Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
FASEB J ; 36(10): e22514, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36106439

RESUMO

Despite several new therapeutic options for acute myeloid leukemia (AML), disease relapse remains a significant challenge. We have previously demonstrated that augmenting ceramides can counter various drug-resistance mechanisms, leading to enhanced cell death in cancer cells and extended survival in animal models. Using a nanoscale delivery system for ceramide (ceramide nanoliposomes, CNL), we investigated the effect of CNL within a standard of care venetoclax/cytarabine (Ara-C) regimen. We demonstrate that CNL augmented the efficacy of venetoclax/cytarabine in in vitro, ex vivo, and in vivo models of AML. CNL treatment induced non-apoptotic cytotoxicity, and augmented cell death induced by Ara-C and venetoclax. Mechanistically, CNL reduced both venetoclax (Mcl-1) and cytarabine (Chk1) drug-resistant signaling pathways. Moreover, venetoclax and Ara-C augmented the generation of endogenous pro-death ceramide species, which was intensified with CNL. Taken together, CNL has the potential to be utilized as an adjuvant therapy to improve outcomes, potentially extending survival, in patients with AML.


Assuntos
Antineoplásicos , Leucemia Mieloide Aguda , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Ceramidas , Citarabina/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Sulfonamidas
2.
Development ; 141(16): 3123-33, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25063451

RESUMO

The orphan nuclear receptor NR5A2 is necessary for the stem-like properties of the epiblast of the pre-gastrulation embryo and for cellular and physiological homeostasis of endoderm-derived organs postnatally. Using conditional gene inactivation, we show that Nr5a2 also plays crucial regulatory roles during organogenesis. During the formation of the pancreas, Nr5a2 is necessary for the expansion of the nascent pancreatic epithelium, for the subsequent formation of the multipotent progenitor cell (MPC) population that gives rise to pre-acinar cells and bipotent cells with ductal and islet endocrine potential, and for the formation and differentiation of acinar cells. At birth, the NR5A2-deficient pancreas has defects in all three epithelial tissues: a partial loss of endocrine cells, a disrupted ductal tree and a >90% deficit of acini. The acinar defects are due to a combination of fewer MPCs, deficient allocation of those MPCs to pre-acinar fate, disruption of acinar morphogenesis and incomplete acinar cell differentiation. NR5A2 controls these developmental processes directly as well as through regulatory interactions with other pancreatic transcriptional regulators, including PTF1A, MYC, GATA4, FOXA2, RBPJL and MIST1 (BHLHA15). In particular, Nr5a2 and Ptf1a establish mutually reinforcing regulatory interactions and collaborate to control developmentally regulated pancreatic genes by binding to shared transcriptional regulatory regions. At the final stage of acinar cell development, the absence of NR5A2 affects the expression of Ptf1a and its acinar specific partner Rbpjl, so that the few acinar cells that form do not complete differentiation. Nr5a2 controls several temporally distinct stages of pancreatic development that involve regulatory mechanisms relevant to pancreatic oncogenesis and the maintenance of the exocrine phenotype.


Assuntos
Células Acinares/citologia , Regulação da Expressão Gênica no Desenvolvimento , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Receptores Citoplasmáticos e Nucleares/fisiologia , Células-Tronco/citologia , Animais , Sequência de Bases , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Fenótipo , Receptores Citoplasmáticos e Nucleares/genética , Transgenes
3.
J Lipid Res ; 57(7): 1231-42, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27140664

RESUMO

The objective of our study was to determine the mechanism of action of the short-chain ceramide analog, C6-ceramide, and the breast cancer drug, tamoxifen, which we show coactively depress viability and induce apoptosis in human acute myelogenous leukemia cells. Exposure to the C6-ceramide-tamoxifen combination elicited decreases in mitochondrial membrane potential and complex I respiration, increases in reactive oxygen species (ROS), and release of mitochondrial proapoptotic proteins. Decreases in ATP levels, reduced glycolytic capacity, and reduced expression of inhibitors of apoptosis proteins also resulted. Cytotoxicity of the drug combination was mitigated by exposure to antioxidant. Cells metabolized C6-ceramide by glycosylation and hydrolysis, the latter leading to increases in long-chain ceramides. Tamoxifen potently blocked glycosylation of C6-ceramide and long-chain ceramides. N-desmethyltamoxifen, a poor antiestrogen and the major tamoxifen metabolite in humans, was also effective with C6-ceramide, indicating that traditional antiestrogen pathways are not involved in cellular responses. We conclude that cell death is driven by mitochondrial targeting and ROS generation and that tamoxifen enhances the ceramide effect by blocking its metabolism. As depletion of ATP and targeting the "Warburg effect" represent dynamic metabolic insult, this ceramide-containing combination may be of utility in the treatment of leukemia and other cancers.


Assuntos
Ceramidas/administração & dosagem , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Tamoxifeno/administração & dosagem , Trifosfato de Adenosina/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Complexo I de Transporte de Elétrons/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
4.
PLoS One ; 18(10): e0291512, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37796967

RESUMO

Proper maintenance of mature cellular phenotypes is essential for stable physiology, suppression of disease states, and resistance to oncogenic transformation. We describe the transcriptional regulatory roles of four key DNA-binding transcription factors (Ptf1a, Nr5a2, Foxa2 and Gata4) that sit at the top of a regulatory hierarchy controlling all aspects of a highly differentiated cell-type-the mature pancreatic acinar cell (PAC). Selective inactivation of Ptf1a, Nr5a2, Foxa2 and Gata4 individually in mouse adult PACs rapidly altered the transcriptome and differentiation status of PACs. The changes most emphatically included transcription of the genes for the secretory digestive enzymes (which conscript more than 90% of acinar cell protein synthesis), a potent anabolic metabolism that provides the energy and materials for protein synthesis, suppressed and properly balanced cellular replication, and susceptibility to transformation by oncogenic KrasG12D. The simultaneous inactivation of Foxa2 and Gata4 caused a greater-than-additive disruption of gene expression and uncovered their collaboration to maintain Ptf1a expression and control PAC replication. A measure of PAC dedifferentiation ranked the effects of the conditional knockouts as Foxa2+Gata4 > Ptf1a > Nr5a2 > Foxa2 > Gata4. Whereas the loss of Ptf1a or Nr5a2 greatly accelerated Kras-mediated transformation of mature acinar cells in vivo, the absence of Foxa2, Gata4, or Foxa2+Gata4 together blocked transformation completely, despite extensive dedifferentiation. A lack of correlation between PAC dedifferentiation and sensitivity to oncogenic KrasG12D negates the simple proposition that the level of differentiation determines acinar cell resistance to transformation.


Assuntos
Pâncreas Exócrino , Neoplasias Pancreáticas , Camundongos , Animais , Células Acinares/metabolismo , Epitélio/metabolismo , Fatores de Transcrição/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Fenótipo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo
5.
Chem Phys Lipids ; 245: 105205, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35483419

RESUMO

Ceramides and diacylglycerols are groups of lipids capable of nucleating and stabilizing ordered lipid domains, structures that have been implicated in a range of biological processes. Previous studies have used fluorescence reporter molecules to explore the influence of ceramide acyl chain structure on sphingolipid-rich ordered phases. Here, we use small-angle neutron scattering (SANS) to examine the ability of ceramides and diacylglycerols to promote lipid domain formation in the well-characterized domain-forming mixture DPPC/DOPC/cholesterol. SANS is a powerful, probe-free technique for interrogating membrane heterogeneity, as it is differentially sensitive to hydrogen's stable isotopes protium and deuterium. Specifically, neutron contrast is generated through selective deuteration of lipid species, thus enabling the detection of nanoscopic domains enriched in deuterated saturated lipids dispersed in a matrix of protiated unsaturated lipids. Using large unilamellar vesicles, we found that upon replacing 10 mol% DPPC with either C16:0 or C18:0 ceramide, or 16:0 diacylglycerol (dag), lipid domains persisted to higher temperatures. However, when DPPC was replaced with short chain (C6:0 or C12:0) or very long chain (C24:0) ceramides, or ceramides with unsaturated acyl chains of any length (C6:1(3), C6:1(5), C18:1, and C24:1), as well as C18:1-dag, lipid domains were destabilized, melting at lower temperatures than those in the DPPC/DOPC/cholesterol system. These results show how ceramide acyl chain length and unsaturation influence lipid domains and have implications for how cell membranes might modify their function through the generation of different ceramide species.


Assuntos
Ceramidas , Diglicerídeos , Ceramidas/química , Colesterol/química , Diglicerídeos/química , Bicamadas Lipídicas/química , Nêutrons , Espalhamento a Baixo Ângulo
6.
Mol Endocrinol ; 20(12): 3133-45, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16901969

RESUMO

The pancreatic and duodenal homeobox factor 1 (Pdx-1) is a Hox-like transcription factor that is responsible for the activation of the insulin gene. Previous studies have demonstrated the interaction in vitro of Pdx-1 with short (20-40 nucleotide) DNA fragments corresponding to A boxes of the insulin promoter. Precisely how Pdx-1 binds to DNA in the complex milieu of chromatin, however, has never been studied. In this study, we explored how Pdx-1-DNA interactions might be influenced by chromatin accessibility at the insulin gene in beta-cells (betaTC3) vs. pancreatic ductal cells (mPAC). We demonstrate that Pdx-1 occupies the endogenous insulin promoter in betaTC3 cells but not in mPAC cells, a finding that is independent of the intracellular Pdx-1 protein concentration. Based on micrococcal nuclease protection assays, the difference in promoter binding between the two cell types appears to be secondary to chromatin accessibility at predicted Pdx-1 binding sites between bp -126 to -296 (relative to the transcriptional start site) of the insulin promoter. Binding studies using purified Pdx-1 and reconstituted chromatin in vitro suggest that the positioning of a nucleosome(s) within this crucial region of the promoter might account for differences in chromatin accessibility. Consistent with these observations, fluorescence colocalization studies show that Pdx-1 does not occupy regions of compacted, nucleosome-rich chromatin within the nucleus. Our findings suggest a model whereby insulin transcription in the beta-cell is at least partially facilitated by enhanced chromatin accessibility within a crucial regulatory region between bp -126 to -296, thereby permitting occupancy by transactivators such as Pdx-1.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/genética , Transativadores/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Cromatina/química , DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Eucromatina/química , Eucromatina/metabolismo , Heterocromatina/química , Heterocromatina/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/análise , Camundongos , Nucleossomos/química , Nucleossomos/metabolismo , Regiões Promotoras Genéticas , Transativadores/análise , Transcrição Gênica , Ativação Transcricional
7.
Mol Cell Biol ; 36(23): 2945-2955, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27644326

RESUMO

Much remains unknown regarding the regulatory networks formed by transcription factors in mature, differentiated mammalian cells in vivo, despite many studies of individual DNA-binding transcription factors. We report a constellation of feed-forward loops formed by the pancreatic transcription factors MIST1 and PTF1 that govern the differentiated phenotype of the adult pancreatic acinar cell. PTF1 is an atypical basic helix-loop-helix transcription factor complex of pancreatic acinar cells and is critical to acinar cell fate specification and differentiation. MIST1, also a basic helix-loop-helix transcription factor, enhances the formation and maintenance of the specialized phenotype of professional secretory cells. The MIST1 and PTF1 collaboration controls a wide range of specialized cellular processes, including secretory protein synthesis and processing, exocytosis, and homeostasis of the endoplasmic reticulum. PTF1 drives Mist1 transcription, and MIST1 and PTF1 bind and drive the transcription of over 100 downstream acinar genes. PTF1 binds two canonical bipartite sites within a 0.7-kb transcriptional enhancer upstream of Mist1 that are essential for the activity of the enhancer in vivo MIST1 and PTF1 coregulate target genes synergistically or additively, depending on the target transcriptional enhancer. The frequent close binding proximity of PTF1 and MIST1 in pancreatic acinar cell chromatin implies extensive collaboration although the collaboration is not dependent on a stable physical interaction.

8.
Mol Cell Biol ; 36(24): 3033-3047, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27697859

RESUMO

Maintenance of cell type identity is crucial for health, yet little is known of the regulation that sustains the long-term stability of differentiated phenotypes. To investigate the roles that key transcriptional regulators play in adult differentiated cells, we examined the effects of depletion of the developmental master regulator PTF1A on the specialized phenotype of the adult pancreatic acinar cell in vivo Transcriptome sequencing and chromatin immunoprecipitation sequencing results showed that PTF1A maintains the expression of genes for all cellular processes dedicated to the production of the secretory digestive enzymes, a highly attuned surveillance of unfolded proteins, and a heightened unfolded protein response (UPR). Control by PTF1A is direct on target genes and indirect through a ten-member transcription factor network. Depletion of PTF1A causes an imbalance that overwhelms the UPR, induces cellular injury, and provokes acinar metaplasia. Compromised cellular identity occurs by derepression of characteristic stomach genes, some of which are also associated with pancreatic ductal cells. The loss of acinar cell homeostasis, differentiation, and identity is directly relevant to the pathologies of pancreatitis and pancreatic adenocarcinoma.


Assuntos
Células Acinares/citologia , Perfilação da Expressão Gênica/métodos , Pâncreas Exócrino/citologia , Fatores de Transcrição/genética , Transcrição Gênica , Células Acinares/metabolismo , Animais , Diferenciação Celular , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Homeostase , Camundongos , Pâncreas Exócrino/metabolismo , Desdobramento de Proteína , Análise de Sequência de RNA/métodos , Fatores de Transcrição/metabolismo , Resposta a Proteínas não Dobradas
9.
Mol Cell Biol ; 36(23): 2931-2944, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27644325

RESUMO

Transcriptional networks that govern secretory cell specialization, including instructing cells to develop a unique cytoarchitecture, amass extensive protein synthesis machinery, and be embodied to respond to endoplasmic reticulum (ER) stress, remain largely uncharacterized. In this study, we discovered that the secretory cell transcription factor MIST1 (Bhlha15), previously shown to be essential for cytoskeletal organization and secretory activity, also functions as a potent ER stress-inducible transcriptional regulator. Genome-wide DNA binding studies, coupled with genetic mouse models, revealed MIST1 gene targets that function along the entire breadth of the protein synthesis, processing, transport, and exocytosis networks. Additionally, key MIST1 targets are essential for alleviating ER stress in these highly specialized cells. Indeed, MIST1 functions as a coregulator of the unfolded protein response (UPR) master transcription factor XBP1 for a portion of target genes that contain adjacent MIST1 and XBP1 binding sites. Interestingly, Mist1 gene expression is induced during ER stress by XBP1, but as ER stress subsides, MIST1 serves as a feedback inhibitor, directly binding the Xbp1 promoter and repressing Xbp1 transcript production. Together, our findings provide a new paradigm for XBP1-dependent UPR regulation and position MIST1 as a potential biotherapeutic for numerous human diseases.

11.
Mol Cell Biol ; 33(16): 3166-79, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23754747

RESUMO

The lineage-specific basic helix-loop-helix transcription factor Ptf1a is a critical driver for development of both the pancreas and nervous system. How one transcription factor controls diverse programs of gene expression is a fundamental question in developmental biology. To uncover molecular strategies for the program-specific functions of Ptf1a, we identified bound genomic regions in vivo during development of both tissues. Most regions bound by Ptf1a are specific to each tissue, lie near genes needed for proper formation of each tissue, and coincide with regions of open chromatin. The specificity of Ptf1a binding is encoded in the DNA surrounding the Ptf1a-bound sites, because these regions are sufficient to direct tissue-restricted reporter expression in transgenic mice. Fox and Sox factors were identified as potential lineage-specific modifiers of Ptf1a binding, since binding motifs for these factors are enriched in Ptf1a-bound regions in pancreas and neural tube, respectively. Of the Fox factors expressed during pancreatic development, Foxa2 plays a major role. Indeed, Ptf1a and Foxa2 colocalize in embryonic pancreatic chromatin and can act synergistically in cell transfection assays. Together, these findings indicate that lineage-specific chromatin landscapes likely constrain the DNA binding of Ptf1a, and they identify Fox and Sox gene families as part of this process.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Tubo Neural/embriologia , Pâncreas/embriologia , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Cromatina/genética , Sequência Consenso , DNA/genética , DNA/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos , Camundongos Transgênicos , Tubo Neural/metabolismo , Pâncreas/metabolismo , Ligação Proteica , Fatores de Transcrição SOXB1/metabolismo
12.
Diabetes ; 58(1): 185-93, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18984737

RESUMO

OBJECTIVE: The activation of beta-cell genes, particularly of those encoding preproinsulin, requires an appropriate euchromatin (or "open") DNA template characterized by hypermethylation of Lys4 of histone H3. We hypothesized that this modification is maintained in islet beta-cells by the action of the histone methyltransferase Set7/9. RESEARCH DESIGN AND METHODS: To identify the role of Set7/9, we characterized its expression pattern and gene regulation and studied its function using RNA interference in both cell lines and primary mouse islets. RESULTS: Within the pancreas, Set7/9 protein shows striking specificity for islet cells, including alpha- and beta-cells, as well as occasional cells within ducts. Consistent with these findings, the Set7/9 gene promoter contained an islet-specific enhancer located between -5,768 and -6,030 base pairs (relative to the transcriptional start site) that exhibited Pdx1-responsive activation in beta-cells. To study Set7/9 function, we depleted insulinoma cells and primary mouse islets of Set7/9 protein using siRNA. Following siRNA treatment, we observed striking repression of genes involved in glucose-stimulated insulin secretion, including Ins1/2, Glut2, and MafA. These changes in transcription were accompanied by loss of dimethylated H3 Lys4 and RNA polymerase II recruitment, particularly at the Ins1/2 and Glut2 genes. Consistent with these data, depletion of Set7/9 in islets led to defects in glucose-stimulated Ca(2+) mobilization and insulin secretion. CONCLUSIONS: We conclude that Set7/9 is required for normal beta-cell function, likely through the maintenance of euchromatin structure at genes necessary for glucose-stimulated insulin secretion.


Assuntos
Eucromatina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas Metiltransferases/genética , Transcrição Gênica/genética , Animais , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Immunoblotting , Imuno-Histoquímica , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Proteínas Metiltransferases/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Mol Cell Biol ; 29(8): 2053-67, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19237535

RESUMO

The nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-gamma) is an important target in diabetes therapy, but its direct role, if any, in the restoration of islet function has remained controversial. To identify potential molecular mechanisms of PPAR-gamma in the islet, we treated diabetic or glucose-intolerant mice with the PPAR-gamma agonist pioglitazone or with a control. Treated mice exhibited significantly improved glycemic control, corresponding to increased serum insulin and enhanced glucose-stimulated insulin release and Ca(2+) responses from isolated islets in vitro. This improved islet function was at least partially attributed to significant upregulation of the islet genes Irs1, SERCA, Ins1/2, and Glut2 in treated animals. The restoration of the Ins1/2 and Glut2 genes corresponded to a two- to threefold increase in the euchromatin marker histone H3 dimethyl-Lys4 at their respective promoters and was coincident with increased nuclear occupancy of the islet methyltransferase Set7/9. Analysis of diabetic islets in vitro suggested that these effects resulting from the presence of the PPAR-gamma agonist may be secondary to improvements in endoplasmic reticulum stress. Consistent with this possibility, incubation of thapsigargin-treated INS-1 beta cells with the PPAR-gamma agonist resulted in the reduction of endoplasmic reticulum stress and restoration of Pdx1 protein levels and Set7/9 nuclear occupancy. We conclude that PPAR-gamma agonists exert a direct effect in diabetic islets to reduce endoplasmic reticulum stress and enhance Pdx1 levels, leading to favorable alterations of the islet gene chromatin architecture.


Assuntos
Retículo Endoplasmático/patologia , Eucromatina/ultraestrutura , Proteínas de Homeodomínio/metabolismo , Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/fisiopatologia , PPAR gama/fisiologia , Transativadores/metabolismo , Animais , Glicemia , Transportador de Glucose Tipo 2/genética , Proteínas de Homeodomínio/análise , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Camundongos , Camundongos Endogâmicos NOD , PPAR gama/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Transativadores/análise , Regulação para Cima/genética
14.
Mol Genet Metab ; 92(1-2): 43-55, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17659992

RESUMO

Emerging evidence over the past decade indicates a central role for transcription factors in the embryonic development of pancreatic islets and the consequent maintenance of normal glucose homeostasis. Pancreatic and duodenal homeobox 1 (Pdx1) is the best studied and perhaps most important of these factors. Whereas deletion or inactivating mutations of the Pdx1 gene causes whole pancreas agenesis in both mice and humans, even haploinsufficiency of the gene or alterations in its expression in mature islet cells causes substantial impairments in glucose tolerance and the development of a late-onset form of diabetes known as maturity onset diabetes of the young. The study of Pdx1 has revealed crucial phenotypic interrelationships of the varied cell types within the pancreas, particularly as these impinge upon cellular differentiation in the embryo and neogenesis and regeneration in the adult. In this review, we describe the actions of Pdx1 in the developing and mature pancreas and attempt to unify these actions with its known roles in modulating transcriptional complex formation and chromatin structure at the molecular genetic level.


Assuntos
Glucose/metabolismo , Proteínas de Homeodomínio/fisiologia , Homeostase , Ilhotas Pancreáticas/crescimento & desenvolvimento , Transativadores/fisiologia , Animais , Humanos , Ilhotas Pancreáticas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA