Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Biochem J ; 433(1): 95-105, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20942803

RESUMO

The SLC30A8 gene encodes the zinc transporter ZnT-8, which provides zinc for insulin-hexamer formation. Genome-wide association studies have shown that a polymorphic variant in SLC30A8 is associated with altered susceptibility to Type 2 diabetes and we recently reported that glucose-stimulated insulin secretion is decreased in islets isolated from Slc30a8-knockout mice. The present study examines the molecular basis for the islet-specific expression of Slc30a8. VISTA analyses identified two conserved regions in Slc30a8 introns 2 and 3, designated enhancers A and B respectively. Transfection experiments demonstrated that enhancer B confers elevated fusion gene expression in both ßTC-3 cells and αTC-6 cells. In contrast, enhancer A confers elevated fusion gene expression selectively in ßTC-3 and not αTC-6 cells. These data suggest that enhancer A is an islet ß-cell-specific enhancer and that the mechanisms controlling Slc30a8 expression in α- and ß-cells are overlapping, but distinct. Gel retardation and ChIP (chromatin immunoprecipitation) assays revealed that the islet-enriched transcription factor Pdx-1 binds enhancer A in vitro and in situ respectively. Mutation of two Pdx-1-binding sites in enhancer A markedly reduces fusion gene expression suggesting that this factor contributes to Slc30a8 expression in ß-cells, a conclusion consistent with developmental studies showing that restriction of Pdx-1 to pancreatic islet ß-cells correlates with the induction of Slc30a8 gene expression and ZnT-8 protein expression in vivo.


Assuntos
Proteínas de Transporte de Cátions/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Ilhotas Pancreáticas/química , Transativadores/fisiologia , Transcrição Gênica , Animais , Sítios de Ligação , Íntrons/genética , Ilhotas Pancreáticas/metabolismo , Camundongos , Distribuição Tecidual , Fatores de Transcrição , Transportador 8 de Zinco
2.
Biochem J ; 421(3): 371-6, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19450229

RESUMO

The Slc30a8 gene encodes the islet-specific zinc transporter ZnT-8, which provides zinc for insulin-hexamer formation. Polymorphic variants in amino acid residue 325 of human ZnT-8 are associated with altered susceptibility to Type 2 diabetes and ZnT-8 autoantibody epitope specificity changes in Type 1 diabetes. To assess the physiological importance of ZnT-8, mice carrying a Slc30a8 exon 3 deletion were analysed histologically and phenotyped for energy metabolism and pancreatic hormone secretion. No gross anatomical or behavioural changes or differences in body weight were observed between wild-type and ZnT-8-/- mice, and ZnT-8-/- mouse islets were indistinguishable from wild-type in terms of their numbers, size and cellular composition. However, total zinc content was markedly reduced in ZnT-8-/- mouse islets, as evaluated both by Timm's histochemical staining of pancreatic sections and direct measurements in isolated islets. Blood glucose levels were unchanged in 16-week-old, 6 h fasted animals of either gender; however, plasma insulin concentrations were reduced in both female (approximately 31%) and male (approximately 47%) ZnT-8-/- mice. Intraperitoneal glucose tolerance tests demonstrated no impairment in glucose clearance in male ZnT-8-/- mice, but glucose-stimulated insulin secretion from isolated islets was reduced approximately 33% relative to wild-type littermates. In summary, Slc30a8 gene deletion is accompanied by a modest impairment in insulin secretion without major alterations in glucose metabolism.


Assuntos
Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Deleção de Sequência , Animais , Glicemia , Diabetes Mellitus/genética , Feminino , Teste de Tolerância a Glucose , Humanos , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Zinco/metabolismo , Transportador 8 de Zinco
3.
Immunol Res ; 55(1-3): 91-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22983906

RESUMO

G6PC2, also known as islet-specific glucose 6-phosphatase catalytic subunit-related protein (IGRP), is a major target of autoreactive CD8(+) T cells in both diabetic human subjects and the non-obese diabetic (NOD) mouse. However, in contrast to the abundant literature regarding the CD8(+) response to this antigen, much less is known about the potential involvement of IGRP-reactive CD4(+) T cells in diabetogenesis. The single previous study that examined this question in NOD mice was based upon a candidate epitope approach and identified three I-A(g7)-restricted epitopes that each elicited spontaneous responses in these animals. However, given the known inaccuracies of MHC class II epitope prediction algorithms, we hypothesized that additional specificities might also be targeted. To address this issue, we immunized NOD mice with membranes from insect cells overexpressing full-length recombinant mouse IGRP and measured recall responses of purified CD4(+) T cells using a library of overlapping peptides encompassing the entire 355-aa primary sequence. Nine peptides representing 8 epitopes gave recall responses, only 1 of which corresponded to any of the previously reported sequences. In each case proliferation was blocked by a monoclonal antibody to I-A(g7), but not the appropriate isotype control. Consistent with a role in diabetogenesis, proliferative responses to 4 of the 9 peptides (3 epitopes) were also detected in CD4(+) T cells purified from the pancreatic draining lymph nodes of pre-diabetic female animals, but not from peripheral lymph nodes or spleens of the same animals. Intriguingly, one of the newly identified spontaneously reactive epitopes (P8 [IGRP(55-72)]) is highly conserved between mice and man, suggesting that it might also be a target of HLA-DQ8-restricted T cells in diabetic human subjects, an hypothesis that we are currently testing.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Glucose-6-Fosfatase/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Proteínas/imunologia , Animais , Drosophila , Epitopos/imunologia , Feminino , Linfonodos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Peptídeos/imunologia
4.
Pancreas ; 41(6): 962-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22450367

RESUMO

OBJECTIVE: This study investigated the utility of advanced computational techniques to large-scale genome-based data to identify novel genes that govern murine pancreatic development. METHODS: An expression data set for mouse pancreatic development was complemented with high-throughput data analyzer to identify and prioritize novel genes. Quantitative real-time polymerase chain reaction, in situ hybridization, and immunohistochemistry were used to validate selected genes. RESULTS: Four new genes whose roles in the development of murine pancreas have not previously been established were identified: cystathionine ß-synthase (Cbs), Meis homeobox 1, growth factor independent 1, and aldehyde dehydrogenase 18 family, member A1. Their temporal expression during development was documented. Cbs was localized in the cytoplasm of the tip cells of the epithelial chords of the undifferentiated progenitor cells at E12.5 and was coexpressed with the pancreatic and duodenal homeobox 1 and pancreas-specific transcription factor, 1a-positive cells. In the adult pancreas, Cbs was localized primarily within the acinar compartment. CONCLUSIONS: In silico analysis of high-throughput microarray data in combination with background knowledge about genes provides an additional reliable method of identifying novel genes. To our knowledge, the expression and localization of Cbs have not been previously documented during mouse pancreatic development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genômica , Morfogênese/genética , Pâncreas/metabolismo , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Animais , Biologia Computacional , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica , Genômica/métodos , Idade Gestacional , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Diabetes ; 61(2): 436-46, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22210319

RESUMO

More than one-half of the ~50 human chemokines have been associated with or implicated in the pathogenesis of type 1 diabetes, yet their actual expression patterns in the islet environment of type 1 diabetic patients remain, at present, poorly defined. Here, we have integrated a human islet culture system, murine models of virus-induced and spontaneous type 1 diabetes, and the histopathological examination of pancreata from diabetic organ donors with the goal of providing a foundation for the informed selection of potential therapeutic targets within the chemokine/receptor family. Chemokine (C-C motif) ligand (CCL) 5 (CCL5), CCL8, CCL22, chemokine (C-X-C motif) ligand (CXCL) 9 (CXCL9), CXCL10, and chemokine (C-X3-C motif) ligand (CX3CL) 1 (CX3CL1) were the major chemokines transcribed (in an inducible nitric oxide synthase-dependent but not nuclear factor-κB-dependent fashion) and translated by human islet cells in response to in vitro inflammatory stimuli. CXCL10 was identified as the dominant chemokine expressed in vivo in the islet environment of prediabetic animals and type 1 diabetic patients, whereas CCL5, CCL8, CXCL9, and CX3CL1 proteins were present at lower levels in the islets of both species. Of importance, additional expression of the same chemokines in human acinar tissues emphasizes an underappreciated involvement of the exocrine pancreas in the natural course of type 1 diabetes that will require consideration for additional type 1 diabetes pathogenesis and immune intervention studies.


Assuntos
Quimiocinas/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Animais , Anti-Inflamatórios/farmacologia , Quimiocinas/genética , Diabetes Mellitus Tipo 1/etiologia , Humanos , Interleucina-1beta/farmacologia , Ilhotas Pancreáticas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , NF-kappa B/fisiologia , Fator de Necrose Tumoral alfa/farmacologia
6.
PLoS One ; 7(7): e40972, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22829903

RESUMO

OBJECTIVE: The SLC30A8 gene encodes the islet-specific transporter ZnT-8, which is hypothesized to provide zinc for insulin-crystal formation. A polymorphic variant in SLC30A8 is associated with altered susceptibility to type 2 diabetes. Several groups have examined the effect of global Slc30a8 gene deletion but the results have been highly variable, perhaps due to the mixed 129SvEv/C57BL/6J genetic background of the mice studied. We therefore sought to remove the conflicting effect of 129SvEv-specific modifier genes. METHODS: The impact of Slc30a8 deletion was examined in the context of the pure C57BL/6J genetic background. RESULTS: Male C57BL/6J Slc30a8 knockout (KO) mice had normal fasting insulin levels and no change in glucose-stimulated insulin secretion (GSIS) from isolated islets in marked contrast to the ∼50% and ∼35% decrease, respectively, in both parameters observed in male mixed genetic background Slc30a8 KO mice. This observation suggests that 129SvEv-specific modifier genes modulate the impact of Slc30a8 deletion. In contrast, female C57BL/6J Slc30a8 KO mice had reduced (∼20%) fasting insulin levels, though this was not associated with a change in fasting blood glucose (FBG), or GSIS from isolated islets. This observation indicates that gender also modulates the impact of Slc30a8 deletion, though the physiological explanation as to why impaired insulin secretion is not accompanied by elevated FBG is unclear. Neither male nor female C57BL/6J Slc30a8 KO mice showed impaired glucose tolerance. CONCLUSIONS: Our data suggest that, despite a marked reduction in islet zinc content, the absence of ZnT-8 does not have a substantial impact on mouse physiology.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Jejum/sangue , Insulina/sangue , Animais , Glicemia/metabolismo , Proteínas de Transporte de Cátions/genética , Feminino , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Insulina/genética , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores Sexuais , Zinco/metabolismo , Transportador 8 de Zinco
7.
J Mol Endocrinol ; 47(3): 251-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21798992

RESUMO

Genome-wide association studies have shown that a polymorphic variant in SLC30A8, which encodes zinc transporter-8, is associated with altered susceptibility to type 2 diabetes (T2D). This association is consistent with the observation that glucose-stimulated insulin secretion is decreased in islets isolated from Slc30a8 knockout mice. In this study, immunohistochemical staining was first used to show that SLC30A8 is expressed specifically in pancreatic islets. Fusion gene studies were then used to examine the molecular basis for the islet-specific expression of SLC30A8. The analysis of SLC30A8-luciferase expression in ßTC-3 cells revealed that the proximal promoter region, located between -6154 and -1, relative to the translation start site, was only active in stable but not transient transfections. VISTA analyses identified three regions in the SLC30A8 promoter and a region in SLC30A8 intron 2 that are conserved in the mouse Slc30a8 gene. Additional fusion gene experiments demonstrated that none of these Slc30a8 promoter regions exhibited enhancer activity when ligated to a heterologous promoter whereas the conserved region in SLC30A8 intron 2 conferred elevated reporter gene expression selectively in ßTC-3 but not in αTC-6 cells. Finally, the functional effects of a single nucleotide polymorphism (SNP), rs62510556, in this conserved intron 2 enhancer were investigated. Gel retardation studies showed that rs62510556 affects the binding of an unknown transcription factor and fusion gene analyses showed that it modulates enhancer activity. However, genetic analyses suggest that this SNP is not a causal variant that contributes to the association between SLC30A8 and T2D, at least in Europeans.


Assuntos
Proteínas de Transporte de Cátions/genética , Elementos Facilitadores Genéticos , Íntrons , Regiões Promotoras Genéticas , Animais , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Sequência Conservada , Diabetes Mellitus Tipo 2/genética , Regulação da Expressão Gênica , Genes Reporter , Células Secretoras de Glucagon/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Camundongos , Ativação Transcricional , Transfecção , Transportador 8 de Zinco
8.
Diabetes ; 60(11): 2922-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21896930

RESUMO

OBJECTIVE: Islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), now known as G6PC2, is a major target of autoreactive T cells implicated in the pathogenesis of type 1 diabetes in both mice and humans. This study aimed to determine whether suppression of G6p2 gene expression might therefore prevent or delay disease progression. RESEARCH DESIGN AND METHODS: G6pc2(-/-) mice were generated on the NOD/ShiLtJ genetic background, and glycemia was monitored weekly up to 35 weeks of age to determine the onset and incidence of diabetes. The antigen specificity of CD8(+) T cells infiltrating islets from NOD/ShiLtJ G6pc2(+/+) and G6pc2(-/-) mice at 12 weeks was determined in parallel. RESULTS: The absence of G6pc2 did not affect the time of onset, incidence, or sex bias of type 1 diabetes in NOD/ShiLtJ mice. Insulitis was prominent in both groups, but whereas NOD/ShiLtJ G6pc2(+/+) islets contained CD8(+) T cells reactive to the G6pc2 NRP peptide, G6pc2 NRP-reactive T cells were absent in NOD/ShiLtJ G6pc2(-/-) islets. CONCLUSIONS: These results demonstrate that G6pc2 is an important driver for the selection and expansion of islet-reactive CD8(+) T cells infiltrating NOD/ShiLtJ islets. However, autoreactivity to G6pc2 is not essential for the emergence of autoimmune diabetes. The results remain consistent with previous studies indicating that insulin may be the primary autoimmune target, at least in NOD/ShiLtJ mice.


Assuntos
Domínio Catalítico , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Progressão da Doença , Deleção de Genes , Glucose-6-Fosfatase/genética , Ilhotas Pancreáticas/metabolismo , Proteínas/genética , Animais , Autoanticorpos/análise , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Feminino , Glucose-6-Fosfatase/química , Humanos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Transgênicos , Proteínas/química , Caracteres Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA