Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genes Dev ; 37(11-12): 451-453, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37399332

RESUMO

The homeodomain transcription factor (TF) Nkx2.2 governs crucial cell fate decisions in several developing organs, including the central nervous system (CNS), pancreas, and intestine. How Nkx2.2 regulates unique targets in these different systems to impact their individual transcriptional programs remains unclear. In this issue of Genes & Development Abarinov and colleagues (pp. 490-504) generated and analyzed mice in which the Nkx2.2 SD is mutated and found that the SD is required for normal pancreatic islet differentiation but dispensable for most aspects of neuronal differentiation.


Assuntos
Proteínas de Homeodomínio , Ilhotas Pancreáticas , Camundongos , Animais , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Peixe-Zebra/genética , Ilhotas Pancreáticas/metabolismo , Diferenciação Celular/genética , Neurônios/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
2.
Development ; 147(12)2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32467237

RESUMO

Thymus function depends on the epithelial compartment of the thymic stroma. Cortical thymic epithelial cells (cTECs) regulate T cell lineage commitment and positive selection, while medullary (m) TECs impose central tolerance on the T cell repertoire. During thymus organogenesis, these functionally distinct sub-lineages are thought to arise from a common thymic epithelial progenitor cell (TEPC). However, the mechanisms controlling cTEC and mTEC production from the common TEPC are not understood. Here, we show that emergence of the earliest mTEC lineage-restricted progenitors requires active NOTCH signaling in progenitor TEC and that, once specified, further mTEC development is NOTCH independent. In addition, we demonstrate that persistent NOTCH activity favors maintenance of undifferentiated TEPCs at the expense of cTEC differentiation. Finally, we uncover a cross-regulatory relationship between NOTCH and FOXN1, a master regulator of TEC differentiation. These data establish NOTCH as a potent regulator of TEPC and mTEC fate during fetal thymus development, and are thus of high relevance to strategies aimed at generating/regenerating functional thymic tissue in vitro and in vivo.


Assuntos
Desenvolvimento Embrionário/genética , Receptores Notch/metabolismo , Timo/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Mutação com Ganho de Função , Regulação da Expressão Gênica no Desenvolvimento , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Organogênese , Receptores Notch/genética , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Timo/citologia , Timo/crescimento & desenvolvimento
3.
Semin Cell Dev Biol ; 92: 77-88, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30142440

RESUMO

The pancreas is a compound gland comprised of both exocrine acinar and duct cells as well as endocrine islet cells. Most notable amongst the latter are the insulin-synthesizing ß-cells, loss or dysfunction of which manifests in diabetes mellitus. All exocrine and endocrine cells derive from multipotent pancreatic progenitor cells arising from the primitive gut epithelium via inductive interactions with adjacent mesodermal tissues. Research in the last two decades has revealed the identity of many of these extrinsic cues and they include signaling molecules used in many other developmental contexts such as retinoic acid, fibroblast growth factors, and members of the TGF-ß superfamily. As important as these inductive cues is the absence of other signaling molecules such as hedgehog family members. Much has been learned about the interactions of extrinsic factors with fate regulators intrinsic to the pancreatic endoderm. This new knowledge has had tremendous impact on the development of directed differentiation protocols for converting pluripotent stem cells to ß-cells in vitro.


Assuntos
Mesoderma/embriologia , Pâncreas/embriologia , Animais , Humanos , Camundongos
4.
Nature ; 564(7734): 114-118, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30487608

RESUMO

The pancreas originates from two epithelial evaginations of the foregut, which consist of multipotent epithelial progenitors that organize into a complex tubular epithelial network. The trunk domain of each epithelial branch consists of bipotent pancreatic progenitors (bi-PPs) that give rise to both duct and endocrine lineages, whereas the tips give rise to acinar cells1. Here we identify the extrinsic and intrinsic signalling mechanisms that coordinate the fate-determining transcriptional events underlying these lineage decisions1,2. Single-cell analysis of pancreatic bipotent pancreatic progenitors derived from human embryonic stem cells reveal that cell confinement is a prerequisite for endocrine specification, whereas spreading drives the progenitors towards a ductal fate. Mechanistic studies identify the interaction of extracellular matrix (ECM) with integrin α5 as the extracellular cue that cell-autonomously, via the F-actin-YAP1-Notch mechanosignalling axis, controls the fate of bipotent pancreatic progenitors. Whereas ECM-integrin α5 signalling promotes differentiation towards the duct lineage, endocrinogenesis is stimulated when this signalling cascade is disrupted. This cascade can be disrupted pharmacologically or genetically to convert bipotent pancreatic progenitors derived from human embryonic stem cells to hormone-producing islet cells. Our findings identify the cell-extrinsic and intrinsic mechanotransduction pathway that acts as gatekeeper in the fate decisions of bipotent pancreatic progenitors in the developing pancreas.


Assuntos
Diferenciação Celular , Linhagem da Célula , Integrinas/metabolismo , Organogênese , Pâncreas/citologia , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Linhagem da Célula/genética , Forma Celular , Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos/genética , Feminino , Fibronectinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Integrina alfa5beta1/metabolismo , Masculino , Camundongos , Proteínas Musculares/metabolismo , Pâncreas/embriologia , Pâncreas/metabolismo , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptor Notch1/genética , Análise de Célula Única , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição HES-1/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transgenes , Proteínas de Sinalização YAP
5.
Diabetes ; 67(1): 58-70, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28986398

RESUMO

Inhibition of notch signaling is known to induce differentiation of endocrine cells in zebrafish and mouse. After performing an unbiased in vivo screen of ∼2,200 small molecules in zebrafish, we identified an inhibitor of Cdk5 (roscovitine), which potentiated the formation of ß-cells along the intrapancreatic duct during concurrent inhibition of notch signaling. We confirmed and characterized the effect with a more selective Cdk5 inhibitor, (R)-DRF053, which specifically increased the number of duct-derived ß-cells without affecting their proliferation. By duct-specific overexpression of the endogenous Cdk5 inhibitors Cdk5rap1 or Cdkal1 (which previously have been linked to diabetes in genome-wide association studies), as well as deleting cdk5, we validated the role of chemical Cdk5 inhibition in ß-cell differentiation by genetic means. Moreover, the cdk5 mutant zebrafish displayed an increased number of ß-cells independently of inhibition of notch signaling, in both the basal state and during ß-cell regeneration. Importantly, the effect of Cdk5 inhibition to promote ß-cell formation was conserved in mouse embryonic pancreatic explants, adult mice with pancreatic ductal ligation injury, and human induced pluripotent stem (iPS) cells. Thus, we have revealed a previously unknown role of Cdk5 as an endogenous suppressor of ß-cell differentiation and thereby further highlighted its importance in diabetes.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ductos Pancreáticos/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Quinase 5 Dependente de Ciclina/genética , Estudo de Associação Genômica Ampla , Genótipo , Larva/citologia , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
6.
Mol Endocrinol ; 30(1): 133-43, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26649805

RESUMO

Diabetes is characterized by insulin insufficiency due to a relative paucity of functional ß-cell mass. Thus, strategies for increasing ß-cell mass in situ are sought-after for therapeutic purposes. Pregnancy is a physiological state capable of inducing robust ß-cell mass expansion, however, the mechanisms driving this expansion are not fully understood. Thus, the aim of this study was to characterize pregnancy-induced changes in the islet proteome at the peak of ß-cell proliferation in mice. Islets from pregnant and nonpregnant littermates were compared via 2 proteomic strategies. In vivo pulsed stable isotope labeling of amino acids in cell culture was used to monitor de novo protein synthesis during the first 14.5 days of pregnancy. In parallel, protein abundance was determined using ex vivo dimethyl labelling at gestational day 14.5. Comparison of the 2 datasets revealed 170 islet proteins to be up regulated as a response to pregnancy. These included several proteins, not previously associated with pregnancy-induced islet expansion, such as CLIC1, STMN1, MCM6, PPIB, NEDD4, and HLTF. Confirming the validity of our approach, we also identified proteins encoded by genes known to be associated with pregnancy-induced islet expansion, such as CHGB, IGFBP5, MATN2, EHHADH, IVD, and BMP1. Bioinformatic analyses demonstrated enrichment and activation of the biological functions: "protein synthesis" and "proliferation," and predicted the transcription factors HNF4α, MYC, MYCN, E2F1, NFE2L2, and HNF1α as upstream regulators of the observed expressional changes. As the first characterization of the islet-proteome during pregnancy, this study provides novel insight into the mechanisms involved in promoting pregnancy-induced ß-cell mass expansion and function.


Assuntos
Proliferação de Células/fisiologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteômica/métodos , Animais , Feminino , Camundongos , Gravidez
7.
Cell Rep ; 13(2): 326-36, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26440894

RESUMO

The generation of pancreas, liver, and intestine from a common pool of progenitors in the foregut endoderm requires the establishment of organ boundaries. How dorsal foregut progenitors activate pancreatic genes and evade the intestinal lineage choice remains unclear. Here, we identify Pdx1 and Sox9 as cooperative inducers of a gene regulatory network that distinguishes the pancreatic from the intestinal lineage. Genetic studies demonstrate dual and cooperative functions for Pdx1 and Sox9 in pancreatic lineage induction and repression of the intestinal lineage choice. Pdx1 and Sox9 bind to regulatory sequences near pancreatic and intestinal differentiation genes and jointly regulate their expression, revealing direct cooperative roles for Pdx1 and Sox9 in gene activation and repression. Our study identifies Pdx1 and Sox9 as important regulators of a transcription factor network that initiates pancreatic fate and sheds light on the gene regulatory circuitry that governs the development of distinct organs from multi-lineage-competent foregut progenitors.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias/metabolismo , Redes Reguladoras de Genes , Proteínas de Homeodomínio/genética , Intestinos/citologia , Fatores de Transcrição SOX9/genética , Transativadores/genética , Animais , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Pâncreas/citologia , Pâncreas/metabolismo , Fatores de Transcrição SOX9/metabolismo , Transativadores/metabolismo , Ativação Transcricional
8.
Development ; 142(13): 2291-303, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25995356

RESUMO

Notochord-derived Sonic Hedgehog (Shh) is essential for dorsoventral patterning of the overlying neural tube. Increasing concentration and duration of Shh signal induces progenitors to acquire progressively more ventral fates. We show that Notch signalling augments the response of neuroepithelial cells to Shh, leading to the induction of higher expression levels of the Shh target gene Ptch1 and subsequently induction of more ventral cell fates. Furthermore, we demonstrate that activated Notch1 leads to pronounced accumulation of Smoothened (Smo) within primary cilia and elevated levels of full-length Gli3. Finally, we show that Notch activity promotes longer primary cilia both in vitro and in vivo. Strikingly, these Notch-regulated effects are Shh independent. These data identify Notch signalling as a novel modulator of Shh signalling that acts mechanistically via regulation of ciliary localisation of key components of its transduction machinery.


Assuntos
Proteínas Aviárias/metabolismo , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Embrião de Galinha , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Neurônios Motores/metabolismo , Células NIH 3T3 , Proteínas do Tecido Nervoso/metabolismo , Placa Neural/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Tubo Neural/metabolismo , Notocorda/metabolismo , Receptores Notch/antagonistas & inibidores , Receptor Smoothened , Proteína Gli3 com Dedos de Zinco
9.
Rev Diabet Stud ; 11(1): 51-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25148367

RESUMO

Over the last decade, it has been discovered that the transcription factor Sox9 plays several critical roles in governing the development of the embryonic pancreas and the homeostasis of the mature organ. While analysis of pancreata from patients affected by the Sox9 haploinsufficiency syndrome campomelic dysplasia initially alluded to a functional role of Sox9 in pancreatic morphogenesis, transgenic mouse models have been instrumental in mechanistically dissecting such roles. Although initially defined as a marker and maintenance factor for pancreatic progenitors, Sox9 is now considered to fulfill additional indispensable functions during pancreogenesis and in the postnatal organ through its interactions with other transcription factors and signaling pathways such as Fgf and Notch. In addition to maintaining both multipotent and bipotent pancreatic progenitors, Sox9 is also required for initiating endocrine differentiation and maintaining pancreatic ductal identity, and it has recently been unveiled as a key player in the initiation of pancreatic cancer. These functions of Sox9 are discussed in this article, with special emphasis on the knowledge gained from various loss-of-function and lineage tracing mouse models. Also, current controversies regarding Sox9 function in healthy and injured adult pancreas and unanswered questions and avenues of future study are discussed.


Assuntos
Homeostase , Modelos Biológicos , Organogênese , Pâncreas/metabolismo , Fatores de Transcrição SOX9/metabolismo , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Pâncreas/citologia , Pâncreas/embriologia , Fatores de Transcrição SOX9/genética
11.
Development ; 139(18): 3363-72, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22874919

RESUMO

All mature pancreatic cell types arise from organ-specific multipotent progenitor cells. Although previous studies have identified cell-intrinsic and -extrinsic cues for progenitor cell expansion, it is unclear how these cues are integrated within the niche of the developing organ. Here, we present genetic evidence in mice that the transcription factor Sox9 forms the centerpiece of a gene regulatory network that is crucial for proper organ growth and maintenance of organ identity. We show that pancreatic progenitor-specific ablation of Sox9 during early pancreas development causes pancreas-to-liver cell fate conversion. Sox9 deficiency results in cell-autonomous loss of the fibroblast growth factor receptor (Fgfr) 2b, which is required for transducing mesenchymal Fgf10 signals. Likewise, Fgf10 is required to maintain expression of Sox9 and Fgfr2 in epithelial progenitors, showing that Sox9, Fgfr2 and Fgf10 form a feed-forward expression loop in the early pancreatic organ niche. Mirroring Sox9 deficiency, perturbation of Fgfr signaling in pancreatic explants or genetic inactivation of Fgf10 also result in hepatic cell fate conversion. Combined with previous findings that Fgfr2b or Fgf10 are necessary for pancreatic progenitor cell proliferation, our results demonstrate that organ fate commitment and progenitor cell expansion are coordinately controlled by the activity of a Sox9/Fgf10/Fgfr2b feed-forward loop in the pancreatic niche. This self-promoting Sox9/Fgf10/Fgfr2b loop may regulate cell identity and organ size in a broad spectrum of developmental and regenerative contexts.


Assuntos
Fator 10 de Crescimento de Fibroblastos/metabolismo , Fígado/embriologia , Fígado/metabolismo , Pâncreas/embriologia , Pâncreas/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição SOX9/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica , Fígado/citologia , Camundongos , Pâncreas/citologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Fatores de Transcrição SOX9/genética
12.
Development ; 139(14): 2488-99, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22675211

RESUMO

In the pancreas, Notch signaling is thought to prevent cell differentiation, thereby maintaining progenitors in an undifferentiated state. Here, we show that Notch renders progenitors competent to differentiate into ductal and endocrine cells by inducing activators of cell differentiation. Notch signaling promotes the expression of Sox9, which cell-autonomously activates the pro-endocrine gene Ngn3. However, at high Notch activity endocrine differentiation is blocked, as Notch also induces expression of the Ngn3 repressor Hes1. At the transition from high to intermediate Notch activity, only Sox9, but not Hes1, is maintained, thus de-repressing Ngn3 and initiating endocrine differentiation. In the absence of Sox9 activity, endocrine and ductal cells fail to differentiate, resulting in polycystic ducts devoid of primary cilia. Although Sox9 is required for Ngn3 induction, endocrine differentiation necessitates subsequent Sox9 downregulation and evasion from Notch activity via cell-autonomous repression of Sox9 by Ngn3. If high Notch levels are maintained, endocrine progenitors retain Sox9 and undergo ductal fate conversion. Taken together, our findings establish a novel role for Notch in initiating both ductal and endocrine development and reveal that Notch does not function in an on-off mode, but that a gradient of Notch activity produces distinct cellular states during pancreas development.


Assuntos
Pâncreas/citologia , Pâncreas/metabolismo , Receptores Notch/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Citometria de Fluxo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores Notch/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição HES-1
13.
PLoS One ; 6(8): e23131, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21829703

RESUMO

The HMG box transcription factor Sox9 plays a critical role in progenitor cell expansion during pancreas organogenesis and is required for proper endocrine cell development in the embryo. Based on in vitro studies it has been suggested that Sox9 controls expression of a network of important developmental regulators, including Tcf2/MODY5, Hnf6, and Foxa2, in pancreatic progenitor cells. Here, we sought to: 1) determine whether Sox9 regulates this transcriptional network in vivo and 2) investigate whether reduced Sox9 gene dosage leads to impaired glucose homeostasis in adult mice. Employing two genetic models of temporally-controlled Sox9 inactivation in pancreatic progenitor cells, we demonstrate that contrary to in vitro findings, Sox9 is not required for Tcf2, Hnf6, or Foxa2 expression in vivo. Moreover, our analysis revealed a novel role for Sox9 in maintaining the expression of Pdx1/MODY4, which is an important transcriptional regulator of beta-cell development. We further show that reduced beta-cell mass in Sox9-haploinsufficient mice leads to glucose intolerance during adulthood. Sox9-haploinsufficient mice displayed 50% reduced beta-cell mass at birth, which recovered partially via a compensatory increase in beta-cell proliferation early postnatally. Endocrine islets from mice with reduced Sox9 gene dosage exhibited normal glucose stimulated insulin secretion. Our findings show Sox9 plays an important role in endocrine development by maintaining Ngn3 and Pdx1 expression. Glucose intolerance in Sox9-haploinsufficient mice suggests that mutations in Sox9 could play a role in diabetes in humans.


Assuntos
Teste de Tolerância a Glucose , Haploinsuficiência , Fatores de Transcrição SOX9/fisiologia , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Primers do DNA , Gorduras na Dieta/administração & dosagem , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Reação em Cadeia da Polimerase , Fatores de Transcrição SOX9/genética , Transativadores/genética
15.
Development ; 138(4): 653-65, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266405

RESUMO

One major unresolved question in the field of pancreas biology is whether ductal cells have the ability to generate insulin-producing ß-cells. Conclusive examination of this question has been limited by the lack of appropriate tools to efficiently and specifically label ductal cells in vivo. We generated Sox9CreER(T2) mice, which, during adulthood, allow for labeling of an average of 70% of pancreatic ductal cells, including terminal duct/centroacinar cells. Fate-mapping studies of the Sox9(+) domain revealed endocrine and acinar cell neogenesis from Sox9(+) cells throughout embryogenesis. Very small numbers of non-ß endocrine cells continue to arise from Sox9(+) cells in early postnatal life, but no endocrine or acinar cell neogenesis from Sox9(+) cells occurs during adulthood. In the adult pancreas, pancreatic injury by partial duct ligation (PDL) has been suggested to induce ß-cell regeneration from a transient Ngn3(+) endocrine progenitor cell population. Here, we identify ductal cells as a cell of origin for PDL-induced Ngn3(+) cells, but fail to observe ß-cell neogenesis from duct-derived cells. Therefore, although PDL leads to activation of Ngn3 expression in ducts, PDL does not induce appropriate cues to allow for completion of the entire ß-cell neogenesis program. In conclusion, although endocrine cells arise from the Sox9(+) ductal domain throughout embryogenesis and the early postnatal period, Sox9(+) ductal cells of the adult pancreas no longer give rise to endocrine cells under both normal conditions and in response to PDL.


Assuntos
Envelhecimento , Diferenciação Celular , Células-Tronco Multipotentes/metabolismo , Ductos Pancreáticos/embriologia , Ductos Pancreáticos/metabolismo , Fatores de Transcrição SOX9/metabolismo , Animais , Células Endócrinas/citologia , Células Endócrinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Pâncreas/embriologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/lesões , Pâncreas/metabolismo , Ductos Pancreáticos/citologia , Fatores de Transcrição SOX9/genética
16.
Proc Natl Acad Sci U S A ; 106(24): 9715-20, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19487660

RESUMO

Neurog3 (Neurogenin 3 or Ngn3) is both necessary and sufficient to induce endocrine islet cell differentiation from embryonic pancreatic progenitors. Since robust Neurog3 expression has not been detected in hormone-expressing cells, Neurog3 is used as an endocrine progenitor marker and regarded as dispensable for the function of differentiated islet cells. Here we used 3 independent lines of Neurog3 knock-in reporter mice and mRNA/protein-based assays to examine Neurog3 expression in hormone-expressing islet cells. Neurog3 mRNA and protein are detected in hormone-producing cells at both embryonic and adult stages. Significantly, inactivating Neurog3 in insulin-expressing beta cells at embryonic stages or in Pdx1-expressing islet cells in adults impairs endocrine function, a phenotype that is accompanied by reduced expression of several Neurog3 target genes that are essential for islet cell differentiation, maturation, and function. These findings demonstrate that Neurog3 is required not only for initiating endocrine cell differentiation, but also for promoting islet cell maturation and maintaining islet function.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Imunofluorescência , Técnicas de Introdução de Genes , Teste de Tolerância a Glucose , Imuno-Histoquímica , Ilhotas Pancreáticas/fisiologia , Camundongos , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Dev Biol ; 323(1): 19-30, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18723011

RESUMO

We have previously shown the transcription factor SOX9 to be required for the maintenance of multipotential pancreatic progenitor cells in the early embryonic pancreas. However, the association of pancreatic endocrine defects with the Sox9-haploinsufficiency syndrome campomelic dysplasia (CD) implies additional later roles for Sox9 in endocrine development. Using short-term lineage tracing in mice, we demonstrate here that SOX9 marks a pool of multipotential pancreatic progenitors throughout the window of major cell differentiation. During mid-pancreogenesis, both endocrine and exocrine cells simultaneously arise from the SOX9(+) epithelial cords. Our analysis of mice with 50%-reduced Sox9 gene dosage in pancreatic progenitors reveals endocrine-specific defects phenocopying CD. By birth, these mice display a specific reduction in endocrine cell mass, while their exocrine compartment and total organ size is normal. The decrease in endocrine cells is caused by reduced generation of endocrine progenitors from the SOX9(+) epithelium. Conversely, formation of exocrine progenitors is insensitive to reduced Sox9 gene dosage, thus explaining the normal organ size at birth. Our results show that not only is SOX9 required for the maintenance of early pancreatic progenitors, but also governs their adoption of an endocrine fate. Our findings therefore suggest that defective endocrine specification might underlie the pancreatic phenotype of individuals with CD.


Assuntos
Células Epiteliais/metabolismo , Dosagem de Genes , Pâncreas Exócrino/metabolismo , Pâncreas/citologia , Fatores de Transcrição SOX9/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Embrião de Mamíferos , Glândulas Endócrinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glucagon/análise , Insulina/análise , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese/genética , Pâncreas/embriologia , Pâncreas/metabolismo , Fatores de Transcrição SOX9/metabolismo
18.
Proc Natl Acad Sci U S A ; 104(6): 1865-70, 2007 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-17267606

RESUMO

The factors necessary to maintain organ-specific progenitor cells are poorly understood and yet of extreme clinical importance. Here, we identify the transcription factor SOX9 as the first specific marker and maintenance factor of multipotential progenitors during pancreas organogenesis. In the developing pancreas, SOX9 expression is restricted to a mitotically active, Notch-responsive subset of PDX1(+) pluripotent progenitors and is absent from committed endocrine precursors or differentiated cells. Similar to Notch mutations, organ-specific Sox9 inactivation in mice causes severe pancreatic hypoplasia resulting from depletion of the progenitor cell pool. We show that Sox9 maintains pancreatic progenitors by stimulating their proliferation, survival, and persistence in an undifferentiated state. Our finding that SOX9 regulates the Notch-effector HES1 suggests a Notch-dependent mechanism and establishes a possible genetic link between SOX factors and Notch. These findings will be of major significance for the development of in vitro protocols for cell replacement therapies.


Assuntos
Proteínas de Grupo de Alta Mobilidade/fisiologia , Pâncreas/citologia , Pâncreas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Feminino , Proteínas de Grupo de Alta Mobilidade/deficiência , Proteínas de Grupo de Alta Mobilidade/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pâncreas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição SOX9 , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
19.
Methods Mol Biol ; 411: 13-23, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18287635

RESUMO

With the recent advances in mouse genetics, it is now possible to mark specific cell types genetically in vivo and to study the fate of cells during development and adulthood. Cells are labeled and followed in vivo through the stable expression of reporter genes in particular cell types. The two most commonly used reporter genes are LacZ, which encodes the enzyme beta-galactosidase (beta-gal), and green fluorescent protein (GFP). beta-Gal expression can be detected enzymatically, using 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside (X-gal) as a substrate, and GFP can be directly visualized by fluorescence microscopy. However, with single detection of beta-gal or GFP, it is often impossible to determine whether expression of the reporter protein is restricted to a particular cell type. To ascertain the identity of individual cells within a multicellular tissue, beta-gal or GFP proteins must be visualized in conjunction with additional cellular markers. For such experiments, specific antibodies raised against beta-gal or GFP can be used in coimmunofluorescence analyses. Such double-staining analyses on tissue sections are a powerful tool to study transgene expression or to trace cells in multicellular tissues.


Assuntos
Genes Reporter , Proteínas de Fluorescência Verde/análise , Imuno-Histoquímica/métodos , beta-Galactosidase/análise , Animais , Imunofluorescência , Galactosídeos/metabolismo , Proteínas de Fluorescência Verde/imunologia , Indóis/metabolismo , Camundongos , Coloração e Rotulagem , beta-Galactosidase/imunologia
20.
Mol Endocrinol ; 18(11): 2765-76, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15297605

RESUMO

The basic helix-loop-helix transcription factor Neurogenin 3 (NGN3) controls endocrine cell fate specification in uncommitted pancreatic progenitor cells. Ngn3-deficient mice do not develop any islet cells and are diabetic. All the major islet cell types, including insulin-producing beta-cells, derive from Ngn3-positive endocrine progenitor cells. Therefore, the characterization of this population of immature cells is of particular interest for the development of novel strategies for cell replacement therapies in type 1 diabetes. To explore further the biology of islet progenitor cells we have generated a mouse in which Ngn3-expressing cells are labeled with the enhanced yellow fluorescent protein (EYFP) using a knock-add-on strategy. In this approach, the EYFP cDNA is introduced into the 3'-untranslated region of the proendocrine transcription factor, Neurogenin 3, without deleting any endogenous coding or regulatory sequences. In Ngn3(EYFP/+) and Ngn3(EYFP/EYFP) mice, the EYFP protein is targeted to Ngn3-expressing progenitors in the developing pancreas, and islets develop normally. Islet progenitors can be purified from whole embryonic pancreas by fluorescence-activated cell sorting from Ngn3(EYFP/+) mice and their development can be monitored in real time in pancreas explant cultures. These experiments showed that endocrine progenitors can form de novo and expand, in vitro, in the absence of signals from the surrounding mesenchyme, suggesting that endocrine commitment is a default pathway. The Ngn3(EYFP) mice represent a valuable tool to study islet cell development and neogenesis in normal and diabetic animals as well as for the determination of the conditions to generate beta-cells in vitro.


Assuntos
Proteínas de Bactérias/genética , Corantes Fluorescentes , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/embriologia , Proteínas Luminescentes/genética , Proteínas do Tecido Nervoso/genética , Células-Tronco/citologia , Animais , Proteínas de Bactérias/análise , Proteínas de Bactérias/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular , Citometria de Fluxo , Ilhotas Pancreáticas/metabolismo , Proteínas Luminescentes/análise , Proteínas Luminescentes/metabolismo , Mesoderma/metabolismo , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/crescimento & desenvolvimento , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células-Tronco/química , Células-Tronco/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...