Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
bioRxiv ; 2023 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-37905040

RESUMO

iPSC-derived human ß-like cells (BLC) hold promise for both therapy and disease modelling, but their generation remains challenging and their functional analyses beyond transcriptomic and morphological assessments remain limited. Here, we validate an approach using multicellular and single cell electrophysiological tools to evaluate BLCs functions. The Multi-Electrode Arrays (MEAs) measuring the extracellular electrical activity revealed that BLCs are electrically coupled, produce slow potential (SP) signals like primary ß-cells that are closely linked to insulin secretion. We also used high-resolution single-cell patch-clamp measurements to capture the exocytotic properties, and characterize voltage-gated sodium and calcium currents. These were comparable to those in primary ß and EndoC-ßH1 cells. The KATP channel conductance is greater than in human primary ß cells which may account for the limited glucose responsiveness observed with MEA. We used MEAs to study the impact of the type 2 diabetes protective SLC30A8 allele (p.Lys34Serfs*50) and found that BLCs with this allele have stronger electrical coupling. Our data suggest that with an adapted approach BLCs from pioneer protocol can be used to evaluate the functional impact of genetic variants on ß-cell function and coupling.

2.
Nat Commun ; 14(1): 6119, 2023 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-37777536

RESUMO

The coding variant (p.Arg192His) in the transcription factor PAX4 is associated with an altered risk for type 2 diabetes (T2D) in East Asian populations. In mice, Pax4 is essential for beta cell formation but its role on human beta cell development and/or function is unknown. Participants carrying the PAX4 p.His192 allele exhibited decreased pancreatic beta cell function compared to homozygotes for the p.192Arg allele in a cross-sectional study in which we carried out an intravenous glucose tolerance test and an oral glucose tolerance test. In a pedigree of a patient with young onset diabetes, several members carry a newly identified p.Tyr186X allele. In the human beta cell model, EndoC-ßH1, PAX4 knockdown led to impaired insulin secretion, reduced total insulin content, and altered hormone gene expression. Deletion of PAX4 in human induced pluripotent stem cell (hiPSC)-derived islet-like cells resulted in derepression of alpha cell gene expression. In vitro differentiation of hiPSCs carrying PAX4 p.His192 and p.X186 risk alleles exhibited increased polyhormonal endocrine cell formation and reduced insulin content that can be reversed with gene correction. Together, we demonstrate the role of PAX4 in human endocrine cell development, beta cell function, and its contribution to T2D-risk.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagon , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina , Humanos , Camundongos , Animais , Proteínas de Homeodomínio/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Estudos Transversais , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Glucagon/metabolismo
3.
Diabetologia ; 66(4): 674-694, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36633628

RESUMO

AIMS/HYPOTHESIS: Genome-wide studies have uncovered multiple independent signals at the RREB1 locus associated with altered type 2 diabetes risk and related glycaemic traits. However, little is known about the function of the zinc finger transcription factor Ras-responsive element binding protein 1 (RREB1) in glucose homeostasis or how changes in its expression and/or function influence diabetes risk. METHODS: A zebrafish model lacking rreb1a and rreb1b was used to study the effect of RREB1 loss in vivo. Using transcriptomic and cellular phenotyping of a human beta cell model (EndoC-ßH1) and human induced pluripotent stem cell (hiPSC)-derived beta-like cells, we investigated how loss of RREB1 expression and activity affects pancreatic endocrine cell development and function. Ex vivo measurements of human islet function were performed in donor islets from carriers of RREB1 type 2 diabetes risk alleles. RESULTS: CRISPR/Cas9-mediated loss of rreb1a and rreb1b function in zebrafish supports an in vivo role for the transcription factor in beta cell mass, beta cell insulin expression and glucose levels. Loss of RREB1 also reduced insulin gene expression and cellular insulin content in EndoC-ßH1 cells and impaired insulin secretion under prolonged stimulation. Transcriptomic analysis of RREB1 knockdown and knockout EndoC-ßH1 cells supports RREB1 as a novel regulator of genes involved in insulin secretion. In vitro differentiation of RREB1KO/KO hiPSCs revealed dysregulation of pro-endocrine cell genes, including RFX family members, suggesting that RREB1 also regulates genes involved in endocrine cell development. Human donor islets from carriers of type 2 diabetes risk alleles in RREB1 have altered glucose-stimulated insulin secretion ex vivo, consistent with a role for RREB1 in regulating islet cell function. CONCLUSIONS/INTERPRETATION: Together, our results indicate that RREB1 regulates beta cell function by transcriptionally regulating the expression of genes involved in beta cell development and function.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animais , Humanos , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Glucose/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/genética , Peixe-Zebra/genética
4.
Mol Metab ; 66: 101621, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36307047

RESUMO

OBJECTIVE: Identifying the transcripts which mediate genetic association signals for type 2 diabetes (T2D) is critical to understand disease mechanisms. Studies in pancreatic islets support the transcription factor ZMIZ1 as a transcript underlying a T2D GWAS signal, but how it influences T2D risk is unknown. METHODS: ß-Cell-specific Zmiz1 knockout (Zmiz1ßKO) mice were generated and phenotypically characterised. Glucose homeostasis was assessed in Zmiz1ßKO mice and their control littermates on chow diet (CD) and high fat diet (HFD). Islet morphology and function were examined by immunohistochemistry and in vitro islet function was assessed by dynamic insulin secretion assay. Transcript and protein expression were assessed by RNA sequencing and Western blotting. In islets isolated from genotyped human donors, we assessed glucose-dependent insulin secretion and islet insulin content by static incubation assay. RESULTS: Male and female Zmiz1ßKO mice were glucose intolerant with impaired insulin secretion, compared with control littermates. Transcriptomic profiling of Zmiz1ßKO islets identified over 500 differentially expressed genes including those involved in ß-cell function and maturity, which we confirmed at the protein level. Upon HFD, Zmiz1ßKO mice fail to expand ß-cell mass and become severely diabetic. Human islets from carriers of the ZMIZ1-linked T2D-risk alleles have reduced islet insulin content and glucose-stimulated insulin secretion. CONCLUSIONS: ß-Cell Zmiz1 is required for normal glucose homeostasis. Genetic variation at the ZMIZ1 locus may influence T2D-risk by reducing islet mass expansion upon metabolic stress and the ability to maintain a mature ß-cell state.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Fatores de Transcrição , Animais , Feminino , Humanos , Masculino , Camundongos , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/metabolismo , Camundongos Knockout , Dieta Hiperlipídica
5.
Diabetes ; 71(12): 2612-2631, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36170671

RESUMO

Transcriptional and functional cellular specialization has been described for insulin-secreting ß-cells of the endocrine pancreas. However, it is not clear whether ß-cell heterogeneity is stable or reflects dynamic cellular states. We investigated the temporal kinetics of endogenous insulin gene activity using live cell imaging, with complementary experiments using FACS and single-cell RNA sequencing, in ß-cells from Ins2GFP knockin mice. In vivo staining and FACS analysis of islets from Ins2GFP mice confirmed that at a given moment, ∼25% of ß-cells exhibited significantly higher activity at the evolutionarily conserved insulin gene, Ins2. Live cell imaging over days captured Ins2 gene activity dynamics in single ß-cells. Autocorrelation analysis revealed a subset of oscillating cells, with mean oscillation periods of 17 h. Increased glucose concentrations stimulated more cells to oscillate and resulted in higher average Ins2 gene activity per cell. Single-cell RNA sequencing showed that Ins2(GFP)HIGH ß-cells were enriched for markers of ß-cell maturity. Ins2(GFP)HIGH ß-cells were also significantly less viable at all glucose concentrations and in the context of endoplasmic reticulum stress. Collectively, our results demonstrate that the heterogeneity of insulin production, observed in mouse and human ß-cells, can be accounted for by dynamic states of insulin gene activity.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Camundongos , Humanos , Animais , Insulina/genética , Estresse do Retículo Endoplasmático , Glucose/farmacologia
6.
Lancet Diabetes Endocrinol ; 9(10): 708-724, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34480875

RESUMO

Type 1 diabetes is characterised by autoimmune-mediated destruction of pancreatic ß-cell mass. With the advent of insulin therapy a century ago, type 1 diabetes changed from a progressive, fatal disease to one that requires lifelong complex self-management. Replacing the lost ß-cell mass through transplantation has proven successful, but limited donor supply and need for lifelong immunosuppression restricts widespread use. In this Review, we highlight incremental advances over the past 20 years and remaining challenges in regenerative medicine approaches to restoring ß-cell mass and function in type 1 diabetes. We begin by summarising the role of endocrine islets in glucose homoeostasis and how this is altered in disease. We then discuss the potential regenerative capacity of the remaining islet cells and the utility of stem cell-derived ß-like cells to restore ß-cell function. We conclude with tissue engineering approaches that might improve the engraftment, function, and survival of ß-cell replacement therapies.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Ilhotas Pancreáticas/fisiologia , Medicina Regenerativa , Animais , Contagem de Células , Proliferação de Células/fisiologia , Diabetes Mellitus Tipo 1/fisiopatologia , História do Século XXI , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/história , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/tendências , Regeneração/fisiologia , Medicina Regenerativa/história , Medicina Regenerativa/métodos , Medicina Regenerativa/tendências , Engenharia Tecidual/história , Engenharia Tecidual/métodos , Engenharia Tecidual/tendências
7.
Nat Rev Endocrinol ; 16(4): 202-212, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32099086

RESUMO

Type 2 diabetes mellitus (T2DM) is an increasingly prevalent multifactorial disease that has both genetic and environmental risk factors, resulting in impaired glucose homeostasis. Genome-wide association studies (GWAS) have identified over 400 genetic signals that are associated with altered risk of T2DM. Human physiology and epigenomic data support a central role for the pancreatic islet in the pathogenesis of T2DM. This Review focuses on the promises and challenges of moving from genetic associations to molecular mechanisms and highlights efforts to identify the causal variant and effector transcripts at T2DM GWAS susceptibility loci. In addition, we examine current human models that are used to study both ß-cell development and function, including EndoC-ß cell lines and human induced pluripotent stem cell-derived ß-like cells. We use examples of four T2DM susceptibility loci (CDKAL1, MTNR1B, SLC30A8 and PAM) to emphasize how a holistic approach involving genetics, physiology, and cellular and developmental biology can disentangle disease mechanisms at T2DM GWAS signals.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Tipo 2/genética , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Humanos , Modelos Biológicos
8.
Nat Genet ; 51(11): 1596-1606, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31676859

RESUMO

A rare loss-of-function allele p.Arg138* in SLC30A8 encoding the zinc transporter 8 (ZnT8), which is enriched in Western Finland, protects against type 2 diabetes (T2D). We recruited relatives of the identified carriers and showed that protection was associated with better insulin secretion due to enhanced glucose responsiveness and proinsulin conversion, particularly when compared with individuals matched for the genotype of a common T2D-risk allele in SLC30A8, p.Arg325. In genome-edited human induced pluripotent stem cell (iPSC)-derived ß-like cells, we establish that the p.Arg138* allele results in reduced SLC30A8 expression due to haploinsufficiency. In human ß cells, loss of SLC30A8 leads to increased glucose responsiveness and reduced KATP channel function similar to isolated islets from carriers of the T2D-protective allele p.Trp325. These data position ZnT8 as an appealing target for treatment aimed at maintaining insulin secretion capacity in T2D.


Assuntos
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/prevenção & controle , Glucose/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Transportador 8 de Zinco/metabolismo , Adolescente , Adulto , Idoso , Diabetes Mellitus Tipo 2/patologia , Feminino , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Ilhotas Pancreáticas/patologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Transportador 8 de Zinco/genética
9.
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
10.
Stem Cell Reports ; 11(6): 1551-1564, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30540962

RESUMO

Human embryonic stem cells (hESCs) are a potential unlimited source of insulin-producing ß cells for diabetes treatment. A greater understanding of how ß cells form during embryonic development will improve current hESC differentiation protocols. All pancreatic endocrine cells, including ß cells, are derived from Neurog3-expressing endocrine progenitors. This study characterizes the single-cell transcriptomes of 6,905 mouse embryonic day (E) 15.5 and 6,626 E18.5 pancreatic cells isolated from Neurog3-Cre; Rosa26mT/mG embryos, allowing for enrichment of endocrine progenitors (yellow; tdTomato + EGFP) and endocrine cells (green; EGFP). Using a NEUROG3-2A-eGFP CyT49 hESC reporter line (N5-5), 4,462 hESC-derived GFP+ cells were sequenced. Differential expression analysis revealed enrichment of markers that are consistent with progenitor, endocrine, or previously undescribed cell-state populations. This study characterizes the single-cell transcriptomes of mouse and hESC-derived endocrine progenitors and serves as a resource (https://lynnlab.shinyapps.io/embryonic_pancreas) for improving the formation of functional ß-like cells from hESCs.


Assuntos
Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Pâncreas/citologia , Análise de Célula Única , Transcriptoma/genética , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Humanos , Camundongos , RNA/metabolismo , Fatores de Tempo
11.
Dev Cell ; 41(2): 129-142.e6, 2017 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-28441528

RESUMO

During pancreatic development, proliferating pancreatic progenitors activate the proendocrine transcription factor neurogenin 3 (NEUROG3), exit the cell cycle, and differentiate into islet cells. The mechanisms that direct robust NEUROG3 expression within a subset of progenitor cells control the size of the endocrine population. Here we demonstrate that NEUROG3 is phosphorylated within the nucleus on serine 183, which catalyzes its hyperphosphorylation and proteosomal degradation. During progression through the progenitor cell cycle, NEUROG3 phosphorylation is driven by the actions of cyclin-dependent kinases 2 and 4/6 at G1/S cell-cycle checkpoint. Using models of mouse and human pancreas development, we show that lengthening of the G1 phase of the pancreatic progenitor cell cycle is essential for proper induction of NEUROG3 and initiation of endocrine cell differentiation. In sum, these studies demonstrate that progenitor cell-cycle G1 lengthening, through its actions on stabilization of NEUROG3, is an essential variable in normal endocrine cell genesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Células-Tronco/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Endócrinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Ilhotas Pancreáticas/citologia , Camundongos , Proteínas do Tecido Nervoso/genética , Fosforilação/fisiologia , Células-Tronco/metabolismo
12.
Diabetologia ; 58(5): 1013-23, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25652387

RESUMO

AIMS/HYPOTHESIS: The sex-determining region Y (SRY)-related high mobility group (HMG) box (SOX) family of transcription factors is essential for normal organismal development. Despite the longstanding knowledge that many SOX family members are expressed during pancreas development, a role for many of these factors in the establishment of insulin-producing beta cell fate remains to be determined. The aim of this study is to elucidate the role of SOX4 during beta cell development. METHODS: We used pancreas and endocrine progenitor mouse knockouts of Sox4 to uncover the roles of SOX4 during pancreas development. Lineage tracing and in vitro models were used to determine how SOX4 regulates beta cell formation and understand the fate of Sox4-null endocrine lineage cells. RESULTS: This study demonstrates a progenitor cell-autonomous role for SOX4 in regulating the genesis of beta cells and shows that it is required at multiple stages of the process. SOX4 deletion in the multipotent pancreatic progenitors resulted in impaired endocrine progenitor cell differentiation. Deletion of SOX4 later in the Neurog3-expressing cells also caused reductions in beta cells. Lineage studies showed loss of Sox4 in endocrine progenitors resulted in a block in terminal islet cell differentiation that was attributed to reduction in the production of key beta cell specification factors. CONCLUSIONS/INTERPRETATION: These results demonstrate that SOX4 is essential for normal endocrine pancreas development both concomitant with, and downstream of, the endocrine fate decision. In conclusion, these studies position Sox4 temporally in the endocrine differentiation programme and provide a new target for improving in vitro differentiation of glucose-responsive pancreatic beta cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Ilhotas Pancreáticas/embriologia , Proteínas do Tecido Nervoso/metabolismo , Organogênese/genética , Fatores de Transcrição SOXC/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição SOXC/genética
13.
PLoS One ; 9(12): e114275, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25474420

RESUMO

Human embryonic stem cells (hESCs) have great promise as a source of unlimited transplantable cells for regenerative medicine. However, current progress on producing the desired cell type for disease treatment has been limited due to an insufficient understanding of the developmental processes that govern their differentiation, as well as a paucity of tools to systematically study differentiation in the lab. In order to overcome these limitations, cell-type reporter hESC lines will be required. Here we outline two strategies using Transcription Activator Like Effector Nucleases (TALENs) and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-CRISPR-Associated protein (Cas) to create OCT4-eGFP knock-in add-on hESC lines. Thirty-one and forty-seven percent of clones were correctly modified using the TALEN and CRISPR-Cas9 systems, respectively. Further analysis of three correctly targeted clones demonstrated that the insertion of eGFP in-frame with OCT4 neither significantly impacted expression from the wild type allele nor did the fusion protein have a dramatically different biological stability. Importantly, the OCT4-eGFP fusion was easily detected using microscopy, flow cytometry and western blotting. The OCT4 reporter lines remained equally competent at producing CXCR4+ definitive endoderm that expressed a panel of endodermal genes. Moreover, the genomic modification did not impact the formation of NKX6.1+/SOX9+ pancreatic progenitor cells following directed differentiation. In conclusion, these findings demonstrate for the first time that CRISPR-Cas9 can be used to modify OCT4 and highlight the feasibility of creating cell-type specific reporter hESC lines utilizing genome-editing tools that facilitate homologous recombination.


Assuntos
Diferenciação Celular/genética , Endoderma/citologia , Células-Tronco Embrionárias Humanas/citologia , Fator 3 de Transcrição de Octâmero/genética , Sistemas CRISPR-Cas , Linhagem Celular , Endoderma/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Genoma Humano , Recombinação Homóloga/genética , Humanos , Fator 3 de Transcrição de Octâmero/biossíntese
14.
Development ; 141(19): 3772-81, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25209250

RESUMO

Although many regulatory networks involved in defining definitive endoderm have been identified, the mechanisms through which these networks interact to pattern the endoderm are less well understood. To explore the mechanisms involved in midgut patterning, we dissected the transcriptional regulatory elements of nephrocan (Nepn), the earliest known midgut specific gene in mice. We observed that Nepn expression is dramatically reduced in Sox17(-/-) and Raldh2(-/-) embryos compared with wild-type embryos. We further show that Nepn is directly regulated by Sox17 and the retinoic acid (RA) receptor via two enhancer elements located upstream of the gene. Moreover, Nepn expression is modulated by Activin signaling, with high levels inhibiting and low levels enhancing RA-dependent expression. In Foxh1(-/-) embryos in which Nodal signaling is reduced, the Nepn expression domain is expanded into the anterior gut region, confirming that Nodal signaling can modulate its expression in vivo. Together, Sox17 is required for Nepn expression in the definitive endoderm, while RA signaling restricts expression to the midgut region. A balance of Nodal/Activin signaling regulates the anterior boundary of the midgut expression domain.


Assuntos
Padronização Corporal/fisiologia , Endoderma/fisiologia , Trato Gastrointestinal/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Redes Reguladoras de Genes/fisiologia , Glicoproteínas/metabolismo , Transdução de Sinais/fisiologia , Ativinas/metabolismo , Aldeído Oxirredutases/metabolismo , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Redes Reguladoras de Genes/genética , Vetores Genéticos/genética , Proteínas HMGB/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Luciferases , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Ácido Retinoico/metabolismo , Fatores de Transcrição SOXF/metabolismo
15.
Diabetes ; 62(8): 2808-20, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23656887

RESUMO

Cellular homeostasis requires intrinsic sensing mechanisms to temper function in the face of prolonged activity. In the pancreatic ß-cell, glucose is likely a physiological trigger that activates an adaptive response to stimulation, thereby maintaining cellular homeostasis. Immediate early genes (IEGs) are activated as a first line of defense in cellular homeostasis and are largely responsible for transmitting an environmental cue to a cellular response. Here we examine the regulation and function of the novel ß-cell IEG, neuronal PAS domain protein 4 (Npas4). Using MIN6 cells, mouse and human islets, as well as in vivo infusions, we demonstrate that Npas4 is expressed within pancreatic islets and is upregulated by ß-cell depolarizing agents. Npas4 tempers ß-cell function through a direct inhibitory interaction with the insulin promoter and by blocking the potentiating effects of GLP-1 without significantly reducing glucose-stimulated secretion. Finally, Npas4 expression is induced by classical endoplasmic reticulum (ER) stressors and can prevent thapsigargin- and palmitate-induced dysfunction and cell death. These results suggest that Npas4 is a key activity-dependent regulator that improves ß-cell efficiency in the face of stress. We posit that Npas4 could be a novel therapeutic target in type 2 diabetes that could both reduce ER stress and cell death and maintain basal cell function.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Citoproteção/genética , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Citoproteção/efeitos dos fármacos , Emulsões/farmacologia , Estresse do Retículo Endoplasmático/fisiologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Glucose/farmacologia , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Fosfolipídeos/farmacologia , Regiões Promotoras Genéticas , Óleo de Soja/farmacologia , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA