Download PDF
ads:
FERNANDA TAÍS MECABÔ
A RESTRIÇÃO PROTÉICA DURANTE A LACTAÇÃO
ALTERA O CONTROLE PELO SISTEMA NERVOSO
AUTÔNOMO DA SECREÇÃO DE INSULINA INDUZIDA
PELA GLICOSE EM RATOS ADULTOS.
“Dissertação apresentada ao Curso de
mestrado em Ciências Biológicas
(áreas de concentração – Biologia Celular),
da Universidade Estadual de Maringá para
a obtenção do grau de Mestre em
Ciências Biológicas.”
Maringá - 2006
ads:
Livros Grátis
http://www.livrosgratis.com.br
Milhares de livros grátis para download.
“PROTEIN RESTRICTION DURING LACTATION
ALTERS THE AUTONOMIC NERVOUS SYSTEM
CONTROL ON GLUCOSE-INDUCED INSULIN
SECRETION IN ADULT RATS.
Orientador: Prof. Dr. Paulo Cezar de Freitas Mathias
Orientanda: Fernanda Taís Mecabô
Maringá – 2006
ads:
AGRADECIMENTOS:
Aos meus pais: Gentil e Rozenete. Minha vovozinha Irma e minha irmã Carol, pelo
apoio incondicional.
Ao meu amor: Antonio, pelo carinho, compreensão e paciência.
Aos colegas de laboratório: Dionizia, Sabrina, Ana Eliza, Fernanda, Diego,
Anderson, Rodrigo, Marcelo e Carioca, pela ajuda nas dificuldades e pelos
momentos de descontração.
Às técnicas: Maroly e Clarice pelo auxílio e amizade.
Ao Prof. Paulo por apostar na minha capacidade.
Aos professores da pós-graduação pelo empenho em transmitir o conhecimento
científico.
Resumo:
1. Objetivo geral: Avaliar a modulação promovida pelo sistema nervoso
autônomo (SNA) na secreção de insulina de ratos adultos que foram
submetidos à desnutrição protéica durante parte da lactação.
Bases teóricas: A desnutrição protéica é um problema importante nos
países em desenvolvimento. Quando mudanças no estado nutricional
ocorrem no início da vida, podem afetar o desenvolvimento dos
mecanismos de controle metabólico desse organismo. Evidências
epidemiológicas indicam que uma relação entre desnutrição protéica e
um aumento na suscetibilidade a alterações na tolerância a glicose. Estudos
em modelos experimentais de desnutrição protéica perinatal documentam
alterações nas ilhotas pancreáticas. De maneira geral, ocorre uma
diminuição na magnitude da resposta secretória da célula β pancreática e
dessa forma, leva a uma permanente hipoinsulinemia. A glicose é
considerada a biomolécula mais importante na estimulação das lulas β
pancreáticas durante o processo de secreção de insulina. Todavia, o SNA
constituído pelo Sistema Nervoso Parassimpático (SNP) e Sistema Nervoso
Simpático (SNS) tamm exercem um importante papel na regulação da
secreção de insulina. O SNP, liberando o seu principal neurotransmissor, a
acetilcolina, através da ligação com receptores muscarínicos nas células β
pancreáticas , potencializa a secreção de insulina estimulada por glicose. O
SNS, liberando ou secretando catecolaminas (adrenalina e noradrenalina),
exerce um papel dual na secreção de insulina, sendo inibitório via
receptores α adrenérgico e estimulatório via receptores β adrenérgico.
Ilhotas pancreáticas de animais que sofreram desnutrição protéica
desenvolvem diversas alterações intrínsecas, como morfologia, expressão
de genes, atividade enzimática, etc. Entretanto, essas evidências não
excluem a possibilidade da participação do SNA como modulador da
secreção deficiente de insulina. Foi observado que a desnutrição protéica
depois do desmame provoca redução na atividade do SNP e aumento da
atividade do SNS. No entanto, é provável que as alterações sofridas por um
mamífero durante a lactação não sejam exatamente as mesmas que
aquelas sofridas pelo animal jovem, levando-se em conta que a lactação é
uma fase em que ocorre uma alta taxa de crescimento e desenvolvimento,
particularmente no que se refere ao Sistema Nervoso.
2. Objetivos específicos: Observar a secreção de insulina in vivo e in vitro,
estimulada por glicose e ainda a modulação exercida pelos agonistas e
antagonistas dos receptores muscarínicos (acetilcolina e atropina) e dos
receptores α adrenérgicos (yohimbina e oxymetazolina) e β adrenérgicos
(isoproterenol).
3. Procedimentos: Ratos Wistar cujas mães receberam nos 14 primeiros dias
da lactação uma dieta isocalórica contendo 4% de proteína (LP) ou uma
dieta contendo 23% de proteína (NP). Aos 21 dias eram desmamados e aos
81dias parte dos animais de ambos os grupos foi submetido ao teste de
tolerância à glicose intravenosa (ivGTT) associada ou não a aceticolina
(ach), atropina (atr), yohimbina (yoh) e oxymetazolina (oxy). O restante dos
animais foram sacrificados. Foi medido o comprimento e o peso corporal. O
índice de Lee foi calculado [peso corporal(g)
1/3
/comprimento naso-anal
(cm). Foram tamm isoladas e pesadas as gorduras periepididimais e
retroperitoneais para estimar a massa de gordura corporal e o cérebro para
aferir possíveis alterações. As ilhotas pancreáticas foram isoladas pelo
método da colagenase. Ilhotas do grupo NP e LP foram incubadas com
glicose na presença ou ausência de epinefrina (100nM) com propranolol
(100nm) ou com carbamilcolina (1mM) com e sem atr (10µM). Tamm
foram feitas incubações com iso (10µM) com e sem yoh (10µM).
4. Resultados: Ratos submetidos à desnutrição protéica durante a lactação
apresentaram o peso corporal reduzido em 23% e das gorduras
periepididimal e retroperitoneal em 30% e 49%, respectivamente, tudo
comparado aos NP. A insulinemia de jejum foi 41% menor nos LP sem,
contudo apresentar diferença significativa na glicemia de jejum. O ivGTT
mostrou que a desnutrição provocou intolerância à glicose e
hipoinsulinemia. Os animais LP apresentaram I (calculado a partir da área
sob a curva - AUC) 31% menor que os NP(p<0.05). Ach afetou os LP
aumentando o I (AUC) 64% e em apenas 18% nos NP. Atr teve efeito
somente nos NP reduzindo o I (AUC) em 22% (p<0.05). Oxy reduziu o I
(AUC) em 25% apenas nos NP. Por outro lado, yoh aumentou o I(AUC)
em 65% nos LP e em somente 32% nos NP (p<0.05). Em ilhotas, a
carbamilcolina potencializou a secreção de insulina em 311% nos LP e em
594% nos NP (p<0.05). Epinefrina inibiu a secreção na mesma magnitude
nos dois grupos. Isso potencializou em 70% a secreção nos LP e apenas
35% nos NP (p<0.05).
5. Conclusões: A desnutrição protéica perinatal induz mudanças no
metabolismo que são mantidas na vida adulta. Dentre as alterações está o
baixo peso e a hipoinsulinemia. Ilhotas pancreáticas de animais LP
secretam menos insulina sob o estímulo da glicose tanto in vivo como in
vitro. A resposta secretória das ilhotas aos moduladores autonômicos
também está alterada. Dessa forma os resultados sugerem que a regulação
da secreção de insulina está comprometida, não o controle pela glicose,
mas também os efeitos insulinotrópicos modulados pelo SNA em ratos
adultos que sofreram desnutrição protéica durante parte da lactação.
Summary:
6. Main Objective: To evaluate the modulation caused by the autonomic
nervous system (SNA) on the insulin secretion in adult rats submitted to
protein undernutrition during part of lactation.
Theoretical foundations: Protein undernutrition is an important problem in
developing countries. When changes in nutritional situation happen in the
beginning of life, they may affect the development of the organism’s
metabolic control mechanisms. Epidemiologic evidences indicate that there
is a relationship between protein undernutrition and increase in susceptibility
to alterations in glucose tolerance. Studies in perinatal protein undernutrition
experimental models show alterations in pancreatic islets. In a general way,
there is a decrease in the magnitude of the secretory response of the β
pancreatic cell, thus leading to permanent hypoinsulinemia. Glucose is
considered to be the most important biomolecule to the β pancreatic cells
stimulation, during the insulin secretion process. However, the SNA,
consisting of the Parasympathetic Nervous System (SNP) and the
Sympathetic Nervous System (SNS), also have an important role in the
regulation of insulin secretion. The SNP, releasing acetylcholine its main
neurotransmitter – through the binding with muscarinic receptors in the
pancreatic β cells, enhances glucose-stimulated insulin secretion. SNS,
releasing or secreting catecholamines (adrenaline and noradrenaline), plays
a dual role in insulin secretion, being an inhibitor through α adrenergic
receptors and a stimulator through the β adrenergic receptors. Pancreatic
islets from animals who have suffered protein undernutrition develop several
intrinsic alterations, e.g. in morphology, genes expression, enzymatic
activity, etc. However, these evidences do not exclude the possibility of the
participation of the SNA as a modulator of the defective insulin secretion. It
has been observed that the protein undernutrition after weaning provokes a
decrease in SNP activity and increase in SNS activity. Nevertheless, it is
possible that the alterations suffered by a mammal during lactation are not
exactly the same as those ones suffered by the young animal, considering
that lactation is a period in which there is a high rate of growth and
development, especially regarding the central nervous system.
7. Specific Objectives: Observe glucose-induced insulin secretion in vivo and
in vitro, and also the modulation induced by agonists and antagonists of the
muscarinic receptors acetylcholine and atropine, respectively and of the
α adrenergic receptors yohimbine and oxymetazoline and one β adrenergic
agonist isoproterenol in adults rats whose were fed with protein restriction
during part of lactation.
8. Procedures: Wistar rats whose mothers received an isocaloric diet
containing 4% protein (LP) or a 23% protein diet (NP), in the 14 days
following birth. They were weaned at the age of 21 days, and at the age of
81 days part of the animals from both groups was submitted to the
intravenous glucose tolerance test (ivGTT), associated or not to
acetylcholine (ach), atropine (atr), yohimbine (yoh) and oxymetazoline (oxy).
The rest of the animals were killed. Length and body weight were measured.
Lee index was calculated [body weight(g)
1/3
/nasoanal length (cm)].
Periepididymal and retroperitoneal fat was also isolated and weighed to
estimate body fat mass, as well as the brain, to assess possible alterations.
Pancreatic islets were isolated by the collagenase method. Islets from the
NP and LP groups were incubated with glucose in the presence and in the
absence of epinephrine (100nM) or with cholinergic agonist carbamylcholine
(1mM) with and without muscarinic antagonist atropine (10µM). Incubations
were made also with a β adrenergic agonist isoproterenol (10µM), with
and without yohimbine (10µM).
9. Results: Rats submitted to protein restriction during lactation presented
their body weight reduced in 23% and their periepididymal and
retroperitoneal fat pads in 30 and 49%, respectively, all when compared to
NP rats. Fasting insulinemia was 41% lower in the LP rats though there
were no significant differences in the fasting glycemia. Protein undernutrition
provoked intolerance to glucose and hypoinsulinemia during ivGTT. LP
animals presented I (calculated from the area under the curve) 31% lower
than NP rats (p<0.05). Ach affected LP rats, provoking a 64% increase in
their I, while increasing only 18% in the NP animals. Atr affected only NP
animals, reducing I (AUC) by 22% (p<0.05). Oxy reduced I by 25% only in
NP rats. On the other hand, yoh increased I by 65% in LP and only 32% in
NP (p<0.05). In islets, carbamylcholine increased insulin secretion by 311%
in the LP rats and 594% in the NP ones (p<0.05). Epinephrine inhibited
secretion in the same rate for both groups. Isoproterenol increased secretion
by 70% in LP and only 35% in NP (p<0.05).
10. Conclusions: Perinatal protein undernutrition induces changes in
metabolism which remain during adult life. Among the alterations is low
weight and hypoinsulinemia. Pancreatic islets from LP animals secrete less
insulin under glucose stimulus both in vivo and in vitro. Results suggest that
the regulation of insulin secretion is impaired, not only regarding glucose
control as well as the insulinotropic effects modulated by the SNA in adult
rats who suffered from protein underutrition during part of their lactation
period.
Protein restriction during lactation alters the autonomic nervous system
control on glucose-induced insulin secretion in adult rats.
CLARICE GRAVENA, ANA ELIZA ANDREAZZI, FERNANDA TAIS
MECABÔ, SABRINA GRASSIOLLI, VIVIANE M. SCANTAMBURLO &
PAULO C. F. MATHIAS
1
.
1
Department of Cell Biology and Genetics, State University of Maringá, 87020-900
Maringá, PR, Brazil.
Abstract
Involvement of autonomic nervous system (ANS) neurotransmitters on insulin secretion
in rats submitted to protein malnutrition during lactation was studied. During the first
2/3 of lactation, mothers received a 4% protein diet (LP). Control group received
normal diet (23% protein) (NP). After protein restriction, mothers received normal
diets. At 81 days rats were submitted to ivGTT. Plasma glucose and insulin
concentration (PIC) were measured. Glucose-induced insulin secretion (GIIS) was
tested in pancreatic islets. Fasting normoglycemia and hypoinsulinemia were observed
in LP rats. Glucose intolerance and low PIC in LP group were detected during ivGTT.
Acetylcholine or blockage of α-adrenoceptors induced high PIC increment in LP rats;
atropine or stimulation of α-adrenoceptors did not change PIC. Insulin secretion of LP
rat islets showed low glucose and carbachol responses. Epinephrine inhibited GIIS in
both islet groups. Hypoinsulinemia observed in lactation-malnourished rats might be
caused by alterations in GIIS regulation, including ANS modulation.
Keywords: Protein malnutrition, lactation, insulin secretion, autonomous nervous
system.
Correspondence: Paulo C. F. Mathias, Laboratory of Secretion Cell Biology, Dept. of
Cell Biology and Genetics, State University of Maringá, Av. Colombo 5790, CEP
87020-900, Maringá, PR, Brazil. Phone/Fax: 55 44 32 63 26 48. E-mail:
pmathias@uem.br
INTRODUCTION
Human perinatal malnutrition has been suggested as a risk to predisposing
metabolic and cardiovascular diseases later in life (Bhargava et al., 2004; Waterland &
Garza, 2002). Protein restriction during gestation and lactation in rodents induce
underweight, growth and development impairment in the offspring, and it is associated
with irreversible metabolic disturbances when the pups turn into adult life (Dollet et al.,
1988; Garofano et al., 1999; Langley-Evans, 1997; Rasschaert et al., 1995). It has been
recognized that central nervous system (CNS) is pivotal for metabolism and body
weight control, and the hypothalamus has been identified as the major area
concentrating many neurons implicated in the reception of signals from the periphery,
which induce efferent signals to correct excessive or insufficient food intake and energy
storage (Steffens et al., 1990). Protein malnutrition during gestation and lactation
resulted in underweight, hypoglycemia and hipoinsulinemia in the offspring at weaning,
and it was associated to structural and functional changes in the hypothalamus
(Plagemann et al., 2000a; Plagemann et al., 2000b).
Low plasma insulin concentration and glucose intolerance were observed in
adult rats which were protein-malnourished only during lactation (Barbosa et al., 1999;
Moura et al., 1996). Hypoinsulinemia was associated to an impairment of glucose-
induced insulin secretion observed in pancreatic islets isolated from protein-
malnourished suckling rats; however, the mechanisms which are injured by early
protein restriction are still unclear. A fluctuation in glucose concentrations is by far the
most important signal that stimulates pancreatic islets beta-cells to release insulin into
the blood stream. Glucose enters into beta-cells and immediately stimulates the cells
metabolism, leading to ionic events that induce a huge increment in intracellular
calcium. The cytosolic free calcium starts exocytosis of insulin granules (Meglasson et
al., 1989). Other nutrient secretagogues, such as amino acids and fatty acids also
induce insulin secretion; however, in spite of a cellular metabolic regulation, insulin
secretion is also under important neural control. The beta-cellsmembrane is equipped
with many receptors to neurotransmitters and neuropeptides. Autonomic nervous
system (ANS), through their neurotransmitters, modulates the nutrient-induced insulin
secretion. While parasympathetic branches potentiate, sympathetic ends in pancreatic
islets inhibit glucose-induced insulin secretion (Gilon & Henquin, 2001).
Lactation is considered a very important phase in development, including that of
the CNS (Lima et al., 1993). Protein restriction in this phase might cause functional
injuries to the brain, leading to altered neural regulation of glucose-induced insulin
secretion. Therefore, we investigated the effect of protein malnutrition during lactation
on insulin secretion modulated by ANS neurotransmitters in adult rats.
MATERIAL AND METHODS
Dietary manipulation of lactating rats
Female Wistar rats were fed a normal laboratory diet throughout pregnancy.
After delivery the rats were distributed into two groups and each lactating dam was kept
with six pups. Since gender differences in insulin levels and glucose tolerance have
been observed in some early poor protein feeding maneuvers (Lopes Da Costa et al.,
2004), only male offspring were used on the experiments. On the normal protein (NP)
group all dams received a normal-protein (23%) diet ad libitum during lactation,
although the protein reduced (4%) diet had the same amount of calories as the normal
one, as described previously (Barbosa et al., 1999). On the low protein (LP) group all
dams received a low-protein diet ad libitum during the first 14 days of lactation,
returning to normal diet for the remaining 1/3 of lactation. After 21 days the pups were
weaned and received normal diet for 60 days at which time they were used for tests, as
indicated by others (Barbosa et al. 1999). The State University of Maringá Ethical
Committee for Animal Experiments approved the described protocol.
Early Protein Effects on Adult Rats
Adult rats with 81-day-old, NP or LP groups were anaesthetized by an
intraperitoneal injection of pentobarbital sodium [5 mg/100 g body weight (BW)] and
killed by cervical dislocation. The Lee index was calculated from the ratio [BW
1/3
(g)/nasoanal length (cm)] × 1000, used as a predictor of obesity in MSG-rodents
(Bernardis and Patterson, 1968). The periepididymal and retroperitoneal fat pads were
removed, washed with saline solution and weighed. The fat mass of these tissues is used as
a simple reliable estimation of body fat in normal and obese rodents (Rogers and Webb,
1980). To obtain rat brains, before killing, animals were perfused transcardially with saline
solution; the brains were removed and weighed.
Glucose Tolerance Test
Under ketamine and xylasine anesthesia (55mg and 8mg/kg BW, respectively), a
silicone cannula was implanted into 81-days-old rats’ left jugular and attached to the
animals’ back. To avoid blood clotting, heparinized saline (50 IU heparin/ml) was
previously injected into the cannula.
Intravenous glucose tolerance tests (ivGTT) were performed at 8:00 h in 81-days-old
LP and NP rats after a 12h fast (19:00-07:00 h) and without any anesthesia rats were
injected with a glucose load (1g glucose/Kg BW) through the cannula. Blood samples
(300 µl) were collected, from the same cannula, sequentially before glucose load (t0)
and 5 (t5), 15 (t15), 30 (t30) and 60 (t60) min after the injection of glucose. They were
then centrifuged, and the plasma was separated and stored at –20
0
C for posterior
dosage of glucose concentration by the glucose oxidase method (Kit-Bio Diagnostic
Chemistry Industry®) and insulin by radioimmunoassay (RIA) (Moura et al., 1996).
The total increments of glycemia (G) and insulinemia (I) were calculated using the
area under glycemia and insulinemia curves for the 60min of ivGTT, subtracting fasting
values. Insulinogenic index was calculated by I (pmol/l/60 min)/ G (mmol/l/60 min).
To investigate whether the decreased insulin response to glucose presented in this
perinatal poor protein nutrition model is related, at least in part, to impairment of
parasympathetic activity, we attempted to reactivate the glucose-induced insulinemia
increase by acetylcholine (Ach) administered exogenously. We used 27 nmoles/kg BW
Ach injected intraperitonealy (ip) 5 min before the ivGTT in LP and NP rats.
To test parasympathetic activity through its muscarinic effect on beta cells we used 20
nmoles/kg BW of muscarinic antagonist atropine (Atr). The drug was ip injected 5 min
before the study of ivGTT in LP and NP rats. Adrenergic adrenoceptors activity was
also investigated, using α
2
- adrenoceptor antagonist yohimbine. We used 20 nmoles/kg
BW yohimbine hydrochloride that was ip injected 5 min before the glucose load in LP
and NP rats. We also studied the influence of an α
2A
- adrenoceptor agonist,
oxymetazoline (16nmoles/kg BW), on glucose-induced insulinemia increase.
Oxymetazoline was selected because it has been proposed that in the pancreatic β-cell,
the α
2A
adrenoceptor subtype is specifically involved in the inhibition of insulin release
(Ullrich & Wollheim, 1985). Oxymetazoline was ip injected 5min before glucose load
in rats.
To select the concentration of each drug used in the protocol of ivGTT a dose response
curve was performed, measuring plasma insulin concentrations (results not shown). The
doses chosen were obtained by values close to EC
50
, which reduces or increases in
about 20% the total glycemia increment induced by glucose load in NP rats.
Preparation of islets of Langerhans
Isolation of islets from rat pancreas was performed as previously described
(Boschero et al., 1995) with adaptations. Male adult Wistar rats, only used to islets
isolation, were deeply anesthetized with ketamine and xylasine (55 and 8 mg/kg BW,
respectively), and the abdominal wall was cut and open. A 10 ml Hank’s buffered saline
solution (HBSS) containing collagenase type V (0.7 mg/ml, Sigma Chemical CO., St.
Louis, MO) was injected into the common bile duct of the rat. The pancreas, swollen
with the collagenase solution, was quickly excised and incubated in a plastic culture
bottle for 15 min at 37 ºC. The suspension obtained was filtered with a 0.5mm metal
mesh and washed with HBSS, including 0.12% bovine serum albumin fraction V (BSA)
in 5 continuous washings. The islets were collected with the aid of a microscope.
Insulin secretion from islets stimulated by glucose and neurotransmitters
To each incubation protocol a pool of islets were used, from, at least, 6 rats of
each group. Groups of 4 islets placed on plastic coverslips were pre-incubated for 60
min in 1.0 ml of Krebs-Ringer bicarbonate-buffered solution containing 0.12% (wt/vol)
bovine albumin (fraction V), and glucose 5.6 mM, pH 7.4. The solution was
equilibrated against a mixture of CO
2
(5%) and O
2
(95%). After the adaptation to a low
glucose concentration solution, islets were submitted to incubations for further 60 min
in glucose 2.8; 5.6; 8.3; 11.1; 16.7 and 20.0 mM with Krebs-Ringer solutions. To test
the response to cholinergic stimulus, islets were incubated with 8.3 mM of glucose plus
1.0 mM of carbachol in the absence or in the presence of the muscarinic antagonist
atropine 10 µM. Isoproterenol 10 µM, a nonspecific β-adrenoceptor agonist was used
to incubate islets stimulated with 16.7 mM of glucose in the presence or in the absence
of a specific α
2A
-adrenergic antagonist, yohimbine 10µM. Samples of incubation media
were taken and stored frozen until assay to measure secreted insulin by RIA.
Islet insulin content
Insulin content was extracted from 8 sets of 4 islets from each animal group.
Islets were incubated with 0.18 mmol/l HCl-ethanol (1:3, vol/vol) overnight at 4 °C
(Kaiser et al., 1991). The incubation was centrifuged at 10 000 rpm for 20 min and the
supernatants were used to measure the insulin content by RIA.
Data Analysis
Results were given as mean ± SEM. Data were submitted to Student t test or
variance analysis (ANOVA®). In the case of analyses with a significant F, the
differences between means were evaluated by Bonferroni t-test. P values less than 0.05
were considered statistically significant. The tests were performed using GraphPad
Prism version 3.02 for Windows (GraphPad Software®). Glucose concentration
secretion response curve was fitted with sigmoidal concentration response equation
(Insulin release = Min + (Max Min)/(1+ 10
(log Ec50 concentration) x Hill slope)
)), using the
same software package.
RESULTS
81 day old rats (LP) that were nursed by dams fed with a low protein diet had
lower body weight and nasoanal length than normal rats (NP) (Table I). LP animals also
presented lower fat storage as indicated by periepididymal and retroperitoneal fat pads.
However, the Lee index was not altered by this early malnutrition model, indicating that
animals were proportionally lean. Nevertheless, fasting plasma glycemia was not altered
by perinatal low protein diet, although the fasting insulinemia was significantly reduced
by 41% (Table I).
Both plasma glucose and insulin level responses to a glucose load (1g/kg BW, iv) were
affected by the low protein treatment (Figure 1). Plasma glucose was increased by 20%
at 5 min in LP rats, compared to the NP rats and remained greater throughout the
ivGTT. Conversely, plasma insulin, which had a fasting state 41% lower in LP animals
than in NP ones, had a further increase, although 46% less than NP animals 5 min after
the glucose load, returning to a state not different from NP at 60 min. The incremental
plasma glucose (G) was enhanced 32% in LP rats (505.78 ± 43.53 mmol/l/min)
compared to NP ones (343.17 ± 23.54 mmol/l/min), p<0.01. However, incremental
plasma insulin (I) decreased 31% in LP rats (69 182.00 ± 6 428.88 pmol/l/min)
compared to NP animals (100 446.36 ± 2 974.00 pmol/l/min), p<0.05. The
insulinogenic index (I/G) was significantly lower in LP (143.77 ± 20.49) than in NP
rats (320.86 ± 14.10), p<0.001.
In NP rats, plasma insulin concentration was affected by the exogenous Ach and
its muscarinic antagonist atropine. Ach induced a 18% increase in I, while Atr reduced
it by 22%, when compared with the I values obtained during the ivGTT without any
drug addition (p<0.05), as shown in “Figure 2”. In LP rats, the intraperitoneal injection
of Ach significantly enhanced by 63% the I; however, Atr failed to alter I (Figure 2,
open bars). The adrenergic antagonist yohimbine increased the I by 32% and 65% in
NP and LP rats, respectively. The α
2A
- agonist oxymetazoline reduced the I by 25% in
NP rats but didn’t alter it in LP animals.
Islet insulin content was not different in both rat groups: 43.33 ± 3.95 ng/islet in
NP rats and 37.11 ± 2.70 ng/islet in LP ones (p = 0.2). The insulinotropic effect of
glucose on isolated islets of low protein rats was also evaluated, as shown in “Figure 3”.
Overall, LP islets secreted less insulin than NP islets; however, the calculated half-
maximal insulin release and the hill slope were not altered by the low protein treatment.
The effect of the cholinergic analogue carbachol (Cch) on islets was also measured.
“Figure 4” shows the results obtained with 8.3 mM of glucose. The insulinotropic effect
of Cch on glucose-stimulated insulin secretion in islets from NP rats was 6 fold when
compared to release stimulated by glucose alone (p<0.001); however, islets from LP
rats showed only a 3 fold increase (p<0.001). In results not shown, similar results were
obtained from concentrations of glucose (2.8 - 20.0 mM) in response to Cch 1.0 mM,
and the maximal response to Cch was obtained by 1.0 mM. Muscarinic antagonist
atropine at 10µM was able to inhibit the Cch insulinotropic effect by 80% and 50% in
islets from NP and LP rats, respectively. Atropine alone did not cause any change in
glucose-induced insulin release in islets from both animal groups (results not shown).
Stimulation of islets’ adrenoceptors is shown in “Figure 5”. Epinephrine, as expected,
inhibited the glucose-induced insulin release through the α-adrenergic receptor;
however, the magnitude of the response was the same (50%) in islets from both animal
groups. Isoproterenol is a non-specific β-adrenergic agonist and it potentiated (30%) the
insulin release stimulated by glucose in NP islets, whereas the insulinotropic effect was
greater (80%) in LP islets, p<0.05. The association of β-adrenergic stimulation with
isoproterenol and α-adrenergic blockage resulted, as expected, in the potentiation of the
isoproterenol effect; it was 30% and 23% in NP and LP islets respectively.
DISCUSSION
It has been shown that low protein treatment during gestation and lactation
caused increases of neural density in the hypothalamus of weaned rats, mostly in the
medial area, but low immunopositivity in neurons that express neuropeptide Y and
galanine has also been
detected (Plagemann et al., 2000b). These neurotransmitters have
been known as potent stimulators of food intake (Clark et al., 1984). Furthermore,
studies reported that electric stimulation of the hypothalamic ventromedial area (VMH)
induced hypophagia (Inoue et al., 1991); however, its lesion causes an increase in food
intake (Balkan et al., 1991). It has been observed that VMH is the origin of sympathetic
fibers. Animals with hypoinsulinemia that undergo protein restriction early in life,
including the lactation phase, show persistent underweight and reduction in food
ingestion, even after diet recovery (Barbosa et al., 1999; Marcelino et al., 2004).
Perinatal protein undernutrition induces changes in metabolism. The metabolic changes
are retained in adult life. It has been shown that metabolic imprinting caused by protein
restriction during perinatal life is associated with several degenerative diseases, diabetes
among them (Waterland & Garza, 1999). Our results indicate that those 81 day rats have
low body weight when compared to animals whose mothers were submitted to regular
protein diet during lactation. The LP rats showed low fat accumulation, without
changing the Lee index. In results not shown, LP animals have the same plasma protein
concentration as NP rats. This indicates that those adult rats, after the diet recovery, are
not malnourished; however, perinatal protein restriction caused enough permanent
metabolic changes to reduce the body weight, the length and fat tissue accumulation.
Metabolic imprinting caused by perinatal protein restriction is followed by alterations in
the neural and endocrine system (Young, 2002). We have previously found fasting
hypoinsulinemia and normoglycemia in LP rats (Moura et al., 1996). Increasing plasma
glucose concentration, during ivGTT, showed that LP rats have glucose intolerance and
low plasma insulin concentration. The insulinogenic index (I/G) of LP rats is low
when compared to NP rats. Those results suggest that this metabolic imprinting
impaired insulin secretion, but increased the tissue glucose uptake induced by insulin
action. We observed high tissue sensitivity to insulin in adult rats that were protein
malnourished during the lactation phase (Moura et al., 1997), even when protein
restriction manipulation was developed during gestation and lactation, tissues showed
increased sensitivity to insulin (Latorraca et al., 1998; Ozanne et al., 1996).
Many different factors contribute to regulate insulin secretion during oscillations
of plasma glucose concentration, among them the neural activity. The ANS, through
parasympathetic branch activity, enhances insulin secretion stimulated by glucose;
whereas sympathetic activity mostly inhibits it (Gilon & Henquin, 2001). It has been
shown that 60 day old rats who were protein malnourished for 4 weeks, presented low
parasympathetic activity and high sympathetic activity, as observed by electric activity
indicated by firing rates recorded from the thoracic branch of the vagus nerve and from
superior cervical ganglion, respectively (Leon-Quinto et al., 1998). This imbalance of
ANS was suggested as a cause of altered plasma insulin levels changes during ivGTT of
protein malnourished adult rats. Exogenous Ach treatment before glucose load induced
high blood insulin concentration in NP animals during ivGTT; although LP rats had
more significant blood insulin concentration increment. The increase of plasma insulin
provoked by exogenous Ach might be attributed to the occupation of the beta cells
muscarinic receptors, leading to the potentiation of glucose-induced insulin secretion
(Gilon & Henquin, 2001). Results recorded from protein malnourished adult rats during
ivGTT with Ach pre treatment showed less effect on plasma insulin than on the control
rats (Leon-Quinto et al., 1998). Muscarinic antagonist atropine treatment caused, as
expected, a decrease in insulinemia as a result of the blocking of muscarinic receptors;
however, LP rats showed no significant changes in plasma insulin. The set of results
obtained by pretreatment with muscarinic agonist and antagonist suggest that LP rats
have low activation of pancreatic cholinergic ends. Alternatively, it could be suggest
that pancreatic beta cells from LP rats have high numbers of cholinergic receptors
and/or high receptor sensitivity; indeed, atropine treatment was not able to significantly
reduce the plasma insulin increment stimulated by the glucose load. This result can also
indicate that the dose of muscarinic antagonist used in this study, was not enough to
bind significant numbers of muscarinic receptors. One very important component of
insulin secretion control during feeding behavior is the increased vagal activity, which
releases Ach on the parasympathetic ends of pancreatic beta cells; while sympathetic
activity, also associated to feeding behavior, mostly inhibited glucose-induced insulin
secretion, by catecholamine action upon α
2A
adrenergic receptors of pancreatic beta
cells (Peterhoff et al., 2003). Yohimbine, an α
2
-adrenergic antagonist, caused a
significant increase in plasma insulin in both animal groups, though the effect was
stronger on LP rats. This result indicates that α-adrenergic receptors of beta cells from
LP animals are more responsive than those of NP rats. Alternatively, the result suggests
that beta cells of LP rats have high number and/or high sensitivity of α-adrenergic
receptors. Alternatively, a low adrenergic activity could also leads parasympathetic
activity to stimulate the insulin secretion by Ach release in the pancreatic
parasympathetic terminals, increasing the plasma insulin more efficiently as shown by
LP rats. Oxymetazoline is a potent α
2A-
adrenergic agonist and causes inhibition of
insulin secretion (Ullrich & Wollheim, 1985). This adrenergic agonist, as expected,
reduced plasma insulin during ivGTT in NP rats; however, no changes were observed in
LP rats. This former observation could support the hypothesis that LP animals have
high number of α-adrenergic receptor, because the dose of oxymetazoline was not
enough to activate a great number of the receptors; however, it did with NP animals.
The results obtained with α-adrenergic agonists and antagonists can suggest that
sympathetic activity, as well as parasympathetic activity, is also impaired in LP rats.
In results not shown, G changes caused by pretreatment of cholinergic and
adrenergic agents showed an inverse relationship with I variations also caused by
drugs during the ivGTT.
Pancreatic islets isolated from LP rats showed a weak insulin secretory response
when stimulated by glucose, although their sensitivity was not changed. It is known that
insulin secretion is regulated by cellular metabolism stimulation, which is able to
provoke ionic changes in the membranes with depolarization and intracellular calcium
increase, through calcium uptake and intracellular calcium mobilization. The increased
intracellular calcium allowed beta cells to secrete insulin (Malaisse et al., 1978). It has
been shown that protein restriction during gestation and post natal life induces reduction
in cell metabolic activity as well as in calcium uptake when their islets are exposed to
high glucose concentration (Latorraca et al., 1999). Recently, it has been shown that rats
who were protein restricted during the 12 first days of lactation were not impaired on
their β- cell glucose stimulated metabolism (Barbosa et al., 2002), although the authors
observed that pancreatic islets from protein malnourished rats showed an impairment in
phosphoinositides metabolism when stimulated by glucose and carbachol. It could be
argued that the beta cells of perinatal protein restricted rats are not able to produce
enough insulin to the secretory demands. In fact, low protein offer during gestation and
lactation decreases total insulin content in pancreatic islets (Bertin et al., 1999; Petrik et
al., 1999); however it was showed that protein restriction only during lactation did not
significantly decrease insulin content of pancreatic islets (Barbosa et al., 2002). Indeed,
in our study islet insulin content of NP and LP was not different. Although for sure that
the metabolic imprinting promoted by perinatal protein restriction maneuver induces
changes in the glucose-induced insulin secretion in beta cells, it is very attractive to
consider that pancreatic islets from LP rats could have changed their secretory responses
to neurotransmitters. It has been shown that obese rodents induced by VMH lesion or by
neonatal monosodium glutamate treatment, present high parasympathetic activity
(Balbo et al., 2002; Campfield et al., 1983; Kiba, 2004; Lucinei Balbo et al., 2000). The
pancreatic islets isolated from the obese animals showed high and low insulin secretion
response to glucose and to cholinergic agonists, respectively; however, bilateral
subdiafragmatic vagotomy restored the secretory responses (Balbo et al., 2002;
Campfield et al., 1983; Kiba, 2004). The authors have concluded that the fasting
hyperinsulinemia observed in those obese animals is attributed to beta-cells high
responsivity to glucose; as well as the low secretory response to cholinergic agonists
was attributed to high parasympathetic activity, which could be caused by decrease of
the number and/or affinity of muscarinic receptors, or alternative changes in cellular
transduction.
Our results showed that the cholinergic secretory response was weak in islets
isolated from rats who were submitted to protein restriction during lactation; however,
we cannot conclude that there is a mechanism of down regulation of muscarinic
receptors. We have not any evidence of increase or decrease in parasympathetic activity
in LP rats.
Stimulation of α-adrenergic receptors of LP islets showed the same inhibition of
glucose-induced insulin secretion than NP islets, indicating that the reception of
adrenergic signals is intact in LP islets; however, β-adrenoceptor stimulation is more
responsive in LP than in NP islets. Whereas, the overall stimulation of adrenoceptors
inhibits the glucose-induced insulin release, as has been shown in well protein
nourished rats (Gilon & Henquin, 2001).
Recently, it was showed that mice with gene knockout that expressed muscarinic
receptor of M3R subfamily are underweight, hypophagic and hypoinsulinemic (Yamada
et al., 2001), as did adult rats who was protein malnourished during lactation. It has
been shown that the insulinotropic effect of cholinergic agonist in pancreatic islets is
majority displaying throughout M3R muscarinic receptors (Boschero et al., 1995;
Miguel et al., 2002). Pancreatic islets from M3R mice (-/-) showed less secretory
response to cholinergic agonist (Duttaroy et al., 2004). Whereas, we have any evidence,
it is possible that metabolic programming induced by early protein malnutrition, causes
reduction of M3R in pancreatic beta cells.
Taken together, our results indicate that lactation protein restriction leads to
underweight, glucose intolerance and hypoinsulinemia in adult rats. This is followed by
alterations on the control of insulin secretion induced by oscillations of glucose
concentration and by modulation of the autonomic nervous system.
Lactation is a very important phase to organism development, including Central
Nervous System. It has been observed that interventions that alter the childhood
nutrition, including lactation phase, induce changes on development of the infantile
(Sawaya et al., 2003) and this condition is a risk to develop chronic degenerative
diseases, such as obesity, hypertension and type 2 diabetes, mostly in developing
countries with nutritional transition (Fjeld et al., 1989; Popkin et al., 1996). Recently, it
was indicated that disturbance on insulin secretion control of stunted children, who were
early malnourished, is related to early protein restriction experimental model presented
in the current work (Martins & Sawaya, 2006). Epidemiologic studies combine with
experimental findings suggests that undernutrition in childhood has enormous impact on
health of this population. Further research is urgently demanded to reduce the morbidity
and mortality due to diseases originated by perinatal undernutrition.
Acknowlegdgments:
We gratefully acknowledge financial support from CNPq (Brazilian Council for
Scietific and Tecnological Development).
LITERATURE CITED
Balbo, S. L., Bonfleur, M. L., Carneiro, E. M., Amaral, M. E., Filiputti, E.,Mathias, P.
C. 2002. Parasympathetic activity changes insulin response to glucose and
neurotransmitters. Diabetes Metab 28: 3S13-17; discussion 13S108-112.
Balkan, B., Steffens, A. B., Bruggink, J. E.,Strubbe, J. H. 1991. Hyperinsulinemia and
glucose tolerance in obese rats with lesions of the ventromedial hypothalamus:
dependence on food intake and route of administration. Metabolism 40: 1092-1100.
Barbosa, F. B., Medina, A. R., Balbo, S. L.,de Freitas Mathias, P. C. 1999. Low protein
diets administered to lactating rats affect in a time-dependent manner the development
of young. Res Commun Mol Pathol Pharmacol 106: 63-76.
Barbosa, F. B., Capito, K., Kofod, H.,Thams, P. 2002. Pancreatic islet insulin secretion
and metabolism in adult rats malnourished during neonatal life. Br J Nutr 87: 147-155.
Bertin, E., Gangnerau, M. N., Bailbe, D.,Portha, B. 1999. Glucose metabolism and beta-
cell mass in adult offspring of rats protein and/or energy restricted during the last week
of pregnancy. Am J Physiol 277: E11-17.
Bhargava, S. K., Sachdev, H. S., Fall, C. H. & other authors 2004. Relation of serial
changes in childhood body-mass index to impaired glucose tolerance in young
adulthood. N Engl J Med 350: 865-875.
Boschero, A. C., Szpak-Glasman, M., Carneiro, E. M., Bordin, S., Paul, I., Rojas,
E.,Atwater, I. 1995. Oxotremorine-m potentiation of glucose-induced insulin release
from rat islets involves M3 muscarinic receptors. Am J Physiol 268: E336-342.
Campfield, L. A., Smith, F. J.,LeMagnen, J. 1983. Altered endocrine pancreatic
function following vagotomy: possible behavioral and metabolic bases for assessing
completeness of vagotomy. J Auton Nerv Syst 9: 283-300.
Clark, J. T., Kalra, P. S., Crowley, W. R.,Kalra, S. P. 1984. Neuropeptide Y and human
pancreatic polypeptide stimulate feeding behavior in rats. Endocrinology 115: 427-429.
Dollet, J. M., Beck, B.,Max, J. P. 1988. Effect of the duration of malnutrition and of
nutritional rehabilitation on blood glucose homeostasis and pancreatic hormones in rats.
Br J Nutr 60: 69-76.
Duttaroy, A., Zimliki, C. L., Gautam, D., Cui, Y., Mears, D.,Wess, J. 2004. Muscarinic
stimulation of pancreatic insulin and glucagon release is abolished in m3 muscarinic
acetylcholine receptor-deficient mice. Diabetes 53: 1714-1720.
Fjeld, C. R., Schoeller, D. A.,Brown, K. H. 1989. Body composition of children
recovering from severe protein-energy malnutrition at two rates of catch-up growth. Am
J Clin Nutr 50: 1266-1275.
Garofano, A., Czernichow, P.,Breant, B. 1999. Effect of ageing on beta-cell mass and
function in rats malnourished during the perinatal period. Diabetologia 42: 711-718.
Gilon, P.,Henquin, J. C. 2001. Mechanisms and physiological significance of the
cholinergic control of pancreatic beta-cell function. Endocr Rev 22: 565-604.
Inoue, S., Nagase, H., Satoh, S., Saito, M., Egawa, M., Tanaka, K.,Takamura, Y. 1991.
Role of the efferent and afferent vagus nerve in the development of ventromedial
hypothalamic (VMH) obesity. Brain Res Bull 27: 511-515.
Kaiser, N., Corcos, A. P., Sarel, I.,Cerasi, E. 1991. Monolayer culture of adult rat
pancreatic islets on extracellular matrix: modulation of B-cell function by chronic
exposure to high glucose. Endocrinology 129: 2067-2076.
Kiba, T. 2004. Relationships between the autonomic nervous system and the pancreas
including regulation of regeneration and apoptosis: recent developments. Pancreas 29:
e51-58.
Langley-Evans, S. C. 1997. Hypertension induced by foetal exposure to a maternal low-
protein diet, in the rat, is prevented by pharmacological blockade of maternal
glucocorticoid synthesis. J Hypertens 15: 537-544.
Latorraca, M. Q., Carneiro, E. M., Boschero, A. C.,Mello, M. A. 1998. Protein
deficiency during pregnancy and lactation impairs glucose-induced insulin secretion but
increases the sensitivity to insulin in weaned rats. Br J Nutr 80: 291-297.
Latorraca, M. Q., Mello, E. M.,Boschero, A. C. 1999. Reduced insulin secretion in
response to nutrients in islets from malnourished young rats is associated with a
diminished calcium uptake. J Nutr Biochem 10: 37-43.
Leon-Quinto, T., Magnan, C.,Portha, B. 1998. Altered activity of the autonomous
nervous system as a determinant of the impaired beta-cell secretory response after
protein-energy restriction in the rat. Endocrinology 139: 3382-3389.
Lima, J. G., Oliveira, L. M., Lachat, J. J., Dal-Bo, C. M.,Almeida, S. S. 1993.
Comparison of the effects of lab chow and casein diets based on body and brain
development of rats. Braz J Med Biol Res 26: 1069-1076.
Lopes Da Costa, C., Sampaio De Freitas, M.,Sanchez Moura, A. 2004. Insulin secretion
and GLUT-2 expression in undernourished neonate rats. J Nutr Biochem 15: 236-241.
Lucinei Balbo, S., Gravena, C., Bonfleur, M. L.,de Freitas Mathias, P. C. 2000. Insulin
secretion and acetylcholinesterase activity in monosodium l-glutamate-induced obese
mice. Horm Res 54: 186-191.
Malaisse, W. J., Herchuelz, A., Devis, G. & other authors 1978. Regulation of calcium
fluxes and their regulatory roles in pancreatic islets. Ann N Y Acad Sci 307: 562-582.
Marcelino, A. A., Moura, A. S., Barradas, P. C.,Tenorio, F. 2004. Hypothalamic nuclei
nitric oxide synthase expression in rats malnourished during early lactation period. Nutr
Neurosci 7: 177-184.
Martins, P. A.,Sawaya, A. L. 2006. Evidence for impaired insulin production and higher
sensitivity in stunted children living in slums. Br J Nutr 95: 996-1001.
Meglasson, M. D., Nelson, J., Nelson, D.,Erecinska, M. 1989. Bioenergetic response of
pancreatic islets to stimulation by fuel molecules. Metabolism 38: 1188-1195.
Miguel, J. C., Abdel-Wahab, Y. H., Mathias, P. C.,Flatt, P. R. 2002. Muscarinic
receptor subtypes mediate stimulatory and paradoxical inhibitory effects on an insulin-
secreting beta cell line. Biochim Biophys Acta 1569: 45-50.
Moura, A. S., Carpinelli, A. R., Barbosa, F. B., Gravena, C.,Mathias, P. C. 1996.
Undernutrition during early lactation as an alternative model to study the onset of
diabetes mellitus type II. Res Commun Mol Pathol Pharmacol 92: 73-84.
Moura, A. S., Caldeira Filho, J. S., de Freitas Mathias, P. C.,de Sa, C. C. 1997. Insulin
secretion impairment and insulin sensitivity improvement in adult rats undernourished
during early lactation. Res Commun Mol Pathol Pharmacol 96: 179-192.
Ozanne, S. E., Wang, C. L., Coleman, N.,Smith, G. D. 1996. Altered muscle insulin
sensitivity in the male offspring of protein-malnourished rats. Am J Physiol 271:
E1128-1134.
Peterhoff, M., Sieg, A., Brede, M., Chao, C. M., Hein, L.,Ullrich, S. 2003. Inhibition of
insulin secretion via distinct signaling pathways in alpha2-adrenoceptor knockout mice.
Eur J Endocrinol 149: 343-350.
Petrik, J., Reusens, B., Arany, E., Remacle, C., Coelho, C., Hoet, J. J.,Hill, D. J. 1999.
A low protein diet alters the balance of islet cell replication and apoptosis in the fetal
and neonatal rat and is associated with a reduced pancreatic expression of insulin-like
growth factor-II. Endocrinology 140: 4861-4873.
Plagemann, A., Harder, T., Rake, A., Melchior, K., Rohde, W.,Dorner, G. 2000a.
Hypothalamic nuclei are malformed in weanling offspring of low protein malnourished
rat dams. J Nutr 130: 2582-2589.
Plagemann, A., Waas, T., Harder, T., Rittel, F., Ziska, T.,Rohde, W. 2000b.
Hypothalamic neuropeptide Y levels in weaning offspring of low-protein malnourished
mother rats. Neuropeptides 34: 1-6.
Popkin, B. M., Richards, M. K.,Montiero, C. A. 1996. Stunting is associated with
overweight in children of four nations that are undergoing the nutrition transition. J Nutr
126: 3009-3016.
Rasschaert, J., Reusens, B., Dahri, S., Sener, A., Remacle, C., Hoet, J. J.,Malaisse, W. J.
1995. Impaired activity of rat pancreatic islet mitochondrial glycerophosphate
dehydrogenase in protein malnutrition. Endocrinology 136: 2631-2634.
Sawaya, A. L., Martins, P., Hoffman, D.,Roberts, S. B. 2003. The link between
childhood undernutrition and risk of chronic diseases in adulthood: a case study of
Brazil. Nutr Rev 61: 168-175.
Steffens, A. B., Strubbe, J. H., Balkan, B.,Scheurink, J. W. 1990. Neuroendocrine
mechanisms involved in regulation of body weight, food intake and metabolism.
Neurosci Biobehav Rev 14: 305-313.
Ullrich, S.,Wollheim, C. B. 1985. Expression of both alpha 1- and alpha 2-
adrenoceptors in an insulin-secreting cell line. Parallel studies of cytosolic free Ca2+
and insulin release. Mol Pharmacol 28: 100-106.
Waterland, R. A.,Garza, C. 1999. Potential mechanisms of metabolic imprinting that
lead to chronic disease. Am J Clin Nutr 69: 179-197.
Waterland, R. A.,Garza, C. 2002. Early postnatal nutrition determines adult pancreatic
glucose-responsive insulin secretion and islet gene expression in rats. J Nutr 132: 357-
364.
Yamada, M., Miyakawa, T., Duttaroy, A. & other authors 2001. Mice lacking the M3
muscarinic acetylcholine receptor are hypophagic and lean. Nature 410: 207-212.
Young, J. B. 2002. Programming of sympathoadrenal function. Trends Endocrinol
Metab 13: 381-385.
NP LP
Body weight (g)
364.36 ± 3.60 278.95 ± 3.16 *
Body Lenght (cm)
23.36 ± 0.08 21.31 ± 0.10 *
Lee Index
300 ± 10.00 300 ± 10.00 NS
Brain weight (g/100g BW)
0.51 ± 0.01 0.55 ± 0.01 *
Epididymal fat (g/100g BW)
0.97 ± 0.04 0.68 ± 0.03 *
Retroperitoneal fat (g/100g BW)
0.81 ± 0.04 0.41 ± 0.03 *
Plasma glucose (mmol/l)
5700 ± 140 6340 ± 300 NS
Plasma insulin (pmol/l) 1480 ± 30 870 ± 30 *
Table I. Effect of hypoproteic diet during part of lactation on 81 days rats. Animals
were sacrificed with 14 h of fasting. The results present the mean±SEM of 28 rats from
each group.
*
p<0,001 (Student’s t test)
0 10 20 30 40 50 60
0
5
10
15
20
25
30
35
40
45
*
*
**
*
NP
LP
*
A
Time (min)
Plasma glucose (mmol/l)
0 10 20 30 40 50 60
0
2000
4000
6000
8000
10000
NP
LP
***
*
***
*
B
Tim e (m in)
Plasma insulim (pmol/l)
Figure 1. Changes in plasma glucose and insulin levels during ivGTT with glucose
load. The upper panel shows the plasma glucose and the low panel shows the plasma
insulin after ivGTT with glucose load 1g/Kg BW. The symbols present the mean of
changes in glycemia and insulinemia during ivGTT, provided by 6 rats from NP and LP
rats. The lines on top of the symbols show the SEM. *p<0.05. **p<0.01, ***p<0.001 in
comparison with the same time (Student’s t test).
0
100
200
NP
LP
*
***
**
*
Ach Atr Yoh
Oxy
bd
ac
ac
bd
bd
bd
ac
ac
ψ
ψψ
I (%)
Figure 2. Changes in plasma insulin levels during ivGTT in rats pretreated with
antagonists and agonists of ANS. Bars represent the mean percentage of insulin
increment (area under curve) obtained on ivGTT with indicated drug. It used 6 different
rats to each drug treatment to NP or LP rats. Dot line represents the total insulin plasma
concentration from area under the curve calculated to all ivGTT (100%), which was
induced by glucose load on NP or LP rats, without any drug pretreatment. The lines on
top of bars indicate the SEM. The letters over the bars refer to differences (p<0.05)
among treatments: (a) acetylcholine, (b) atropine, (c) yohimbine, (d) oxymetazoline to
each group.
*p<0.05;**p<0.01;***p<0.001, comparing with NP rats.
p<0.05;
ΩΩ
p<0.01;
ΩΩΩ
p<0.001, NP comparing with results obtained without any drug.
Ψ
p<0.01;
ΨΨ
p<0.001, LP comparing with results obtained without any drug. Data were
analyzed with ANOVA.
10
-2.75
10
-2.50
10
-2.25
10
-2.00
10
-1.75
10
-1.50
0
1
2
3
4
5
6
7
NP
LP
Glucose [log M]
***
***
***
*
**
*
insulin release (ng/islet/60min)
Figure 3. Dose response curve of glucose-induced insulin release in isolated
pancreatic islets . Symbols represent the mean of 20 different incubations batches of 4
islets incubated during 60 min to each glucose concentration of the curve. Lines on the
symbols represent the respective SEM. (*) p<0.05, (**)p<0.01, (***)p<0.001 when
compared with LP group (Student's t test).
0
2
4
6
8
10
NP LP
Glucose + + + + + +
Cch - + + - + +
Atr - - + - - +
bc
ac
ab
bc
ac
ab
insulin release(ng/islet/60min)
Figure 4. Effect of cholinergic activation on glucose-induced insulin secretion in
pancreatic islets. The bars represent the mean of insulin released from 10 diffrent
batches of islets isolated from NP or LP rats, incubated for 60 min, as indicated over
the bars. Islets were stimulated by 8.3 mM of glucose in the presence or in the absence
of carbachol (1mM) and/or atropine (10 µM). The lines on the top of bars represent
SEM. Letters over the bars represent significant differences (p<0,05) among results
from intra group by ANOVA. (a) glucose only, (b) glucose+Cch, (c) glucose+Cch+ Atr.
0
1
2
3
4
5
6
7
8
9
Glucose + + + + + + + +
Epi - + - - - + - -
Iso - - + + - - + +
Yoh - - - + - - - +
NP LP
bcd
acd
abd
abc
bcd
acd
abd
abc
insuilin release (ng/islet/60min)
Figure 5. Effect of adrenoceptors activation in pancreatic islets stimulated by
glucose 16.7 mM. The bars represent the mean of insulin released obtained from 10
batches of islets incubation with glucose in the presence or in the absence of adrenergic-
agonists and/or antagonist during 60 min. Line on the top of bars represent SEM.
Letters over the bars represent significant differences (p<0,05) among results from intra
group by ANOVA. (a) glucose 16,7mM only, (b) glucose+epinephrine 100nM, (c)
glucose+isoproterenol 10µM and (d) glucose+isoproterenol+yohimbine 10µM.
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