Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Diabetologia ; 66(6): 1097-1115, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36912927

RESUMO

AIMS/HYPOTHESIS: Beta cells control glucose homeostasis via regulated production and secretion of insulin. This function arises from a highly specialised gene expression programme that is established during development and then sustained, with limited flexibility, in terminally differentiated cells. Dysregulation of this programme is seen in type 2 diabetes but mechanisms that preserve gene expression or underlie its dysregulation in mature cells are not well resolved. This study investigated whether methylation of histone H3 lysine 4 (H3K4), a marker of gene promoters with unresolved functional importance, is necessary for the maintenance of mature beta cell function. METHODS: Beta cell function, gene expression and chromatin modifications were analysed in conditional Dpy30 knockout mice, in which H3K4 methyltransferase activity is impaired, and in a mouse model of diabetes. RESULTS: H3K4 methylation maintains expression of genes that are important for insulin biosynthesis and glucose responsiveness. Deficient methylation of H3K4 leads to a less active and more repressed epigenome profile that locally correlates with gene expression deficits but does not globally reduce gene expression. Instead, developmentally regulated genes and genes in weakly active or suppressed states particularly rely on H3K4 methylation. We further show that H3K4 trimethylation (H3K4me3) is reorganised in islets from the Leprdb/db mouse model of diabetes in favour of weakly active and disallowed genes at the expense of terminal beta cell markers with broad H3K4me3 peaks. CONCLUSIONS/INTERPRETATION: Sustained methylation of H3K4 is critical for the maintenance of beta cell function. Redistribution of H3K4me3 is linked to gene expression changes that are implicated in diabetes pathology.


Assuntos
Diabetes Mellitus Tipo 2 , Insulinas , Camundongos , Animais , Histonas/metabolismo , Metilação , Lisina/metabolismo , Diabetes Mellitus Tipo 2/genética , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo
2.
Diabetologia ; 63(5): 1090, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32172312

RESUMO

Unfortunately, the human islet checklist was omitted from the electronic supplementary material (ESM) linked to this paper.

3.
Diabetologia ; 63(3): 561-576, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31984442

RESUMO

AIMS/HYPOTHESIS: Peptide hormones are first synthesised as larger, inactive precursors that are converted to their active forms by endopeptidase cleavage and post-translational modifications, such as amidation. Recent, large-scale genome-wide studies have suggested that two coding variants of the amidating enzyme, peptidylglycine α-amidating monooxygenase (PAM), are associated with impaired insulin secretion and increased type 2 diabetes risk. We aimed to elucidate the role of PAM in modulating beta cell peptide amidation, beta cell function and the development of diabetes. METHODS: PAM transcript and protein levels were analysed in mouse islets following induction of endoplasmic reticulum (ER) or cytokine stress, and PAM expression patterns were examined in human islets. To study whether haploinsufficiency of PAM accelerates the development of diabetes, Pam+/- and Pam+/+ mice were fed a low-fat diet (LFD) or high-fat diet (HFD) and glucose homeostasis was assessed. Since aggregates of the PAM substrate human islet amyloid polypeptide (hIAPP) lead to islet inflammation and beta cell failure, we also investigated whether PAM haploinsufficiency accelerated hIAPP-induced diabetes and islet amyloid formation in Pam+/- and Pam+/+ mice with beta cell expression of hIAPP. RESULTS: Immunostaining revealed high expression of PAM in alpha, beta and delta cells in human pancreatic islets. Pam mRNA and PAM protein expression were reduced in mouse islets following administration of an HFD, and in isolated islets following induction of ER stress with thapsigargin, or cytokine stress with IL-1ß, IFN-γ and TFN-α. Despite Pam+/- only having 50% PAM expression and enzyme activity as compared with Pam+/+ mice, glucose tolerance and body mass composition were comparable in the two models. After 24 weeks of HFD, both Pam+/- and Pam+/+ mice had insulin resistance and impaired glucose tolerance, but no differences in glucose tolerance, insulin sensitivity or plasma insulin levels were observed in PAM haploinsufficient mice. Islet amyloid formation and beta cell function were also similar in Pam+/- and Pam+/+ mice with beta cell expression of hIAPP. CONCLUSIONS/INTERPRETATION: Haploinsufficiency of PAM in mice does not accelerate the development of diet-induced obesity or hIAPP transgene-induced diabetes.


Assuntos
Amidina-Liases/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Oxigenases de Função Mista/genética , Amidina-Liases/fisiologia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Progressão da Doença , Epistasia Genética/fisiologia , Feminino , Predisposição Genética para Doença , Haploinsuficiência , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxigenases de Função Mista/fisiologia , Ratos , Ratos Endogâmicos Lew , Fatores de Risco
4.
Genome Res ; 20(8): 1037-51, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20551221

RESUMO

The liver and pancreas share a common origin and coexpress several transcription factors. To gain insight into the transcriptional networks regulating the function of these tissues, we globally identify binding sites for FOXA2 in adult mouse islets and liver, PDX1 in islets, and HNF4A in liver. Because most eukaryotic transcription factors bind thousands of loci, many of which are thought to be inactive, methods that can discriminate functionally active binding events are essential for the interpretation of genome-wide transcription factor binding data. To develop such a method, we also generated genome-wide H3K4me1 and H3K4me3 localization data in these tissues. By analyzing our binding and histone methylation data in combination with comprehensive gene expression data, we show that H3K4me1 enrichment profiles discriminate transcription factor occupied loci into three classes: those that are functionally active, those that are poised for activation, and those that reflect pioneer-like transcription factor activity. Furthermore, we demonstrate that the regulated presence of H3K4me1-marked nucleosomes at transcription factor occupied promoters and enhancers controls their activity, implicating both tissue-specific transcription factor binding and nucleosome remodeling complex recruitment in determining tissue-specific gene expression. Finally, we apply these approaches to generate novel insights into how FOXA2, PDX1, and HNF4A cooperate to drive islet- and liver-specific gene expression.


Assuntos
Loci Gênicos , Fator 3-beta Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/genética , Histonas/genética , Proteínas de Homeodomínio/genética , Ilhotas Pancreáticas/metabolismo , Fígado/metabolismo , Nucleossomos/genética , Transativadores/genética , Animais , Sequência de Bases , Sítios de Ligação , Perfilação da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos , Dados de Sequência Molecular , Nucleossomos/metabolismo , Sequências Reguladoras de Ácido Nucleico , Transativadores/metabolismo
5.
FEBS J ; 288(12): 3683-3693, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32926557

RESUMO

Pancreatic ß-cells are critical mediators of glucose homeostasis in the body, and proper cellular nutrient metabolism is critical to ß-cell function. Several interacting signaling networks that uniquely control ß-cell metabolism produce essential substrates and co-factors for catalytic reactions, including reactions that modify chromatin. Chromatin modifications, in turn, regulate gene expression. The reactions that modify chromatin are therefore well-positioned to adjust gene expression programs according to nutrient availability. It follows that dysregulation of nutrient metabolism in ß-cells may impact chromatin state and gene expression through altering the availability of these substrates and co-factors. Metabolic disorders such as type 2 diabetes (T2D) can significantly alter metabolite levels in cells. This suggests that a driver of ß-cell dysfunction during T2D may be the altered availability of substrates or co-factors necessary to maintain ß-cell chromatin state. Induced changes in the ß-cell chromatin modifications may then lead to dysregulation of gene expression, in turn contributing to the downward cascade of events that leads to the loss of functional ß-cell mass, and loss of glucose homeostasis, that occurs in T2D.


Assuntos
Cromatina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Processamento de Proteína Pós-Traducional , Acetilcoenzima A/metabolismo , Cromatina/química , Ciclo do Ácido Cítrico/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Glucose/metabolismo , Histonas/genética , Homeostase/genética , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ácidos Cetoglutáricos/metabolismo , Mitocôndrias/patologia , NAD/metabolismo , Fosforilação Oxidativa , Estresse Oxidativo , S-Adenosilmetionina/metabolismo
6.
Diabetes ; 70(11): 2568-2579, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34376477

RESUMO

During pancreas development, endocrine progenitors differentiate into the islet cell subtypes, which undergo further functional maturation in postnatal islet development. In islet ß-cells, genes involved in glucose-stimulated insulin secretion are activated, and glucose exposure increases the insulin response as ß-cells mature. We investigated the role of H3K4 trimethylation in endocrine cell differentiation and functional maturation by disrupting TrxG complex histone methyltransferase activity in mouse endocrine progenitors. In the embryo, genetic inactivation of TrxG component Dpy30 in NEUROG3+ cells did not affect the number of endocrine progenitors or endocrine cell differentiation. H3K4 trimethylation was progressively lost in postnatal islets, and the mice displayed elevated nonfasting and fasting glycemia as well as impaired glucose tolerance by postnatal day 24. Although postnatal endocrine cell proportions were equivalent to controls, islet RNA sequencing revealed a downregulation of genes involved in glucose-stimulated insulin secretion and an upregulation of immature ß-cell genes. Comparison of histone modification enrichment profiles in NEUROG3+ endocrine progenitors and mature islets suggested that genes downregulated by loss of H3K4 trimethylation more frequently acquire active histone modifications during maturation. Taken together, these findings suggest that H3K4 trimethylation is required for the activation of genes involved in the functional maturation of pancreatic islet endocrine cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Glicemia , Intolerância à Glucose , Humanos , Hiperglicemia , Metilação , Camundongos , Proteínas do Tecido Nervoso/genética
7.
Mol Cancer Ther ; 7(4): 749-58, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18375822

RESUMO

Molecular mechanisms responsible for lymphoma resistance to apoptosis often involve the bcl-2 pathway. In this study, we investigated the cell signaling pathways activated in bcl-2-overexpressing human mantle cell lymphoma cell lines (JVM-2 and Z-138) that have been treated with oblimersen, a molecular gene silencing strategy that effectively suppresses bcl-2 in vitro and in vivo. Z-138 cells expressed higher levels of bcl-2 and were more sensitive to the effects of bcl-2 silencing, mediated by oblimersen or bcl-2 small interfering RNA, in vitro. Tumors derived following injection of Z-138 cells were sensitive to oblimersen as judged by decreases in tumor growth rate and decreases in cell proliferation (as measured by Ki-67). Immunohistochemistry and Western blot analysis of oblimersen-treated Z-138 tumors revealed a dose-dependent decrease in bcl-2 levels and an associated increase in the proapoptotic proteins caspase-3 and caspase-9. Silencing bcl-2 in Z-138 xenografts revealed an associated dose-dependent suppression of bax, a decrease in nuclear factor-kappaB and phospho-nuclear factor-kappaB, and transient loss of p53 levels. Coimmunoprecipitation studies suggest that the latter observation is mediated by an association between bcl-2 and phospho-mdm2. Bcl-2 silencing also led to p27 down-regulation and coimmunoprecipitation studies point to a role for bcl-2 in regulation of p27 localization/degradation. Bcl-2 silencing was also correlated with loss of cyclin D1a protein levels but not cyclin D1b levels. Coimmunoprecipitation studies indicate that bcl-2 may mediate its effects on cyclin D1a via interaction with p38 mitogen-activated protein kinase as well as a previously unreported interaction between bcl-2 and cyclin D1a.


Assuntos
Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Inativação Gênica , Linfoma de Célula do Manto/genética , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Animais , Apoptose/fisiologia , Western Blotting , Proliferação de Células , Ciclina D , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Ligação a DNA/fisiologia , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Linfoma de Célula do Manto/metabolismo , Linfoma de Célula do Manto/prevenção & controle , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas de Neoplasias/genética , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tionucleotídeos/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
8.
Cell Rep ; 28(7): 1830-1844.e6, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31412250

RESUMO

Appropriate regulation of genes that coordinate pancreas progenitor proliferation and differentiation is required for pancreas development. Here, we explore the role of H3K4 methylation and the Trithorax group (TrxG) complexes in mediating gene expression during pancreas development. Disruption of TrxG complex assembly, but not catalytic activity, prevented endocrine cell differentiation in pancreas progenitor spheroids. In vivo loss of TrxG catalytic activity in PDX1+ cells increased apoptosis and the fraction of progenitors in the G1 phase of the cell cycle. Pancreas progenitors were reallocated to the acinar lineage, primarily at the expense of NEUROG3+ endocrine progenitors. Later in development, acinar and endocrine cell numbers were decreased, and increased gene expression variance and reduced terminal marker activation in acinar cells led to their incomplete differentiation. These findings demonstrate that TrxG co-activator activity is required for gene induction, whereas TrxG catalytic activity and H3K4 methylation help maintain transcriptional stability.


Assuntos
Diferenciação Celular , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Organogênese , Pâncreas/citologia , Células-Tronco/citologia , Tiorredoxinas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Feminino , Proteínas de Homeodomínio/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/metabolismo , Células-Tronco/metabolismo , Tiorredoxinas/genética , Transativadores/fisiologia , Fatores de Transcrição/fisiologia
9.
BMC Dev Biol ; 8: 81, 2008 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-18778483

RESUMO

BACKGROUND: The full-length mammalian homologs of groucho, Tle1, 2, 3, and 4, act as transcriptional corepressors and are recruited by transcription factors containing an eh1 or WRPW/Y domain. Many transcription factors critical to pancreas development contain a Gro/TLE interaction domain and several have been shown to require Gro/TLE interactions for proper function during neuronal development. However, a detailed analysis of the expression patterns of the Gro/TLE proteins in pancreas development has not been performed. Moreover, little is known about the ability of Gro/TLE proteins to interact with transcription factors in the pancreas. RESULTS: We describe the expression of Gro/TLE family members, and of 34 different transcription factors that contain a Gro/TLE interaction motif, in the pancreas utilizing nine SAGE libraries created from the developing and adult pancreas, as well as the GenePaint database. Next, we show the dynamic expression of Tle1, 2, 3, 4, 5 and 6 during pancreas development by qRT-PCR. To further define the cell-type specificity of the expression of these proteins we use immunofluorescence to co-localize them with Pdx1 at embryonic day 12.5 (E12.5), Ngn3 at E14.5, Pdx1, Nkx2-2, Insulin, Glucagon, Pancreatic polypeptide and Somatostatin at E18.5, as well as Insulin and Glucagon in the adult. We then show that Tle2 can interact with Nkx2-2, Hes1, Arx, and Nkx6-1 which are all critical factors in pancreas development. Finally, we demonstrate that Tle2 modulates the repressive abilities of Arx in a beta-cell line. CONCLUSION: Although Tle1, 2, 3, and 4 show overlapping expression in pancreatic progenitors and in the adult islet, the expression of these factors is restricted to different cell types during endocrine cell maturation. Of note, Tle2 and Tle3 are co-expressed with Gro/TLE interaction domain containing transcription factors that are essential for endocrine pancreas development. We further demonstrate that Tle2 can interact with several of these factors and that Tle2 modulate Arx's repressive activity. Taken together our studies suggest that Gro/TLE proteins play a role in the repression of target genes during endocrine cell specification.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Pâncreas/embriologia , Pâncreas/metabolismo , Proteínas Repressoras/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Linhagem Celular , Proteínas Correpressoras , Feminino , Proteína Homeobox Nkx-2.2 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas/genética , Proteínas/metabolismo , Ratos , Proteínas Repressoras/biossíntese
10.
Cell Metab ; 27(6): 1294-1308.e7, 2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29754954

RESUMO

To date, it remains largely unclear to what extent chromatin machinery contributes to the susceptibility and progression of complex diseases. Here, we combine deep epigenome mapping with single-cell transcriptomics to mine for evidence of chromatin dysregulation in type 2 diabetes. We find two chromatin-state signatures that track ß cell dysfunction in mice and humans: ectopic activation of bivalent Polycomb-silenced domains and loss of expression at an epigenomically unique class of lineage-defining genes. ß cell-specific Polycomb (Eed/PRC2) loss of function in mice triggers diabetes-mimicking transcriptional signatures and highly penetrant, hyperglycemia-independent dedifferentiation, indicating that PRC2 dysregulation contributes to disease. The work provides novel resources for exploring ß cell transcriptional regulation and identifies PRC2 as necessary for long-term maintenance of ß cell identity. Importantly, the data suggest a two-hit (chromatin and hyperglycemia) model for loss of ß cell identity in diabetes.


Assuntos
Cromatina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica , Inativação Gênica , Células Secretoras de Insulina/metabolismo , Complexo Repressor Polycomb 2/fisiologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Mapeamento Cromossômico , Diabetes Mellitus Tipo 2/genética , Epigenômica , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Hiperglicemia/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Complexo Repressor Polycomb 2/genética , Análise de Célula Única
11.
Gene Expr Patterns ; 6(3): 310-24, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16378759

RESUMO

To identify genes and pathways not previously implicated in the mesenchymal-epithelial (M/E) interactions that are critical for normal mouse prostate development, we constructed six serial analysis of gene expression (SAGE) libraries. Bioinformatic analyses revealed expression of various members of numerous signalling pathways and the differential expression of several members of the wingless-related MMTV integration site (Wnt) signalling pathway. This pathway has not been previously implicated in prostate development thus expression of selected Wnt pathway members in the developing prostate was confirmed by RT-qPCR. Of particular interest, an antagonist of the Wnt pathway, secreted frizzled related protein 2 (Sfrp2), was highly expressed in the early prostate libraries and down regulated at later developmental stages. The expression levels of four Wnt ligands reported to interact with Sfrp2 were, therefore, examined by RT-qPCR. We found that only Wnt4 transcripts were detectable in the developing prostate. Expression of Sfrp2 was validated using RT-qPCR and localization of Sfrp2 transcripts and protein was carried out using in situ hybridization and immunofluorescence, respectively. These studies provide the first evidence that Wnt pathway members are expressed in the developing prostate. Functional analyses are now required to establish the biological significance of this observation.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Próstata/embriologia , Transdução de Sinais , Proteínas Wnt/metabolismo , Animais , Biologia Computacional , Imunofluorescência , Hibridização In Situ , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Próstata/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Trends Endocrinol Metab ; 27(3): 142-152, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26783078

RESUMO

The chromatin landscape of a cell is dynamic and can be altered by chromatin regulators that control nucleosome placement and DNA or histone modifications. Together with transcription factors, these complexes help dictate the transcriptional output of a cell and, thus, balance cell proliferation and differentiation while restricting tissue-specific gene expression. In this review, we describe current research on chromatin regulators and their roles in pancreas development and the maintenance of mature ß cell function, which, once elucidated, will help us better understand how ß cell differentiation occurs and is maintained. These studies have so far implicated proteins from several complexes that regulate DNA methylation, nucleosome remodeling, and histone acetylation and methylation that could become promising targets for diabetes therapy and stem cell differentiation.


Assuntos
Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Histonas/metabolismo , Ilhotas Pancreáticas/metabolismo , Modelos Biológicos , Animais , Diferenciação Celular , Proliferação de Células , Proteínas Cromossômicas não Histona/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/patologia , Nucleossomos/metabolismo , Processamento de Proteína Pós-Traducional
13.
Mol Endocrinol ; 29(9): 1254-68, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26177052

RESUMO

Myt3 is a prosurvival factor in pancreatic islets; however, its role in islet-cell development is not known. Here, we demonstrate that myelin transcription factor 3 (Myt3) is expressed in migrating islet cells in the developing and neonatal pancreas and thus sought to determine whether Myt3 plays a role in this process. Using an ex vivo model of islet-cell migration, we demonstrate that Myt3 suppression significantly inhibits laminin-V/integrin-ß1-dependent α- and ß-cell migration onto 804G, and impaired 804G-induced F-actin and E-cadherin redistribution. Exposure of islets to proinflammatory cytokines, which suppress Myt3 expression, had a similar effect, whereas Myt3 overexpression partially rescued the migratory ability of the islet cells. We show that loss of islet-cell migration, due to Myt3 suppression or cytokine exposure, is independent of effects on islet-cell survival or proliferation. Myt3 suppression also had no effect on glucose-induced calcium influx, F-actin remodeling or insulin secretion by ß-cells. RNA-sequencing (RNA-seq) analysis of transduced islets showed that Myt3 suppression results in the up-regulation of Tgfbi, a secreted diabetogenic factor thought to impair cellular adhesion. Exposure of islets to exogenous transforming growth factor ß-induced (Tgfbi) impaired islet-cell migration similar to Myt3 suppression. Taken together, these data suggest a model by which cytokine-induced Myt3 suppression leads to Tgfbi de-repression and subsequently to impaired islet-cell migration, revealing a novel role for Myt3 in regulating islet-cell migration.


Assuntos
Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Células Secretoras de Insulina/metabolismo , Integrina beta1/metabolismo , Fatores de Transcrição/metabolismo , Actinas/metabolismo , Animais , Caderinas/metabolismo , Cálcio/metabolismo , Adesão Celular/fisiologia , Proliferação de Células/fisiologia , Citocinas/farmacologia , Proteínas da Matriz Extracelular/biossíntese , Proteínas da Matriz Extracelular/farmacologia , Feminino , Insulina/metabolismo , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição/antagonistas & inibidores , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/farmacologia , Calinina
14.
PLoS One ; 10(10): e0141470, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26505193

RESUMO

To better understand how ß-cells respond to proinflammatory cytokines we mapped the locations of histone 3 lysine 4 monomethylation (H3K4me1), a post-translational histone modification enriched at active and poised cis-regulatory regions, in IFNγ, Il-1ß, and TNFα treated pancreatic islets. We identified 96,721 putative cis-regulatory loci, of which 3,590 were generated de novo, 3,204 had increased H3K4me1, and 5,354 had decreased H3K4me1 in IFNγ, Il-1ß, and TNFα exposed islets. Roughly 10% of the de novo and increased regions were enriched for the repressive histone modification histone 3 lysine 27 trimethylation (H3K27me3) in untreated cells, and these were frequently associated with chemokine genes. We show that IFNγ, Il-1ß, and TNFα exposure overcomes this repression and induces chemokine gene activation in as little as three hours, and that this expression persists for days in absence of continued IFNγ, Il-1ß, and TNFα exposure. We implicate trithorax group (TrxG) complexes as likely players in the conversion of these repressed loci to an active state. To block the activity of these complexes, we suppressed Wdr5, a core component of the TrxG complexes, and used the H3K27me3 demethylase inhibitor GSK-J4. We show that GSK-J4 is particularly effective in blunting IFNγ, Il-1ß, and TNFα-induced chemokine gene expression in ß-cells; however, it induced significant islet-cell apoptosis and ß-cell dysfunction. Wdr5 suppression also reduced IFNγ, Il-1ß, and TNFα induced chemokine gene expression in ß-cells without affecting islet-cell survival or ß-cell function after 48hrs, but did begin to increase islet-cell apoptosis and ß-cell dysfunction after four days of treatment. Taken together these data suggest that the TrxG complex is potentially a viable target for preventing cytokine induced chemokine gene expression in ß-cells.


Assuntos
Histonas/genética , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas/metabolismo , Adenoviridae/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Benzazepinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/biossíntese , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/administração & dosagem , Interleucina-1beta/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Proteína de Leucina Linfoide-Mieloide/química , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas/genética , Pirimidinas/farmacologia , Fator de Necrose Tumoral alfa/administração & dosagem
15.
PLoS One ; 7(2): e32095, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22393380

RESUMO

We describe a model-based method, PING, for predicting nucleosome positions in MNase-Seq and MNase- or sonicated-ChIP-Seq data. PING compares favorably to NPS and TemplateFilter in scalability, accuracy and robustness to low read density. To demonstrate that PING predictions from widely available sonicated data can have sufficient spatial resolution to be to be useful for biological inference, we use Illumina H3K4me1 ChIP-seq data to detect changes in nucleosome positioning around transcription factor binding sites due to tamoxifen stimulation, to discriminate functional and non-functional transcription factor binding sites more effectively than with enrichment profiles, and to confirm that the pioneer transcription factor Foxa2 associates with the accessible major groove of nucleosomal DNA.


Assuntos
Biologia Computacional/métodos , Histonas/química , Algoritmos , Animais , Área Sob a Curva , Sítios de Ligação , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Ilhotas Pancreáticas/metabolismo , Camundongos , Nuclease do Micrococo/química , Modelos Estatísticos , Nucleossomos/metabolismo , Probabilidade , Reprodutibilidade dos Testes , Tamoxifeno/química , Transativadores/metabolismo , Fatores de Transcrição/química
16.
PLoS One ; 7(12): e51501, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23236509

RESUMO

AIMS/HYPOTHESIS: We previously identified the transcription factor Myt3 as specifically expressed in pancreatic islets. Here, we sought to determine the expression and regulation of Myt3 in islets and to determine its significance in regulating islet function and survival. METHODS: Myt3 expression was determined in embryonic pancreas and adult islets by qPCR and immunohistochemistry. ChIP-seq, ChIP-qPCR and luciferase assays were used to evaluate regulation of Myt3 expression. Suppression of Myt3 was used to evaluate gene expression, insulin secretion and apoptosis in islets. RESULTS: We show that Myt3 is the most abundant MYT family member in adult islets and that it is expressed in all the major endocrine cell types in the pancreas after E18.5. We demonstrate that Myt3 expression is directly regulated by Foxa2, Pdx1, and Neurod1, which are critical to normal ß-cell development and function, and that Ngn3 induces Myt3 expression through alterations in the Myt3 promoter chromatin state. Further, we show that Myt3 expression is sensitive to both glucose and cytokine exposure. Of specific interest, suppressing Myt3 expression reduces insulin content and increases ß-cell apoptosis, at least in part, due to reduced Pdx1, Mafa, Il-6, Bcl-xl, c-Iap2 and Igfr1 levels, while over-expression of Myt3 protects islets from cytokine induced apoptosis. CONCLUSION/INTERPRETATION: We have identified Myt3 as a novel transcriptional regulator with a critical role in ß-cell survival. These data are an important step in clarifying the regulatory networks responsible for ß-cell survival, and point to Myt3 as a potential therapeutic target for improving functional ß-cell mass.


Assuntos
Sobrevivência Celular/genética , Regulação da Expressão Gênica/fisiologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/metabolismo , Dedos de Zinco/genética , Animais , Imunoprecipitação da Cromatina , Primers do DNA/genética , Imunofluorescência , Regulação da Expressão Gênica/genética , Imuno-Histoquímica , Hibridização In Situ , Luciferases , Camundongos , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/genética , Fatores de Transcrição/genética
17.
PLoS One ; 5(11): e14146, 2010 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21152387

RESUMO

Pluripotent embryonic stem cells hold a great promise as an unlimited source of tissue for treatment of chronic diseases such as Type 1 diabetes. Herein, we describe a protocol using all-trans-retinoic acid, basic fibroblast growth factor and dibutyryl cAMP (DBcAMP) in the absence of embryoid body formation, for differentiation of murine embryonic stem cells into definitive endoderm that may serve as pancreatic precursors. The produced cells were analyzed by quantitative PCR, immunohistochemistry and static insulin release assay for markers of trilaminar embryo, and pancreas. Differentiated cells displayed increased Sox17 and Foxa2 expression consistent with definitive endoderm production. There was minimal production of Sox7, an extraembryonic endoderm marker, and Oct4, a marker of pluripotency. There was minimal mesoderm or neuroectoderm formation based on expression levels of the markers brachyury and Sox1, respectively. Various assays revealed that the cell clusters generated by this protocol express markers of the pancreatic lineage including insulin I, insulin II, C-peptide, PDX-1, carboxypeptidase E, pan-cytokeratin, amylase, glucagon, PAX6, Ngn3 and Nkx6.1. This protocol using all-trans-retinoic acid, DBcAMP, in the absence of embryoid bodies, generated cells that have features of definitive endoderm that may serve as pancreatic endocrine precursors.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Animais , Células Cultivadas , CMP Cíclico/análogos & derivados , CMP Cíclico/farmacologia , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica/efeitos dos fármacos , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Imuno-Histoquímica , Insulina/genética , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Fatores de Tempo , Tretinoína/farmacologia
18.
J Endocrinol ; 201(1): 1-13, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19136617

RESUMO

The transcriptional networks underlying mammalian cell development and function are largely unknown. The recently described use of flow cell sequencing devices in combination with chromatin immunoprecipitation (ChIP-seq) stands to revolutionize the identification of DNA-protein interactions. As such, ChIP-seq is rapidly becoming the method of choice for the genome-wide localization of histone modifications and transcription factor binding sites. As further studies are performed, the information generated by ChIP-seq is expected to allow the development of a framework for networks describing the transcriptional regulation of cellular development and function. However, to date, this technology has been applied only to a small number of cell types, and even fewer tissues, suggesting a huge potential for novel discovery in this field.


Assuntos
Imunoprecipitação da Cromatina/métodos , Mapeamento Cromossômico/métodos , Proteínas de Ligação a DNA/metabolismo , Análise de Sequência de DNA/métodos , Animais , Sítios de Ligação/genética , Humanos , Modelos Biológicos , Ligação Proteica
19.
PLoS One ; 3(9): e3205, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18787709

RESUMO

BACKGROUND: Serial Analysis of Gene Expression (SAGE) is a DNA sequencing-based method for large-scale gene expression profiling that provides an alternative to microarray analysis. Most analyses of SAGE data aimed at identifying co-expressed genes have been accomplished using various versions of clustering approaches that often result in a number of false positives. PRINCIPAL FINDINGS: Here we explore the use of seriation, a statistical approach for ordering sets of objects based on their similarity, for large-scale expression pattern discovery in SAGE data. For this specific task we implement a seriation heuristic we term 'progressive construction of contigs' that constructs local chains of related elements by sequentially rearranging margins of the correlation matrix. We apply the heuristic to the analysis of simulated and experimental SAGE data and compare our results to those obtained with a clustering algorithm developed specifically for SAGE data. We show using simulations that the performance of seriation compares favorably to that of the clustering algorithm on noisy SAGE data. CONCLUSIONS: We explore the use of a seriation approach for visualization-based pattern discovery in SAGE data. Using both simulations and experimental data, we demonstrate that seriation is able to identify groups of co-expressed genes more accurately than a clustering algorithm developed specifically for SAGE data. Our results suggest that seriation is a useful method for the analysis of gene expression data whose applicability should be further pursued.


Assuntos
Perfilação da Expressão Gênica/métodos , Algoritmos , Animais , Análise por Conglomerados , Simulação por Computador , Mapeamento de Sequências Contíguas , Reações Falso-Positivas , Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Pâncreas/metabolismo , Retina/metabolismo , Fatores de Transcrição/metabolismo
20.
Genome Biol ; 9(6): R99, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18554416

RESUMO

BACKGROUND: Despite recent advances, the transcriptional hierarchy driving pancreas organogenesis remains largely unknown, in part due to the paucity of comprehensive analyses. To address this deficit we generated ten SAGE libraries from the developing murine pancreas spanning Theiler stages 17-26, making use of available Pdx1 enhanced green fluorescent protein (EGFP) and Neurog3 EGFP reporter strains, as well as tissue from adult islets and ducts. RESULTS: We used a specificity metric to identify 2,536 tags with pancreas-enriched expression compared to 195 other mouse SAGE libraries. We subsequently grouped co-expressed transcripts with differential expression during pancreas development using K-means clustering. We validated the clusters first using quantitative real time PCR and then by analyzing the Theiler stage 22 pancreas in situ hybridization staining patterns of over 600 of the identified genes using the GenePaint database. These were then categorized into one of the five expression domains within the developing pancreas. Based on these results we identified a cascade of transcriptional regulators expressed in the endocrine pancreas lineage and, from this, we developed a predictive regulatory network describing beta-cell development. CONCLUSION: Taken together, this work provides evidence that the SAGE libraries generated here are a valuable resource for continuing to elucidate the molecular mechanisms regulating pancreas development. Furthermore, our studies provide a comprehensive analysis of pancreas development, and insights into the regulatory networks driving this process are revealed.


Assuntos
Perfilação da Expressão Gênica , Pâncreas/embriologia , Animais , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Organogênese , Pâncreas/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa