Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 39(5): 636-649, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33480126

RESUMEN

Angiotensin-converting enzyme (ACE), a key element of the renin-angiotensin system (RAS), has recently been identified as a new marker of both adult and embryonic human hematopoietic stem/progenitor cells (HSPCs). However, whether a full renin-angiotensin pathway is locally present during the hematopoietic emergence is still an open question. In the present study, we show that this enzyme is expressed by hematopoietic progenitors in the developing mouse embryo. Furthermore, ACE and the other elements of RAS-namely angiotensinogen, renin, and angiotensin II type 1 (AT1) and type 2 (AT2) receptors-are expressed in the paraaortic splanchnopleura (P-Sp) and in its derivative, the aorta-gonad-mesonephros region, both in human and mouse embryos. Their localization is compatible with the existence of a local autocrine and/or paracrine RAS in these hemogenic sites. in vitro perturbation of the RAS by administration of a specific AT1 receptor antagonist inhibits almost totally the generation of blood CD45-positive cells from dissected P-Sp, implying that angiotensin II signaling is necessary for the emergence of hematopoietic cells. Conversely, addition of exogenous angiotensin II peptide stimulates hematopoiesis in culture, with an increase in the number of immature c-Kit+ CD41+ CD31+ CD45+ hematopoietic progenitors, compared to the control. These results highlight a novel role of local-RAS during embryogenesis, suggesting that angiotensin II, via activation of AT1 receptor, promotes the emergence of undifferentiated hematopoietic progenitors.


Asunto(s)
Angiotensina II/genética , Angiotensinógeno/genética , Células Madre Hematopoyéticas/citología , Receptor de Angiotensina Tipo 1/genética , Sistema Renina-Angiotensina/genética , Animales , Aorta/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/genética , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Antígenos Comunes de Leucocito/genética , Ratones , Péptidos/farmacología , Peptidil-Dipeptidasa A/genética , Receptor de Angiotensina Tipo 2/genética , Renina/genética , Transducción de Señal/efectos de los fármacos , Células Madre/citología
2.
Blood ; 119(16): 3712-23, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-22282502

RESUMEN

Adult-type lympho-myeloid hematopoietic progenitors are first generated in the aorta-gonad-mesonephros region between days 27 and 40 of human embryonic development, but an elusive blood forming potential is present earlier in the underlying splanchnopleura. In the present study, we show that angiotensin-converting enzyme (ACE, also known as CD143), a recently identified cell-surface marker of adult human hematopoietic stem cells, is already expressed in all presumptive and developing blood-forming tissues of the human embryo and fetus: para-aortic splanchnopleura, yolk sac, aorta-gonad-mesonephros, liver, and bone marrow (BM). Fetal liver and BM-derived CD34(+)ACE(+) cells, but not CD34(+)ACE(-) cells, are endowed with long-term culture-initiating cell potential and sustain multilineage hematopoietic cell engraftment when transplanted into NOD/SCID mice. Furthermore, from 23-26 days of development, ACE expression characterizes rare CD34(-)CD45(-) cells concentrated in the hemogenic portion of the para-aortic splanchnopleura. ACE(+) cells sorted from the splanchnopleura generated colonies of hematopoietic cells more than 40 times more frequently than ACE(-) cells. These data suggest that, in addition to being a marker of adult human hematopoietic stem cells, ACE identifies embryonic mesodermal precursors responsible for definitive hematopoiesis, and we propose that this enzyme is involved in the regulation of human blood formation.


Asunto(s)
Médula Ósea/embriología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hígado/embriología , Peptidil-Dipeptidasa A/metabolismo , Animales , Antígenos CD34/metabolismo , Linfocitos B/citología , Linaje de la Célula/fisiología , Femenino , Granulocitos/citología , Trasplante de Células Madre Hematopoyéticas , Humanos , Células Asesinas Naturales/citología , Antígenos Comunes de Leucocito/metabolismo , Hígado/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T/citología , Trasplante Heterólogo
3.
Cell Death Dis ; 15(5): 305, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693109

RESUMEN

Zebrafish is widely adopted as a grafting model for studying human development and diseases. Current zebrafish xenotransplantations are performed using embryo recipients, as the adaptive immune system, responsible for host versus graft rejection, only reaches maturity at juvenile stage. However, transplanted primary human hematopoietic stem/progenitor cells (HSC) rapidly disappear even in zebrafish embryos, suggesting that another barrier to transplantation exists before the onset of adaptive immunity. Here, using a labelled macrophage zebrafish line, we demonstrated that engraftment of human HSC induces a massive recruitment of macrophages which rapidly phagocyte transplanted cells. Macrophages depletion, by chemical or pharmacological treatments, significantly improved the uptake and survival of transplanted cells, demonstrating the crucial implication of these innate immune cells for the successful engraftment of human cells in zebrafish. Beyond identifying the reasons for human hematopoietic cell engraftment failure, this work images the fate of human cells in real time over several days in macrophage-depleted zebrafish embryos.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Macrófagos , Pez Cebra , Pez Cebra/embriología , Animales , Macrófagos/metabolismo , Humanos , Células Madre Hematopoyéticas/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Embrión no Mamífero/metabolismo , Trasplante Heterólogo , Fagocitosis
4.
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
5.
Stem Cells Transl Med ; 11(1): 35-43, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35641167

RESUMEN

The vascular wall is comprised of distinct layers controlling angiogenesis, blood flow, vessel anchorage within organs, and cell and molecule transit between blood and tissues. Moreover, some blood vessels are home to essential stem-like cells, a classic example being the existence in the embryo of hemogenic endothelial cells at the origin of definitive hematopoiesis. In recent years, microvascular pericytes and adventitial perivascular cells were observed to include multi-lineage progenitor cells involved not only in organ turnover and regeneration but also in pathologic remodeling, including fibrosis and atherosclerosis. These perivascular mesodermal elements were identified as native forerunners of mesenchymal stem cells. We have presented in this brief review our current knowledge on vessel wall-associated tissue remodeling cells with respect to discriminating phenotypes, functional diversity in health and disease, and potential therapeutic interest.


Asunto(s)
Células Madre Mesenquimatosas , Células Madre de Sangre Periférica , Células Endoteliales , Humanos , Células Madre Mesenquimatosas/fisiología , Pericitos , Células Madre/fisiología
6.
J Vis Exp ; (171)2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34096917

RESUMEN

Bone marrow megakaryocytes are large polyploid cells that ensure the production of blood platelets. They arise from hematopoietic stem cells through megakaryopoiesis. The final stages of this process are complex and classically involve the bipotent Megakaryocyte-Erythrocyte Progenitors (MEP) and the unipotent Megakaryocyte Progenitors (MKp). These populations precede the formation of bona fide megakaryocytes and, as such, their isolation and characterization could allow for the robust and unbiased analysis of megakaryocyte formation. This protocol presents in detail the procedure to collect hematopoietic cells from mouse bone marrow, the enrichment of hematopoietic progenitors through magnetic depletion and finally a cell sorting strategy that yield highly purified MEP and MKp populations. First, bone marrow cells are collected from the femur, the tibia, and also the iliac crest, a bone that contains a high number of hematopoietic progenitors. The use of iliac crest bones drastically increases the total cell number obtained per mouse and thus contributes to a more ethical use of animals. A magnetic lineage depletion was optimized using 450 nm magnetic beads allowing a very efficient cell sorting by flow cytometry. Finally, the protocol presents the labeling and gating strategy for the sorting of the two highly purified megakaryocyte progenitor populations: MEP (Lin-Sca-1-c-Kit+CD16/32-CD150+CD9dim) and MKp (Lin- Sca-1-c-Kit+CD16/32-CD150+CD9bright). This technique is easy to implement and provides enough cellular material to perform i) molecular characterization for a deeper knowledge of their identity and biology, ii) in vitro differentiation assays, that will provide a better understanding of the mechanisms of maturation of megakaryocytes, or iii) in vitro models of interaction with their microenvironment.


Asunto(s)
Células Progenitoras de Megacariocitos , Megacariocitos , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Separación Celular/métodos , Células Madre Hematopoyéticas/citología , Células Progenitoras de Megacariocitos/citología , Megacariocitos/citología , Ratones
7.
Mol Oncol ; 15(9): 2318-2329, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33960108

RESUMEN

The intestine-specific caudal-related homeobox gene-2 (CDX2) homeobox gene, while being a tumor suppressor in the gut, is ectopically expressed in a large proportion of acute leukemia and is associated with poor prognosis. Here, we report that turning on human CDX2 expression in the hematopoietic lineage of mice induces acute monoblastic leukemia, characterized by the decrease in erythroid and lymphoid cells at the benefit of immature monocytic and granulocytic cells. One of the highly stimulated genes in leukemic bone marrow cells was BMP and activin membrane-bound inhibitor (Bambi), an inhibitor of transforming growth factor-ß (TGF-ß) signaling. The CDX2 protein was shown to bind to and activate the transcription of the human BAMBI promoter. Moreover, in a leukemic cell line established from CDX2-expressing mice, reducing the levels of CDX2 or Bambi stimulated the TGF-ß-dependent expression of Cd11b, a marker of monocyte maturation. Taken together, this work demonstrates the strong oncogenic potential of the homeobox gene CDX2 in the hematopoietic lineage, in contrast with its physiological tumor suppressor activity exerted in the gut. It also reveals, through BAMBI and TGF-ß signaling, the involvement of CDX2 in the perturbation of the interactions between leukemia cells and their microenvironment.


Asunto(s)
Factor de Transcripción CDX2/genética , Leucemia Monocítica Aguda/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Antígeno CD11b/genética , Linaje de la Célula , Humanos , Leucemia Monocítica Aguda/patología , Proteínas de la Membrana/genética , Ratones , Transducción de Señal , Microambiente Tumoral
8.
Leukemia ; 35(5): 1463-1474, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33833385

RESUMEN

B-cell receptor (BCR) signaling is crucial for the pathophysiology of most mature B-cell lymphomas/leukemias and has emerged as a therapeutic target whose effectiveness remains limited by the occurrence of mutations. Therefore, deciphering the cellular program activated downstream this pathway has become of paramount importance for the development of innovative therapies. Using an original ex vivo model of BCR-induced proliferation of chronic lymphocytic leukemia cells, we generated 108 temporal transcriptional and proteomic profiles from 1 h up to 4 days after BCR activation. This dataset revealed a structured temporal response composed of 13,065 transcripts and 4027 proteins, comprising a leukemic proliferative signature consisting of 430 genes and 374 proteins. Mathematical modeling of this complex cellular response further highlighted a transcriptional network driven by 14 early genes linked to proteins involved in cell proliferation. This group includes expected genes (EGR1/2, NF-kB) and genes involved in NF-kB signaling modulation (TANK, ROHF) and immune evasion (KMO, IL4I1) that have not yet been associated with leukemic cells proliferation. Our study unveils the BCR-activated proliferative genetic program in primary leukemic cells. This approach combining temporal measurements with modeling allows identifying new putative targets for innovative therapy of lymphoid malignancies and also cancers dependent on ligand-receptor interactions.


Asunto(s)
Linfocitos B/metabolismo , Proliferación Celular/genética , Leucemia Linfocítica Crónica de Células B/genética , Receptores de Antígenos de Linfocitos B/genética , Anciano , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Activación de Linfocitos/genética , Masculino , Persona de Mediana Edad , Proteoma/genética , Proteómica/métodos , Transducción de Señal/genética , Transcripción Genética/genética
9.
Brain Res ; 1230: 50-60, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18671952

RESUMEN

Human embryonic stem cells (hESC) have been directed to differentiate into CNS cells with clinical importance. However, for study of development and regeneration of the human PNS, and peripheral neuropathies, it would be useful to have a source of human PNS derivatives. We have demonstrated that peripheral sensory neuron-like cells (PSN) can also be derived from hESC via neural crest-like (NC) intermediates, and from neural progenitors induced from hESC using noggin. Here we report the generation of higher purity PSN from passagable neurospheres (NSP) induced by murine PA6 stromal cells. hESC were cultured with PA6, and colonies that developed a specific morphology were cut from the plates. Culture of these colonies under non-adhesive conditions yielded NSPs. Several NC marker genes were expressed in the NSP, and these were also detected in 3-5week gestation human embryos containing migrating NC. These NSPs passaged for 2-8weeks and re-plated on PA6 gave rise to many Brn3a+/peripherin+ cells, characteristic of early sensory-like neurons. Re-culturing PA6-induced NSP cells with PA6 resulted in about 25% of the human cells in the co-cultures differentiating to PSN after 1week, compared to only about 10% PSN obtained after 3 weeks when noggin-induced NSP were used. Two month adherent cultures of PA6-induced NSP cells contained neurons expressing several PSN neuropeptides, and voltage-dependent currents and action potentials were obtained from a molecularly identified PSN. hESC-derived PA6-induced NSP cells are therefore an excellent potential source of human PSN for study of differentiation and modeling of PNS disease.


Asunto(s)
Células Madre Embrionarias/fisiología , Cresta Neural/fisiología , Células Receptoras Sensoriales/fisiología , Biomarcadores , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Adhesión Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Análisis Citogenético , Electrofisiología , Células Madre Embrionarias/metabolismo , Humanos , Inmunohistoquímica , Cresta Neural/citología , Cresta Neural/metabolismo , Neuropéptidos/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Receptoras Sensoriales/metabolismo
11.
Ann N Y Acad Sci ; 1106: 223-32, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17360801

RESUMEN

Human hematopoiesis proceeds transiently in the extraembryonic yolk sac and embryonic, then fetal liver before being stabilized in the bone marrow during the third month of gestation. In addition to this classic developmental sequence, we have previously shown that the aorta-gonad-mesonephros (AGM) embryonic territory produces stem cells for definitive hematopoiesis from 27 to 40 days of human development, through an intermediate blood-forming endothelium stage. These studies have relied on the use of traditional markers of human hematopoietic and endothelial cells. In addition, we have recently identified and characterized a novel surface molecule, BB9, which typifies the earliest founders of the human angiohematopoietic system. BB9, which was initially identified with a monoclonal antibody raised to Stro-1(+) bone marrow stromal cells, recognizes in the adult the most primitive Thy-1(+) CD133(+) Lin(-), non-obese diabetic--severe combined immunodeficiency disease (NOD-SCID) mouse engrating hematopoietic stem cells (HSCs). In the 3- to 4-week embryo, BB9 expression typifies a subset of splanchnopleural mesodermal cells that migrate dorsally and colonize the ventral aspect of the aorta where they establish a population of hemogenic endothelial cells. We have indeed confirmed that hematopoietic potential in the human embryo, as assessed by long-term culture-initiating cell (LTC-IC) and SCID mouse reconstituting cell (SRC) activities, is confined to BB9-expressing cells. We have further validated these results in the model of human embryonic stem cells (hESCs) in which we have modeled, through the development of hematopoietic embryoid bodies (EBs), primitive and definitive hematopoieses. In this setting, we have documented the emergence of BB9(+) hemangioblast-like clonogenic angiohematopoietic progenitors that currently represent the earliest known founders of the human vascular and blood systems.


Asunto(s)
Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Neovascularización Fisiológica , Antígeno AC133 , Animales , Antígenos CD/biosíntesis , Movimiento Celular , Células Endoteliales/citología , Glicoproteínas/biosíntesis , Hematopoyesis , Humanos , Hígado/embriología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Biológicos , Péptidos
12.
Trends Cardiovasc Med ; 16(3): 95-101, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16546690

RESUMEN

During the early weeks of human gestation, hematopoietic cells first emerge within the extraembryonic yolk sac (primitive hematopoiesis) and secondarily within the truncal arteries of the embryo. This second wave includes the stem cells giving rise to adult-type lymphohematopoiesis. In both yolk sac blood islands and embryonic aorta, hematopoietic cells arise in the immediate vicinity of vascular endothelial cells. In vitro hematopoietic differentiation of endothelial cells stringently sorted from human embryonic and fetal blood-forming tissues has demonstrated that primitive endothelium lies at the origin of incipient hematopoiesis. These anatomically and temporally localized blood-forming endothelial cells are ultimately derived from a rare subset of mesodermal angio-hematopoietic stem cells, or hemangioblasts. The evidence for an early progenitor of blood-forming cells within the walls of human embryonic blood vessels concurs with parallel data obtained from lower vertebrate, avian, and murine models. Importantly, converging results have recently been obtained with in vitro differentiated human embryonic stem cells, in which we have modeled primitive and definitive hematopoiesis via an endothelium-like developmental intermediate.


Asunto(s)
Endotelio/citología , Hematopoyesis , Células Madre/citología , Saco Vitelino/crecimiento & desarrollo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/citología , Endotelio/embriología , Humanos
13.
Exp Hematol ; 33(9): 1062-9, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16140155

RESUMEN

The hematopoietic system is indispensable from the earliest stages of development and adapts to the rapidly changing anatomy of the embryo and fetus; this takes place in such different anatomic locations as the yolk sac blood island, hepatic parenchyme, aorta-gonads-mesonephros paravascular mesenchyme, and bone marrow primary logette. We herein summarize our investigation of these serial blood-forming events in the human embryo and fetus. The access to early stages of human development, availability of a large panoply of molecular markers for human blood cell lineages, and recent development of robust assays for the earliest human hematopoietic stem cells have allowed us to gain relatively clear insight into the developmental sequence that underlies the ontogeny of human blood cells. Conversely, the control exerted by these diverse cellular environments on the emergence of human hematopoietic cells remains elusive, as is the case in animal models. We nonetheless present preliminary attempts to decipher the structure and molecular characteristics of the distinct cellular "niches" in which blood cells are produced during human gestation.


Asunto(s)
Desarrollo Embrionario , Hematopoyesis/fisiología , Biomarcadores , Linaje de la Célula , Inducción Embrionaria , Femenino , Humanos , Embarazo , Saco Vitelino/irrigación sanguínea
14.
Int J Dev Biol ; 49(2-3): 243-50, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15906238

RESUMEN

Human hematopoiesis is initiated in the yolk sac during the third week of development. At the same time the capacity to produce blood cells also arises in the embryo, within the splanchnopleura, but this potential is not expressed before day 27, when clustered hematopoietic stem cells emerge from the ventral wall of the aorta and vitelline artery. Budding of hematopoietic cells from vessel walls reflects the re-differentiation of local endothelial cells, which are likely derived from angio-hematopoietic mesodermal ancestors emigrated from the splanchnopleura. Yolk sac-derived stem cells are limited to myelo-erythroid development, whereas those born in the embryo are, in addition, lymphopoietic and therefore represent the first multi-potent, adult-type blood progenitors that appear in human ontogeny, preceding shortly the onset of liver hematopoiesis. These results allowed the establishment of a novel hierarchy of blood-forming tissues in human development and induced an in depth reconsideration of the very origin of definitive human hematopoiesis. These results also fully corroborate the outcome of experiments performed in parallel in avian and mouse embryos and point to the conservation in all higher vertebrates of an ancestral route of blood cell production via embryonic vessel walls.


Asunto(s)
Desarrollo Embrionario , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Diferenciación Celular , Células Madre Hematopoyéticas/fisiología , Humanos , Hígado/embriología , Modelos Biológicos , Fenotipo , Saco Vitelino/fisiología
15.
FEBS Lett ; 590(22): 3987-4001, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27597316

RESUMEN

The continuous generation of blood cells throughout life relies on the existence of hematopoietic stem cells (HSC) generated during embryogenesis. Given the importance of HSC transplantation in cell-based therapeutic approaches, considerable efforts have been made toward understanding the developmental origins of embryonic HSC. Adult-type HSC are first generated in the aorta-gonad-mesonephros (AGM) region between days 27 and 40 of human embryonic development, but an elusive blood-forming potential is present earlier in the underlying splanchnopleura. It is relatively well accepted that the HSC emerge in the AGM through a hemogenic endothelium, but the direct precursor of this cell type remains to be clearly identified. This review is intended to summarize the recent advances made to understand the origins of hematopoietic stem cells in the early human embryo. In addition, we discuss in detail the discovery of the angiotensin-converting enzyme (ACE) as a novel marker of human HSC and of prehematopoietic precursors inside the embryo.


Asunto(s)
Desarrollo Embrionario/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Peptidil-Dipeptidasa A/genética , Adulto , Animales , Aorta/crecimiento & desarrollo , Embrión de Mamíferos , Femenino , Gónadas/crecimiento & desarrollo , Humanos , Ratones , Embarazo
17.
Ann N Y Acad Sci ; 1044: 41-50, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15958696

RESUMEN

We have characterized the emerging hematopoietic system in the human embryo and fetus. Two embryonic organs, the yolk sac and aorta, support the primary emergence of hematopoietic stem cells (HSCs), but only the latter contributes lymphomyeloid stem cells for definitive, adult-type hematopoiesis. A common feature of intra- and extraembryonic hematopoiesis is that in both locations hematopoietic cells emerge in close vicinity to vascular endothelial cells. We have provided evidence that a population of angiohematopoietic mesodermal stem cells, marked by the expression of flk-1 and the novel BB9/ACE antigen, migrate from the paraaortic splanchnopleura into the ventral part of the aorta, where they give rise to hemogenic endothelial cells and, in turn, hematopoietic cells. HSCs also appear to develop from endothelium in the embryonic liver and fetal bone marrow, albeit at a much lower frequency. This would imply that the organism does not function during its whole life on a stock of hematopoietic stem cells established in the early embryo, as is usually accepted. We next examined whether the vessel wall can contribute stem cells for other cell lineages, primarily in the model of adult skeletal muscle regeneration. By immunohistochemistry and flow cytometry, we documented the existence in skeletal muscle, besides genuine endothelial and myogenic cells, of a subset of satellite cells that coexpress endothelial cell markers. This suggested the existence of a continuum of differentiation from vascular cells to endothelial cells that was confirmed in long-term culture. The regenerating capacity of these cells expressing both myogenic and endothelial markers is being investigated in skeletal and cardiac muscle, and the results are being compared with those generated by satellite cells. Altogether, these results point to a generalized progenitor potential of a subset of endothelial, or endothelium-like, cells in blood vessel walls, in pre- and postnatal life.


Asunto(s)
Endotelio Vascular/citología , Células Madre Hematopoyéticas/citología , Anticuerpos Monoclonales/metabolismo , Aorta/citología , Células Cultivadas , Humanos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Saco Vitelino/citología
18.
Methods Mol Med ; 105: 413-24, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15492411

RESUMEN

We describe here diverse methods used to study the onset of hematopoiesis in the human embryo and fetus. In the first part of this chapter, the criteria for estimating developmental stages in human embryos are discussed. This section also presents in detail a refined method for embedding and freezing intact human embryonic tissues that are destined to be analyzed by histology and immunostaining. In the second section, several protocols for the microdissection of human embryos are described in detail, with special attention given to differences encountered between tissues at different ages of gestation. Because of the limited number of cells available at the early stages of human gestation, we have established a miniaturized cell amplification system permitting further development of intact organ rudiments dissected from a human embryo and cultured in toto. The last part of the chapter is devoted to the study of myeloid and lymphoid potentials using, respectively, a mouse bone marrow-derived stromal cell line and cultured mouse embryonic thymus rudiments.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Embrión de Mamíferos/embriología , Células Madre Hematopoyéticas/fisiología , Hematopoyesis , Humanos , Técnicas de Cultivo de Órganos/métodos
19.
Nat Commun ; 6: 6227, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25686881

RESUMEN

In vertebrates, haematopoietic stem/progenitor cells (HSPCs) first emerge in the aorta-gonad-mesonephros (AGM) before colonizing transitory and subsequently definitive haematopoietic organs allowing haematopoiesis throughout adult life. Here we identify an unexpected primitive macrophage population accumulated in the dorsal mesenteric mesoderm surrounding the dorsal aorta of the human embryo and study its function in the transparent zebrafish embryo. Our study reveals dynamic interactions occurring between the HSPCs and primitive macrophages in the AGM. Specific chemical and inducible genetic depletion of macrophages or inhibition of matrix metalloproteinases (Mmps) leads to an accumulation of HSPCs in the AGM and a decrease in the colonization of haematopoietic organs. Finally, in vivo zymography demonstrates the function of primitive macrophages in extracellular matrix degradation, which allows HSPC migration through the AGM stroma, their intravasation, leading to the colonization of haematopoietic organs and the establishment of definitive haematopoiesis.


Asunto(s)
Aorta/embriología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Macrófagos/citología , Células Madre/citología , Animales , Animales Modificados Genéticamente , Linaje de la Célula , Biología Evolutiva , Matriz Extracelular/metabolismo , Gónadas/embriología , Humanos , Macrófagos/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Mesonefro/embriología , Microscopía Fluorescente , Pez Cebra
20.
Ann N Y Acad Sci ; 996: 132-40, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12799291

RESUMEN

Two waves of hematopoietic stem cell generation take place in the first month of human gestation. The first one has long been known to occur in the yolk sac; only recently was another one identified that results in the development, from the 27th day, of clusters of hematopoietic cells on the ventral endothelium of the aorta and vitelline artery. This latter, intra-embryonic phase of blood cell progenitor production is undoubtedly local since its presumptive territory of occurrence, the para-aortic splanchnopleura, is endowed with blood-forming potential in vitro from, at least, day 19 of development. The first multipotent, lympho-myeloid stem cells emerge within that intra-embryonic territory, whereas the yolk sac only produces myeloid precursor cells devoid of lymphoid potential. The forerunners of the hematopoietic stem cells born to intra-embryonic arteries appear to migrate from the splanchnopleura as KDR(+) angio-hematopoietic cells that colonize the ventral wall of the aorta to give rise to hematogenous endothelium. All these results indicate that stem cells for human definitive hematopoiesis emerge within the embryo, in the walls of truncal arteries, from splanchnopleural ancestors and through a vascular endothelial intermediate stage. Less expectedly, blood-forming endothelial cells were also encoutered in the embryonic and fetal liver and in the fetal and, even, adult bone marrow, suggesting that a filiation between vascular and hematopoietic cells may persist during the whole life.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/citología , Feto/citología , Células Madre Hematopoyéticas/citología , Antígenos CD34/metabolismo , Desarrollo Embrionario y Fetal , Endotelio Vascular/citología , Femenino , Humanos , Antígenos Comunes de Leucocito/metabolismo , Mesodermo/citología , Embarazo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA