Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 35(10): e21938, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34547143

RESUMO

Uterine glands are essential for the establishment of pregnancy and have critical roles in endometrial receptivity to blastocyst implantation, stromal cell decidualization, and placentation. Uterine gland dysfunction is considered a major contributing factor to pregnancy loss, however our understanding of how glands impact embryo survival and stromal cell decidualization is incomplete. Forkhead box A2 (FOXA2) is expressed only in the glandular epithelium and regulates its development and function. Mice with a conditional deletion of FOXA2 in the uterus are infertile due to defective embryo implantation arising from a lack of leukemia inhibitory factor (LIF), a critical factor of uterine gland origin. Here, a glandless FOXA2-deficient mouse model, coupled with LIF repletion to rescue the implantation defect, was used to investigate the roles of uterine glands in embryo survival and decidualization. Studies found that embryo survival and decidualization were compromised in glandless FOXA2-deficient mice on gestational day 6.5, resulting in abrupt pregnancy loss by day 7.5. These findings strongly support the hypothesis that uterine glands secrete factors other than LIF that impact embryo survival and stromal cell decidualization for pregnancy success.


Assuntos
Decídua/metabolismo , Perda do Embrião , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Células Estromais/metabolismo , Útero/metabolismo , Animais , Decídua/imunologia , Perda do Embrião/imunologia , Embrião de Mamíferos/imunologia , Desenvolvimento Embrionário/imunologia , Feminino , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator Inibidor de Leucemia , Camundongos , Gravidez , Resultado da Gravidez , Células Estromais/imunologia , Transcriptoma , Útero/imunologia
2.
Cell Death Dis ; 12(1): 103, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33473118

RESUMO

FOXA2 has been identified as an essential factor for pancreas development and emerging evidence supports an association between FOXA2 and diabetes. Although the role of FOXA2 during pancreatic development is well-studied in animal models, its role during human islet cell development remains unclear. Here, we generated induced pluripotent stem cells (iPSCs) from a patient with FOXA2 haploinsufficiency (FOXA2+/- iPSCs) followed by beta-cell differentiation to understand the role of FOXA2 during pancreatic beta-cell development. Our results showed that FOXA2 haploinsufficiency resulted in aberrant expression of genes essential for the differentiation and proper functioning of beta cells. At pancreatic progenitor (PP2) and endocrine progenitor (EPs) stages, transcriptome analysis showed downregulation in genes associated with pancreatic development and diabetes and upregulation in genes associated with nervous system development and WNT signaling pathway. Knockout of FOXA2 in control iPSCs (FOXA2-/- iPSCs) led to severe phenotypes in EPs and beta-cell stages. The expression of NGN3 and its downstream targets at EPs as well as INSUILIN and GLUCAGON at the beta-cell stage, were almost absent in the cells derived from FOXA2-/- iPSCs. These findings indicate that FOXA2 is crucial for human pancreatic endocrine development and its defect may lead to diabetes based on FOXA2 dosage.


Assuntos
Diabetes Mellitus/genética , Fator 3-beta Nuclear de Hepatócito/deficiência , Células Secretoras de Insulina/metabolismo , Pâncreas/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Transfecção
3.
Aging (Albany NY) ; 11(21): 9280-9294, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31689237

RESUMO

BACKGROUND: Intrahepatic cholangiocarcinoma (ICC) is characterized by a highly aggressive nature and a dismal outcome. FOXA2 is an archetypal transcription factor involved in cholangiocyte proliferation. RESULTS: FOXA2 expression was negatively correlated with tumor stage (p = 0.024). Univariate and multivariate analyses showed that low FoxA2 expression was associated with tumor relapse and survival. At 20 weeks after TAA administration, FoxA2-/- mice displayed significant manifestations of neoplasia, while WT mice did not.RNA sequencing analysis showed that the expression of genes in the MAPK signaling pathway was significantly higher in FoxA2-/- mice. IHC and Western blot results showed that p-ERK1/2, CREB1 and RAS were highly expressed in FoxA2-/- mice. Furthermore, using in vitro experiments with siRNA, we found that low expression of FoxA2 could exacerbate the metastatic potential of ICC. The expression of p-ERK1/2 and RAS, which are key mediators of the MAPK signaling pathway, was significantly increased. CONCLUSION: Low FOXA2 expression negatively affected the prognosis of patients with ICC. Loss of FoxA2 expression could promote intrahepatic bile duct neoplasia partly via activation of the MAPK signaling pathway. MATERIALS AND METHODS: In all, the data of 85 patients with ICC were retrospectively collected and analyzed. TAA was used to induce ICC in FoxA2-/- mice and WT mice. RNA-sequencing analysis was used to identify the expression of different genes.


Assuntos
Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Fator 3-beta Nuclear de Hepatócito/deficiência , Sistema de Sinalização das MAP Quinases , Adulto , Idoso , Animais , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Proliferação de Células , Transformação Celular Neoplásica , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Progressão da Doença , Feminino , Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/genética , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Tioacetamida/toxicidade
4.
J Autoimmun ; 93: 131-138, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30061015

RESUMO

The Foxa1 and Foxa2 transcription factors are essential for mouse development. Here we show that they are expressed in thymic epithelial cells (TEC) where they regulate TEC development and function, with important consequences for T-cell development. TEC are essential for T-cell differentiation, lineage decisions and repertoire selection. Conditional deletion of Foxa1 and Foxa2 from murine TEC led to a smaller thymus with a greater proportion of TEC and a greater ratio of medullary to cortical TEC. Cell-surface MHCI expression was increased on cortical TEC in the conditional Foxa1Foxa2 knockout thymus, and MHCII expression was reduced on both cortical and medullary TEC populations. These changes in TEC differentiation and MHC expression led to a significant reduction in thymocyte numbers, reduced positive selection of CD4+CD8+ cells to the CD4 lineage, and increased CD8 cell differentiation. Conditional deletion of Foxa1 and Foxa2 from TEC also caused an increase in the medullary TEC population, and increased expression of Aire, but lower cell surface MHCII expression on Aire-expressing mTEC, and increased production of regulatory T-cells. Thus, Foxa1 and Foxa2 in TEC promote positive selection of CD4SP T-cells and modulate regulatory T-cell production and activity, of importance to autoimmunity.


Assuntos
Células Epiteliais/imunologia , Fator 3-alfa Nuclear de Hepatócito/imunologia , Fator 3-beta Nuclear de Hepatócito/imunologia , Linfócitos T Reguladores/imunologia , Timócitos/imunologia , Timo/imunologia , Animais , Autoimunidade , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Células Epiteliais/citologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/deficiência , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Ativação Linfocitária , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho do Órgão , Transdução de Sinais , Linfócitos T Reguladores/citologia , Timócitos/citologia , Timo/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteína AIRE
5.
Am J Respir Cell Mol Biol ; 49(6): 960-70, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23822876

RESUMO

Foxa2 is a member of the Forkhead family of nuclear transcription factors that is highly expressed in respiratory epithelial cells of the developing and mature lung. Foxa2 is required for normal airway epithelial differentiation, and its deletion causes goblet-cell metaplasia and Th2-mediated pulmonary inflammation during postnatal development. Foxa2 expression is inhibited during aeroallergen sensitization and after stimulation with Th2 cytokines, when its loss is associated with goblet-cell metaplasia. Mechanisms by which Foxa2 controls airway epithelial differentiation and Th2 immunity are incompletely known. During the first 2 weeks after birth, the loss of Foxa2 increases the production of leukotrienes (LTs) and Th2 cytokines in the lungs of Foxa2 gene-targeted mice. Foxa2 expression inhibited 15-lipoxygenase (Alox15) and increased Alox5 transcription, each encoding key lipoxygenases associated with asthma. The inhibition of the cysteinyl LT (CysLT) signaling pathway by montelukast inhibited IL-4, IL-5, eotaxin-2, and regulated upon activation normal T cell expressed and presumably secreted expression in the developing lungs of Foxa2 gene-targeted mice. Montelukast inhibited the expression of genes regulating mucus metaplasia, including Spdef, Muc5ac, Foxa3, and Arg2. Foxa2 plays a cell-autonomous role in the respiratory epithelium, and is required for the suppression of Th2 immunity and mucus metaplasia in the developing lung in a process determined in part by its regulation of the CysLT pathway.


Assuntos
Fator 3-beta Nuclear de Hepatócito/imunologia , Leucotrienos/imunologia , Pneumonia/imunologia , Células Th2/imunologia , Acetatos/farmacologia , Animais , Araquidonato 12-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/genética , Ciclopropanos , Cisteína/imunologia , Modelos Animais de Doenças , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/imunologia , Células Caliciformes/patologia , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Mediadores da Inflamação/imunologia , Antagonistas de Leucotrienos/farmacologia , Metaplasia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pneumonia/etiologia , Pneumonia/patologia , Quinolinas/farmacologia , Transdução de Sinais/imunologia , Sulfetos
6.
J Neurosci ; 33(18): 8022-34, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23637192

RESUMO

The maintained expression of transcription factors throughout the development of mesodiencephalic dopaminergic (mDA) neurons suggests multiple roles at various stages in development. Two members of the forkhead/winged helix transcription factor family, Foxa1 and Foxa2, have been recently shown to have an important influence in the early development of mDA neurons. Here we present data demonstrating that these genes are also involved in the later maintenance of the mDA system. We conditionally removed both genes in postmitotic mDA neurons using the dopamine transporter-cre mouse. Deletion of both Foxa1 and Foxa2 resulted in a significant reduction in the number of tyrosine hydroxylase (TH)-positive mDA neurons. The decrease was predominantly observed in the substantia nigra region of the mDA system, which led to a loss of TH+ fibers innervating the striatum. Further analysis demonstrated that the reduction in the number of TH+ cells in the mutant mice was not due to apoptosis or cell-fate change. Using reporter mouse lines, we found that the mDA neurons were still present in the ventral midbrain, but that they had lost much of their dopaminergic phenotype. The majority of these neurons remained in the ventral mesencephalon until at least 18 months of age. Chromatin immunoprecipitation suggested that the loss of the mDA phenotype is due to a reduction in the binding of the nuclear orphan receptor, Nurr-1 to the promoter region of TH. These results extend previous findings and demonstrate a later role for Foxa genes in regulating the maintenance of dopaminergic phenotype in mDA neurons.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Mesencéfalo/citologia , Análise de Variância , Animais , Proteínas de Bactérias/metabolismo , Morte Celular/genética , Diferenciação Celular/genética , Movimento Celular/genética , Tamanho Celular , Imunoprecipitação da Cromatina , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Embrião de Mamíferos , Marcha/genética , Fator 3-alfa Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/deficiência , Marcação In Situ das Extremidades Cortadas , Proteínas Luminescentes/metabolismo , Mesencéfalo/embriologia , Camundongos , Camundongos Transgênicos , Mutação/genética , Fibras Nervosas/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica/genética , Proteínas/genética , Proteínas/metabolismo , RNA não Traduzido , Tirosina 3-Mono-Oxigenase/metabolismo
7.
Dev Cell ; 22(5): 927-39, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22595668

RESUMO

During endochondral ossification, small, immature chondrocytes enlarge to form hypertrophic chondrocytes, which express collagen X. In this work, we demonstrate that FoxA factors are induced during chondrogenesis, bind to conserved binding sites in the collagen X enhancer, and can promote the expression of a collagen X-luciferase reporter in both chondrocytes and fibroblasts. In addition, we demonstrate by both gain- and loss-of-function analyses that FoxA factors play a crucial role in driving the expression of both endogenous collagen X and other hypertrophic chondrocyte-specific genes. Mice engineered to lack expression of both FoxA2 and FoxA3 in their chondrocytes display defects in chondrocyte hypertrophy, alkaline phosphatase expression, and mineralization in their sternebrae and, in addition, exhibit postnatal dwarfism that is coupled to significantly decreased expression of both collagen X and MMP13 in their growth plates. Our findings indicate that FoxA family members are crucial regulators of the hypertrophic chondrocyte differentiation program.


Assuntos
Crescimento Celular , Condrócitos/metabolismo , Condrogênese/genética , Colágeno Tipo X/metabolismo , Nanismo/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 3-gama Nuclear de Hepatócito/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular/genética , Células Cultivadas , Embrião de Galinha , Condrócitos/citologia , Colágeno Tipo X/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Nanismo/embriologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Genes Reporter , Lâmina de Crescimento/metabolismo , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-gama Nuclear de Hepatócito/deficiência , Fator 3-gama Nuclear de Hepatócito/genética , Metaloproteinase 13 da Matriz/genética , Ossos do Metatarso/citologia , Ossos do Metatarso/metabolismo , Camundongos , Camundongos Mutantes , Fatores de Regulação Miogênica/metabolismo , Proteína Smad1/metabolismo
8.
Circ Res ; 110(1): 34-46, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22076636

RESUMO

RATIONALE: Positive signals, such as vascular endothelial growth factor, direct endothelial cells (ECs) to specific locations during blood vessel formation. Less is known about repulsive signal contribution to shaping vessels. Recently, "neuronal guidance cues" have been shown to influence EC behavior, particularly in directing sprouting angiogenesis by repelling ECs. However, their role during de novo blood vessel formation remains unexplored. OBJECTIVE: To identify signals that guide and pattern the first mammalian blood vessels. METHODS AND RESULTS: Using genetic mouse models, we show that blood vessels are sculpted through the generation of stereotyped avascular zones by EC-repulsive cues. We demonstrate that Semaphorin3E (Sema3E) is a key factor that shapes the paired dorsal aortae in mouse, as sema3E(-/-) embryos develop an abnormally branched aortic plexus with a markedly narrowed avascular midline. In vitro cultures and avian grafting experiments show strong repulsion of ECs by Sema3E-expressing cells. We further identify the mouse notochord as a rich source of multiple redundant neuronal guidance cues. Mouse embryos that lack notochords fail to form cohesive aortic vessels because of loss of the avascular midline, yet maintain lateral avascular zones. We demonstrate that lateral avascular zones are directly generated by the lateral plate mesoderm, a critical source of Sema3E. CONCLUSIONS: These findings demonstrate that Sema3E-generated avascular zones are critical regulators of mammalian cardiovascular patterning and are the first to identify a repulsive role for the lateral plate mesoderm. Integration of multiple, and in some cases redundant, repulsive cues from various tissues is critical to patterning the first embryonic blood vessels.


Assuntos
Vasos Sanguíneos/embriologia , Embrião de Mamíferos/irrigação sanguínea , Endotélio Vascular/embriologia , Glicoproteínas/fisiologia , Proteínas de Membrana/fisiologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Animais , Aorta/citologia , Aorta/embriologia , Vasos Sanguíneos/citologia , Células Cultivadas , Proteínas do Citoesqueleto , Endotélio Vascular/citologia , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Glicoproteínas/deficiência , Glicoproteínas/genética , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/fisiologia , Técnicas In Vitro , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Mesoderma/citologia , Mesoderma/embriologia , Camundongos , Camundongos Knockout , Modelos Animais , Notocorda/citologia , Notocorda/embriologia , Semaforinas
9.
Dev Biol ; 337(2): 471-81, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19896480

RESUMO

The definitive endoderm forms during gastrulation and is rapidly transformed into the gut tube which is divided along the anterior-posterior axis into the foregut, midgut, and hindgut. Lineage tracing and genetic analysis have examined the origin of the definitive endoderm during gastrulation and demonstrated that the majority of definitive endoderm arises at the anterior end of the primitive streak (APS). Foxh1 and Foxa2 have been shown to play a role in specification of the APS and definitive endoderm. However, prior studies have focused on the role of these factors in specification of foregut definitive endoderm, while their role in the specification of midgut and hindgut definitive endoderm is less understood. Furthermore, previous analyses of these mutants have utilized definitive endoderm markers that are restricted to the anterior endoderm, expressed in extraembryonic endoderm, or present in other germ layers. Here, we characterized the expression of several novel definitive and visceral endoderm markers in Foxh1 and Foxa2 null embryos. In accordance with previous studies, we observed a deficiency of foregut definitive endoderm resulting in incorporation of visceral endoderm into the foregut. Interestingly, this analysis revealed that formation of midgut and hindgut definitive endoderm is unaffected by loss of Foxh1 or Foxa2. This finding represents a significant insight into specification and regionalization of mouse definitive endoderm.


Assuntos
Sistema Digestório/embriologia , Endoderma/embriologia , Endoderma/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Animais , Diferenciação Celular , Sistema Digestório/citologia , Sistema Digestório/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Endoderma/citologia , Fatores de Transcrição Forkhead/deficiência , Fator 3-beta Nuclear de Hepatócito/deficiência , Peroxidase do Rábano Silvestre/metabolismo , Camundongos
10.
Gastroenterology ; 137(6): 2052-62, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19737569

RESUMO

BACKGROUND & AIMS: The winged helix transcription factors Foxa1 and Foxa2 are expressed in all epithelia of the gastrointestinal tract from its embryonic origin into adulthood. In vitro studies have shown that Foxa1/a2 can transactivate the promoters of Mucin 2 (Muc2), which is expressed in goblet cells, and of preproglucagon, which is expressed in enteroendocrine cells. These findings suggest Foxa1/a2 as critical factors in the differentiation of gut epithelial cells. METHODS: Mice with intestine-specific simultaneous deletion of Foxa1 and Foxa2 were derived using the Cre-loxP system and analyzed using histologic and molecular means. RESULTS: Both Foxa1 and Foxa2 were deleted successfully in the epithelia of the small intestine and colon using Villin-Cre mice. Immunohistochemical staining showed that Foxa1/a2 mutants lack glucagon-like peptide-1- and peptide-2-expressing cells (L-cells), and have reduced numbers of somatostatin (D-cells) and peptide YY-expressing cells (L-cells). Preproglucagon, somatostatin, and peptide YY messenger RNA (mRNA) levels also were reduced significantly in Foxa1/a2 mutants. Thus, Foxa1 and Foxa2 are essential regulators of these enteroendocrine lineages in vivo. The mRNA levels of transcription factors Islet-1 and Pax6 were reduced significantly in the small intestine, showing that Foxa1 and Foxa2 impact on a transcription factor network in the enteroendocrine lineage. In addition, deletion of Foxa1/a2 caused a reduction in goblet cell number with altered expression of the secretory mucins Muc2, Mucin5b, Mucin5ac, and Mucin 6. CONCLUSIONS: The winged helix factors Foxa1 and Foxa2 are essential members of the transcription factor network that govern secretory cell differentiation in the mammalian gastrointestinal tract.


Assuntos
Diferenciação Celular , Células Enteroendócrinas/metabolismo , Células Caliciformes/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Intestino Delgado/metabolismo , Células Secretoras de Somatostatina/metabolismo , Animais , Células Enteroendócrinas/patologia , Proteínas do Olho/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Células Caliciformes/patologia , Fator 3-alfa Nuclear de Hepatócito/deficiência , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Intestino Delgado/patologia , Proteínas com Homeodomínio LIM , Masculino , Camundongos , Camundongos Knockout , Mucina-5AC/metabolismo , Mucina-2/metabolismo , Mucina-5B/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Peptídeo YY/metabolismo , Proglucagon/metabolismo , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Somatostatina/metabolismo , Células Secretoras de Somatostatina/patologia , Fatores de Transcrição
11.
J Clin Invest ; 119(6): 1537-45, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19436110

RESUMO

The forkhead box proteins A1 and A2 (Foxa1 and Foxa2) are transcription factors with critical roles in establishing the developmental competence of the foregut endoderm and in initiating liver specification. Using conditional gene ablation during a later phase of liver development, we show here that deletion of both Foxa1 and Foxa2 (Foxa1/2) in the embryonic liver caused hyperplasia of the biliary tree. Abnormal bile duct formation in Foxa1/2-deficient liver was due, at least in part, to activation of IL-6 expression, a proliferative signal for cholangiocytes. The glucocorticoid receptor is a negative regulator of IL-6 transcription; in the absence of Foxa1/2, the glucocorticoid receptor failed to bind to the IL-6 promoter, causing enhanced IL-6 expression. Thus, after liver specification, Foxa1/2 are required for normal bile duct development through prevention of excess cholangiocyte proliferation. Our data suggest that Foxa1/2 function as terminators of bile duct expansion in the adult liver through inhibition of IL-6 expression.


Assuntos
Ductos Biliares/crescimento & desenvolvimento , Ductos Biliares/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Animais , Doenças dos Ductos Biliares/genética , Doenças dos Ductos Biliares/metabolismo , Doenças dos Ductos Biliares/patologia , Ductos Biliares/citologia , Diferenciação Celular , Proliferação de Células , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Fator 3-alfa Nuclear de Hepatócito/deficiência , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Hepatócitos/citologia , Hepatócitos/metabolismo , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/patologia , Interleucina-6/genética , Interleucina-6/metabolismo , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Receptores de Glucocorticoides/metabolismo
12.
PLoS Biol ; 5(12): e325, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18076286

RESUMO

Parkinson disease affects more than 1% of the population over 60 y old. The dominant models for Parkinson disease are based on the use of chemical toxins to kill dopamine neurons, but do not address the risk factors that normally increase with age. Forkhead transcription factors are critical regulators of survival and longevity. The forkhead transcription factor, foxa2, is specifically expressed in adult dopamine neurons and their precursors in the medial floor plate. Gain- and loss-of-function experiments show this gene, foxa2, is required to generate dopamine neurons during fetal development and from embryonic stem cells. Mice carrying only one copy of the foxa2 gene show abnormalities in motor behavior in old age and an associated progressive loss of dopamine neurons. Manipulating forkhead function may regulate both the birth of dopamine neurons and their spontaneous death, two major goals of regenerative medicine.


Assuntos
Envelhecimento/fisiologia , Dopamina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Parto/metabolismo , Animais , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Camundongos , Camundongos Knockout , Degeneração Neural/genética , Parto/genética
13.
Cell Metab ; 6(4): 267-79, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17908556

RESUMO

The winged-helix transcription factor Foxa2 regulates Pdx1 gene expression and fetal endocrine pancreas development. We show here by inducible gene ablation that Foxa2 inactivation in mature beta cells induces hyperinsulinemic hypoglycemia in Foxa2(loxP/loxP),Pdx1-CreERT2 adult mice. Mutant beta cells exhibited a markedly increased pool of docked insulin granules, some of which were engaged in sequential or compound exocytosis, consistent with increased first-phase glucose-stimulated insulin secretion. Expression of multiple genes involved in vesicular trafficking, membrane targeting, and fuel-secretion pathways is dependent on Foxa2. In addition, impaired cytosolic Ca(2+) oscillations and elevated intracellular cyclic AMP production accompanied this secretory defect and were likely contributors to the sensitization of the exocytotic machinery. Thus, in the absence of Foxa2, alterations in intracellular second-messenger signaling redistribute the insulin granules into the readily releasable pool. We conclude that Foxa2 is required for both fetal pancreas development and the function of mature beta cells.


Assuntos
Fator 3-beta Nuclear de Hepatócito/fisiologia , Hiperinsulinismo/genética , Hipoglicemia/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Sinalização do Cálcio/genética , Deleção de Genes , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Secreção de Insulina , Células Secretoras de Insulina/ultraestrutura , Camundongos , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura
14.
Dev Biol ; 296(2): 458-75, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16839541

RESUMO

A critical question in mammalian development is how the forebrain is established. In amphibians, bone morphogenetic protein (BMP) antagonism emanating from the gastrula organizer is key. Roles of BMP antagonism and the organizer in mammals remain unclear. Anterior visceral endoderm (AVE) promotes early mouse head development, but its function is controversial. Here, we explore the timing and regulation of forebrain establishment in the mouse. Forebrain specification requires tissue interaction through the late streak stage of gastrulation. Foxa2(-/-) embryos lack both the organizer and its BMP antagonists, yet about 25% show weak forebrain gene expression. A similar percentage shows ectopic AVE gene expression distally. The distal VE may thus be a source of forebrain promoting signals in these embryos. In wild-type ectoderm explants, AVE promoted forebrain specification, while anterior mesendoderm provided maintenance signals. Embryological and molecular data suggest that the AVE is a source of active BMP antagonism in vivo. In prespecification ectoderm explants, exogenous BMP antagonists triggered forebrain gene expression and inhibited posterior gene expression. Conversely, BMP inhibited forebrain gene expression, an effect that could be antagonized by anterior mesendoderm, and promoted expression of some posterior genes. These results lead to a model in which BMP antagonism supplied by exogenous tissues promotes forebrain establishment and maintenance in the murine ectoderm.


Assuntos
Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Organizadores Embrionários/fisiologia , Prosencéfalo/embriologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Ectoderma/metabolismo , Ectoderma/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/deficiência , Glicoproteínas/genética , Glicoproteínas/fisiologia , Fator 3-beta Nuclear de Hepatócito/deficiência , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Mutação , Organizadores Embrionários/embriologia , Prosencéfalo/metabolismo , Transdução de Sinais/genética , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...