Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci ; 291: 120239, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-34942163

RESUMO

Aim Investigate whether inheritance of improved skeletal muscle mitochondrial function and its association with glycemic control are multigenerational benefits of exercise. MAIN METHODS: Male Swiss mice were subjected to 8 weeks of endurance training and mated with untrained females. KEY FINDINGS: Trained fathers displayed typical endurance training-induced adaptations. Remarkably, offspring from trained fathers also exhibited higher endurance performance, mitochondrial oxygen consumption, glucose tolerance and insulin sensitivity. However, PGC-1α expression was not increased in the offspring. In the offspring, the expression of the co-repressor NCoR1 was reduced, increasing activation of PGC-1α target genes. These effects correlated with higher DNA methylation at the NCoR1 promoter in both, the sperm of trained fathers and in the skeletal muscle of their offspring. SIGNIFICANCE: Higher skeletal muscle mitochondrial function is inherited by epigenetic de-activation of a key PGC-1α co-repressor.


Assuntos
Mitocôndrias/metabolismo , Condicionamento Físico Animal/fisiologia , Esforço Físico/fisiologia , Animais , Metilação de DNA , Epigênese Genética/genética , Feminino , Masculino , Camundongos , Mitocôndrias/fisiologia , Músculo Esquelético/fisiologia , Correpressor 1 de Receptor Nuclear/metabolismo , Consumo de Oxigênio/fisiologia , Herança Paterna/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Condicionamento Físico Animal/métodos , RNA Mensageiro/genética
2.
Diabetes ; 70(9): 2026-2041, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34183374

RESUMO

Most obese and insulin-resistant individuals do not develop diabetes. This is the result of the capacity of ß-cells to adapt and produce enough insulin to cover the needs of the organism. The underlying mechanism of ß-cell adaptation in obesity, however, remains unclear. Previous studies have suggested a role for STAT3 in mediating ß-cell development and human glucose homeostasis, but little is known about STAT3 in ß-cells in obesity. We observed enhanced cytoplasmic expression of STAT3 in severely obese subjects with diabetes. To address the functional role of STAT3 in adult ß-cells, we generated mice with tamoxifen-inducible partial or full deletion of STAT3 in ß-cells and fed them a high-fat diet before analysis. Interestingly, ß-cell heterozygous and homozygous STAT3-deficient mice showed glucose intolerance when fed a high-fat diet. Gene expression analysis with RNA sequencing showed that reduced expression of mitochondrial genes in STAT3 knocked down human EndoC-ß1H cells, confirmed in FACS-purified ß-cells from obese STAT3-deficient mice. Moreover, silencing of STAT3 impaired mitochondria activity in EndoC-ß1H cells and human islets, suggesting a mechanism for STAT3-modulated ß-cell function. Our study postulates STAT3 as a novel regulator of ß-cell function in obesity.


Assuntos
Intolerância à Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Obesidade/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Glicemia/metabolismo , Dieta Hiperlipídica , Genes Mitocondriais , Intolerância à Glucose/genética , Humanos , Insulina/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Obesidade/genética , Fator de Transcrição STAT3/genética
3.
J Innate Immun ; 11(4): 375-390, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30799417

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease characterized by islet inflammation and progressive pancreatic ß cell destruction. The disease is triggered by a combination of genetic and environmental factors, but the mechanisms leading to the triggering of early innate and late adaptive immunity and consequent progressive pancreatic ß cell death remain unclear. The insulin-producing ß cells are active secretory cells and are thus particularly sensitive to endoplasmic reticulum (ER) stress. ER stress plays an important role in the pathologic pathway leading to autoimmunity, islet inflammation, and ß cell death. We show here that group B coxsackievirus (CVB) infection, a putative causative factor for T1D, induces a partial ER stress in rat and human ß cells. The activation of the PERK/ATF4/CHOP branch is blunted while the IRE1α branch leads to increased spliced XBP1 expression and c-Jun N-terminal kinase (JNK) activation. Interestingly, JNK1 activation is essential for CVB amplification in both human and rat ß cells. Furthermore, a chemically induced ER stress preceding viral infection increases viral replication, in a process dependent on IRE1α activation. Our findings show that CVB tailors the unfolded protein response in ß cells to support their replication, preferentially triggering the pro-viral IRE1α/XBP1s/JNK1 pathway while blocking the pro-apoptotic PERK/ATF4/CHOP pathway.


Assuntos
Infecções por Coxsackievirus/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Enterovirus Humano B/fisiologia , Células Secretoras de Insulina/imunologia , Resposta a Proteínas não Dobradas/imunologia , Animais , Linhagem Celular , Infecções por Coxsackievirus/imunologia , Diabetes Mellitus Tipo 1/imunologia , Endorribonucleases/genética , Endorribonucleases/metabolismo , Humanos , Evasão da Resposta Imune , Células Secretoras de Insulina/virologia , MAP Quinase Quinase 4/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Transdução de Sinais , Replicação Viral , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
4.
EBioMedicine ; 36: 367-375, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30269996

RESUMO

BACKGROUND: Antibodies targeting PD-1 and its ligand PDL1 are used in cancer immunotherapy but may lead to autoimmune diseases, including type 1 diabetes (T1D). It remains unclear whether PDL1 is expressed in pancreatic islets of people with T1D and how is it regulated. METHODS: The expression of PDL1, IRF1, insulin and glucagon was evaluated in samples of T1D donors by immunofluorescence. Cytokine-induced PDL1 expression in the human beta cell line, EndoC-ßH1, and in primary human pancreatic islets was determined by real-time RT-PCR, flow cytometry and Western blot. Specific and previously validated small interference RNAs were used to inhibit STAT1, STAT2, IRF1 and JAK1 signaling. Key results were validated using the JAK inhibitor Ruxolitinib. FINDINGS: PDL1 was present in insulin-positive cells from twelve T1D individuals (6 living and 6 deceased donors) but absent from insulin-deficient islets or from the islets of six non-diabetic controls. Interferons-α and -γ, but not interleukin-1ß, induced PDL1 expression in vitro in human islet cells and EndoC-ßH1 cells. Silencing of STAT1 or STAT2 individually did not prevent interferon-α-induced PDL1, while blocking of JAKs - a proposed therapeutic strategy for T1D - or IRF1 prevented PDL1 induction. INTERPRETATION: These findings indicate that PDL1 is expressed in beta cells from people with T1D, possibly to attenuate the autoimmune assault, and that it is induced by both type I and II interferons via IRF1.


Assuntos
Antígeno B7-H1/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Regulação da Expressão Gênica , Fator Regulador 1 de Interferon/metabolismo , Interferon-alfa/metabolismo , Interferon gama/metabolismo , Ilhotas Pancreáticas/metabolismo , Adolescente , Adulto , Biomarcadores , Linhagem Celular , Criança , Pré-Escolar , Humanos , Células Secretoras de Insulina/metabolismo , Pessoa de Meia-Idade , Adulto Jovem
5.
Mol Cell Endocrinol ; 473: 186-193, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29409957

RESUMO

It has been reported that the incretin system, including regulated GLP-1 secretion and locally expressed DPP-4, is present in pancreatic islets. In this study we comprehensively evaluated the expression and role of DPP-4 in islet alpha and beta cells from non-diabetic (ND) and type 2 diabetic (T2D) individuals, including the effects of its inhibition on beta cell function and survival. Isolated islets were prepared from 25 ND and 18 T2D organ donors; studies were also performed with the human insulin-producing EndoC-ßH1 cells. Morphological (including confocal microscopy), ultrastructural (electron microscopy, EM), functional (glucose-stimulated insulin secretion), survival (EM and nuclear dyes) and molecular (RNAseq, qPCR and western blot) studies were performed under several different experimental conditions. DPP-4 co-localized with glucagon and was also expressed in human islet insulin-containing cells. Furthermore, DPP-4 was expressed in EndoC-ßH1 cells. The proportions of DPP-4 positive alpha and beta cells and DPP-4 gene expression were significantly lower in T2D islets. A DPP-4 inhibitor protected ND human beta cells and EndoC-ßH1 cells against cytokine-induced toxicity, which was at least in part independent from GLP1 and associated with reduced NFKB1 expression. Finally, DPP-4 inhibition augmented glucose-stimulated insulin secretion, reduced apoptosis and improved ultrastructure in T2D beta cells. These results demonstrate the presence of DPP-4 in human islet alpha and beta cells, with reduced expression in T2D islets, and show that DPP-4 inhibition has beneficial effects on human ND and T2D beta cells. This suggests that DPP-4, besides playing a role in incretin effects, directly affects beta cell function and survival.


Assuntos
Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Dipeptidil Peptidase 4/metabolismo , Células Secretoras de Insulina/enzimologia , Idoso , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/toxicidade , Citoproteção/efeitos dos fármacos , Inibidores da Dipeptidil Peptidase IV/farmacologia , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/ultraestrutura , Masculino , Pessoa de Meia-Idade
6.
J Cell Physiol ; 233(1): 486-496, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28370189

RESUMO

In the present study, we investigated the relationship between early life protein malnutrition-induced redox imbalance, and reduced glucose-stimulated insulin secretion. After weaning, male Wistar rats were submitted to a normal-protein-diet (17%-protein, NP) or to a low-protein-diet (6%-protein, LP) for 60 days. Pancreatic islets were isolated and hydrogen peroxide (H2 O2 ), oxidized (GSSG) and reduced (GSH) glutathione content, CuZn-superoxide dismutase (SOD1), glutathione peroxidase (GPx1) and catalase (CAT) gene expression, as well as enzymatic antioxidant activities were quantified. Islets that were pre-incubated with H2 O2 and/or N-acetylcysteine, were subsequently incubated with glucose for insulin secretion measurement. Protein malnutrition increased CAT mRNA content by 100%. LP group SOD1 and CAT activities were 50% increased and reduced, respectively. H2 O2 production was more than 50% increased whereas GSH/GSSG ratio was near 60% lower in LP group. Insulin secretion was, in most conditions, approximately 50% lower in LP rat islets. When islets were pre-incubated with H2 O2 (100 µM), and incubated with glucose (33 mM), LP rats showed significant decrease of insulin secretion. This effect was attenuated when LP islets were exposed to N-acetylcysteine.


Assuntos
Glicemia/metabolismo , Dieta com Restrição de Proteínas , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Estresse Oxidativo , Desnutrição Proteico-Calórica/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Animais , Antioxidantes/farmacologia , Catalase/genética , Catalase/metabolismo , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Glutationa/metabolismo , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Estado Nutricional , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Desnutrição Proteico-Calórica/sangue , Desnutrição Proteico-Calórica/genética , Desnutrição Proteico-Calórica/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Fatores de Tempo
7.
FASEB J ; 32(3): 1524-1536, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29133342

RESUMO

Prolonged exercise has positive metabolic effects in obese or diabetic individuals. These effects are usually ascribed to improvements in insulin sensitivity. We evaluated whether exercise also generates circulating signals that protect human and rodent ß cells against endoplasmic reticulum (ER) stress and apoptosis. For this purpose, we obtained serum from humans or mice before and after an 8 wk training period. Exposure of human islets or mouse or rat ß cells to human or rodent sera, respectively, obtained from trained individuals reduced cytokine (IL-1ß+IFN-γ)- or chemical ER stressor-induced ß-cell ER stress and apoptosis, at least in part via activation of the transcription factor STAT3. These findings indicate that exercise training improves human and rodent ß-cell survival under diabetogenic conditions and support lifestyle interventions as a protective approach for both type 1 and 2 diabetes.-Paula, F. M. M., Leite, N. C., Borck, P. C., Freitas-Dias, R., Cnop, M., Chacon-Mikahil, M. P. T., Cavaglieri, C. R., Marchetti, P., Boschero, A. C., Zoppi, C. C., Eizirik, D. L. Exercise training protects human and rodent ß cells against endoplasmic reticulum stress and apoptosis.


Assuntos
Apoptose/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Exercício Físico/fisiologia , Células Secretoras de Insulina/metabolismo , Condicionamento Físico Animal/fisiologia , Animais , Feminino , Humanos , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Ratos , Ratos Wistar
8.
FASEB J ; 29(5): 1805-16, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25609426

RESUMO

Type 1 diabetes (T1D) is provoked by an autoimmune assault against pancreatic ß cells. Exercise training enhances ß-cell mass in T1D. Here, we investigated how exercise signals ß cells in T1D condition. For this, we used several approaches. Wild-type and IL-6 knockout (KO) C57BL/6 mice were exercised. Afterward, islets from control and trained mice were exposed to inflammatory cytokines (IL-1ß plus IFN-γ). Islets from control mice and ß-cell lines (INS-1E and MIN6) were incubated with serum from control or trained mice or medium obtained from 5-aminoimidazole-4 carboxamide1-ß-d-ribofuranoside (AICAR)-treated C2C12 skeletal muscle cells. Subsequently, islets and ß cells were exposed to IL-1ß plus IFN-γ. Proteins were assessed by immunoblotting, apoptosis was determined by DNA-binding dye propidium iodide fluorescence, and NO(•) was estimated by nitrite. Exercise reduced 25, 75, and 50% of the IL-1ß plus IFN-γ-induced iNOS, nitrite, and cleaved caspase-3 content, respectively, in pancreatic islets. Serum from trained mice and medium from AICAR-treated C2C12 cells reduced ß-cell death, induced by IL-1ß plus IFN-γ treatment, in 15 and 38%, respectively. This effect was lost in samples treated with IL-6 inhibitor or with serum from exercised IL-6 KO mice. In conclusion, muscle contraction signals ß-cell survival in T1D through IL-6.


Assuntos
Apoptose , Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/patologia , Interleucina-6/fisiologia , Ilhotas Pancreáticas/patologia , Músculo Esquelético/patologia , Condicionamento Físico Animal , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Glucose/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Óxido Nítrico/metabolismo , RNA Mensageiro/genética , Radioimunoensaio , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
9.
Amino Acids ; 47(4): 745-55, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25575490

RESUMO

Endurance exercise training as well as leucine supplementation modulates glucose homeostasis and protein turnover in mammals. Here, we analyze whether leucine supplementation alters the effects of endurance exercise on these parameters in healthy mice. Mice were distributed into sedentary (C) and exercise (T) groups. The exercise group performed a 12-week swimming protocol. Half of the C and T mice, designated as the CL and TL groups, were supplemented with leucine (1.5 % dissolved in the drinking water) throughout the experiment. As well known, endurance exercise training reduced body weight and the retroperitoneal fat pad, increased soleus mass, increased VO2max, decreased muscle proteolysis, and ameliorated peripheral insulin sensitivity. Leucine supplementation had no effect on any of these parameters and worsened glucose tolerance in both CL and TL mice. In the soleus muscle of the T group, AS-160(Thr-642) (AKT substrate of 160 kDa) and AMPK(Thr-172) (AMP-Activated Protein Kinase) phosphorylation was increased by exercise in both basal and insulin-stimulated conditions, but it was reduced in TL mice with insulin stimulation compared with the T group. Akt phosphorylation was not affected by exercise but was lower in the CL group compared with the other groups. Leucine supplementation increased mTOR phosphorylation at basal conditions, whereas exercise reduced it in the presence of insulin, despite no alterations in protein synthesis. In trained groups, the total FoxO3a protein content and the mRNA for the specific isoforms E2 and E3 ligases were reduced. In conclusion, leucine supplementation did not potentiate the effects of endurance training on protein turnover, and it also reduced its positive effects on glucose homeostasis.


Assuntos
Suplementos Nutricionais/análise , Glucose/metabolismo , Leucina/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Feminino , Homeostase , Humanos , Insulina/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Resistência Física , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Natação , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
10.
PLoS Genet ; 9(5): e1003532, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23737756

RESUMO

Mutations in human Gli-similar (GLIS) 3 protein cause neonatal diabetes. The GLIS3 gene region has also been identified as a susceptibility risk locus for both type 1 and type 2 diabetes. GLIS3 plays a role in the generation of pancreatic beta cells and in insulin gene expression, but there is no information on the role of this gene on beta cell viability and/or susceptibility to immune- and metabolic-induced stress. GLIS3 knockdown (KD) in INS-1E cells, primary FACS-purified rat beta cells, and human islet cells decreased expression of MafA, Ins2, and Glut2 and inhibited glucose oxidation and insulin secretion, confirming the role of this transcription factor for the beta cell differentiated phenotype. GLIS3 KD increased beta cell apoptosis basally and sensitized the cells to death induced by pro-inflammatory cytokines (interleukin 1ß + interferon-γ) or palmitate, agents that may contribute to beta cell loss in respectively type 1 and 2 diabetes. The increased cell death was due to activation of the intrinsic (mitochondrial) pathway of apoptosis, as indicated by cytochrome c release to the cytosol, Bax translocation to the mitochondria and activation of caspases 9 and 3. Analysis of the pathways implicated in beta cell apoptosis following GLIS3 KD indicated modulation of alternative splicing of the pro-apoptotic BH3-only protein Bim, favouring expression of the pro-death variant BimS via inhibition of the splicing factor SRp55. KD of Bim abrogated the pro-apoptotic effect of GLIS3 loss of function alone or in combination with cytokines or palmitate. The present data suggest that altered expression of the candidate gene GLIS3 may contribute to both type 1 and 2 type diabetes by favouring beta cell apoptosis. This is mediated by alternative splicing of the pro-apoptotic protein Bim and exacerbated formation of the most pro-apoptotic variant BimS.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , Idoso , Processamento Alternativo/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Proteínas de Ligação a DNA , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 2/etiologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Pessoa de Meia-Idade , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Proteínas Repressoras , Transativadores
11.
Mol Cell Endocrinol ; 374(1-2): 56-64, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23623867

RESUMO

Peroxiredoxins are a family of six antioxidant enzymes (PRDX1-6), and may be an alternative system for the pancreatic beta cells to cope with oxidative stress. This study investigated whether the main diabetogenic pro-inflammatory cytokines or the anti-inflammatory cytokine IL-4 modulate PRDXs levels and putative intracellular pathways important for this process in the insulin-producing RINm5F cells. RINm5F cells expressed significant amounts of PRDX1, PRDX3 and PRDX6 enzymes. Only PRDX6 was modulated by cytokines, showing both mRNA and protein down-regulation following incubation of RINm5F cells with TNF-alpha and IFN-gamma but not with IL-1beta. Separately IFN-gamma or TNF-alpha decreased PRDX6 protein but not mRNA levels. The blockage of the JNK signalling and of the calpains and proteasome proteolysis systems restored PRDX6 protein levels. IL-4 alone did not modulate PRDXs levels. However, pre/co-incubation with IL-4 substantially prevented the decrease in PRDX6 induced by pro-inflammatory cytokines. Knockdown of PRDX6 increased susceptibility of RINm5F cells to the deleterious effects of pro-inflammatory cytokines and to oxidative stress. These results show that, from the PRDXs significantly expressed in RINm5F cells, only PRDX6 is modulated by the diabetogenic cytokines IFN-gamma and TNF-alpha. This PRDX6 down-regulation depends on the calpain and proteasome systems and JNK signalling. PRDX6 is an important enzyme for protection against oxidative stress and the interaction between pro- and anti-inflammatory cytokines might be important to determine the antioxidant capacity of the cells.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Interferon gama/farmacologia , Peroxirredoxina VI/genética , Fator de Necrose Tumoral alfa/farmacologia , Animais , Calpaína/antagonistas & inibidores , Calpaína/genética , Calpaína/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1beta/farmacologia , Interleucina-4/farmacologia , MAP Quinase Quinase 4/antagonistas & inibidores , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Estresse Oxidativo , Peroxirredoxina VI/antagonistas & inibidores , Peroxirredoxina VI/metabolismo , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais
12.
J Appl Physiol (1985) ; 112(5): 711-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22174407

RESUMO

Endurance training has been shown to increase pancreatic ß-cell function and mass. However, whether exercise modulates ß-cell growth and survival pathways signaling is not completely understood. This study investigated the effects of exercise on growth and apoptotic markers levels in rat pancreatic islets. Male Wistar rats were randomly assigned to 8-wk endurance training or to a sedentary control group. After that, pancreatic islets were isolated; gene expression and the total content and phosphorylation of several proteins related to growth and apoptotic pathways as well as the main antioxidant enzymes were determined by real-time polymerase chain reaction and Western blot analysis, respectively. Reactive oxygen species (ROS) production was measured by fluorescence. Endurance training increased the time to reach fatigue by 50%. Endurance training resulted in increased protein phosphorylation content of AKT (75%), AKT substrate (AS160; 100%), mTOR (60%), p70s6k (90%), and ERK1/2 (50%), compared with islets from control group. Catalase protein content was 50% higher, whereas ROS production was 49 and 77% lower in islets from trained rats under basal and stimulating glucose conditions, respectively. Bcl-2 mRNA and protein levels increased by 46 and 100%, respectively. Bax and cleaved caspase-3 protein contents were reduced by 25 and 50% in islets from trained rats, respectively. In conclusion, these results demonstrate that endurance training favors the ß-cell growth and survival by activating AKT and ERK1/2 pathways, enhancing antioxidant capacity, and reducing ROS production and apoptotic proteins content.


Assuntos
Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Resistência Física/fisiologia , Transdução de Sinais/fisiologia , Animais , Antioxidantes/metabolismo , Apoptose/fisiologia , Peso Corporal , Fadiga/genética , Fadiga/metabolismo , Fadiga/fisiopatologia , Expressão Gênica , Glucose/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Oxirredução , Fosforilação , Condicionamento Físico Animal , Resistência Física/genética , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
13.
Biol Res ; 44(3): 251-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22688912

RESUMO

Chronic administration of glucocorticoids induces insulin resistance that is compensated by an increase in p-cell function and mass. Since insulin signaling is involved in the control of p-cell function and mass, we investigated the content of insulin pathway proteins in pancreatic islets. Rats were made insulin resistant by daily administration of dexamethasone (1mg/kg, b.w., i.p.) for 5 consecutive days (DEX), whilst control rats received saline (CTL). Circulating insulin and insulin released from isolated islets were measured by radioimmunoassay whereas the content of proteins was analyzed by Western blotting. DEX rats were hyperinsulinemic and exhibited augmented insulin secretion in response to glucose (P < 0.01). The IRa-subunit, IRS-1, Shc, AKT, p-p70S6K, ERK1/2, p-ERK1/2, and glucocorticoid receptor protein levels were similar between DEX and CTL islets. However, the IRp-subunit, p-IRp-subunit, IRS-2, PI3-K, p-AKT and p70S6K protein contents were increased in DEX islets (P < 0.05). We conclude that IRS-2 may have a major role, among the immediate substrates of the insulin receptor, to link activated receptors to downstream signaling components related to islet function and growth in this insulin-resistant rat model.


Assuntos
Dexametasona/efeitos adversos , Glucocorticoides/efeitos adversos , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Ratos , Ratos Wistar , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Transdução de Sinais
14.
Eur J Pharmacol ; 642(1-3): 37-46, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20541544

RESUMO

The pentadecapeptide comprising the 104-118 amino acid sequence of the ilotropin-derived Reg3-related islet neogenesis-associated protein (INGAP-PP) has been implicated in beta cell neogenesis and enhancement of insulin secretion in pancreatic islets. The aim of this study was to investigate intracellular pathways by which INGAP-PP signals in insulin-producing cells. Treatment with INGAP-PP increased insulin secretion and intracellular calcium levels in MIN6 cells. INGAP-PP exposure activated c-Myc, serum and particularly nuclear factor-kappaB (NF-kappaB) response elements in insulin-producing cells (1.7+/-0.1, 1.8+/-0.1, 2.4+/-0.3 for RINm5F, and 1.3+/-0.1, 1.3+/-0.1 and 1.6+/-0.1 fold for MIN6 cells compared to controls, respectively). There was an increase in the proliferation rate of viable cells (162+/-17% for RINm5F and 155+/-13% for MIN6) that was accompanied by an increase in proliferating cell nuclear antigen (PCNA) protein expression (187+/-19% and 170+/-8% for RINm5F and MIN6 cells respectively) following INGAP-PP treatment. INGAP-PP increased the expression of the muscarinic M(3) receptor subtype (169+/-4% for RINm5F and 222+/-20% for MIN6 cells). Activation of multiple serum response elements by foetal calf serum also increased muscarinic M(3) receptor expression (173+/-9% for RINm5F and 140+/-7% for MIN6 cells). The blockade of NF-kappaB signalling pathway strongly decreased muscarinic M(3) receptor expression in response to both stimuli. In summary, a network of intracellular signals that includes activation of c-Myc signalling pathway and increased PCNA expression might be related to the increased proliferation rate of insulin-producing cells following incubation with INGAP-PP. NF-kappaB signalling plays an essential role in controlling the expression of the muscarinic M(3) receptor.


Assuntos
Acetilcolina/metabolismo , Citocinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Fragmentos de Peptídeos/farmacologia , Receptor Muscarínico M3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sequência de Bases , Carbacol/farmacologia , Domínio Catalítico , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Citocinas/química , Técnicas de Silenciamento de Genes , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Dados de Sequência Molecular , Proteínas Associadas a Pancreatite , Fragmentos de Peptídeos/química , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Interferente Pequeno/genética , Ratos , Reprodutibilidade dos Testes , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Elemento de Resposta Sérica/genética , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/genética , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...