involved in Calcineurin-Dependent
Ca2+ Tolerance and Virulence in Cryptococcus neoformans.. Eukaryotic Cell , v. 10, p. 12, 2010.
Oliveira, et al. Biogenesis of extracellular vesicles in yeast Many questions with few answers, v. 3, p.
1-4, 2010.
6. Oliveira, Débora L. et al. Cryptococcus neoformans cryoultramicrotomy and vesicle fractionation
reveals an intimate association between membrane lipids and glucuronoxylomannan. Fungal
Genetics and Biology, v. 46, p. 956-963, 2009.
7. RODRIGUES, M. L. ; et al. Extracellular vesicles produced by Cryptococcus neoformans contain
protein components associated with virulence. Eukaryotic Cell , v. 7, p. 58-67, 2008
8. RODRIGUES, M. L. et al. Vesicular Trans-Cell Wall Transport in Fungi: A Mechanism for the Delivery of
Virulence-Associated Macromolecules?. Lipid Insights , v. 2, p. 27-40, 2008.
9. RODRIGUES, M. L. et al. Vesicular Polysaccharide Export in Cryptococcus neoformans Is a
Eukaryotic Solution to the Problem of Fungal Trans-Cell Wall Transport (M.L.R., L.N., and D.L.O.
contributed equally to this work.). Eukaryotic Cell , v. 6, p. 48-59, 2007.
Livros Grátis
( http://www.livrosgratis.com.br )
Milhares de Livros para Download:
Baixar livros de Administração
Baixar livros de Agronomia
Baixar livros de Arquitetura
Baixar livros de Artes
Baixar livros de Astronomia
Baixar livros de Biologia Geral
Baixar livros de Ciência da Computação
Baixar livros de Ciência da Informação
Baixar livros de Ciência Política
Baixar livros de Ciências da Saúde
Baixar livros de Comunicação
Baixar livros do Conselho Nacional de Educação - CNE
Baixar livros de Defesa civil
Baixar livros de Direito
Baixar livros de Direitos humanos
Baixar livros de Economia
Baixar livros de Economia Doméstica
Baixar livros de Educação
Baixar livros de Educação - Trânsito
Baixar livros de Educação Física
Baixar livros de Engenharia Aeroespacial
Baixar livros de Farmácia
Baixar livros de Filosofia
Baixar livros de Física
Baixar livros de Geociências
Baixar livros de Geografia
Baixar livros de História
Baixar livros de Línguas
Baixar livros de Literatura
Baixar livros de Literatura de Cordel
Baixar livros de Literatura Infantil
Baixar livros de Matemática
Baixar livros de Medicina
Baixar livros de Medicina Veterinária
Baixar livros de Meio Ambiente
Baixar livros de Meteorologia
Baixar Monografias e TCC
Baixar livros Multidisciplinar
Baixar livros de Música
Baixar livros de Psicologia
Baixar livros de Química
Baixar livros de Saúde Coletiva
Baixar livros de Serviço Social
Baixar livros de Sociologia
Baixar livros de Teologia
Baixar livros de Trabalho
Baixar livros de Turismo