Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Development ; 140(18): 3799-808, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23924634

RESUMO

Genetic regulation of the cell fate transition from lateral plate mesoderm to the specification of cardiomyocytes requires suppression of Wnt/ß-catenin signaling, but the mechanism for this is not well understood. By analyzing gene expression and chromatin dynamics during directed differentiation of human embryonic stem cells (hESCs), we identified a suppressor of Wnt/ß-catenin signaling, transmembrane protein 88 (TMEM88), as a potential regulator of cardiovascular progenitor cell (CVP) specification. During the transition from mesoderm to the CVP, TMEM88 has a chromatin signature of genes that mediate cell fate decisions, and its expression is highly upregulated in advance of key cardiac transcription factors in vitro and in vivo. In early zebrafish embryos, tmem88a is expressed broadly in the lateral plate mesoderm, including the bilateral heart fields. Short hairpin RNA targeting of TMEM88 during hESC cardiac differentiation increases Wnt/ß-catenin signaling, confirming its role as a suppressor of this pathway. TMEM88 knockdown has no effect on NKX2.5 or GATA4 expression, but 80% of genes most highly induced during CVP development have reduced expression, suggesting adoption of a new cell fate. In support of this, analysis of later stage cell differentiation showed that TMEM88 knockdown inhibits cardiomyocyte differentiation and promotes endothelial differentiation. Taken together, TMEM88 is crucial for heart development and acts downstream of GATA factors in the pre-cardiac mesoderm to specify lineage commitment of cardiomyocyte development through inhibition of Wnt/ß-catenin signaling.


Assuntos
Proteínas de Membrana/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Linhagem da Célula/genética , Regulação para Baixo/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Membrana/genética , Camundongos , Modelos Biológicos , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Regulação para Cima/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , beta Catenina/metabolismo
2.
Blood ; 121(5): 717-8, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23372149

RESUMO

In this issue of Blood, Rafii et al present an elegant study of human embryonic stem cell (ESC)­derived hematopoiesis incorporating live imaging at the single-cell level to track hematopoietic lineage potential during the endothelial to hematopoietic transition.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Transdução Genética , Humanos
3.
J Vis Exp ; (135)2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29806841

RESUMO

The ability to study hematopoietic stem cell (HSC) genesis during embryonic development has been limited by the rarity of HSC precursors in the early embryo and the lack of assays that functionally identify the long-term multilineage engraftment potential of individual putative HSC precursors. Here, we describe methodology that enables the isolation and characterization of functionally validated HSC precursors at the single cell level. First, we utilize index sorting to catalog the precise phenotypic parameter of each individually sorted cell, using a combination of phenotypic markers to enrich for HSC precursors with additional markers for experimental analysis. Second, each index-sorted cell is co-cultured with vascular niche stroma from the aorta-gonad-mesonephros (AGM) region, which supports the maturation of non-engrafting HSC precursors to functional HSC with multilineage, long-term engraftment potential in transplantation assays. This methodology enables correlation of phenotypic properties of clonal hemogenic precursors with their functional engraftment potential or other properties such as transcriptional profile, providing a means for the detailed analysis of HSC precursor development at the single cell level.


Assuntos
Técnicas de Cocultura/métodos , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Separação Celular , Células Cultivadas , Células Endoteliais/citologia , Feminino , Células-Tronco Hematopoéticas/citologia , Humanos , Gravidez
4.
J Exp Med ; 215(2): 645-659, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29282253

RESUMO

Hematopoietic stem cells (HSCs) mature from pre-HSCs that originate in the major arteries of the embryo. To identify HSCs from in vitro sources, it will be necessary to refine markers of HSCs matured ex vivo. We purified and compared the transcriptomes of pre-HSCs, HSCs matured ex vivo, and fetal liver HSCs. We found that HSC maturation in vivo or ex vivo is accompanied by the down-regulation of genes involved in embryonic development and vasculogenesis, and up-regulation of genes involved in hematopoietic organ development, lymphoid development, and immune responses. Ex vivo matured HSCs more closely resemble fetal liver HSCs than pre-HSCs, but are not their molecular equivalents. We show that ex vivo-matured and fetal liver HSCs express programmed death ligand 1 (PD-L1). PD-L1 does not mark all pre-HSCs, but cell surface PD-L1 was present on HSCs matured ex vivo. PD-L1 signaling is not required for engraftment of embryonic HSCs. Hence, up-regulation of PD-L1 is a correlate of, but not a requirement for, HSC maturation.


Assuntos
Antígeno B7-H1/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Antígeno B7-H1/deficiência , Antígeno B7-H1/genética , Diferenciação Celular , Feminino , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transplante de Células-Tronco Hematopoéticas , Fígado/citologia , Fígado/embriologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Gravidez , Regulação para Cima
5.
Nat Commun ; 9(1): 1828, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29739946

RESUMO

NOTCH signaling is required for the arterial specification and formation of hematopoietic stem cells (HSCs) and lympho-myeloid progenitors in the embryonic aorta-gonad-mesonephros region and extraembryonic vasculature from a distinct lineage of vascular endothelial cells with hemogenic potential. However, the role of NOTCH signaling in hemogenic endothelium (HE) specification from human pluripotent stem cell (hPSC) has not been studied. Here, using a chemically defined hPSC differentiation system combined with the use of DLL1-Fc and DAPT to manipulate NOTCH, we discover that NOTCH activation in hPSC-derived immature HE progenitors leads to formation of CD144+CD43-CD73-DLL4+Runx1 + 23-GFP+ arterial-type HE, which requires NOTCH signaling to undergo endothelial-to-hematopoietic transition and produce definitive lympho-myeloid and erythroid cells. These findings demonstrate that NOTCH-mediated arterialization of HE is an essential prerequisite for establishing definitive lympho-myeloid program and suggest that exploring molecular pathways that lead to arterial specification may aid in vitro approaches to enhance definitive hematopoiesis from hPSCs.


Assuntos
Artérias/citologia , Endotélio Vascular/citologia , Hemangioblastos/citologia , Hematopoese , Neovascularização Fisiológica , Células-Tronco Pluripotentes/citologia , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Antígenos CD/imunologia , Artérias/metabolismo , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Rastreamento de Células/instrumentação , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Endotélio Vascular/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/imunologia , Hemangioblastos/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Células-Tronco Pluripotentes/imunologia
6.
Stem Cell Reports ; 8(6): 1563-1572, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28479303

RESUMO

Recent evidence points to the embryonic emergence of some tissue-resident innate immune cells, such as B-1a lymphocytes, prior to and independently of hematopoietic stem cells (HSCs). However, whether the full hematopoietic repertoire of embryonic HSCs initially includes these unique lineages of innate immune cells has been difficult to assess due to lack of clonal assays that identify and assess HSC precursor (pre-HSC) potential. Here, by combining index sorting of single embryonic hemogenic precursors with in vitro HSC maturation and transplantation assays, we analyze emerging pre-HSCs at the single-cell level, revealing their unique stage-specific properties and clonal lineage potential. Remarkably, clonal pre-HSCs detected between E9.5 and E11.5 contribute to the complete B cell repertoire, including B-1a lymphocytes, revealing a previously unappreciated common precursor for all B cell lineages at the pre-HSC stage and a second embryonic origin for B-1a lymphocytes.


Assuntos
Linfócitos B/metabolismo , Células-Tronco Hematopoéticas/citologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos CD/metabolismo , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/citologia , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio , Células Cultivadas , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Feminino , Citometria de Fluxo , Fatores de Troca do Nucleotídeo Guanina/genética , Células-Tronco Hematopoéticas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
7.
Cell Rep ; 17(12): 3178-3192, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28009288

RESUMO

Hematopoietic stem cell (HSC) transplantation is curative for malignant and genetic blood disorders, but is limited by donor availability and immune-mismatch. Deriving HSCs from patient-matched embryonic/induced-pluripotent stem cells (ESCs/iPSCs) could address these limitations. Prior efforts in murine models exploited ectopic HoxB4 expression to drive self-renewal and enable multi-lineage reconstitution, yet fell short in delivering robust lymphoid engraftment. Here, by titrating exposure of HoxB4-ESC-HSC to Notch ligands, we report derivation of engineered HSCs that self-renew, repopulate multi-lineage hematopoiesis in primary and secondary engrafted mice, and endow adaptive immunity in immune-deficient recipients. Single-cell analysis shows that following engraftment in the bone marrow niche, these engineered HSCs further specify to a hybrid cell type, in which distinct gene regulatory networks of hematopoietic stem/progenitors and differentiated hematopoietic lineages are co-expressed. Our work demonstrates engineering of fully functional HSCs via modulation of genetic programs that govern self-renewal and lineage priming.


Assuntos
Imunidade Adaptativa/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Autorrenovação Celular/genética , Redes Reguladoras de Genes/genética , Hematopoese/genética , Hematopoese/imunologia , Células-Tronco Hematopoéticas/imunologia , Proteínas de Homeodomínio/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Camundongos , Receptores Notch/genética , Receptores Notch/imunologia , Análise de Célula Única , Fatores de Transcrição/imunologia
8.
J Clin Invest ; 125(5): 2032-45, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25866967

RESUMO

Hematopoietic stem cells (HSCs) first emerge during embryonic development within vessels such as the dorsal aorta of the aorta-gonad-mesonephros (AGM) region, suggesting that signals from the vascular microenvironment are critical for HSC development. Here, we demonstrated that AGM-derived endothelial cells (ECs) engineered to constitutively express AKT (AGM AKT-ECs) can provide an in vitro niche that recapitulates embryonic HSC specification and amplification. Specifically, nonengrafting embryonic precursors, including the VE-cadherin-expressing population that lacks hematopoietic surface markers, cocultured with AGM AKT-ECs specified into long-term, adult-engrafting HSCs, establishing that a vascular niche is sufficient to induce the endothelial-to-HSC transition in vitro. Subsequent to hematopoietic induction, coculture with AGM AKT-ECs also substantially increased the numbers of HSCs derived from VE-cadherin⁺CD45⁺ AGM hematopoietic cells, consistent with a role in supporting further HSC maturation and self-renewal. We also identified conditions that included NOTCH activation with an immobilized NOTCH ligand that were sufficient to amplify AGM-derived HSCs following their specification in the absence of AGM AKT-ECs. Together, these studies begin to define the critical niche components and resident signals required for HSC induction and self-renewal ex vivo, and thus provide insight for development of defined in vitro systems targeted toward HSC generation for therapeutic applications.


Assuntos
Aorta/embriologia , Células Endoteliais/fisiologia , Endotélio Vascular/embriologia , Gônadas/embriologia , Sistema Hematopoético/embriologia , Mesonefro/embriologia , Receptor Notch1/fisiologia , Receptor Notch2/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Antígenos CD/análise , Caderinas/análise , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Endotélio Vascular/citologia , Feminino , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Antígenos Comuns de Leucócito/análise , Masculino , Proteínas de Membrana/fisiologia , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Quimera por Radiação , Transdução de Sinais , Células Estromais/fisiologia
9.
Cell Stem Cell ; 13(4): 459-70, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24094326

RESUMO

Human pluripotent stem cells (hPSCs) represent a promising source of patient-specific cells for disease modeling, drug screens, and cellular therapies. However, the inability to derive engraftable human hematopoietic stem and progenitor cells (HSPCs) has limited their characterization to in vitro assays. We report a strategy to respecify lineage-restricted CD34(+)CD45(+) myeloid precursors derived from hPSCs into multilineage progenitors that can be expanded in vitro and engrafted in vivo. HOXA9, ERG, and RORA conferred self-renewal and multilineage potential in vitro and maintained primitive CD34(+)CD38(-) cells. Screening cells via transplantation revealed that two additional factors, SOX4 and MYB, conferred engraftment. Progenitors specified with all five factors gave rise to reproducible short-term engraftment with myeloid and erythroid lineages. Erythroid precursors underwent hemoglobin switching in vivo, silencing embryonic and activating adult globin expression. Our combinatorial screening approach establishes a strategy for obtaining transcription-factor-mediated engraftment of blood progenitors from human pluripotent cells.


Assuntos
Linhagem da Célula , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Pluripotentes/citologia , Antígenos CD34/metabolismo , Humanos , Antígenos Comuns de Leucócito/metabolismo , Células-Tronco Multipotentes/metabolismo , Células-Tronco Pluripotentes/metabolismo
10.
J Clin Oncol ; 27(25): 4217-26, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19636005

RESUMO

Erythropoietin-stimulating agents (ESAs) were originally designed to replace endogenous erythropoietin in patients with anemia secondary to renal failure. Their use has subsequently been expanded to include patients with anemia of other causes, including cancer patients, in whom deficiency of erythropoietin, per se, is not the primary cause of anemia. Although early studies showed promise of ESA administration in reducing the need for transfusions and improving the quality of life in cancer patients, several large randomized clinical trials have recently shown a potential detrimental effect of ESA administration on tumor progression and survival in these patients. These studies have called into question the safety of ESAs as supportive therapy in patients being treated for oncologic conditions. However, numerous questions remain to be addressed regarding the design of these studies, the effect of various targeted hemoglobin levels, and the potential biologic mechanisms proposed to explain promotion of tumor progression and reduced survival.


Assuntos
Anemia/tratamento farmacológico , Células Eritroides/efeitos dos fármacos , Hematínicos/efeitos adversos , Neoplasias/complicações , Anemia/sangue , Anemia/etiologia , Anemia/mortalidade , Animais , Proliferação de Células/efeitos dos fármacos , Intervalo Livre de Doença , Células Eritroides/metabolismo , Hemoglobinas/metabolismo , Humanos , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Neoplasias/mortalidade , Neoplasias/patologia , Neovascularização Patológica/induzido quimicamente , Receptores da Eritropoetina/agonistas , Receptores da Eritropoetina/sangue , Medição de Risco , Transdução de Sinais/efeitos dos fármacos , Trombose/induzido quimicamente , Resultado do Tratamento
11.
Genesis ; 40(1): 40-4, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15354292

RESUMO

In the past decade we have witnessed an epidemic of obesity in developed countries. Therefore, understanding the mechanisms involved in regulation of body weight is becoming an increasingly important goal shared by the public and the scientific community. The key to fat deposition is the adipocyte, a specialized cell that plays a critical role in energy balance and appetite regulation. Much of our knowledge of adipogenesis comes from studies using preadipocytic cell lines that have provided important information regarding molecular control of adipocyte differentiation. However, they fall short of revealing how naive cells acquire competence for adipogenesis. Studies in preadipocytes indicate that the Notch pathway plays a role in regulating adipogenesis (Garces et al.: J Biol Chem 272:29729-29734, 1997). Given the known biological functions of Notch in mediating cell fate decisions (Artavanis-Tsakonas et al.: Science 284:770-776, 1999), we wished to test the hypothesis that the Notch pathway is required for this cellular program by examining adipogenesis in several genetic loss-of-function models that encompass the entire pathway. We conclude that the "canonical" Notch signaling pathway is dispensable for adipocyte specification and differentiation from either mesenchymal or epithelial progenitors.


Assuntos
Adipócitos/citologia , Proteínas de Membrana/fisiologia , Células-Tronco/citologia , Adipócitos/fisiologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Células Epiteliais/citologia , Fibroblastos/citologia , Proteínas de Membrana/genética , Mesoderma/citologia , Camundongos , Receptores Notch , Células-Tronco/efeitos dos fármacos , Tretinoína/farmacologia
12.
Blood ; 104(10): 3097-105, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15251982

RESUMO

Notch1 is known to play a critical role in regulating fates in numerous cell types, including those of the hematopoietic lineage. Multiple defects exhibited by Notch1-deficient embryos confound the determination of Notch1 function in early hematopoietic development in vivo. To overcome this limitation, we examined the developmental potential of Notch1(-/-) embryonic stem (ES) cells by in vitro differentiation and by in vivo chimera analysis. Notch1 was found to affect primitive erythropoiesis differentially during ES cell differentiation and in vivo, and this result reflected an important difference in the regulation of Notch1 expression during ES cell differentiation relative to the developing mouse embryo. Notch1 was dispensable for the onset of definitive hematopoiesis both in vitro and in vivo in that Notch1(-/-) definitive progenitors could be detected in differentiating ES cells as well as in the yolk sac and early fetal liver of chimeric mice. Despite the fact that Notch1(-/-) cells can give rise to multiple types of definitive progenitors in early development, Notch1(-/-) cells failed to contribute to long-term definitive hematopoiesis past the early fetal liver stage in the context of a wild-type environment in chimeric mice. Thus, Notch1 is required, in a cell-autonomous manner, for the establishment of long-term, definitive hematopoietic stem cells (HSCs).


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Óperon Lac , Fígado/citologia , Fígado/embriologia , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Receptor Notch1 , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Saco Vitelino/citologia , Saco Vitelino/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA