Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(10): 1470-1483, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36138183

RESUMEN

Traditionally viewed as poorly plastic, neutrophils are now recognized as functionally diverse; however, the extent and determinants of neutrophil heterogeneity in humans remain unclear. We performed a comprehensive immunophenotypic and transcriptome analysis, at a bulk and single-cell level, of neutrophils from healthy donors and patients undergoing stress myelopoiesis upon exposure to growth factors, transplantation of hematopoietic stem cells (HSC-T), development of pancreatic cancer and viral infection. We uncover an extreme diversity of human neutrophils in vivo, reflecting the rates of cell mobilization, differentiation and exposure to environmental signals. Integrated control of developmental and inducible transcriptional programs linked flexible granulopoietic outputs with elicitation of stimulus-specific functional responses. In this context, we detected an acute interferon (IFN) response in the blood of patients receiving HSC-T that was mirrored by marked upregulation of IFN-stimulated genes in neutrophils but not in monocytes. Systematic characterization of human neutrophil plasticity may uncover clinically relevant biomarkers and support the development of diagnostic and therapeutic tools.


Asunto(s)
Mielopoyesis , Neutrófilos , Biomarcadores/metabolismo , Humanos , Interferones/genética , Interferones/metabolismo , Neutrófilos/metabolismo , Plásticos/metabolismo
2.
Nat Rev Mol Cell Biol ; 18(1): 56-67, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27876786

RESUMEN

Human pluripotent stem cells (hPSCs) provide an unparalleled opportunity to establish in vitro differentiation models that will transform our approach to the study of human development. In the case of the blood system, these models will enable investigation of the earliest stages of human embryonic haematopoiesis that was previously not possible. In addition, they will provide platforms for studying the origins of human blood cell diseases and for generating de novo haematopoietic stem and progenitor cell populations for cell-based regenerative therapies.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Pluripotentes/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Células Madre Pluripotentes/fisiología
3.
Cell ; 155(1): 215-27, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24074870

RESUMEN

Hematopoietic stem cells (HSCs) develop from a specialized subpopulation of endothelial cells known as hemogenic endothelium (HE). Although the HE origin of HSCs is now well established in different species, the signaling pathways that control this transition remain poorly understood. Here, we show that activation of retinoic acid (RA) signaling in aorta-gonad-mesonephros-derived HE ex vivo dramatically enhanced its HSC potential, whereas conditional inactivation of the RA metabolizing enzyme retinal dehydrogenase 2 in VE-cadherin expressing endothelial cells in vivo abrogated HSC development. Wnt signaling completely blocked the HSC inductive effects of RA modulators, whereas inhibition of the pathway promoted the development of HSCs in the absence of RA signaling. Collectively, these findings position RA and Wnt signaling as key regulators of HSC development and in doing so provide molecular insights that will aid in developing strategies for their generation from pluripotent stem cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Tretinoina/metabolismo , Aldehído Oxidorreductasas/metabolismo , Animales , Aorta/citología , Aorta/embriología , Regulación hacia Abajo , Embrión de Mamíferos , Gónadas/citología , Gónadas/embriología , Células Madre Hematopoyéticas/metabolismo , Mesonefro/citología , Ratones , Receptores de Ácido Retinoico/metabolismo , Vía de Señalización Wnt
4.
Development ; 148(15)2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34370006

RESUMEN

B1 lymphocytes are a small but unique component of the innate immune-like cells. However, their ontogenic origin is still a matter of debate. Although it is widely accepted that B1 cells originate early in fetal life, whether or not they arise from hematopoietic stem cells (HSCs) is still unclear. In order to shed light on the B1 cell origin, we set out to determine whether their lineage specification is dependent on Notch signaling, which is essential for the HSC generation and, therefore, all derivatives lineages. Using mouse embryonic stem cells (mESCs) to recapitulate murine embryonic development, we have studied the requirement for Notch signaling during the earliest B-cell lymphopoiesis and found that Rbpj-deficient mESCs are able to generate B1 cells. Their Notch independence was confirmed in ex vivo experiments using Rbpj-deficient embryos. In addition, we found that upregulation of Notch signaling induced the emergence of B2 lymphoid cells. Taken together, these findings indicate that control of Notch signaling dose is crucial for different B-cell lineage specification from endothelial cells and provides pivotal information for their in vitro generation from PSCs for therapeutic applications. This article has an associated 'The people behind the papers' interview.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Desarrollo Embrionario/inmunología , Receptores Notch/inmunología , Transducción de Señal/inmunología , Animales , Diferenciación Celular/inmunología , Células Endoteliales/inmunología , Células Madre Hematopoyéticas/inmunología , Ratones , Ratones Endogámicos C57BL
5.
Nature ; 545(7655): 432-438, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28514439

RESUMEN

A variety of tissue lineages can be differentiated from pluripotent stem cells by mimicking embryonic development through stepwise exposure to morphogens, or by conversion of one differentiated cell type into another by enforced expression of master transcription factors. Here, to yield functional human haematopoietic stem cells, we perform morphogen-directed differentiation of human pluripotent stem cells into haemogenic endothelium followed by screening of 26 candidate haematopoietic stem-cell-specifying transcription factors for their capacity to promote multi-lineage haematopoietic engraftment in mouse hosts. We recover seven transcription factors (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1 and SPI1) that are sufficient to convert haemogenic endothelium into haematopoietic stem and progenitor cells that engraft myeloid, B and T cells in primary and secondary mouse recipients. Our combined approach of morphogen-driven differentiation and transcription-factor-mediated cell fate conversion produces haematopoietic stem and progenitor cells from pluripotent stem cells and holds promise for modelling haematopoietic disease in humanized mice and for therapeutic strategies in genetic blood disorders.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Animales , Reprogramación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Endotelio/citología , Femenino , Trasplante de Células Madre Hematopoyéticas , Proteínas Homeobox A10 , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Regulador Transcripcional ERG/metabolismo
6.
Methods ; 101: 65-72, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26439174

RESUMEN

The generation of hematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) remains a major goal for regenerative medicine and disease modeling. However, hPSC differentiation cultures produce mostly hematopoietic progenitors belonging to the embryonic HSC-independent hematopoietic program, which may not be relevant or accurate for modeling normal and disease-state adult hematopoietic processes. Through a stage-specific directed differentiation approach, it is now possible to generate exclusively definitive hematopoietic progenitors from hPSCs showing characteristics of the more developmentally advanced fetal hematopoiesis. Here, we summarize recent efforts at generating hPSC-derived definitive hematopoiesis through embryoid body differentiation under defined conditions. Embryoid bodies are generated through enzymatic dissociation of hPSCs from matrigel-coated plasticware, followed by recombinant BMP4, driving mesoderm specification. Definitive hematopoiesis is specified by a GSK3ß-inhibitor, followed by recombinant VEGF and supportive hematopoietic cytokines. The CD34+ cells obtained using this method are then suitable for hematopoietic assays for definitive hematopoietic potential.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/fisiología , Células Madre Pluripotentes/fisiología , Antígenos CD34/metabolismo , Proteína Morfogenética Ósea 4/fisiología , Técnicas de Cultivo de Célula , Línea Celular , Citocinas/fisiología , Cuerpos Embrioides/citología , Cuerpos Embrioides/fisiología , Hemangioblastos/fisiología , Hematopoyesis , Humanos , Factor A de Crecimiento Endotelial Vascular/fisiología
7.
Nat Cell Biol ; 26(5): 719-730, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38594587

RESUMEN

During embryonic development, blood cells emerge from specialized endothelial cells, named haemogenic endothelial cells (HECs). As HECs are rare and only transiently found in early developing embryos, it remains difficult to distinguish them from endothelial cells. Here we performed transcriptomic analysis of 28- to 32-day human embryos and observed that the expression of Fc receptor CD32 (FCGR2B) is highly enriched in the endothelial cell population that contains HECs. Functional analyses using human embryonic and human pluripotent stem cell-derived endothelial cells revealed that robust multilineage haematopoietic potential is harboured within CD32+ endothelial cells and showed that 90% of CD32+ endothelial cells are bona fide HECs. Remarkably, these analyses indicated that HECs progress through different states, culminating in FCGR2B expression, at which point cells are irreversibly committed to a haematopoietic fate. These findings provide a precise method for isolating HECs from human embryos and human pluripotent stem cell cultures, thus allowing the efficient generation of haematopoietic cells in vitro.


Asunto(s)
Desarrollo Embrionario , Receptores de IgG , Humanos , Desarrollo Embrionario/genética , Receptores de IgG/metabolismo , Receptores de IgG/genética , Hemangioblastos/metabolismo , Hemangioblastos/citología , Diferenciación Celular , Células Endoteliales/metabolismo , Células Endoteliales/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Linaje de la Célula , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Transcriptoma , Perfilación de la Expresión Génica , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología
8.
Cell Stem Cell ; 29(11): 1512-1514, 2022 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-36332568

RESUMEN

A recent study1 demonstrates how hematopoietic stem cells (HSCs) contribute minimally to blood and immune cell production during development and only become active postnatally. The work also reveals how Mecom expression can be used to distinguish rare HSCs from the more abundant progenitors that arise to maintain embryonic hematopoiesis.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Desarrollo Embrionario , Factores de Transcripción/metabolismo , Diferenciación Celular
9.
J Exp Med ; 219(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-34928315

RESUMEN

In the mouse, the first hematopoietic cells are generated in the yolk sac from the primitive, erythro-myeloid progenitor (EMP) and lymphoid programs that are specified before the emergence of hematopoietic stem cells. While many of the yolk sac-derived populations are transient, specific immune cell progeny seed developing tissues, where they function into adult life. To access the human equivalent of these lineages, we modeled yolk sac hematopoietic development using pluripotent stem cell differentiation. Here, we show that the combination of Activin A, BMP4, and FGF2 induces a population of KDR+CD235a/b+ mesoderm that gives rise to the spectrum of erythroid, myeloid, and T lymphoid lineages characteristic of the mouse yolk sac hematopoietic programs, including the Vδ2+ subset of γ/δ T cells that develops early in the human embryo. Through clonal analyses, we identified a multipotent hematopoietic progenitor with erythroid, myeloid, and T lymphoid potential, suggesting that the yolk sac EMP and lymphoid lineages may develop from a common progenitor.


Asunto(s)
Hematopoyesis , Modelos Biológicos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Saco Vitelino/citología , Animales , Biomarcadores , Diferenciación Celular/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Linfopoyesis/genética , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
10.
Nat Cell Biol ; 24(5): 616-624, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35484246

RESUMEN

The generation of haematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) is a major goal for regenerative medicine. During embryonic development, HSCs derive from haemogenic endothelium (HE) in a NOTCH- and retinoic acid (RA)-dependent manner. Although a WNT-dependent (WNTd) patterning of nascent hPSC mesoderm specifies clonally multipotent intra-embryonic-like HOXA+ definitive HE, this HE is functionally unresponsive to RA. Here we show that WNTd mesoderm, before HE specification, is actually composed of two distinct KDR+ CD34neg populations. CXCR4negCYP26A1+ mesoderm gives rise to HOXA+ multilineage definitive HE in an RA-independent manner, whereas CXCR4+ ALDH1A2+ mesoderm gives rise to HOXA+ multilineage definitive HE in a stage-specific, RA-dependent manner. Furthermore, both RA-independent (RAi) and RA-dependent (RAd) HE harbour transcriptional similarity to distinct populations found in the early human embryo, including HSC-competent HE. This revised model of human haematopoietic development provides essential resolution to the regulation and origins of the multiple waves of haematopoiesis. These insights provide the basis for the generation of specific haematopoietic populations, including the de novo specification of HSCs.


Asunto(s)
Hemangioblastos , Células Madre Pluripotentes , Diferenciación Celular/fisiología , Linaje de la Célula , Femenino , Hematopoyesis , Humanos , Embarazo , Tretinoina/farmacología
11.
Blood ; 113(17): 3953-60, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19221036

RESUMEN

We have isolated c-Kit(+)Lin(-) cells from both human and murine amniotic fluid (AF) and investigated their hematopoietic potential. In vitro, the c-Kit(+)Lin(-) population in both species displayed a multilineage hematopoietic potential, as demonstrated by the generation of erythroid, myeloid, and lymphoid cells. In vivo, cells belonging to all 3 hematopoietic lineages were found after primary and secondary transplantation of murine c-Kit(+)Lin(-) cells into immunocompromised hosts, thus demonstrating the ability of these cells to self-renew. Gene expression analysis of c-Kit(+) cells isolated from murine AF confirmed these results. The presence of cells with similar characteristics in the surrounding amnion indicates the possible origin of AF c-Kit(+)Lin(-) cells. This is the first report showing that cells isolated from the AF do have hematopoietic potential; our results support the idea that AF may be a new source of stem cells for therapeutic applications.


Asunto(s)
Líquido Amniótico/metabolismo , Linaje de la Célula , Hematopoyesis , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
12.
Neuromuscul Disord ; 18(8): 597-605, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18602263

RESUMEN

Antisense-mediated exon skipping holds great potential for the treatment of DMD. In mdx mice, functional recovery of skeletal muscle has been obtained upon systemic delivery of "naked" oligonucleotides or viral vectors encoding for antisense snRNAs. However, amongst the studies reported so far, which used either neonatal or young adult animals--only one achieved dystrophin restoration in cardiac muscle, using an adeno-associated vector. Here we report the in vivo delivery of morpholino oligos in aged mdx mice, both in skeletal muscle, via intra-arterial injection, and in cardiac muscle, via intra-muscular injection. Localized intra-arterial delivery yielded high levels of dystrophin restoration and just two doses of 100 microg each resulted into detectable force recovery in the EDL muscles of treated limbs. On the other hand, upon intra-cardiac injections in the left ventricle wall the skipping effect was much lower than what obtained in tibialis anterior muscles injected with comparable amounts of oligos. This latter finding suggests that even upon direct delivery antisense-mediated dystrophin restoration in cardiac muscle might suffer from limitations that do not exist in skeletal muscle.


Asunto(s)
Distrofina/biosíntesis , Distrofina/genética , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Oligonucleótidos Antisentido/farmacología , Envejecimiento/fisiología , Animales , Western Blotting , Interpretación Estadística de Datos , Exones/genética , Corazón/efectos de los fármacos , Corazón/fisiología , Ventrículos Cardíacos/efectos de los fármacos , Inmunohistoquímica , Inyecciones Intraarteriales , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Contracción Muscular/fisiología , Músculo Esquelético/fisiopatología , Oligonucleótidos Antisentido/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Stem Cell Reports ; 11(6): 1324-1326, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30540960

RESUMEN

Being able to summarize properly your work is not an easy task. But learning the skill of writing a good abstract is very important, as it can open many doors, including the possibility to be selected as a speaker at conferences. As meeting abstract reviewers, here we are writing to give you insights into the abstract review process and insiders' tips to help increase your chances of landing on that podium.


Asunto(s)
Indización y Redacción de Resúmenes , Congresos como Asunto , Escritura
15.
Cardiovasc Res ; 65(2): 366-73, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15639475

RESUMEN

OBJECTIVE: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a genetically heterogeneous disorder characterized by fibro-fatty replacement of the right ventricular myocardium, associated with high risk of sudden death. The objective of this study is to identify the gene involved in ARVD1, which has been elusive ever since its locus was mapped to chromosome 14q24.3. METHODS AND RESULTS: Mutation screening of the promoter and untranslated regions (UTRs) of the transforming growth factor-beta3 (TGFbeta3) gene was performed by direct sequencing of genomic DNA of one index case belonging to an ARVD1 family including 38 members in four generations. We detected a nucleotide substitution (c.-36G>A) in 5' UTR of TGFbeta3 gene, invariably associated with the typical ARVC clinical phenotype in the affected family members, according to the established diagnostic criteria. Investigation extended to 30 unrelated ARVC patients, performed by denaturing high-performance liquid chromatography (DHPLC), led to the identification of an additional mutation (c.1723C>T) in the 3' UTR of one proband. Neither nucleotide change was found in 300 control subjects. In vitro expression assays with constructs containing the mutations showed that mutated UTRs were twofold more active than wild-types. CONCLUSION: We identified TGFbeta3 as the disease gene involved in ARVD1. The identification of a novel ARVC gene will increase the power of the genetic screening for early diagnosis of asymptomatic carriers among relatives of ARVC patients.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Factor de Crecimiento Transformador beta/genética , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Animales , Displasia Ventricular Derecha Arritmogénica/complicaciones , Displasia Ventricular Derecha Arritmogénica/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Niño , Cromatografía Líquida de Alta Presión , Análisis Mutacional de ADN , Muerte Súbita Cardíaca/etiología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Miocitos Cardíacos , Linaje , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta3
16.
Nat Cell Biol ; 17(5): 580-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25915127

RESUMEN

The generation of haematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) will depend on the accurate recapitulation of embryonic haematopoiesis. In the early embryo, HSCs develop from the haemogenic endothelium (HE) and are specified in a Notch-dependent manner through a process named endothelial-to-haematopoietic transition (EHT). As HE is associated with arteries, it is assumed that it represents a subpopulation of arterial vascular endothelium (VE). Here we demonstrate at a clonal level that hPSC-derived HE and VE represent separate lineages. HE is restricted to the CD34(+)CD73(-)CD184(-) fraction of day 8 embryoid bodies and it undergoes a NOTCH-dependent EHT to generate RUNX1C(+) cells with multilineage potential. Arterial and venous VE progenitors, in contrast, segregate to the CD34(+)CD73(med)CD184(+) and CD34(+)CD73(hi)CD184(-) fractions, respectively. Together, these findings identify HE as distinct from VE and provide a platform for defining the signalling pathways that regulate their specification to functional HSCs.


Asunto(s)
Arterias/fisiología , Diferenciación Celular , Linaje de la Célula , Células Progenitoras Endoteliales/fisiología , Células Madre Hematopoyéticas/fisiología , Células Madre Multipotentes/fisiología , Células Madre Pluripotentes/fisiología , 5'-Nucleotidasa/deficiencia , Antígenos CD34/metabolismo , Arterias/citología , Arterias/metabolismo , Biomarcadores/metabolismo , Línea Celular , Separación Celular/métodos , Técnicas de Cocultivo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Progenitoras Endoteliales/metabolismo , Proteínas Ligadas a GPI/deficiencia , Células Madre Hematopoyéticas/metabolismo , Humanos , Microscopía por Video , Células Madre Multipotentes/metabolismo , Fenotipo , Células Madre Pluripotentes/metabolismo , Células Precursoras de Linfocitos T/fisiología , Receptores CXCR5/deficiencia , Receptores Notch/metabolismo , Transducción de Señal , Factores de Tiempo , Venas/citología , Venas/fisiología
17.
Nat Biotechnol ; 32(6): 554-61, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24837661

RESUMEN

Efforts to derive hematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) are complicated by the fact that embryonic hematopoiesis consists of two programs, primitive and definitive, that differ in developmental potential. As only definitive hematopoiesis generates HSCs, understanding how this program develops is essential for being able to produce this cell population in vitro. Here we show that both hematopoietic programs transition through hemogenic endothelial intermediates and develop from KDR(+)CD34(-)CD144(-) progenitors that are distinguished by CD235a expression. Generation of primitive progenitors (KDR(+)CD235a(+)) depends on stage-specific activin-nodal signaling and inhibition of the Wnt-ß-catenin pathway, whereas specification of definitive progenitors (KDR(+)CD235a(-)) requires Wnt-ß-catenin signaling during this same time frame. Together, these findings establish simple selective differentiation strategies for the generation of primitive or definitive hematopoietic progenitors by Wnt-ß-catenin manipulation, and in doing so provide access to enriched populations for future studies on hPSC-derived hematopoietic development.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Vía de Señalización Wnt/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Humanos , Ratones
18.
Cell Rep ; 2(6): 1722-35, 2012 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-23219550

RESUMEN

The efficient generation of hematopoietic stem cells from human pluripotent stem cells is dependent on the appropriate specification of the definitive hematopoietic program during differentiation. In this study, we used T lymphocyte potential to track the onset of definitive hematopoiesis from human embryonic and induced pluripotent stem cells differentiated with specific morphogens in serum- and stromal-free cultures. We show that this program develops from a progenitor population with characteristics of hemogenic endothelium, including the expression of CD34, VE-cadherin, GATA2, LMO2, and RUNX1. Along with T cells, these progenitors display the capacity to generate myeloid and erythroid cells. Manipulation of Activin/Nodal signaling during early stages of differentiation revealed that development of the definitive hematopoietic progenitor population is not dependent on this pathway, distinguishing it from primitive hematopoiesis. Collectively, these findings demonstrate that it is possible to generate T lymphoid progenitors from pluripotent stem cells and that this lineage develops from a population whose emergence marks the onset of human definitive hematopoiesis.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Pluripotentes/metabolismo , Células Precursoras de Linfocitos T/metabolismo , Transducción de Señal/fisiología , Linfocitos T/metabolismo , Línea Celular , Humanos , Células Madre Pluripotentes/citología , Células Precursoras de Linfocitos T/citología , Linfocitos T/citología
19.
Dev Cell ; 23(1): 45-57, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22749417

RESUMEN

Primitive erythropoiesis defines the onset of hematopoiesis in the yolk sac of the early embryo and is initiated by the emergence of progenitors assayed as colony-forming cells (EryP-CFCs). EryP-CFCs are detected for only a narrow window during embryonic development, suggesting that both their initiation and termination are tightly controlled. Using the embryonic stem differentiation system to model primitive erythropoiesis, we found that miR-126 regulates the termination of EryP-CFC development. Analyses of miR-126 null embryos revealed that this miR also regulates EryP-CFCs in vivo. We identified vascular cell adhesion molecule-1 (Vcam-1) expressed by a mesenchymal cell population as a relevant target of miR-126. Interaction of EryP-CFCs with Vcam-1 accelerated their maturation to ßh1-globin(+) and Ter119(+) cells through a Src family kinase. These findings uncover a cell nonautonomous regulatory pathway for primitive erythropoiesis that may provide insight into the mechanism(s) controlling the developmental switch from primitive to definitive hematopoiesis.


Asunto(s)
Células Madre Embrionarias/citología , Células Precursoras Eritroides/citología , Eritropoyesis/fisiología , MicroARNs/fisiología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , Células Madre Embrionarias/fisiología , Eritroblastos/citología , Eritroblastos/fisiología , Células Precursoras Eritroides/fisiología , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Osteoblastos/citología , Osteoblastos/fisiología , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
PLoS One ; 5(1): e8523, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20049087

RESUMEN

Satellite cells (SCs) represent a distinct lineage of myogenic progenitors responsible for the postnatal growth, repair and maintenance of skeletal muscle. Distinguished on the basis of their unique position in mature skeletal muscle, SCs were considered unipotent stem cells with the ability of generating a unique specialized phenotype. Subsequently, it was demonstrated in mice that opposite differentiation towards osteogenic and adipogenic pathways was also possible. Even though the pool of SCs is accepted as the major, and possibly the only, source of myonuclei in postnatal muscle, it is likely that SCs are not all multipotent stem cells and evidences for diversities within the myogenic compartment have been described both in vitro and in vivo. Here, by isolating single fibers from rat flexor digitorum brevis (FDB) muscle we were able to identify and clonally characterize two main subpopulations of SCs: the low proliferative clones (LPC) present in major proportion (approximately 75%) and the high proliferative clones (HPC), present instead in minor amount (approximately 25%). LPC spontaneously generate myotubes whilst HPC differentiate into adipocytes even though they may skip the adipogenic program if co-cultured with LPC. LPC and HPC differ also for mitochondrial membrane potential (DeltaPsi(m)), ATP balance and Reactive Oxygen Species (ROS) generation underlying diversities in metabolism that precede differentiation. Notably, SCs heterogeneity is retained in vivo. SCs may therefore be comprised of two distinct, though not irreversibly committed, populations of cells distinguishable for prominent differences in basal biological features such as proliferation, metabolism and differentiation. By these means, novel insights on SCs heterogeneity are provided and evidences for biological readouts potentially relevant for diagnostic purposes described.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Clonales , Músculo Esquelético/citología , Adenosina Trifosfato/metabolismo , Animales , Animales Modificados Genéticamente , Músculo Esquelético/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA