Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Nanomedicine ; 14: 371-382, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30662261

RESUMO

BACKGROUND: Islet cell transplantation is one of the key treatments for type 1 diabetes. Understanding the mechanisms of insulin fusion and exocytosis are of utmost importance for the improvement of the current islet cell transplantation and treatment of diabetes. These phenomena have not been fully evaluated due either to the lack of proper dynamic imaging, or the lack of proper cell preservation during imaging at nanoscales. METHODS: By maintaining the native environment of pancreatic ß-cells between two graphene monolayer sheets, we were able to monitor the subcellular events using in situ graphene liquid cell (GLC)-transmission electron microscopy (TEM) with both high temporal and high spatial resolution. RESULTS: For the first time, the nucleation and growth of insulin particles until the later stages of fusion were imaged at nanometer scales. The release of insulin from plasma membrane involves the degradation of plasma membrane and drastic reductions in the shorter axis of the insulin particles. Sequential exocytosis results indicated the nucleation, growth and attachment of the new insulin particles to the already anchored ones, which is thermodynamically favorable due to the reduction in total surface, further reducing the Gibbs free energy. The retraction of the already anchored insulin toward the cell is also monitored for the first time live at nanoscale resolution. CONCLUSION: Investigation of insulin granule dynamics in ß-cells can be investigated via GLC-TEM. Our findings with this technology open new realms for the development of novel drugs on pathological pancreatic ß-cells, because this approach facilitates observing the effects of the stimuli on the live cells and insulin granules.


Assuntos
Grafite/química , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Microscopia Eletrônica de Transmissão , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Exocitose , Insulina/metabolismo , Fusão de Membrana , Camundongos
2.
Diabetes ; 67(12): 2626-2639, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30305365

RESUMO

Syntaxin 4 (Stx4) enrichment in human and mouse islet grafts improves the success of transplants in reversing streptozotocin (STZ)-induced diabetes in mice, although the underlying molecular mechanisms remain elusive. Toward a further understanding of this, human islets and inducible transgenic mice that selectively overexpress Stx4 in islet ß-cells (ßTG-Stx4) were challenged with proinflammatory stressors in vitro and in vivo. Remarkably, ßTG-Stx4 mice resisted the loss of ß-cell mass and the glucose intolerance that multiple low doses of STZ induce. Under standard conditions, glucose tolerance was enhanced and mice maintained normal fasting glycemia and insulinemia. Conversely, Stx4 heterozygous knockout mice succumbed rapidly to STZ-induced glucose intolerance compared with their wild-type littermates. Human islet ß-cells overexpressing Stx4 exhibited enhanced insulin secretory capability; resilience against proinflammatory cytokine-induced apoptosis; and reduced expression of the CXCL9, CXCL10, and CXCL11 genes coordinate with decreased activation/nuclear localization of nuclear factor-κB. Finding ways to boost Stx4 expression presents a novel potential therapeutic avenue for promoting islet function and preserving ß-cell mass.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Intolerância à Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Qa-SNARE/metabolismo , Animais , Apoptose/fisiologia , Citocinas/metabolismo , Diabetes Mellitus Experimental/genética , Intolerância à Glucose/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas Qa-SNARE/genética
4.
Genome Med ; 9(1): 42, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28511717

RESUMO

Recent advances in ß-cell regeneration in vivo are providing insights into the mechanisms involved in the conversion of distinct pancreatic cell lineages into ß cells. These mechanisms mostly involve reactivation of the gene encoding the pancreatic endocrine cell-specifying transcription factor neurogenin-3.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Células Secretoras de Insulina/fisiologia , Proteínas do Tecido Nervoso , Regeneração , Transdução de Sinais , Humanos , Células Secretoras de Insulina/metabolismo
5.
Mol Cell Endocrinol ; 445: 85-94, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-27838399

RESUMO

The adult pancreas is only capable of limited regeneration. Unlike highly regenerative tissues such as the skin, intestinal crypts and hematopoietic system, no dedicated adult stem cells or stem cell niche have so far been identified within the adult pancreas. New ß cells have been shown to form in the adult pancreas, in response to high physiological demand or experimental ß-cell ablation, mostly by replication of existing ß cells. The possibility that new ß cells are formed from other sources is currently a point of major controversy. Under particular injury conditions, fully differentiated pancreatic duct and acinar cells have been shown to dedifferentiate into a progenitor-like state, however the extent, to which ductal, acinar or other endocrine cells contribute to restoring pancreatic ß-cell mass remains to be resolved. In this review we focus on regenerative events in the pancreas with emphasis on the restoration of ß-cell mass. We present an overview of regenerative responses noted within the different pancreatic lineages, following injury. We also highlight the intrinsic plasticity of the adult pancreas that allows for inter-conversion of fully differentiated pancreatic lineages through manipulation of few genes or growth factors. Taken together, evidence from a number of studies suggest that differentiated pancreatic lineages could act as facultative progenitor cells, but the extent to which these contribute to ß-cell regeneration in vivo is still a matter of contention.


Assuntos
Células-Tronco Adultas/citologia , Células Secretoras de Insulina/fisiologia , Regeneração , Animais , Desdiferenciação Celular , Plasticidade Celular , Humanos , Células Secretoras de Insulina/citologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-31660541

RESUMO

Islet transplantation effectively treats diabetes but relies on immune suppression and is practically limited by the number of cadaveric islets available. An alternative cellular source is insulin-producing cells derived from pluripotent cell sources. Three animal cohorts were used in the current study to evaluate whether an oxygen-providing macro-encapsulation device, 'ßAIR', could function in conjunction with human embryonic stem cells (hESCs) and their derivatives. The first cohort received macro-encapsulated undifferentiated hESCs, a second cohort received hESCs differentiated to a pancreatic progenitor state with limited endocrine differentiation. A reference cohort received human islets. Macro-encapsulation devices were implanted subcutaneously and monitored for up to 4 months. Undifferentiated pluripotent stem cells did not form teratoma but underwent cell death following implantation. Human C-peptide (hC- peptide) was detectable in host serum one week after implantation for both other cohorts. hC-peptide levels decreasing over time but remained detectable up to the end of the study. Key factors associated with mature endocrine cells were observed in grafts recovered from cohorts containing islets and hESC-derivatives including C-peptide, insulin, glucagon and urocortin 3. We conclude that the 'ßAIR' macroencapsulation device is compatible with both human islets and pluripotent derivatives, but has a limited capability of sustaining undifferentiated pluripotent cells.

7.
Dev Biol ; 399(2): 204-17, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25576928

RESUMO

Wnt signaling is a well conserved pathway critical for growth, patterning and differentiation of multiple tissues and organs. Previous studies on Wnt signaling in the pancreas have been based predominantly on downstream pathway effector genes such as ß-catenin. We here provide evidence that the canonical-pathway member Wnt7b is a physiological regulator of pancreatic progenitor cell growth. Genetic deletion of Wnt7b in the developing pancreas leads to pancreatic hypoplasia due to reduced proliferation of pancreatic progenitor cells during the phase of pancreas development marked by rapid progenitor cell growth. While the differentiation potential of pancreatic progenitor cells is unaffected by Wnt7b deletion, through a gain-of-function analysis, we find that early pancreatic progenitor cells are highly sensitive to Wnt7b expression, but later lose such competence. By modulating the level and the temporal windows of Wnt7b expression we demonstrate a significant impact on organ growth and morphogenesis particularly during the early branching stages of the organ, which negatively affects generation of the pro-endocrine (Ngn3(+)/Nkx6.1(+)), and pro-acinar (Ptf1A(+)) fields. Consequently, Wnt7b gain-of-function results in failed morphogenesis and almost complete abrogation of the differentiation of endocrine and acinar cells, leading to cystic epithelial metaplasia expressing ductal markers including Sox9, Hnf6 and Hnf1ß. While Wnt7b is expressed exclusively in the developing pancreatic epithelium, adjacent mesenchymal cells in the organ display a direct trophic response to elevated Wnt7b and increase expression of Lef1, cFos and desmin. Of note, in contrast to the pancreatic epithelium, the pancreatic mesenchyme remains competent to respond to Wnt7b ligand, at later stages in development. We conclude that Wnt7b helps coordinate pancreatic development through autocrine, as well as paracrine mechanisms, and as such represents a novel bi-modal morphogen ligand.


Assuntos
Células Epiteliais/citologia , Transição Epitelial-Mesenquimal/fisiologia , Morfogênese/fisiologia , Pâncreas/embriologia , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Animais , Pesos e Medidas Corporais , Diferenciação Celular/fisiologia , Cílios/química , Células Epiteliais/fisiologia , Imunofluorescência , Perfilação da Expressão Gênica , Hibridização In Situ , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Tamanho do Órgão , Pâncreas/metabolismo , Células-Tronco/fisiologia , Tubulina (Proteína)/análise
8.
Wiley Interdiscip Rev Dev Biol ; 2(4): 531-44, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24014421

RESUMO

Notch signaling is an evolutionarily conserved mechanism adapted to control binary fate decisions. The first evidence of Notch in pancreatic development focused on its critical role in controlling endocrine fate decisions. Since then, we have come to understand that this signaling system operates iteratively in the pancreas, and is not limited to the control of endocrine fate decision. Notch appears to play a role in early organ development, then during organ domain patterning, and only during a final refinement process, in the control of terminal cell fates. In so doing, Notch receptors and their ligands are under the influence of a wealth of genetic components that together help orchestrate the building of a complex, glandular organ.


Assuntos
Diferenciação Celular , Linhagem da Célula , Pâncreas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Humanos , Pâncreas/citologia , Pâncreas/embriologia , Receptores Notch/genética
9.
Nucleic Acids Res ; 41(8): 4459-69, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23449223

RESUMO

SOX9 encodes a transcription factor that presides over the specification and differentiation of numerous progenitor and differentiated cell types, and although SOX9 haploinsufficiency and overexpression cause severe diseases in humans, including campomelic dysplasia, sex reversal and cancer, the mechanisms underlying SOX9 transcription remain largely unsolved. We identify here an evolutionarily conserved enhancer located 70-kb upstream of mouse Sox9 and call it SOM because it specifically activates a Sox9 promoter reporter in most Sox9-expressing somatic tissues in transgenic mice. Moreover, SOM-null fetuses and pups reduce Sox9 expression by 18-37% in the pancreas, lung, kidney, salivary gland, gut and liver. Weanlings exhibit half-size pancreatic islets and underproduce insulin and glucagon, and adults slowly recover from acute pancreatitis due to a 2-fold impairment in Sox9 upregulation. Molecular and genetic experiments reveal that Sox9 protein dimers bind to multiple recognition sites in the SOM sequence and are thereby both necessary and sufficient for enhancer activity. These findings thus uncover that Sox9 directly enhances its functions in somatic tissue development and adult regeneration through SOM-mediated positive auto-regulation. They provide thereby novel insights on molecular mechanisms controlling developmental and disease processes and suggest new strategies to improve disease treatments.


Assuntos
Elementos Facilitadores Genéticos , Regeneração , Fatores de Transcrição SOX9/genética , Animais , Linhagem Celular , Embrião de Mamíferos/metabolismo , Homeostase , Camundongos , Camundongos Transgênicos , Pâncreas/crescimento & desenvolvimento , Pâncreas/fisiologia , Pancreatite/patologia , Ratos , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/fisiologia
10.
Dev Biol ; 376(1): 1-12, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23370147

RESUMO

Ngn3 is recognized as a regulator of pancreatic endocrine formation, and Notch signaling as an important negative regulator Ngn3 gene expression. By conditionally controlling expression of Ngn3 in the pancreas, we find that these two signaling components are dynamically linked. This connection involves transcriptional repression as previously shown, but also incorporates a novel post-translational mechanism. In addition to its ability to promote endocrine fate, we provide evidence of a competing ability of Ngn3 in the patterning of multipotent progenitor cells in turn controlling the formation of ducts. On one hand, Ngn3 cell-intrinsically activates endocrine target genes; on the other, Ngn3 cell-extrinsically promotes lateral signaling via the Dll1>Notch>Hes1 pathway which substantially limits its ability to sustain endocrine formation. Prior to endocrine commitment, the Ngn3-mediated activation of the Notch>Hes1 pathway impacts formation of the trunk domain in the pancreas causing multipotent progenitors to lose acinar, while gaining endocrine and ductal, competence. The subsequent selection of fate from such bipotential progenitors is then governed by lateral inhibition, where Notch>Hes1-mediated Ngn3 protein destabilization serves to limit endocrine differentiation by reducing cellular levels of Ngn3. This system thus allows for rapid dynamic changes between opposing bHLH proteins in cells approaching a terminal differentiation event. Inhibition of Notch signaling leads to Ngn3 protein stabilization in the normal mouse pancreas explants. We conclude that the mutually exclusive expression pattern of Ngn3/Hes1 proteins in the mammalian pancreas is partially controlled through Notch-mediated post-translational regulation and we demonstrate that the formation of insulin-producing beta-cells can be significantly enhanced upon induction of a pro-endocrine drive combined with the inhibition of Notch processing.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Morfogênese/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/embriologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Dipeptídeos , Técnicas Histológicas , Imuno-Histoquímica , Camundongos , Pâncreas/metabolismo , Estabilidade Proteica , Reação em Cadeia da Polimerase em Tempo Real
11.
Semin Cell Dev Biol ; 23(6): 711-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22743232

RESUMO

This review summarizes our current understanding of exocrine pancreas development, including the formation of acinar, ductal and centroacinar cells. We discuss the transcription factors associated with various stages of exocrine differentiation, from multipotent progenitor cells to fully differentiated acinar and ductal cells. Within the branching epithelial tree of the embryonic pancreas, this involves the progressive restriction of multipotent pancreatic progenitor cells to either a central "trunk" domain giving rise to the islet and ductal lineages, or a peripheral "tip" domain giving rise to acinar cells. This review also discusses the soluble morphogens and other signaling pathways that influence these events. Finally, we examine centroacinar cells as an enigmatic pancreatic cell type whose lineage remains uncertain, and whose possible progenitor capacities continue to be explored.


Assuntos
Células Acinares/citologia , Morfogênese , Pâncreas Exócrino/embriologia , Ductos Pancreáticos/embriologia , Células Acinares/metabolismo , Animais , Humanos , Organogênese , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas Exócrino/citologia , Pâncreas Exócrino/metabolismo , Ductos Pancreáticos/citologia , Ductos Pancreáticos/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo
12.
Proc Natl Acad Sci U S A ; 109(29): E2001-9, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22745161

RESUMO

In eukaryotic cells, newly synthesized secretory proteins require COPII (coat protein complex II) to exit the endoplasmic reticulum (ER). COPII contains five core components: SAR1, SEC23, SEC24, SEC13, and SEC31. SEC23 is a GTPase-activating protein that activates the SAR1 GTPase and also plays a role in cargo recognition. Missense mutations in the human COPII paralogues SEC23A and SEC23B result in craniolenticulosutural dysplasia and congenital dyserythropoietic anemia type II, respectively. We now report that mice completely deficient for SEC23B are born with no apparent anemia phenotype, but die shortly after birth, with degeneration of professional secretory tissues. In SEC23B-deficient embryonic pancreas, defects occur in exocrine and endocrine tissues shortly after differentiation. Pancreatic acini are completely devoid of zymogen granules, and the ER is severely distended. Similar ultrastructural alterations are also observed in salivary glands, but not in liver. Accumulation of proteins in the ER lumen activates the proapoptotic pathway of the unfolded protein response, suggesting a central role for apoptosis in the degeneration of these tissues in SEC23B-deficient embryos. Although maintenance of the secretory pathway should be required by all cells, our findings reveal a surprising tissue-specific dependence on SEC23B for the ER exit of highly abundant cargo, with high levels of SEC23B expression observed in professional secretory tissues. The disparate phenotypes in mouse and human could result from residual SEC23B function associated with the hypomorphic mutations observed in humans, or alternatively, might be explained by a species-specific shift in function between the closely related SEC23 paralogues.


Assuntos
Apoptose/fisiologia , Retículo Endoplasmático/metabolismo , Pâncreas/metabolismo , Via Secretória/fisiologia , Proteínas de Transporte Vesicular/deficiência , Azul Alciano , Animais , Antraquinonas , Apoptose/genética , Imunofluorescência , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Imunoeletrônica , Mutação/genética , Pâncreas/embriologia , Pâncreas/ultraestrutura , Reação em Cadeia da Polimerase em Tempo Real , Via Secretória/genética , Especificidade da Espécie , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
13.
Development ; 139(10): 1744-53, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22461559

RESUMO

Early pancreatic morphogenesis is characterized by the transformation of an uncommitted pool of pancreatic progenitor cells into a branched pancreatic epithelium that consists of 'tip' and 'trunk' domains. These domains have distinct molecular signatures and differentiate into distinct pancreatic cell lineages. Cells at the branched tips of the epithelium develop into acinar cells, whereas cells in the trunk subcompartment differentiate into endocrine and duct cells. Recent genetic analyses have highlighted the role of key transcriptional regulators in the specification of these subcompartments. Here, we analyzed in mice the role of Notch signaling in the patterning of multipotent pancreatic progenitor cells through mosaic overexpression of a Notch signaling antagonist, dominant-negative mastermind-like 1, resulting in a mixture of wild-type and Notch-suppressed pancreatic progenitor cells. We find that attenuation of Notch signaling has pronounced patterning effects on multipotent pancreatic progenitor cells prior to terminal differentiation. Relative to the wild-type cells, the Notch-suppressed cells lose trunk marker genes and gain expression of tip marker genes. The Notch-suppressed cells subsequently differentiate into acinar cells, whereas duct and endocrine populations are formed predominantly from the wild-type cells. Mechanistically, these observations could be explained by a requirement of Notch for the expression of the trunk determination gene Nkx6.1. This was supported by the finding of direct binding of RBP-jκ to the Nkx6.1 proximal promoter.


Assuntos
Pâncreas/citologia , Receptores Notch/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Citometria de Fluxo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Imuno-Histoquímica , Camundongos , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/genética
14.
BMC Dev Biol ; 7: 110, 2007 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-17910758

RESUMO

BACKGROUND: In recent years, considerable knowledge has been gained on the molecular mechanisms underlying retinal cell fate specification. However, hitherto studies focused primarily on the six major retinal cell classes (five types of neurons of one type of glial cell), and paid little attention to the specification of different neuronal subtypes within the same cell class. In particular, the molecular machinery governing the specification of the two most abundant neurotransmitter phenotypes in the retina, GABAergic and glutamatergic, is largely unknown. In the spinal cord and cerebellum, the transcription factor Ptf1a is essential for GABAergic neuron production. In the mouse retina, Ptf1a has been shown to be involved in horizontal and most amacrine neurons differentiation. RESULTS: In this study, we examined the distribution of neurotransmitter subtypes following Ptf1a gain and loss of function in the Xenopus retina. We found cell-autonomous dramatic switches between GABAergic and glutamatergic neuron production, concomitant with profound defects in the genesis of amacrine and horizontal cells, which are mainly GABAergic. Therefore, we investigated whether Ptf1a promotes the fate of these two cell types or acts directly as a GABAergic subtype determination factor. In ectodermal explant assays, Ptf1a was found to be a potent inducer of the GABAergic subtype. Moreover, clonal analysis in the retina revealed that Ptf1a overexpression leads to an increased ratio of GABAergic subtypes among the whole amacrine and horizontal cell population, highlighting its instructive capacity to promote this specific subtype of inhibitory neurons. Finally, we also found that within bipolar cells, which are typically glutamatergic interneurons, Ptf1a is able to trigger a GABAergic fate. CONCLUSION: Altogether, our results reveal for the first time in the retina a major player in the GABAergic versus glutamatergic cell specification genetic pathway.


Assuntos
Linhagem da Célula , Neurônios/citologia , Retina/citologia , Fatores de Transcrição/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Sequência de Bases , Primers do DNA , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Xenopus laevis
15.
Genes Dev ; 20(11): 1441-6, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16751182

RESUMO

Patterning of the embryonic endoderm into distinct sets of precursor cells involves the precisely regulated activities of key transcription regulators. Ectopic, pan-endodermal activation of XPtf1a/p48 during pancreas precursor cell stages of Xenopus embryogenesis results in an expansion of the pancreatic territory, precisely within the borders of XlHbox8 expression. A combination of both activities is sufficient to expand the pancreatic precursor cell population also into more posterior portions of the endoderm. Both treatments result in the formation of a giant pancreas that persists up to late tadpole stages of development and carries both supernumerary endocrine and exocrine cells. A combination of XPtf1a/p48 and XlHbox8 is thus sufficient to convert nonpancreatic endodermal cells into pancreatic precursor cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Endoderma , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Pâncreas/embriologia , Transativadores/genética , Fatores de Transcrição/genética , Proteínas de Xenopus/genética , Animais , Sequência de Bases , Primers do DNA , Xenopus
16.
Dev Biol ; 271(1): 144-60, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15196957

RESUMO

How and when the vertebrate endoderm is first subdivided into discrete progenitor cell populations that will give rise to the different major organs, including pancreas and liver, are only poorly understood. We have used Xenopus laevis as a model system to characterize these events, since it is particularly suited to study the early embryonic patterning in vertebrates. Our experimental results support the notion that retinoic acid (RA) functions as an essential endodermal patterning signal in Xenopus and that it acts as early as during gastrulation. As a result of RA treatment, the expression of Sonic Hedgehog (Shh), a known inhibitor of pancreas development in other vertebrate systems, is negatively regulated in the dorsal prepancreatic endoderm. Furthermore, RA is found to promote endocrine at the expense of exocrine differentiation in the dorsal pancreas, correlating with a specific inhibition of Notch signaling activities in this territory. Conversely, RA enhances exocrine marker gene expression in the ventral pancreas.


Assuntos
Diferenciação Celular/fisiologia , Epigênese Genética , Pâncreas/embriologia , Transdução de Sinais/fisiologia , Tretinoína/fisiologia , Proteínas de Xenopus , Animais , Contagem de Células , Primers do DNA , Endoderma/fisiologia , Células Enteroendócrinas/fisiologia , Glândulas Exócrinas/embriologia , Proteínas Hedgehog , Hibridização In Situ , Insulina/metabolismo , Microinjeções , Morfogênese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/metabolismo , Transativadores/metabolismo , Tretinoína/metabolismo , Xenopus laevis
17.
Gene Expr Patterns ; 4(1): 71-6, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14678831

RESUMO

The pancreas develops from dorsal and ventral epithelial extensions at the foregut/midgut boundary in Xenopus embryos. Endocrine and exocrine specification is thought to occur from a pool of uniform precursor cells. While the genetic network controlling endocrine specification and differentiation has been the object of extensive investigations, the corresponding mechanism leading to the exocrine pancreas is much less understood. Here, we report on the identification and characterisation of a novel molecular marker for the early exocrine pancreas in Xenopus embryos. Xenopus pancreatic protein disulfide isomerase is expressed in both dorsal and ventral pancreatic buds. By whole mount in situ hybridization it is detected as early as stage 39 in the exocrine lineage of the developing pancreas; RT-PCR reveals onset of expression as early as stage 35/36.


Assuntos
Pâncreas/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Marcadores Genéticos , Hibridização In Situ , Dados de Sequência Molecular , Pâncreas/embriologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Xenopus laevis/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...