Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Development ; 144(13): 2402-2414, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28576773

RESUMO

While much is known about the molecular pathways that regulate embryonic development and adult homeostasis of the endocrine pancreas, little is known about what regulates early postnatal development and maturation of islets. Given that birth marks the first exposure to enteral nutrition, we investigated how nutrient-regulated signaling pathways influence postnatal islet development in mice. We performed loss-of-function studies of mechanistic target of rapamycin (mTOR), a highly conserved kinase within a nutrient-sensing pathway known to regulate cellular growth, morphogenesis and metabolism. Deletion of Mtor in pancreatic endocrine cells had no significant effect on their embryonic development. However, within the first 2 weeks after birth, mTOR-deficient islets became dysmorphic, ß-cell maturation and function were impaired, and animals lost islet mass. Moreover, we discovered that these distinct functions of mTOR are mediated by separate downstream branches of the pathway, in that mTORC1 (with adaptor protein Raptor) is the main complex mediating the maturation and function of islets, whereas mTORC2 (with adaptor protein Rictor) impacts islet mass and architecture. Taken together, these findings suggest that nutrient sensing may be an essential trigger for postnatal ß-cell maturation and islet development.


Assuntos
Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Morfogênese , Complexos Multiproteicos/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Recém-Nascidos , Agregação Celular , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Modelos Biológicos , Mutação/genética
2.
Diabetologia ; 62(12): 2245-2251, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31511930

RESUMO

AIMS/HYPOTHESIS: Type 1 diabetes is an autoimmune disorder characterised by loss of insulin-producing beta cells of the pancreas. Progress in understanding the cellular and molecular mechanisms underlying the human disease has been hampered by a dearth of appropriate human experimental models. We previously reported the characterisation of islet-infiltrating CD4+ T cells from a deceased organ donor who had type 1 diabetes. METHODS: Induced pluripotent stem cell (iPSC) lines derived from the above donor were differentiated into CD14+ macrophages and tested for their capacity to present antigen to T cell receptors (TCRs) derived from islet-infiltrating CD4+ T cells from the same donor. RESULTS: The iPSC macrophages displayed typical macrophage morphology, surface markers (CD14, CD86, CD16 and CD11b) and were phagocytic. In response to IFNγ treatment, iPSC macrophages upregulated expression of HLA class II, a characteristic that correlated with their capacity to present epitopes derived from proinsulin C-peptide to a T cell line expressing TCRs derived from islet-infiltrating CD4+ T cells of the original donor. T cell activation was specifically blocked by anti-HLA-DQ antibodies but not by antibodies directed against HLA-DR. CONCLUSIONS/INTERPRETATION: This study provides a proof of principle for the use of iPSC-derived immune cells for modelling key cellular interactions in human type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Ilhotas Pancreáticas/metabolismo , Macrófagos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Ilhotas Pancreáticas/imunologia , Macrófagos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
3.
Blood ; 119(26): 6243-54, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22611158

RESUMO

Transcriptional profiling of differentiating human embryonic stem cells (hESCs) revealed that MIXL1-positive mesodermal precursors were enriched for transcripts encoding the G-protein-coupled APELIN receptor (APLNR). APLNR-positive cells, identified by binding of the fluoresceinated peptide ligand, APELIN (APLN), or an anti-APLNR mAb, were found in both posterior mesoderm and anterior mesendoderm populations and were enriched in hemangioblast colony-forming cells (Bl-CFC). The addition of APLN peptide to the media enhanced the growth of embryoid bodies (EBs), increased the expression of hematoendothelial genes in differentiating hESCs, and increased the frequency of Bl-CFCs by up to 10-fold. Furthermore, APLN peptide also synergized with VEGF to promote the growth of hESC-derived endothelial cells. These studies identified APLN as a novel growth factor for hESC-derived hematopoietic and endothelial cells.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Apelina , Receptores de Apelina , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Endoderma/fisiologia , Perfilação da Expressão Gênica , Hemangioblastos/efeitos dos fármacos , Hemangioblastos/metabolismo , Hemangioblastos/fisiologia , Hematopoese/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Mesoderma/fisiologia , Análise em Microsséries , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
4.
Stem Cell Res ; 56: 102547, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34619644

RESUMO

Insulin expressing beta cells and glucagon expressing alpha cells are the two most abundant endocrine cell types of the human pancreatic islet. Both alpha and beta cells can be generated in vitro via the differentiation of pluripotent stem cells (PSCs), affording the opportunity to study their ontogeny and to examine their developmental inter-relationship. To aid these analyses, we have generated a PSC line in which insulin expression is reported by GFP and glucagon expression is reported by mCherry. This cell line enables viable isolation of cells expressing each hormone and optimisation of methods that lead to their generation.


Assuntos
Células Endócrinas , Ilhotas Pancreáticas , Células-Tronco Pluripotentes , Diferenciação Celular , Glucagon , Proteínas de Fluorescência Verde , Humanos , Insulina , Ilhotas Pancreáticas/citologia , Proteínas Luminescentes , Pâncreas , Proteína Vermelha Fluorescente
5.
Sci Rep ; 11(1): 8315, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33859325

RESUMO

The identification of cell surface markers specific to pancreatic beta cells is important for both the study of islet biology and for investigating the pathophysiology of diseases in which this cell type is lost or damaged. Following analysis of publicly available RNAseq data, we identified specific integrin subunits, integrin αv and integrin ß5, that were expressed in beta cells. This finding was further elaborated using immunofluorescence analysis of histological sections derived from donor human pancreas. Despite the broad expression of specific integrin subunits, we found that expression of integrin αvß5 heterodimers was restricted to beta cells and that this complex persisted in islet remnants of some type 1 diabetic individuals from which insulin expression had been lost. This study identifies αvß5 heterodimers as a novel cell surface marker of human pancreatic beta cells, a finding that will aid in the identification and characterisation of this important cell type.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Receptores de Vitronectina/metabolismo , Adulto , Biomarcadores/metabolismo , Feminino , Expressão Gênica , Humanos , Insulina/genética , Insulina/metabolismo , Masculino , Pessoa de Meia-Idade , Receptores de Vitronectina/genética
6.
Nat Cell Biol ; 22(1): 60-73, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31907413

RESUMO

Defining the ontogeny of the human adaptive immune system during embryogenesis has implications for understanding childhood diseases including leukaemias and autoimmune conditions. Using RAG1:GFP human pluripotent stem cell reporter lines, we examined human T-cell genesis from pluripotent-stem-cell-derived haematopoietic organoids. Under conditions favouring T-cell development, RAG1+ cells progressively upregulated a cohort of recognized T-cell-associated genes, arresting development at the CD4+CD8+ stage. Sort and re-culture experiments showed that early RAG1+ cells also possessed B-cell, myeloid and erythroid potential. Flow cytometry and single-cell-RNA-sequencing data showed that early RAG1+ cells co-expressed the endothelial/haematopoietic progenitor markers CD34, VECAD and CD90, whereas imaging studies identified RAG1+ cells within CD31+ endothelial structures that co-expressed SOX17+ or the endothelial marker CAV1. Collectively, these observations provide evidence for a wave of human T-cell development that originates directly from haemogenic endothelium via a RAG1+ intermediate with multilineage potential.


Assuntos
Endotélio/citologia , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Linhagem Celular , Desenvolvimento Embrionário/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Organoides/citologia
7.
Stem Cell Res ; 34: 101380, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30605840

RESUMO

We describe the generation and characterization of 5 human induced pluripotent stem cell (iPSC) lines derived from peripheral blood mononuclear cells (PBMCs) of healthy adult individuals. The PBMCs were reprogrammed using non-integrating Sendai viruses containing the reprogramming factors POU5F1 (OCT4), SOX2, KLF4 and MYC. The iPSC lines exhibited a normal karyotype, expressed pluripotency markers and differentiated into cells representative of the three embryonic germ layers. These iPSC lines can be used as controls in studying disease mechanisms.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/patologia , Leucócitos Mononucleares/patologia , Adulto , Linhagem Celular , Feminino , Humanos , Fator 4 Semelhante a Kruppel , Masculino , Pessoa de Meia-Idade , Adulto Jovem
8.
Stem Cell Reports ; 7(3): 518-526, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27594589

RESUMO

The ability to reliably express fluorescent reporters or other genes of interest is important for using human pluripotent stem cells (hPSCs) as a platform for investigating cell fates and gene function. We describe a simple expression system, designated GAPTrap (GT), in which reporter genes, including GFP, mCherry, mTagBFP2, luc2, Gluc, and lacZ are inserted into the GAPDH locus in hPSCs. Independent clones harboring variations of the GT vectors expressed remarkably consistent levels of the reporter gene. Differentiation experiments showed that reporter expression was reliably maintained in hematopoietic cells, cardiac mesoderm, definitive endoderm, and ventral midbrain dopaminergic neurons. Similarly, analysis of teratomas derived from GT-lacZ hPSCs showed that ß-galactosidase expression was maintained in a spectrum of cell types representing derivatives of the three germ layers. Thus, the GAPTrap vectors represent a robust and straightforward tagging system that enables indelible labeling of PSCs and their differentiated derivatives.


Assuntos
Expressão Gênica , Genes Reporter , Vetores Genéticos/genética , Células-Tronco Pluripotentes/metabolismo , Transgenes , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Humanos , Células-Tronco Pluripotentes/citologia , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição
9.
Ann N Y Acad Sci ; 1311: 124-37, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24611778

RESUMO

In 1998, the landmark paper describing the isolation and culture of human embryonic stem cells (ESCs) was published. Since that time, the main goal of many diabetes researchers has been to derive ß cells from ESCs as a renewable cell-based therapy for the treatment of patients with diabetes. In working toward this goal, numerous protocols that attempt to recapitulate normal pancreatic development have been published that result in the formation of pancreatic cell types from human pluripotent cells. This review examines stem cell differentiation methods and places them within the context of pancreatic development. We additionally compare strategies that are currently being used to generate pancreatic cell types and contrast them with approaches that have been used to generate functional cell types in different lineages. In doing this, we aim to identify how new approaches might be used to improve yield and functionality of in vitro-derived pancreatic ß cells as an eventual cell-based therapy for type 1 diabetes.


Assuntos
Diferenciação Celular , Células Secretoras de Insulina/citologia , Pâncreas/citologia , Células-Tronco Pluripotentes/citologia , Humanos
10.
Rev Diabet Stud ; 11(1): 6-18, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25148364

RESUMO

Human embryonic stem cells have been advanced as a source of insulin-producing cells that could potentially replace cadaveric-derived islets in the treatment of type 1 diabetes. To this end, protocols have been developed that promote the formation of pancreatic progenitors and endocrine cells from human pluripotent stem cells, encompassing both embryonic stem cells and induced pluripotent stem cells. In this review, we examine these methods and place them in the context of the developmental and embryological studies upon which they are based. In particular, we outline the stepwise differentiation of cells towards definitive endoderm, pancreatic endoderm, endocrine lineages and the emergence of functional beta-cells. In doing so, we identify key factors common to many such protocols and discuss the proposed action of these factors in the context of cellular differentiation and ongoing development. We also compare strategies that entail transplantation of progenitor populations with those that seek to develop fully functional hormone expressing cells in vitro. Overall, our survey of the literature highlights the significant progress already made in the field and identifies remaining deficiencies in developing a pluripotent stem cell based treatment for type 1 diabetes.


Assuntos
Diferenciação Celular , Células Secretoras de Insulina/citologia , Modelos Biológicos , Células-Tronco Pluripotentes/citologia , Animais , Células Cultivadas , Células-Tronco Embrionárias/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células Secretoras de Insulina/metabolismo , Cultura Primária de Células/tendências
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA