Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Stem Cell ; 30(4): 488-497.e3, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-37028408

RESUMO

Understanding the origin of pancreatic ß cells has profound implications for regenerative therapies in diabetes. For over a century, it was widely held that adult pancreatic duct cells act as endocrine progenitors, but lineage-tracing experiments challenged this dogma. Gribben et al. recently used two existing lineage-tracing models and single-cell RNA sequencing to conclude that adult pancreatic ducts contain endocrine progenitors that differentiate to insulin-expressing ß cells at a physiologically important rate. We now offer an alternative interpretation of these experiments. Our data indicate that the two Cre lines that were used directly label adult islet somatostatin-producing ∂ cells, which precludes their use to assess whether ß cells originate from duct cells. Furthermore, many labeled ∂ cells, which have an elongated neuron-like shape, were likely misclassified as ß cells because insulin-somatostatin coimmunolocalizations were not used. We conclude that most evidence so far indicates that endocrine and exocrine lineage borders are rarely crossed in the adult pancreas.


Assuntos
Células Secretoras de Insulina , Lacunas de Evidências , Diferenciação Celular , Pâncreas/fisiologia , Ductos Pancreáticos , Insulina , Somatostatina
2.
Nat Cell Biol ; 24(10): 1528-1540, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36202974

RESUMO

The biological purpose of long non-coding RNAs (lncRNAs) is poorly understood. Haploinsufficient mutations in HNF1A homeobox A (HNF1A), encoding a homeodomain transcription factor, cause diabetes mellitus. Here, we examine HASTER, the promoter of an lncRNA antisense to HNF1A. Using mouse and human models, we show that HASTER maintains cell-specific physiological HNF1A concentrations through positive and negative feedback loops. Pancreatic ß cells from Haster mutant mice consequently showed variegated HNF1A silencing or overexpression, resulting in hyperglycaemia. HASTER-dependent negative feedback was essential to prevent HNF1A binding to inappropriate genomic regions. We demonstrate that the HASTER promoter DNA, rather than the lncRNA, modulates HNF1A promoter-enhancer interactions in cis and thereby regulates HNF1A transcription. Our studies expose a cis-regulatory element that is unlike classic enhancers or silencers, it stabilizes the transcription of its target gene and ensures the fidelity of a cell-specific transcription factor program. They also show that disruption of a mammalian lncRNA promoter can cause diabetes mellitus.


Assuntos
Fator 1-alfa Nuclear de Hepatócito , Regiões Promotoras Genéticas , RNA Longo não Codificante , Animais , Humanos , Camundongos , Fator 1-alfa Nuclear de Hepatócito/genética , Mamíferos , RNA Longo não Codificante/genética , Transcrição Gênica/genética , Transcrição Gênica/fisiologia
3.
Dev Cell ; 57(16): 1922-1936.e9, 2022 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-35998583

RESUMO

Sequence variants in cis-acting enhancers are important for polygenic disease, but their role in Mendelian disease is poorly understood. Redundancy between enhancers that regulate the same gene is thought to mitigate the pathogenic impact of enhancer mutations. Recent findings, however, have shown that loss-of-function mutations in a single enhancer near PTF1A cause pancreas agenesis and neonatal diabetes. Using mouse and human genetic models, we show that this enhancer activates an entire PTF1A enhancer cluster in early pancreatic multipotent progenitors. This leading role, therefore, precludes functional redundancy. We further demonstrate that transient expression of PTF1A in multipotent progenitors sets in motion an epigenetic cascade that is required for duct and endocrine differentiation. These findings shed insights into the genome regulatory mechanisms that drive pancreas differentiation. Furthermore, they reveal an enhancer that acts as a regulatory master key and is thus vulnerable to pathogenic loss-of-function mutations.


Assuntos
Diabetes Mellitus , Fatores de Transcrição , Animais , Diferenciação Celular/genética , Diabetes Mellitus/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Recém-Nascido , Camundongos , Mutação/genética , Pâncreas/metabolismo , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/metabolismo
4.
Genes Dev ; 35(17-18): 1229-1242, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34385258

RESUMO

Multiple transcription factors have been shown to promote pancreatic ß-cell differentiation, yet much less is known about negative regulators. Earlier epigenomic studies suggested that the transcriptional repressor REST could be a suppressor of endocrinogenesis in the embryonic pancreas. However, pancreatic Rest knockout mice failed to show abnormal numbers of endocrine cells, suggesting that REST is not a major regulator of endocrine differentiation. Using a different conditional allele that enables profound REST inactivation, we observed a marked increase in pancreatic endocrine cell formation. REST inhibition also promoted endocrinogenesis in zebrafish and mouse early postnatal ducts and induced ß-cell-specific genes in human adult duct-derived organoids. We also defined genomic sites that are bound and repressed by REST in the embryonic pancreas. Our findings show that REST-dependent inhibition ensures a balanced production of endocrine cells from embryonic pancreatic progenitors.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Peixe-Zebra , Animais , Diferenciação Celular/genética , Camundongos , Organogênese/genética , Pâncreas , Peixe-Zebra/genética
5.
Islets ; 13(5-6): 134-139, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34282714

RESUMO

The Hnf1b-CreERT2 BAC transgenic (Tg(Hnf1b-cre/ERT2)1Jfer) has been used extensively to trace the progeny of pancreatic ducts in developmental, regeneration, or cancer models. Hnf1b-CreERT2 transgenics have been used to show that the cells that form the embryonic pancreas duct-like plexus are bipotent duct-endocrine progenitors, whereas adult mouse duct cells are not a common source of ß cells in various regenerative settings. The interpretation of such genetic lineage tracing studies is critically dependent on a correct understanding of the cell type specificity of recombinase activity with each reporter system. We have reexamined the performance of Hnf1b-CreERT2 with a Rosa26-RFP reporter transgene. This showed inducible recombination of up to 96% adult duct cells, a much higher efficiency than previously used reporter transgenes. Despite this high duct-cell excision, recombination in α and ß cells remained very low, similar to previously used reporters. However, nearly half of somatostatin-expressing δ cells showed reporter activation, which was due to Cre expression in δ cells rather than to duct to δ cell conversions. The high recombination efficiency in duct cells indicates that the Hnf1b-CreERT2 model can be useful for both ductal fate mapping and genetic inactivation studies. The recombination in δ cells does not modify the interpretation of studies that failed to show duct conversions to other cell types, but needs to be considered if this model is used in studies that aim to modify the plasticity of pancreatic duct cells.


Assuntos
Ilhotas Pancreáticas , Células Secretoras de Somatostatina , Animais , Fator 1-beta Nuclear de Hepatócito/genética , Integrases/genética , Camundongos , Camundongos Transgênicos , Recombinação Genética , Transgenes
6.
Cell Rep ; 35(2): 108981, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852861

RESUMO

Despite the central role of chromosomal context in gene transcription, human noncoding DNA variants are generally studied outside of their genomic location. This limits our understanding of disease-causing regulatory variants. INS promoter mutations cause recessive neonatal diabetes. We show that all INS promoter point mutations in 60 patients disrupt a CC dinucleotide, whereas none affect other elements important for episomal promoter function. To model CC mutations, we humanized an ∼3.1-kb region of the mouse Ins2 gene. This recapitulated developmental chromatin states and cell-specific transcription. A CC mutant allele, however, abrogated active chromatin formation during pancreas development. A search for transcription factors acting through this element revealed that another neonatal diabetes gene product, GLIS3, has a pioneer-like ability to derepress INS chromatin, which is hampered by the CC mutation. Our in vivo analysis, therefore, connects two human genetic defects in an essential mechanism for developmental activation of the INS gene.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Diabetes Mellitus/genética , Insulina/genética , Pâncreas/metabolismo , Mutação Puntual , Proteínas Repressoras/genética , Transativadores/genética , Alelos , Animais , Cromatina/química , Cromatina/patologia , Proteínas de Ligação a DNA/deficiência , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Recém-Nascido , Doenças do Recém-Nascido , Insulina/deficiência , Camundongos , Camundongos Transgênicos , Pâncreas/crescimento & desenvolvimento , Pâncreas/patologia , Regiões Promotoras Genéticas , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Proteínas Repressoras/deficiência , Transativadores/deficiência , Transcrição Gênica
7.
EMBO J ; 39(9): e102808, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32154941

RESUMO

Defects in transcriptional regulators of pancreatic exocrine differentiation have been implicated in pancreatic tumorigenesis, but the molecular mechanisms are poorly understood. The locus encoding the transcription factor HNF1A harbors susceptibility variants for pancreatic ductal adenocarcinoma (PDAC), while KDM6A, encoding Lysine-specific demethylase 6A, carries somatic mutations in PDAC. Here, we show that pancreas-specific Hnf1a null mutant transcriptomes phenocopy those of Kdm6a mutations, and both defects synergize with KrasG12D to cause PDAC with sarcomatoid features. We combine genetic, epigenomic, and biochemical studies to show that HNF1A recruits KDM6A to genomic binding sites in pancreatic acinar cells. This remodels the acinar enhancer landscape, activates differentiated acinar cell programs, and indirectly suppresses oncogenic and epithelial-mesenchymal transition genes. We also identify a subset of non-classical PDAC samples that exhibit the HNF1A/KDM6A-deficient molecular phenotype. These findings provide direct genetic evidence that HNF1A deficiency promotes PDAC. They also connect the tumor-suppressive role of KDM6A deficiency with a cell-specific molecular mechanism that underlies PDAC subtype definition.


Assuntos
Células Acinares/metabolismo , Carcinoma Ductal Pancreático/genética , Fator 1-alfa Nuclear de Hepatócito/genética , Histona Desmetilases/genética , Neoplasias Pancreáticas/genética , Animais , Carcinoma Ductal Pancreático/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Histona Desmetilases/metabolismo , Humanos , Camundongos , Mutação , Especificidade de Órgãos , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo
8.
Nat Cell Biol ; 17(5): 615-626, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25915126

RESUMO

The genomic regulatory programmes that underlie human organogenesis are poorly understood. Pancreas development, in particular, has pivotal implications for pancreatic regeneration, cancer and diabetes. We have now characterized the regulatory landscape of embryonic multipotent progenitor cells that give rise to all pancreatic epithelial lineages. Using human embryonic pancreas and embryonic-stem-cell-derived progenitors we identify stage-specific transcripts and associated enhancers, many of which are co-occupied by transcription factors that are essential for pancreas development. We further show that TEAD1, a Hippo signalling effector, is an integral component of the transcription factor combinatorial code of pancreatic progenitor enhancers. TEAD and its coactivator YAP activate key pancreatic signalling mediators and transcription factors, and regulate the expansion of pancreatic progenitors. This work therefore uncovers a central role for TEAD and YAP as signal-responsive regulators of multipotent pancreatic progenitors, and provides a resource for the study of embryonic development of the human pancreas.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Multipotentes/metabolismo , Proteínas Nucleares/metabolismo , Pâncreas/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Biologia Computacional , Proteínas de Ligação a DNA/genética , Bases de Dados Genéticas , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Organogênese , Pâncreas/embriologia , Fenótipo , Fosfoproteínas/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição de Domínio TEA , Fatores de Tempo , Fatores de Transcrição/genética , Proteínas de Sinalização YAP , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
9.
Diabetologia ; 58(3): 558-65, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25500700

RESUMO

AIMS/HYPOTHESIS: Pancreatic beta cells play a central role in the control of glucose homeostasis by secreting insulin to stimulate glucose uptake by peripheral tissues. Understanding the molecular mechanisms that control beta cell function and plasticity has critical implications for the pathophysiology and therapy of major forms of diabetes. Selective gene inactivation in pancreatic beta cells, using the Cre-lox system, is a powerful approach to assess the role of particular genes in beta cells and their impact on whole body glucose homeostasis. Several Cre recombinase (Cre) deleter mice have been established to allow inactivation of genes in beta cells, but many show non-specific recombination in other cell types, often in the brain. METHODS: We describe the generation of Ins1(Cre) and Ins1(CreERT2) mice in which the Cre or Cre-oestrogen receptor fusion protein (CreERT2) recombinases have been introduced at the initiation codon of the Ins1 gene. RESULTS: We show that Ins1(Cre) mice induce efficient and selective recombination of floxed genes in beta cells from the time of birth, with no recombination in the central nervous system. These mice have normal body weight and glucose homeostasis. Furthermore, we show that tamoxifen treatment of adult Ins1(CreERT2) mice crossed with Rosa26-tdTomato mice induces efficient recombination in beta cells. CONCLUSIONS/INTERPRETATION: These two strains of deleter mice are useful new resources to investigate the molecular physiology of pancreatic beta cells.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Feminino , Teste de Tolerância a Glucose , Insulina/genética , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Mutantes
10.
Hepatology ; 60(4): 1367-77, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24700364

RESUMO

UNLABELLED: Severe liver diseases are characterized by expansion of liver progenitor cells (LPC), which correlates with disease severity. However, the origin and role of LPC in liver physiology and in hepatic injury remains a contentious topic. We found that ductular reaction cells in human cirrhotic livers express hepatocyte nuclear factor 1 homeobox B (HNF1ß). However, HNF1ß expression was not present in newly generated epithelial cell adhesion molecule (EpCAM)-positive hepatocytes. In order to investigate the role of HNF1ß-expressing cells we used a tamoxifen-inducible Hnf1ßCreER/R26R(Yfp/LacZ) mouse to lineage-trace Hnf1ß(+) biliary duct cells and to assess their contribution to LPC expansion and hepatocyte generation. Lineage tracing demonstrated no contribution of HNF1ß(+) cells to hepatocytes during liver homeostasis in healthy mice or after loss of liver mass. After acute acetaminophen or carbon tetrachloride injury no contribution of HNF1ß(+) cells to hepatocyte was detected. We next assessed the contribution of Hnf1ß(+) -derived cells following two liver injury models with LPC expansion, a diethoxycarbonyl-1,4-dihydro-collidin (DDC)-diet and a choline-deficient ethionine-supplemented (CDE)-diet. The contribution of Hnf1ß(+) cells to liver regeneration was dependent on the liver injury model. While no contribution was observed after DDC-diet treatment, mice fed with a CDE-diet showed a small population of hepatocytes derived from Hnf1ß(+) cells that were expanded to 1.86% of total hepatocytes after injury recovery. Genome-wide expression profile of Hnf1ß(+) -derived cells from the DDC and CDE models indicated that no contribution of LPC to hepatocytes was associated with LPC expression of genes related to telomere maintenance, inflammation, and chemokine signaling pathways. CONCLUSION: HNF1ß(+) biliary duct cells are the origin of LPC. HNF1ß(+) cells do not contribute to hepatocyte turnover in the healthy liver, but after certain liver injury, they can differentiate to hepatocytes contributing to liver regeneration.


Assuntos
Ductos Biliares/patologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Células Epiteliais/patologia , Hepatócitos/patologia , Regeneração Hepática/fisiologia , Fígado/patologia , Células-Tronco/patologia , Acetaminofen/efeitos adversos , Animais , Ductos Biliares/metabolismo , Tetracloreto de Carbono/efeitos adversos , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Dieta/efeitos adversos , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Fator 1-beta Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Homeostase/fisiologia , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco/metabolismo
11.
Nat Genet ; 46(2): 136-143, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24413736

RESUMO

Type 2 diabetes affects over 300 million people, causing severe complications and premature death, yet the underlying molecular mechanisms are largely unknown. Pancreatic islet dysfunction is central in type 2 diabetes pathogenesis, and understanding islet genome regulation could therefore provide valuable mechanistic insights. We have now mapped and examined the function of human islet cis-regulatory networks. We identify genomic sequences that are targeted by islet transcription factors to drive islet-specific gene activity and show that most such sequences reside in clusters of enhancers that form physical three-dimensional chromatin domains. We find that sequence variants associated with type 2 diabetes and fasting glycemia are enriched in these clustered islet enhancers and identify trait-associated variants that disrupt DNA binding and islet enhancer activity. Our studies illustrate how islet transcription factors interact functionally with the epigenome and provide systematic evidence that the dysregulation of islet enhancers is relevant to the mechanisms underlying type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Bases , Cromatina/genética , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Diabetes Mellitus Tipo 2/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Formaldeído , Estudo de Associação Genômica Ampla , Humanos , Dados de Sequência Molecular , Análise de Sequência de RNA , Fatores de Transcrição/genética , Navegador
12.
Genes Dev ; 27(1): 52-63, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23271347

RESUMO

Polycomb-mediated gene repression is essential for embryonic development, yet its precise role in lineage-specific programming is poorly understood. Here we inactivated Ring1b, encoding a polycomb-repressive complex 1 subunit, in pancreatic multipotent progenitors (Ring1b(progKO)). This caused transcriptional derepression of a subset of direct Ring1b target genes in differentiated pancreatic islet cells. Unexpectedly, Ring1b inactivation in differentiated islet ß cells (Ring1b(ßKO)) did not cause derepression, even after multiple rounds of cell division, suggesting a role for Ring1b in the establishment but not the maintenance of repression. Consistent with this notion, derepression in Ring1b(progKO) islets occurred preferentially in genes that were targeted de novo by Ring1b during pancreas development. The results support a model in which Ring1b bookmarks its target genes during embryonic development, and these genes are maintained in a repressed state through Ring1b-independent mechanisms in terminally differentiated cells. This work provides novel insights into how epigenetic mechanisms contribute to shaping the transcriptional identity of differentiated lineages.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Metilação de DNA , Embrião de Mamíferos , Epigênese Genética , Masculino , Camundongos , Neurônios/metabolismo , Complexo Repressor Polycomb 1/genética , Ubiquitina-Proteína Ligases/genética
13.
Mol Endocrinol ; 25(2): 339-47, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21193557

RESUMO

The expression pattern of genes important for pancreatic islet cell function requires the actions of cell-enriched transcription factors. Musculoaponeurotic fibrosarcoma homolog A (MafA) is a ß-cell-specific transcriptional activator critical to adult islet ß-cell function, with MafA mutant mice manifesting symptoms associated with human type 2 diabetes. Here, we describe that MafA expression is controlled by hepatocyte nuclear factor 1-α (Hnf1α), the transcription factor gene mutated in the most common monoallelic form of maturity onset diabetes of the young. There are six conserved sequence domains in the 5'-flanking MafA promoter, of which one, region 3 (R3) [base pair (bp) -8118/-7750] is principally involved in controlling the unique developmental and adult islet ß-cell-specific expression pattern. Chromatin immunoprecipitation analysis demonstrated that Hnf1α bound specifically within R3. Furthermore, in vitro DNA-binding experiments localized an Hnf1α regulatory element between bp -7822 and -7793, an area previously associated with stimulation by the islet developmental regulator, Islet1. However, site-directed mutational studies showed that Hnf1α was essential to R3-driven reporter activation through bp -7816/-7811. Significantly, MafA levels were dramatically reduced in the insulin(+) cell population remaining in embryonic and adult Hnf1α(-/-) pancreata. Our results demonstrate that Hnf1α regulates MafA in ß-cells and suggests that compromised MafA expression contributes to ß-cell dysfunction in maturity onset diabetes of the young.


Assuntos
Fator 1-alfa Nuclear de Hepatócito/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição Maf Maior/genética , Animais , Animais Geneticamente Modificados , Western Blotting , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/genética , Humanos , Fatores de Transcrição Maf Maior/metabolismo , Camundongos , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico
14.
Genome Res ; 20(6): 722-32, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395405

RESUMO

The epigenome changes that underlie cellular differentiation in developing organisms are poorly understood. To gain insights into how pancreatic beta-cells are programmed, we profiled key histone methylations and transcripts in embryonic stem cells, multipotent progenitors of the nascent embryonic pancreas, purified beta-cells, and 10 differentiated tissues. We report that despite their endodermal origin, beta-cells show a transcriptional and active chromatin signature that is most similar to ectoderm-derived neural tissues. In contrast, the beta-cell signature of trimethylated H3K27, a mark of Polycomb-mediated repression, clusters with pancreatic progenitors, acinar cells and liver, consistent with the epigenetic transmission of this mark from endoderm progenitors to their differentiated cellular progeny. We also identified two H3K27 methylation events that arise in the beta-cell lineage after the pancreatic progenitor stage. One is a wave of cell-selective de novo H3K27 trimethylation in non-CpG island genes. Another is the loss of bivalent and H3K27me3-repressed chromatin in a core program of neural developmental regulators that enables a convergence of the gene activity state of beta-cells with that of neural cells. These findings reveal a dynamic regulation of Polycomb repression programs that shape the identity of differentiated beta-cells.


Assuntos
Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Ilhotas Pancreáticas/metabolismo , Pâncreas/embriologia , Proteínas Repressoras/genética , Animais , Separação Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Epigênese Genética , Citometria de Fluxo , Histonas/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/citologia , Proteínas do Grupo Polycomb
15.
Proc Natl Acad Sci U S A ; 107(7): 3105-10, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20133622

RESUMO

Heterozygous coding mutations in the INS gene that encodes preproinsulin were recently shown to be an important cause of permanent neonatal diabetes. These dominantly acting mutations prevent normal folding of proinsulin, which leads to beta-cell death through endoplasmic reticulum stress and apoptosis. We now report 10 different recessive INS mutations in 15 probands with neonatal diabetes. Functional studies showed that recessive mutations resulted in diabetes because of decreased insulin biosynthesis through distinct mechanisms, including gene deletion, lack of the translation initiation signal, and altered mRNA stability because of the disruption of a polyadenylation signal. A subset of recessive mutations caused abnormal INS transcription, including the deletion of the C1 and E1 cis regulatory elements, or three different single base-pair substitutions in a CC dinucleotide sequence located between E1 and A1 elements. In keeping with an earlier and more severe beta-cell defect, patients with recessive INS mutations had a lower birth weight (-3.2 SD score vs. -2.0 SD score) and were diagnosed earlier (median 1 week vs. 10 weeks) compared to those with dominant INS mutations. Mutations in the insulin gene can therefore result in neonatal diabetes as a result of two contrasting pathogenic mechanisms. Moreover, the recessively inherited mutations provide a genetic demonstration of the essential role of multiple sequence elements that regulate the biosynthesis of insulin in man.


Assuntos
Diabetes Mellitus/genética , Insulina/biossíntese , Mutação/genética , Precursores de Proteínas/genética , Análise Mutacional de DNA , Primers do DNA/genética , Dosagem de Genes , Genes Recessivos/genética , Humanos , Recém-Nascido , Insulina/genética , Masculino , Sondas de Oligonucleotídeos
16.
Mol Cell Biol ; 29(11): 2945-59, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19289501

RESUMO

Heterozygous HNF1A mutations cause pancreatic-islet beta-cell dysfunction and monogenic diabetes (MODY3). Hnf1alpha is known to regulate numerous hepatic genes, yet knowledge of its function in pancreatic islets is more limited. We now show that Hnf1a deficiency in mice leads to highly tissue-specific changes in the expression of genes involved in key functions of both islets and liver. To gain insights into the mechanisms of tissue-specific Hnf1alpha regulation, we integrated expression studies of Hnf1a-deficient mice with identification of direct Hnf1alpha targets. We demonstrate that Hnf1alpha can bind in a tissue-selective manner to genes that are expressed only in liver or islets. We also show that Hnf1alpha is essential only for the transcription of a minor fraction of its direct-target genes. Even among genes that were expressed in both liver and islets, the subset of targets showing functional dependence on Hnf1alpha was highly tissue specific. This was partly explained by the compensatory occupancy by the paralog Hnf1beta at selected genes in Hnf1a-deficient liver. In keeping with these findings, the biological consequences of Hnf1a deficiency were markedly different in islets and liver. Notably, Hnf1a deficiency led to impaired large-T-antigen-induced growth and oncogenesis in beta cells yet enhanced proliferation in hepatocytes. Collectively, these findings show that Hnf1alpha governs broad, highly tissue-specific genetic programs in pancreatic islets and liver and reveal key consequences of Hnf1a deficiency relevant to the pathophysiology of monogenic diabetes.


Assuntos
Fator 1-alfa Nuclear de Hepatócito/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Fígado/citologia , Fígado/metabolismo , Transcrição Gênica , Região 5'-Flanqueadora/genética , Animais , Sequência de Bases , Proliferação de Células , Biologia Computacional , Sequência Conservada , Regulação da Expressão Gênica , Genoma/genética , Fator 1-alfa Nuclear de Hepatócito/deficiência , Fator 1-beta Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Ligação Proteica , Sequências Reguladoras de Ácido Nucleico/genética
17.
Dev Cell ; 17(6): 849-60, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20059954

RESUMO

A longstanding unsettled question is whether pancreatic beta cells originate from exocrine duct cells. We have now used genetic labeling to fate map embryonic and adult pancreatic duct cells. We show that Hnf1beta+ cells of the trunk compartment of the early branching pancreas are precursors of acinar, duct, and endocrine lineages. Hnf1beta+ cells subsequent form the embryonic duct epithelium, which gives rise to both ductal and endocrine lineages, but not to acinar cells. By the end of gestation, the fate of Hnf1beta+ duct cells is further restrained. We provide compelling evidence that the ductal epithelium does not make a significant contribution to acinar or endocrine cells during neonatal growth, during a 6 month observation period, or during beta cell growth triggered by ligation of the pancreatic duct or by cell-specific ablation with alloxan followed by EGF/gastrin treatment. Thus, once the ductal epithelium differentiates it has a restricted plasticity, even under regenerative settings.


Assuntos
Células Secretoras de Insulina/citologia , Pâncreas/embriologia , Animais , Feminino , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pâncreas/citologia , Pâncreas Exócrino/embriologia
18.
Endocr Dev ; 12: 33-45, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17923767

RESUMO

Mutations in the genes encoding transcriptional regulators HNF1beta (TCF2), HNF1alpha (TCF1), and HNF4alpha cause autosomal dominant diabetes (also known as maturity-onset diabetes of the young). Herein, we review what we have learnt during recent years concerning the functions of these regulators in the developing and adult pancreas. Mouse studies have revealed that HNF1beta is a critical regulator of a transcriptional network that controls the specification, growth, and differentiation of the embryonic pancreas. HNF1beta mutations in humans accordingly often cause pancreas hypoplasia. By contrast, HNF1alpha and HNF4alpha have been shown to regulate the function of differentiated beta-cells. HNF1alpha and HNF4alpha mutations in patients thus cause decreased glucose-induced insulin secretion that leads to a progressive form of diabetes. HNF4alpha mutations paradoxically also cause in utero and neonatal hyperinsulinism, which later evolves to decreased glucose-induced secretion. Recent studies show that Hnf4alpha deficiency in mice causes not only abnormal insulin secretion, but also an impairment of the expansion of beta-cell mass that normally occurs during pregnancy. In line with this finding, we present data that Hnf1alpha-/- beta-cells expressing SV40 large T antigen show a severe impairment of proliferation and failure to form tumours. Collectively, these findings implicate HNF1beta as a regulator of pancreas organogenesis and differentiation, whereas HNF1alpha and HNF4alpha primarily regulate both growth and function of islet beta-cells.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Fator 1-alfa Nuclear de Hepatócito/fisiologia , Fator 1-beta Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Pâncreas/fisiologia , Divisão Celular , Diabetes Mellitus Tipo 2/genética , Regulação da Expressão Gênica no Desenvolvimento , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/genética , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Pâncreas/crescimento & desenvolvimento , Fenótipo
19.
J Org Chem ; 69(23): 8053-61, 2004 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-15527290

RESUMO

Here we describe the synthesis and use of the Pulegone-derived bidentate P,S ligands PuPHOS and CyPuPHOS in the intermolecular Pauson-Khand reaction. Ligand exchange reaction of hexacarbonyldicobalt-alkyne complexes with PuPHOS provides a diasteromeric mixture of complexes (up to 4.5:1) from which the major isomers can be conveniently separated by simple crystallization. An isomerization-crystallization sequence of the original mixture results in a dynamic resolution that allows the preparation of the pure major Co(2)(mu-TMSC(2)H)(CO)(4)-PuPHOS (15a) in a multigram scale. Pauson-Khand reaction of 15a with norbornadiene provided, for the first time, the corresponding enone 18 with up to 93% yield and 97% ee. The use of (+)-18 as a surrogate of chiral cyclopentadienone is also demonstrated. Copper-catalyzed Michael addition of a Grignard reagent followed by removal of the TMS group with TBAF were the most reliable methods to transform (+)-18 into valuable starting materials 20a-e for the enantioselective synthesis of cyclopentenoid systems.

20.
Inorg Chem ; 42(19): 5890-9, 2003 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-12971757

RESUMO

The new fluorinated rigid ligand L, 1,4-bis(4-pyridyl)tetrafluorobenzene, was used in combination with different diphosphine Pd(II) and Pt(II) triflates to build metallosupramolecular assemblies. Complex equilibria between triangular and square entities were detected for all the cases. Characterization of the equilibria was accomplished by 1H, 31P(1H), 19F, and 195Pt(1H) NMR in combination with mass spectrometry. The square/triangle ratio was seen to depend on several factors, such as the nature of the metal corners, the concentration, and the solvent. The relative stability of the square and triangular complexes was explored by using force field methods. A GIAO-DFT study was carried out to analyze the changes of the 31P and 1H NMR data with the geometry of the complexes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...