Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Diabetes ; 69(5): 902-914, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31896553

RESUMO

Studies implicating sodium-glucose cotransporter 2 (SGLT2) inhibitors in glucagon secretion by pancreatic α-cells reported controversial results. We hypothesized that interindividual heterogeneity in SGLT2 expression and regulation may affect glucagon secretion by human α-cells in response to SGLT2 inhibitors. An unbiased RNA-sequencing analysis of 207 donors revealed an unprecedented level of heterogeneity of SLC5A2 expression. To determine heterogeneity of SGLT2 expression at the protein level, the anti-SGLT2 antibody was first rigorously evaluated for specificity, followed by Western blot and immunofluorescence analysis on islets from 10 and 12 donors, respectively. The results revealed a high interdonor variability of SGLT2 protein expression. Quantitative analysis of 665 human islets showed a significant SGLT2 protein colocalization with glucagon but not with insulin or somatostatin. Moreover, glucagon secretion by islets from 31 donors at low glucose (1 mmol/L) was also heterogeneous and correlated with dapagliflozin-induced glucagon secretion at 6 mmol/L glucose. Intriguingly, islets from three donors did not secrete glucagon in response to either 1 mmol/L glucose or dapagliflozin, indicating a functional impairment of the islets of these donors to glucose sensing and SGLT2 inhibition. Collectively, these data suggest that heterogeneous expression of SGLT2 protein and variability in glucagon secretory responses contribute to interindividual differences in response to SGLT2 inhibitors.


Assuntos
Compostos Benzidrílicos/farmacologia , Glucosídeos/farmacologia , Ilhotas Pancreáticas/metabolismo , Transportador 2 de Glucose-Sódio/metabolismo , Anticorpos , Glicemia , Bases de Dados de Ácidos Nucleicos , Glucagon/metabolismo , Glucose/administração & dosagem , Glucose/farmacologia , Células HEK293 , Humanos , RNA Interferente Pequeno , Transportador 2 de Glucose-Sódio/genética , Transportador 2 de Glucose-Sódio/imunologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia
2.
Cell Rep ; 28(6): 1447-1454.e4, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31390560

RESUMO

The newest classes of anti-diabetic agents include sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 receptor (GLP1R) agonists. The SGLT2 inhibitor dapagliflozin reduces glucotoxicity by glycosuria but elevates glucagon secretion. The GLP1R agonist liraglutide inhibits glucagon; therefore, we hypothesize that the cotreatment of dapagliflozin with liraglutide could reduce hyperglucagonemia and hyperglycemia. Here we use five complementary models: human islet cultures, healthy mice, db/db mice, diet-induced obese (DIO) mice, and somatostatin receptor-2 (SSTR2) KO mice. A single administration of liraglutide and dapagliflozin in combination improves glycemia and reduces dapagliflozin-induced glucagon secretion in diabetic mice. Chronic treatment with liraglutide and dapagliflozin produces a sustainable reduction of glycemia compared with each drug alone. Moreover, liraglutide reduces dapagliflozin-induced glucagon secretion by enhancing somatostatin release, as demonstrated by SSTR2 inhibition in human islets and in mice. Collectively, these data provide mechanistic insights into how intra-islet GLP1R activation is critical for the regulation of glucose homeostasis.


Assuntos
Compostos Benzidrílicos/efeitos adversos , Diabetes Mellitus Experimental/tratamento farmacológico , Glucagon/efeitos dos fármacos , Glucosídeos/efeitos adversos , Liraglutida/uso terapêutico , Somatostatina/efeitos dos fármacos , Animais , Humanos , Liraglutida/farmacologia , Masculino , Camundongos
3.
Cell Transplant ; 26(2): 309-317, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-27938471

RESUMO

Human islet transplantation is a viable treatment option for type 1 diabetes mellitus (T1DM). However, pancreatic islet inflammation after transplantation induced by innate immune responses is likely to hinder graft function. This is mediated by incompatibility between islets and the blood interface, known as instant blood-mediated inflammatory reaction (IBMIR). Herein we hypothesized that portal venous administration of islet cells with human recombinant antithrombin (ATryn®), a serine protease inhibitor (serpin), which plays a central role in the physiological regulation of coagulation and exerts indirect anti-inflammatory activities, may offset coagulation abnormalities such as disseminated intravascular coagulation (DIC) and IBMIR. The current prospective, randomized experiment was conducted using an established preclinical pig model. Three groups were constituted for digested pancreatic tissue transplantation (0.15 ml/kg): control, NaCl 0.9% (n = 7); gold standard, heparin (25 UI/kg) (n = 7); and human recombinant ATryn® (500 UI/kg) (n = 7). Blood samples were collected over time (T0 to 24 h), and biochemical, coagulation, and inflammatory parameters were evaluated. In both the control and heparin groups, one animal died after a portal thrombosis, while no deaths occurred in the ATryn®-treated group. As expected, islet transplantation was associated with an increase in plasma IL-6 or TNF-α levels in all three groups. However, DIC was only observed in the control group, an effect that was suppressed after ATryn® administration. ATryn® administration increased antithrombin activity by 800%, which remained at 200% for the remaining period of the study, without any hemorrhagic complications. These studies suggest that coadministration of ATryn® and pancreatic islets via intraportal transplantation may be a valuable therapeutic approach for DIC without risk for islets and subjects.


Assuntos
Antitrombina III/uso terapêutico , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Feminino , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Interleucina-6/metabolismo , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/imunologia , Estimativa de Kaplan-Meier , Estudos Prospectivos , Distribuição Aleatória , Suínos , Fator de Necrose Tumoral alfa/metabolismo
4.
Nat Med ; 21(5): 512-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25894829

RESUMO

Type 2 diabetes (T2D) is characterized by chronic hyperglycemia resulting from a deficiency in insulin signaling, because of insulin resistance and/or defects in insulin secretion; it is also associated with increases in glucagon and endogenous glucose production (EGP). Gliflozins, including dapagliflozin, are a new class of approved oral antidiabetic agents that specifically inhibit sodium-glucose co-transporter 2 (SGLT2) function in the kidney, thus preventing renal glucose reabsorption and increasing glycosuria in diabetic individuals while reducing hyperglycemia. However, gliflozin treatment in subjects with T2D increases both plasma glucagon and EGP by unknown mechanisms. In spite of the rise in EGP, T2D patients treated with gliflozin have lower blood glucose levels than those receiving placebo, possibly because of increased glycosuria; however, the resulting increase in plasma glucagon levels represents a possible concerning side effect, especially in a patient population already affected by hyperglucagonemia. Here we demonstrate that SGLT2 is expressed in glucagon-secreting alpha cells of the pancreatic islets. We further found that expression of SLC5A2 (which encodes SGLT2) was lower and glucagon (GCG) gene expression was higher in islets from T2D individuals and in normal islets exposed to chronic hyperglycemia than in islets from non-diabetics. Moreover, hepatocyte nuclear factor 4-α (HNF4A) is specifically expressed in human alpha cells, in which it controls SLC5A2 expression, and its expression is downregulated by hyperglycemia. In addition, inhibition of either SLC5A2 via siRNA-induced gene silencing or SGLT2 via dapagliflozin treatment in human islets triggered glucagon secretion through KATP channel activation. Finally, we found that dapagliflozin treatment further promotes glucagon secretion and hepatic gluconeogenesis in healthy mice, thereby limiting the decrease of plasma glucose induced by fasting. Collectively, these results identify a heretofore unknown role of SGLT2 and designate dapagliflozin an alpha cell secretagogue.


Assuntos
Compostos Benzidrílicos/química , Regulação da Expressão Gênica , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Glucosídeos/química , Transportador 2 de Glucose-Sódio/metabolismo , Administração Oral , Adulto , Animais , Glicemia/química , Separação Celular , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Inativação Gênica , Glucagon/sangue , Gluconeogênese , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , RNA Interferente Pequeno/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo
5.
J Endocrinol ; 214(2): 225-32, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22582094

RESUMO

Zinc ions are essential for the formation of insulin crystals in pancreatic ß cells, thereby contributing to packaging efficiency of stored insulin. Zinc fluxes are regulated through the SLC30A (zinc transporter, ZNT) family. Here, we investigated the effect of metabolic stress associated with the prediabetic state (zinc depletion, glucotoxicity, and lipotoxicity) on ZNT expression and human pancreatic islet function. Both zinc depletion and lipotoxicity (but not glucotoxicity) downregulated ZNT8 (SLC30A8) expression and altered the glucose-stimulated insulin secretion index (GSIS). ZNT8 overexpression in human islets protected them from the decrease in GSIS induced by tetrakis-(2-pyridylmethyl) ethylenediamine and palmitate but not from cell death. In addition, zinc supplementation decreased palmitate-induced human islet cell death without restoring GSIS. Altogether, we showed that ZNT8 expression responds to variation in zinc and lipid levels in human ß cells, with repercussions on insulin secretion. Prospects for increasing ZNT8 expression and/or activity may prove beneficial in type 2 diabetes in humans.


Assuntos
Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/fisiologia , Ilhotas Pancreáticas/metabolismo , Adulto , Proteínas de Transporte de Cátions/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Células Cultivadas , Quelantes/farmacologia , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Lipídeos/farmacologia , Lipídeos/toxicidade , Ácido Palmítico/farmacologia , Ácido Palmítico/toxicidade , Transfecção , Zinco/farmacologia , Transportador 8 de Zinco
6.
Mol Endocrinol ; 26(3): 399-413, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22301783

RESUMO

The NR4A orphan nuclear receptors Nur77, Nurr1, and Nor1 exert multiple cellular and metabolic functions. These transcriptional regulators are activated in response to extracellular stresses, including lipotoxic fatty acids (FA) and proinflammatory cytokines. The contribution of NR4As to ß-cell pathophysiology is, however, unknown. We have therefore examined the role of NR4As as downstream contributors to FA-induced ß-cell dysfunctions. Human pancreatic islets and insulinoma ß-cells were used to determine transcriptional programs elicited by NR4A, which were compared to those triggered by palmitate treatment. Functional studies evaluated the consequence of an increased NR4A expression on insulin biosynthesis and secretion and cell viability in insulinoma ß-cells. FA and cytokine treatment increased NR4A expression in pancreatic ß-cells, with Nur77 being most highly inducible in murine ß-cells. Nur77, Nurr1, or Nor1 modulated common and distinct clusters of genes involved notably in cation homeostasis and insulin gene transcription. By altering zinc homeostasis, insulin gene transcription, and secretion, Nur77 was found to be a major transcriptional mediator of part of FA-induced ß-cell dysfunctions. The repressive role of Nur77 in insulin gene regulation was tracked down to protein-protein interaction with FoxO1, a pivotal integrator of the insulin gene regulatory network. The present study identifies a member of the NR4A nuclear receptor subclass, Nur77/NR4A1, as a modulator of pancreatic ß-cell biology. Together with its previously documented role in liver and muscle, its role in ß-cells establishes Nur77 as an important integrator of glucose metabolism.


Assuntos
Glucose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/fisiologia , Animais , Linhagem Celular , Cromogranina A/metabolismo , Ácidos Graxos/fisiologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Glucose/metabolismo , Humanos , Insulina/genética , Secreção de Insulina , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Camundongos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Pâncreas/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , Estresse Fisiológico
7.
BMC Biotechnol ; 10: 28, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20353585

RESUMO

BACKGROUND: In view of the importance of beta cells in glucose homeostasis and the profound repercussions of beta cell pathology on human health, the acquisition of tools to study pancreatic islet function is essential for the design of alternative novel therapies for diabetes. One promising approach toward this goal involves the modification of gene expression profile of beta cells. RESULTS: This study describes a new method of gene and siRNA delivery into human pancreatic islets by microporation technology. We demonstrated that mild islet distention with accutase greatly enhanced the transfection efficiency without compromising in vitro function (secretion, apoptosis and viability). As an example, the recently identified gene involved in type 2 diabetes, ZnT8, can be over-expressed or silenced by RNA interference using this technology. Microporation can also be used on rodent islets. CONCLUSIONS: Taken together, our results demonstrate that microporation technology can be used to modify gene expression in whole rodent and human islets without altering their in vitro function and will be key to the elucidation of the factors responsible for proper islet function.


Assuntos
Inativação Gênica , Células Secretoras de Insulina/metabolismo , RNA Interferente Pequeno/genética , Transfecção , Animais , Apoptose , Proteínas de Transporte de Cátions/genética , Sobrevivência Celular , Células Cultivadas , Eletroporação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transportador 8 de Zinco
8.
Transplantation ; 89(9): 1154-60, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20098354

RESUMO

BACKGROUND.: The original Edmonton protocol used fresh islets, but for obvious logistic advantages most transplant centers have implemented pretransplant culture in human albumin. The aim of this study was to improve current pretransplant human islet culture techniques. METHODS.: Clinical-grade purified human islets from a total of 24 donors were directly resuspended after isolation in CMRL 1066-based media at 37 degrees C, and media additions and renewal were tested. At days 1 and 5 of culture, in vitro quality controls included islet viability, insulin content and function, apoptosis, and in vivo islet potency assay in nude mice. RESULTS.: Replacing human albumin with human AB serum improved 1- and 5-day preservation of islet function and viability which was further enhanced with antioxidant Stem Ease, leading to the iCulture medium (enriched CMRL: pyruvate, zinc sulfate, insulin, transferrin, selenium, 2.5% human AB serum and Stem Ease). Major damage occurs in the first day of culture and frequent media renewal (25% vol/hr) in this period further improved viability, apoptosis, islet recovery, and function in vitro and in vivo, compared with only changing medium after overnight culture. CONCLUSIONS.: The described human islet culture technique (iCulture medium+renewal) seems to be the best choice for clinical human islet culture when short (1 day) or long (5 days) periods are used. Media choice and dilution play a major role in the function and survival of human islets in culture.


Assuntos
Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/citologia , Animais , Apoptose , Sangue , Contagem de Células , Técnicas de Cultura de Células/métodos , Técnicas de Cultura de Células/normas , Divisão Celular , Sobrevivência Celular , Meios de Cultura , Glucose/farmacologia , Humanos , Insulina/análise , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Nus , Albumina Sérica/fisiologia
9.
Biochem Biophys Res Commun ; 366(4): 1096-101, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18155663

RESUMO

We explored the in vitro effects of Rosiglitazone (RZG), a PPARgamma agonist, on human pancreatic islet dysfunctions induced by chronic free fatty acid exposure. We demonstrated that RZG beneficial effects on insulin secretion and apoptosis did not imply PDX-1 or insulin gene modulation. It rather involved, through a PPARgamma-dependent mechanism, a reduction of iNOS overexpressed in lipotoxic islets. This reduction likely led to the restoration of ATP level and insulin secretion as well as the decrease in apoptosis. More interestingly, we also demonstrated that RZG beneficial effects involved PPARgamma-independent mechanisms. RZG treatment led to a limitation of oxidative stress exemplified by an increase of GPx and SOD expression. It also increased UCP2 expression that seemed to display antioxidant action in this model. Thus, RZG did not appear to exert a direct action on insulin expression but rather an indirect action on insulin secretion and apoptosis, through PPARgamma-dependent and -independent mechanisms, via regulation of nitrogen and oxygen reactive species injury.


Assuntos
Ácidos Graxos não Esterificados/toxicidade , Ilhotas Pancreáticas/efeitos dos fármacos , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Adulto , Perfilação da Expressão Gênica , Humanos , Ilhotas Pancreáticas/metabolismo , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Ácido Palmítico/farmacologia , Rosiglitazona
10.
Transplantation ; 83(5): 532-8, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17353769

RESUMO

BACKGROUND: The effects of sirolimus on insulin secretion are still debated. Our aim was to investigate the effects of sirolimus, both (1) in vivo in healthy minipigs; and (2) in vitro on human islets. METHODS: (1) Ten minipigs were evaluated during three successive stages: (a) basal; (b) at the end of a 4-week period of treatment with sirolimus; and (c) after a 4-week period of wash-out. We evaluated insulin secretion with the acute insulin response (AIR), and glucose tolerance with the glucose disposal rate (GDR). (2) Insulin content, stimulation index, adenosine triphosphate (ATP), and apoptosis were measured in human islets treated in vitro with sirolimus at therapeutic and supratherapeutic concentrations. RESULTS: (1) Basal and stimulated insulin levels and GDR increased during sirolimus administration and returned to baseline after a wash-out period; (2) regardless of culture duration, sirolimus dose-dependently decreased apoptosis and increased insulin content. Stimulation indexes and ATP were also significantly enhanced but only at therapeutic concentrations. CONCLUSIONS: This study suggests that sirolimus, at plasma-drug concentrations usually targeted in clinical practice, (1) increases basal and stimulated insulin levels in vivo and insulin content in vitro regardless of culture duration; (2) is able to reduce apoptosis. These findings may partly underlie the improved results of islet transplantation.


Assuntos
Glicemia/metabolismo , Teste de Tolerância a Glucose , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Sirolimo/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Células Cultivadas , DNA/metabolismo , Glucose/fisiologia , Humanos , Insulina/sangue , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Modelos Animais , Suínos , Porco Miniatura
11.
Biochem Biophys Res Commun ; 333(4): 1153-9, 2005 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15979049

RESUMO

In vivo lineage tracing experiments in mice have recently cast doubt on the potential islet neogenesis from ductal precursors in adult mammals. We examined, in human obesity, a model for pancreatic endocrine tissue plasticity, the gene and protein expression of PBX-1-a transcription factor expressed in regenerating rat ductules and potentially implicated in the pancreatic development, alone or in association with PDX-1. When comparing gene expression, by quantitative real-time RT-PCR, in pancreatic exocrine tissue from obese non-diabetic subjects with increased islet mass, we found that Pbx-1 and Pdx-1 were up-regulated (5.9+/-1.2 and 2.4+/-0.6 versus non-obese). Immunohistochemistry confirmed PBX-1 over-expression and its cytoplasmic sequestration in ductal cells of obese subjects, associated with pronounced islet neogenesis (cytokeratin 19/chromogranin A double labeling). cDNA microarray analysis also showed up-regulation of other genes implicated in islet regeneration, including betacellulin, laminin, TGFa, NeuroD1, Pax6, substantiating the role of the islet neogenesis pathway in human obesity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Ductos Pancreáticos/metabolismo , Ductos Pancreáticos/patologia , Proteínas Proto-Oncogênicas/metabolismo , Regeneração , Transativadores/metabolismo , Adaptação Fisiológica , Cadáver , Células Cultivadas , Citoplasma/metabolismo , Citoplasma/patologia , Regulação da Expressão Gênica , Humanos , Técnicas In Vitro , Fator de Transcrição 1 de Leucemia de Células Pré-B , Distribuição Tecidual
12.
Gene Expr ; 12(2): 83-98, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15892450

RESUMO

Functional pancreatic beta cell mass is dynamic and although fully differentiated, beta cells are capable of reentering the cell cycle upon appropriate stimuli. Stimulating regeneration-competent cells in situ is clearly the most desirable way to restore damaged tissue. Regeneration by dedifferentiation and transdifferentiation is a potential source of cells exhibiting a more developmentally immature phenotype and a wide differentiation potential. In this context and to gain a better understanding of the transformation induced in human beta cells during forced in vitro expansion, we focused on identifying differences in gene expression along with phenotypical transformation between proliferating and quiescent human beta cells. FACS-purified beta cells from three different human pancreata were cultured during 3-4 months (8-10 subcultures) on HTB-9 cell matrix with hepatocyte growth factor. Gene expression profiling was performed on cells from each subculture on "in-house" pancreas-specific microarrays consisting of 218 genes and concomitant morphological transformations were studied by immunocytochemistry. Immunocytochemical studies indicated a shift from epithelial to neuroepithelial cell phenotype, including progenitor cell features such as protein gene product 9.5 (PGP 9.5), Reg, vimentin, and neurogenin 3 protein expression. The expression of 49 genes was downregulated, including several markers of endocrine differentiation while 76 were induced by cell expansion including several markers of progenitor cells. Their pattern also argues for the transdifferentiation of beta cells into progenitor cells, demonstrating neuroepithelial features and overexpressing both PBX1, a homeodomain protein that can bind as a heterodimer with PDX1 and could switch the nature of its transcriptional activity, and neurogenin 3, a key factor for the generation of endocrine islet cells. Our study of the machinery that regulates human beta cell expansion and dedifferentiation may help elucidate some of the critical genes that control the formation of adult pancreatic progenitor cells and hence design targets to modify their expression in view of the production of insulin-secreting cells.


Assuntos
Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Expressão Gênica , Ilhotas Pancreáticas/metabolismo , Células-Tronco/metabolismo , Adulto , Diferenciação Celular , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Imunoensaio , Ilhotas Pancreáticas/citologia , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Biochem J ; 383(Pt. 3): 573-80, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15233628

RESUMO

HNF4alpha (hepatocyte nuclear factor 4alpha) belongs to a complex transcription factor network that is crucial for the function of hepatocytes and pancreatic beta-cells. In these cells, it activates the expression of a very large number of genes, including genes involved in the transport and metabolism of glucose and lipids. Mutations in the HNF4alpha gene correlate with MODY1 (maturity-onset diabetes of the young 1), a form of type II diabetes characterized by an impaired glucose-induced insulin secretion. The MODY1 G115S (Gly115-->Ser) HNF4alpha mutation is located in the DNA-binding domain of this nuclear receptor. We show here that the G115S mutation failed to affect HNF4alpha-mediated transcription on apolipoprotein promoters in HepG2 cells. Conversely, in pancreatic beta-cell lines, this mutation resulted in strong impairments of HNF4alpha transcriptional activity on the promoters of LPK (liver pyruvate kinase) and HNF1alpha, with this transcription factor playing a key role in endocrine pancreas. We show as well that the G115S mutation creates a PKA (protein kinase A) phosphorylation site, and that PKA-mediated phosphorylation results in a decreased transcriptional activity of the mutant. Moreover, the G115E (Gly115-->Glu) mutation mimicking phosphorylation reduced HNF4alpha DNA-binding and transcriptional activities. Our results may account for the 100% penetrance of diabetes in human carriers of this mutation. In addition, they suggest that introduction of a phosphorylation site in the DNA-binding domain may represent a new mechanism by which a MODY1 mutation leads to loss of HNF4alpha function.


Assuntos
Idade de Início , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/genética , Diabetes Mellitus/genética , Glicina/genética , Mutação de Sentido Incorreto/genética , Fosfoproteínas/genética , Serina/genética , Fatores de Transcrição/genética , Substituição de Aminoácidos/genética , Sítios de Ligação/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proteína Quinase Tipo II Dependente de AMP Cíclico , DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/fisiologia , Células HeLa/química , Células HeLa/metabolismo , Células HeLa/patologia , Fator 4 Nuclear de Hepatócito , Histidina/biossíntese , Histidina/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Fosfoproteínas/biossíntese , Fosfoproteínas/fisiologia , Fosforilação , Serina/metabolismo , Especificidade por Substrato , Fatores de Transcrição/biossíntese , Fatores de Transcrição/fisiologia
14.
Endocrinology ; 143(12): 4809-19, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12446608

RESUMO

Transplantation of islets of Langerhans is a potential cure for type 1 diabetes, but its success is hampered by destruction of the islets. The data presented herein suggest that the active metabolite of vitamin D3 [1,25-(OH)2D3] may promote islet cell survival by modulating the effects of inflammatory cytokines, which contribute to beta-cell demise. We investigated some of the mechanisms triggering the apoptotic machinery in rat insulinoma RINm5F cells and human islets treated with IL-1beta plus interferon-gamma plus TNFalpha and assessed the effects of 1,25-(OH)2D3 in these processes. Mitochondrial transmembrane permeability and apoptotic features, determined by percentage of sub-G1 cells, quantitation of DNA strand breaks, and Hoechst staining, were significantly increased by cytokines and reverted toward control values by 1,25-(OH)2D3 cotreatment. The cytoprotection of cells correlated with the abrogation of cytokine-induced nitric oxide production. The activation of nuclear factor-kappaB plays a key role in the different pathways implicated in nitric oxide generation. We demonstrated for the first time, in both RINm5F cells and human islets, that 1,25-(OH)2D3 was able to induce and maintain high levels of A20, an antiapoptotic protein known to block nuclear factor-kappaB activation. Our study showed a clear efficiency of 1,25-(OH)2D3 on the apoptotic machinery triggered by cytokines in beta-cells and suggests that 1,25-(OH)2D3 could help overcome a major obstacle encountered in the cellular therapy of diabetes, such as nonfunction in the immediate posttransplantation period.


Assuntos
Apoptose/efeitos dos fármacos , Calcitriol/farmacologia , Insulinoma/patologia , Ilhotas Pancreáticas/ultraestrutura , Neoplasias Pancreáticas/patologia , Proteínas/farmacologia , Adulto , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cromatina/efeitos dos fármacos , Cromatina/ultraestrutura , Fragmentação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA , Expressão Gênica/efeitos dos fármacos , Humanos , Quinase I-kappa B , Imuno-Histoquímica , Interferon gama/farmacologia , Interleucina-1/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Mitocôndrias/ultraestrutura , NF-kappa B/metabolismo , Proteínas Nucleares , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/genética , RNA Mensageiro/análise , Ratos , Células Tumorais Cultivadas , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/farmacologia
15.
J Mol Med (Berl) ; 80(7): 423-30, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12110948

RESUMO

Mutations in the HNF4alpha gene have been correlated with maturity-onset diabetes of the young, which is characterized mainly by pancreatic beta-cell dysfunction and is also associated with mild liver abnormalities. HNF4alpha D126Y and D126H mutations were found in a patient with early-onset type 2 diabetes, and the R324H mutation was found in a common type 2 diabetic nephropathic patient. We investigated whether these mutations, which have not yet been functionally characterized, impair HNF4alpha function in three cell models: HEK 293 embryonal kidney cells, HepG2 hepatoma cells, and betaTC3 pancreatic beta-cells. The R324H mutation had no effect on HNF4alpha function with either the HNF1alpha and L-type pyruvate kinase (LPK) promoters, but the D126Y and D126H mutations impaired HNF4alpha transcriptional activities in all tested cell lines. These impairments by D126Y and D126H mutations, which are located in the T box, are not due to a loss of dimerization but to a loss of DNA binding. Interestingly, the strongest functional consequences of these mutations were observed on the HNF1alpha promoter in betaTC3 cells. Given the key role of the transcription factor HNF1alpha in pancreatic beta-cell function, it can be inferred that impairment of HNF4alpha function by these mutations affects metabolic pathways in pancreatic beta-cells and contributes to development of diabetes. Moreover, the HNF4alpha-mediated activation of the apolipoprotein CIII promoter in HepG2 cells was significantly impaired by D126Y and D126H mutations. These results support clinical findings that liver function can also be impaired in diabetic patients having HNF4alpha mutations.


Assuntos
Apolipoproteínas C/genética , Diabetes Mellitus Tipo 2/genética , Ilhotas Pancreáticas/metabolismo , Mutação , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Apolipoproteína C-III , Apolipoproteínas C/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dimerização , Fator 4 Nuclear de Hepatócito , Humanos , Fosfoproteínas/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA