Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Diabetologia ; 67(6): 1066-1078, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38630142

RESUMO

AIMS/HYPOTHESIS: Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS: We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS: We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION: This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.


Assuntos
Diferenciação Celular , Proliferação de Células , Imageamento Tridimensional , Pâncreas , Humanos , Pâncreas/embriologia , Pâncreas/citologia , Pâncreas/metabolismo , Diferenciação Celular/fisiologia , Feminino , Células-Tronco/citologia , Células-Tronco/metabolismo , Gravidez , Insulina/metabolismo
2.
Cell ; 139(4): 791-801, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19914171

RESUMO

Understanding how cells polarize and coordinate tubulogenesis during organ formation is a central question in biology. Tubulogenesis often coincides with cell-lineage specification during organ development. Hence, an elementary question is whether these two processes are independently controlled, or whether proper cell specification depends on formation of tubes. To address these fundamental questions, we have studied the functional role of Cdc42 in pancreatic tubulogenesis. We present evidence that Cdc42 is essential for tube formation, specifically for initiating microlumen formation and later for maintaining apical cell polarity. Finally, we show that Cdc42 controls cell specification non-cell-autonomously by providing the correct microenvironment for proper control of cell-fate choices of multipotent progenitors. For a video summary of this article, see the PaperFlick file with the Supplemental Data available online.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Organogênese , Pâncreas/embriologia , Animais , Polaridade Celular , Células Epiteliais/metabolismo , Laminina/metabolismo , Camundongos , Camundongos Knockout , Pâncreas/citologia , Pâncreas/metabolismo , Pâncreas Exócrino/citologia , Pâncreas Exócrino/embriologia , Pâncreas Exócrino/metabolismo , Células-Tronco/metabolismo , Quinases Associadas a rho/metabolismo
3.
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
4.
Development ; 141(3): 685-96, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24449844

RESUMO

Delamination plays a pivotal role during normal development and cancer. Previous work has demonstrated that delamination and epithelial cell movement within the plane of an epithelium are associated with a change in cellular phenotype. However, how this positional change is linked to differentiation remains unknown. Using the developing mouse pancreas as a model system, we show that ß cell delamination and differentiation are two independent events, which are controlled by Cdc42/N-WASP signaling. Specifically, we show that expression of constitutively active Cdc42 in ß cells inhibits ß cell delamination and differentiation. These processes are normally associated with junctional actin and cell-cell junction disassembly and the expression of fate-determining transcription factors, such as Isl1 and MafA. Mechanistically, we demonstrate that genetic ablation of N-WASP in ß cells expressing constitutively active Cdc42 partially restores both delamination and ß cell differentiation. These findings elucidate how junctional actin dynamics via Cdc42/N-WASP signaling cell-autonomously control not only epithelial delamination but also cell differentiation during mammalian organogenesis.


Assuntos
Actinas/metabolismo , Diferenciação Celular , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Transdução de Sinais , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Animais Recém-Nascidos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Epitélio/metabolismo , Humanos , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Junções Intercelulares/metabolismo , Junções Intercelulares/patologia , Camundongos , Ratos , Imagem com Lapso de Tempo
5.
Development ; 140(21): 4452-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24130330

RESUMO

In the context of a cellular therapy for diabetes, methods for pancreatic progenitor expansion and subsequent differentiation into insulin-producing beta cells would be extremely valuable. Here we establish three-dimensional culture conditions in Matrigel that enable the efficient expansion of dissociated mouse embryonic pancreatic progenitors. By manipulating the medium composition we generate either hollow spheres, which are mainly composed of pancreatic progenitors, or complex organoids that spontaneously undergo pancreatic morphogenesis and differentiation. The in vitro maintenance and expansion of pancreatic progenitors require active Notch and FGF signaling, thus recapitulating in vivo niche signaling interactions. Our experiments reveal new aspects of pancreas development, such as a community effect by which small groups of cells better maintain progenitor properties and expand more efficiently than isolated cells, as well as the requirement for three-dimensionality. Finally, growth conditions in chemically defined biomaterials pave the way for testing the biophysical and biochemical properties of the niche that sustains pancreatic progenitors.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Morfogênese/fisiologia , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Células-Tronco/citologia , Animais , Colágeno , Combinação de Medicamentos , Hidrogel de Polietilenoglicol-Dimetacrilato , Imuno-Histoquímica , Laminina , Camundongos , Microscopia de Fluorescência , Proteoglicanas , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Estatísticas não Paramétricas , Imagem com Lapso de Tempo
6.
Methods ; 59(1): 59-70, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22503774

RESUMO

Characterization of directed differentiation of pluripotent stem cells towards therapeutically relevant cell types, including pancreatic beta-cells and hepatocytes, depends on molecular markers and assays that resolve the signature of individual cells. Pancreas and liver both have a common origin of anterior definitive endoderm (DE). Here, we differentiated human embryonic stem cells towards DE using three different activin A based treatments. Differentiation efficiencies were evaluated by gene expression profiling over time at cell population level. A panel of key markers was used to study DE formation. Final DE differentiation was also analyzed with immunocytochemistry and single-cell gene expression profiling. We found that cells treated with activin A in combination with sodium butyrate and B27 serum-free supplement medium generated the most mature DE cells. Cell population studies were useful to monitor the temporal expression of genes involved in primitive streak formation and endoderm formation, while single-cell analysis allowed us to study cell culture heterogeneity and fingerprint individual cells. In addition, single-cell analysis revealed distinct gene expression patterns for the three activin A based protocols applied. Our data provide novel insights in DE gene expression at the cellular level of in vitro differentiated human embryonic stem cells, and illustrate the power of using single-cell gene expression profiling to study differentiation heterogeneity and to characterize cell types and subpopulations.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Análise de Célula Única , Ativinas/farmacologia , Ativinas/fisiologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Forma Celular , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Endoderma/citologia , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Transcriptoma
7.
Cell Stem Cell ; 30(1): 38-51.e8, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36563694

RESUMO

MODY3 is a monogenic hereditary form of diabetes caused by mutations in the transcription factor HNF1A. The patients progressively develop hyperglycemia due to perturbed insulin secretion, but the pathogenesis is unknown. Using patient-specific hiPSCs, we recapitulate the insulin secretion sensitivity to the membrane depolarizing agent sulfonylurea commonly observed in MODY3 patients. Unexpectedly, MODY3 patient-specific HNF1A+/R272C ß cells hypersecrete insulin both in vitro and in vivo after transplantation into mice. Consistently, we identified a trend of increased birth weight in human HNF1A mutation carriers compared with healthy siblings. Reduced expression of potassium channels, specifically the KATP channel, in MODY3 ß cells, increased calcium signaling, and rescue of the insulin hypersecretion phenotype by pharmacological targeting ATP-sensitive potassium channels or low-voltage-activated calcium channels suggest that more efficient membrane depolarization underlies the hypersecretion of insulin in MODY3 ß cells. Our findings identify a pathogenic mechanism leading to ß cell failure in MODY3.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Camundongos , Animais , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Tipo 2/genética , Fenótipo
8.
Dev Biol ; 352(2): 267-77, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21281624

RESUMO

Endoderm development is dependent on inductive signals from different structures in close vicinity, including the notochord, lateral plate mesoderm and endothelial cells. Recently, we demonstrated that a functional vascular system is necessary for proper pancreas development, and that sphingosine-1-phosphate (S1P) exhibits the traits of a blood vessel-derived molecule involved in early pancreas morphogenesis. To examine whether S1P(1)-signaling plays a more general role in endoderm development, S1P(1)-deficient mice were analyzed. S1P(1) ablation results in compromised growth of several foregut-derived organs, including the stomach, dorsal and ventral pancreas and liver. Within the developing pancreas the reduction in organ size was due to deficient proliferation of Pdx1(+) pancreatic progenitors, whereas endocrine cell differentiation was unaffected. Ablation of endothelial cells in vitro did not mimic the S1P(1) phenotype, instead, increased organ size and hyperbranching were observed. Consistent with a negative role for endothelial cells in endoderm organ expansion, excessive vasculature was discovered in S1P(1)-deficient embryos. Altogether, our results show that endothelial cell hyperplasia negatively influences organ development in several foregut-derived organs.


Assuntos
Endoderma/embriologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Desenvolvimento Embrionário , Endoderma/citologia , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese/genética , Morfogênese/fisiologia , Pâncreas/irrigação sanguínea , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/deficiência , Receptores de Lisoesfingolipídeo/genética , Receptores de Esfingosina-1-Fosfato , Transativadores/metabolismo
9.
Stem Cells ; 28(1): 45-56, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19890880

RESUMO

Fibroblast growth factor (FGF) signaling controls axis formation during endoderm development. Studies in lower vertebrates have demonstrated that FGF2 primarily patterns the ventral foregut endoderm into liver and lung, whereas FGF4 exhibits broad anterior-posterior and left-right patterning activities. Furthermore, an inductive role of FGF2 during dorsal pancreas formation has been shown. However, whether FGF2 plays a similar role during human endoderm development remains unknown. Here, we show that FGF2 specifies hESC-derived definitive endoderm (DE) into different foregut lineages in a dosage-dependent manner. Specifically, increasing concentrations of FGF2 inhibits hepatocyte differentiation, whereas intermediate concentration of FGF2 promotes differentiation toward a pancreatic cell fate. At high FGF2 levels specification of midgut endoderm into small intestinal progenitors is increased at the expense of PDX1(+) pancreatic progenitors. High FGF2 concentrations also promote differentiation toward an anterior foregut pulmonary cell fate. Finally, by dissecting the FGF receptor intracellular pathway that regulates pancreas specification, we demonstrate for the first time to the best of our knowledge that induction of PDX1(+) pancreatic progenitors relies on FGF2-mediated activation of the MAPK signaling pathway. Altogether, these observations suggest a broader gut endodermal patterning activity of FGF2 that corresponds to what has previously been advocated for FGF4, implying a functional switch from FGF4 to FGF2 during evolution. Thus, our results provide new knowledge of how cell fate specification of human DE is controlled-facts that will be of great value for future regenerative cell therapies.


Assuntos
Diferenciação Celular , Linhagem da Célula , Sistema Digestório/metabolismo , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Gástrula/metabolismo , Ativinas/metabolismo , Animais , Evolução Biológica , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Sistema Digestório/efeitos dos fármacos , Sistema Digestório/embriologia , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/efeitos dos fármacos , Endoderma/citologia , Endoderma/efeitos dos fármacos , Gástrula/citologia , Gástrula/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Hepatócitos/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Intestino Delgado/embriologia , Intestino Delgado/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Pâncreas/embriologia , Pâncreas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Tempo , Transativadores/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt3
10.
STAR Protoc ; 2(4): 100806, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34632415

RESUMO

We have developed a protocol to quantify the position of a cell in a branched structure based on two-dimensional microscopy images of tissue sections. Biological branched structures include organs such as the lungs, kidneys, and pancreas. In these organs, cell fate has been correlated with position, based on a qualitative estimate. However, a quantitative means of evaluating the cell position has been lacking. With this protocol, the correlation between cell fate and cell position was measured in mouse embryonic pancreas. For complete details on the use and execution of this protocol, please refer to Nyeng et al. (2019).


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Imuno-Histoquímica/métodos , Microscopia/métodos , Animais , Embrião de Mamíferos/química , Embrião de Mamíferos/citologia , Feminino , Rim/citologia , Pulmão/citologia , Masculino , Camundongos , Pâncreas/citologia
11.
STAR Protoc ; 2(3): 100636, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34258596

RESUMO

We here report a flow-cytometry-based protocol to measure single-cell protein expression in small samples. The protocol is optimized for simultaneous detection of fluorescent proteins and intracellular and surface antigens in the embryonic pancreas from the mouse. Owing to low cell numbers, current protocols for flow cytometric analysis of embryonic tissues rely on tissue pooling. Our protocol enables analysis of one pancreas per sample, thereby facilitating detection of biological variation and minimizing the number of experimental animals needed. For complete details on the use and execution of this protocol, please refer to Nyeng et al (2019).


Assuntos
Antígenos de Superfície/análise , Antígenos/análise , Embrião de Mamíferos/imunologia , Citometria de Fluxo/métodos , Pâncreas/imunologia , Animais , Feminino , Masculino , Camundongos , Análise de Célula Única/métodos
12.
Genesis ; 48(6): 374-81, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20533404

RESUMO

The cadherin family of cell adhesion molecules mediates adhesive interactions that are required for the formation and maintenance of tissues. Previously, we demonstrated that N-cadherin, which is required for numerous morphogenetic processes, is expressed in the pancreatic epithelium at E9.5, but later becomes restricted to endocrine aggregates in mice. To study the role of N-cadherin during pancreas formation and function we generated a tissue-specific knockout of N-cadherin in the early pancreatic epithelium by inter-crossing N-cadherin-floxed mice with Pdx1Cre mice. Analysis of pancreas-specific ablation of N-cadherin demonstrates that N-cadherin is dispensable for pancreatic development, but required for beta-cell granule turnover. The number of insulin secretory granules is significantly reduced in N-cadherin-deficient beta-cells, and as a consequence insulin secretion is decreased.


Assuntos
Caderinas/fisiologia , Grânulos Citoplasmáticos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/fisiologia , Células Secretoras de Insulina/metabolismo , Pâncreas/crescimento & desenvolvimento , Transativadores/fisiologia , Animais , Feminino , Imunofluorescência , Immunoblotting , Integrases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pâncreas/metabolismo
13.
J Lipid Res ; 51(6): 1535-45, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20026666

RESUMO

We investigated a family from northern Sweden in which three of four siblings have congenital chylomicronemia. LPL activity and mass in pre- and postheparin plasma were low, and LPL release into plasma after heparin injection was delayed. LPL activity and mass in adipose tissue biopsies appeared normal. [(35)S]Methionine incorporation studies on adipose tissue showed that newly synthesized LPL was normal in size and normally glycosylated. Breast milk from the affected female subjects contained normal to elevated LPL mass and activity levels. The milk had a lower than normal milk lipid content, and the fatty acid composition was compatible with the milk lipids being derived from de novo lipogenesis, rather than from the plasma lipoproteins. Given the delayed release of LPL into the plasma after heparin, we suspected that the chylomicronemia might be caused by mutations in GPIHBP1. Indeed, all three affected siblings were compound heterozygotes for missense mutations involving highly conserved cysteines in the Ly6 domain of GPIHBP1 (C65S and C68G). The mutant GPIHBP1 proteins reached the surface of transfected Chinese hamster ovary cells but were defective in their ability to bind LPL (as judged by both cell-based and cell-free LPL binding assays). Thus, the conserved cysteines in the Ly6 domain are crucial for GPIHBP1 function.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/genética , Quilomícrons/metabolismo , Sequência Conservada , Cisteína , Transtornos do Metabolismo dos Lipídeos/genética , Mutação , Tecido Adiposo/enzimologia , Tecido Adiposo/patologia , Adolescente , Adulto , Alelos , Animais , Apolipoproteína C-II/deficiência , Sequência de Bases , Células CHO , Proteínas de Transporte/metabolismo , Pré-Escolar , Cricetinae , Cricetulus , Feminino , Regulação da Expressão Gênica , Heparina/administração & dosagem , Heparina/farmacologia , Heterozigoto , Humanos , Transtornos do Metabolismo dos Lipídeos/enzimologia , Transtornos do Metabolismo dos Lipídeos/metabolismo , Transtornos do Metabolismo dos Lipídeos/patologia , Lipase Lipoproteica/sangue , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Masculino , Pessoa de Meia-Idade , Leite Humano/enzimologia , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína , Receptores de Lipoproteínas , Irmãos , Transfecção
14.
BMC Dev Biol ; 9: 2, 2009 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-19126201

RESUMO

BACKGROUND: Pancreatic islets of Langerhans originate from endocrine progenitors within the pancreatic ductal epithelium. Concomitant with differentiation of these progenitors into hormone-producing cells such cells delaminate, aggregate and migrate away from the ductal epithelium. The cellular and molecular mechanisms regulating islet cell delamination and cell migration are poorly understood. Extensive biochemical and cell biological studies using cultured cells demonstrated that Rac1, a member of the Rho family of small GTPases, acts as a key regulator of cell migration. RESULTS: To address the functional role of Rac1 in islet morphogenesis, we generated transgenic mice expressing dominant negative Rac1 under regulation of the Rat Insulin Promoter. Blocking Rac1 function in beta cells inhibited their migration away from the ductal epithelium in vivo. Consistently, transgenic islet cell spreading was compromised in vitro. We also show that the EGF-receptor ligand betacellulin induced actin remodelling and cell spreading in wild-type islets, but not in transgenic islets. Finally, we demonstrate that cell-cell contact E-cadherin increased as a consequence of blocking Rac1 activity. CONCLUSION: Our data support a model where Rac1 signalling controls islet cell migration by modulating E-cadherin-mediated cell-cell adhesion. Furthermore, in vitro experiments show that betacellulin stimulated islet cell spreading and actin remodelling is compromised in transgenic islets, suggesting that betacellulin may act as a regulator of Rac1 activity and islet migration in vivo. Our results further emphasize Rac1 as a key regulator of cell migration and cell adhesion during tissue and organ morphogenesis.


Assuntos
Movimento Celular/fisiologia , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Morfogênese/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Betacelulina , Glicemia/metabolismo , Peso Corporal , Caderinas/metabolismo , Receptores ErbB/metabolismo , Matriz Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Transgênicos , Tamanho do Órgão , RNA Mensageiro/metabolismo , Ratos , Proteínas rac1 de Ligação ao GTP/genética
15.
J Clin Invest ; 116(3): 642-51, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16470244

RESUMO

Previously we observed that neural cell adhesion molecule (NCAM) deficiency in beta tumor cells facilitates metastasis into distant organs and local lymph nodes. Here, we show that NCAM-deficient beta cell tumors grew leaky blood vessels with perturbed pericyte-endothelial cell-cell interactions and deficient perivascular deposition of ECM components. Conversely, tumor cell expression of NCAM in a fibrosarcoma model (T241) improved pericyte recruitment and increased perivascular deposition of ECM molecules. Together, these findings suggest that NCAM may limit tumor cell metastasis by stabilizing the microvessel wall. To directly address whether pericyte dysfunction increases the metastatic potential of solid tumors, we studied beta cell tumorigenesis in primary pericyte-deficient Pdgfb(ret/ret) mice. This resulted in beta tumor cell metastases in distant organs and local lymph nodes, demonstrating a role for pericytes in limiting tumor cell metastasis. These data support a new model for how tumor cells trigger metastasis by perturbing pericyte-endothelial cell-cell interactions.


Assuntos
Adenoma de Células das Ilhotas Pancreáticas/patologia , Metástase Neoplásica/patologia , Metástase Neoplásica/prevenção & controle , Neoplasias Pancreáticas/patologia , Pericitos/fisiologia , Adenoma de Células das Ilhotas Pancreáticas/irrigação sanguínea , Adenoma de Células das Ilhotas Pancreáticas/genética , Adenoma de Células das Ilhotas Pancreáticas/metabolismo , Animais , Comunicação Celular/genética , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Fibrossarcoma/irrigação sanguínea , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/metabolismo , Moléculas de Adesão de Célula Nervosa/deficiência , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/fisiologia , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Pericitos/metabolismo
16.
Clin Chem ; 55(12): 2162-70, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19815608

RESUMO

BACKGROUND: Human embryonic stem cells (hESCs) require expression of transcription factor genes POU5F1 (POU class 5 homeobox 1), NANOG (Nanog homeobox), and SOX2 [SRY (sex determining region Y)-box 2] to maintain their capacity for self-renewal and pluripotency. Because of the heterogeneous nature of cell populations, it is desirable to study the gene regulation in single cells. Large and potentially important fluctuations in a few cells cannot be detected at the population scale with microarrays or sequencing technologies. We used single-cell gene expression profiling to study cell heterogeneity in hESCs. METHODS: We collected 47 single hESCs from cell line SA121 manually by glass capillaries and 57 single hESCs from cell line HUES3 by flow cytometry. Single hESCs were lysed and reverse-transcribed. Reverse-transcription quantitative real-time PCR was then used to measure the expression POU5F1, NANOG, SOX2, and the inhibitor of DNA binding genes ID1, ID2, and ID3. A quantitative noise model was used to remove measurement noise when pairwise correlations were estimated. RESULTS: The numbers of transcripts per cell varied >100-fold between cells and showed lognormal features. POU5F1 expression positively correlated with ID1 and ID3 expression (P < 0.05) but not with NANOG or SOX2 expression. When we accounted for measurement noise, SOX2 expression was also correlated with ID1, ID2, and NANOG expression (P < 0.05). CONCLUSIONS: We demonstrate an accurate method for transcription profiling of individual hESCs. Cell-to-cell variability is large and is at least partly nonrandom because we observed correlations between core transcription factors. High fluctuations in gene expression may explain why individual cells in a seemingly undifferentiated cell population have different susceptibilities for inductive cues.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição/biossíntese , Diferenciação Celular , Linhagem Celular , Separação Celular , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Proteína 1 Inibidora de Diferenciação/biossíntese , Proteína 1 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/biossíntese , Proteína 2 Inibidora de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/biossíntese , Proteínas Inibidoras de Diferenciação/genética , Modelos Genéticos , Proteína Homeobox Nanog , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição/genética
17.
J Mol Med (Berl) ; 86(2): 135-44, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17891366

RESUMO

Tumour cells use two major routes to spread during metastasis, e.g. lymph vessels and blood vessels within or surrounding the primary tumour. The growth rate of the primary tumour often correlates with the quantity of new blood vessels that form within the tumour. However, qualitative abnormalities of the tumour vasculature profoundly affect the perfusion of the primary tumour and the escape of tumour cells into the circulation. In this paper, we review recent evidence for a novel role of the supporting mural cells in limiting blood-borne metastasis.


Assuntos
Neoplasias/metabolismo , Células Neoplásicas Circulantes/metabolismo , Neovascularização Patológica/metabolismo , Pericitos/metabolismo , Transdução de Sinais , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Becaplermina , Adesão Celular , Movimento Celular , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Células Neoplásicas Circulantes/efeitos dos fármacos , Células Neoplásicas Circulantes/patologia , Neovascularização Patológica/prevenção & controle , Moléculas de Adesão de Célula Nervosa/metabolismo , Pericitos/efeitos dos fármacos , Pericitos/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis , Transdução de Sinais/efeitos dos fármacos , Células Estromais/metabolismo
18.
Dev Cell ; 49(1): 31-47.e9, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30853440

RESUMO

The mechanism of how organ shape emerges and specifies cell fate is not understood. Pancreatic duct and endocrine lineages arise in a spatially distinct domain from the acinar lineage. Whether these lineages are pre-determined or settle once these niches have been established remains unknown. Here, we reconcile these two apparently opposing models, demonstrating that pancreatic progenitors re-localize to establish the niche that will determine their ultimate fate. We identify a p120ctn-regulated mechanism for coordination of organ architecture and cellular fate mediated by differential E-cadherin based cell sorting. Reduced p120ctn expression is necessary and sufficient to re-localize a subset of progenitors to the peripheral tip domain, where they acquire an acinar fate. The same mechanism is used re-iteratively during endocrine specification, where it balances the choice between the alpha and beta cell fates. In conclusion, organ patterning is regulated by p120ctn-mediated cellular positioning, which precedes and determines pancreatic progenitor fate.


Assuntos
Padronização Corporal/genética , Cateninas/genética , Pâncreas/crescimento & desenvolvimento , Ductos Pancreáticos/crescimento & desenvolvimento , Animais , Caderinas/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Movimento Celular/genética , Desenvolvimento Embrionário/genética , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/metabolismo , Camundongos , Pâncreas/metabolismo , Receptores Notch/genética , Transdução de Sinais/genética , Células-Tronco/metabolismo , delta Catenina
19.
BMC Cell Biol ; 9: 37, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18620564

RESUMO

BACKGROUND: FUS, EWS and TAF15 are structurally similar multifunctional proteins that were first discovered upon characterization of fusion oncogenes in human sarcomas and leukemias. The proteins belong to the FET (previously TET) family of RNA-binding proteins and are implicated in central cellular processes such as regulation of gene expression, maintenance of genomic integrity and mRNA/microRNA processing. In the present study, we investigated the expression and cellular localization of FET proteins in multiple human tissues and cell types. RESULTS: FUS, EWS and TAF15 were expressed in both distinct and overlapping patterns in human tissues. The three proteins showed almost ubiquitous nuclear expression and FUS and TAF15 were in addition present in the cytoplasm of most cell types. Cytoplasmic EWS was more rarely detected and seen mainly in secretory cell types. Furthermore, FET expression was downregulated in differentiating human embryonic stem cells, during induced differentiation of neuroblastoma cells and absent in terminally differentiated melanocytes and cardiac muscle cells. The FET proteins were targeted to stress granules induced by heat shock and oxidative stress and FUS required its RNA-binding domain for this translocation. Furthermore, FUS and TAF15 were detected in spreading initiation centers of adhering cells. CONCLUSION: Our results point to cell-specific expression patterns and functions of the FET proteins rather than the housekeeping roles inferred from earlier studies. The localization of FET proteins to stress granules suggests activities in translational regulation during stress conditions. Roles in central processes such as stress response, translational control and adhesion may explain the FET proteins frequent involvement in human cancer.


Assuntos
Adesão Celular/genética , Regulação da Expressão Gênica/fisiologia , Proteínas Oncogênicas/genética , Estresse Oxidativo/genética , Forma Celular , Resposta ao Choque Térmico , Humanos , Proteína EWS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/genética , Fatores Associados à Proteína de Ligação a TATA/genética
20.
Mol Cell Biol ; 22(5): 1474-87, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11839813

RESUMO

The distinct expression of R-cadherin in the induced aggregating metanephric mesenchyme suggests that it may regulate the mesenchymal-epithelial transition during kidney development. To address whether R-cadherin is required for kidney ontogeny, R-cadherin-deficient mice were generated. These mice appeared to be healthy and were fertile, demonstrating that R-cadherin is not essential for embryogenesis. The only kidney phenotype of adult mutant animals was the appearance of dilated proximal tubules, which was associated with an accumulation of large intracellular vacuoles. Morphological analysis of nephrogenesis in R-cadherin(-/-) mice in vivo and in vitro revealed defects in the development of both ureteric bud-derived cells and metanephric mesenchyme-derived cells. First, the morphology and organization of the proximal parts of the ureteric bud epithelium were altered. Interestingly, these morphological changes correlated with an increased rate of apoptosis and were further supported by perturbed branching and patterning of the ureteric bud epithelium during in vitro differentiation. Second, during in vitro studies of mesenchymal-epithelial conversion, significantly fewer epithelial structures developed from R-cadherin(-/-) kidneys than from wild-type kidneys. These data suggest that R-cadherin is functionally involved in the differentiation of both mesenchymal and epithelial components during metanephric kidney development. Finally, to investigate whether the redundant expression of other classic cadherins expressed in the kidney could explain the rather mild kidney defects in R-cadherin-deficient mice, we intercrossed R-cadherin(-/-) mice with cadherin-6(-/-), P-cadherin(-/-), and N-cadherin(+/-) mice. Surprisingly, however, in none of the compound knockout strains was kidney development affected to a greater extent than within the individual cadherin knockout strains.


Assuntos
Caderinas/genética , Néfrons/embriologia , Animais , Apoptose , Diferenciação Celular , Indução Embrionária , Células Epiteliais/citologia , Fertilidade , Rim/fisiologia , Rim/ultraestrutura , Testes de Função Renal , Túbulos Renais Proximais/patologia , Mesoderma/citologia , Camundongos , Camundongos Knockout , Morfogênese , Ureter/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA