Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36747642

RESUMO

Post-transcriptional regulation of gene expression by RNA-binding proteins helps facilitate fast, clean transitions from one cell state to the next during germ cell differentiation. Previously we showed that the RNA helicase YTHDC2 is required for germ cells to properly switch from mitosis to meiosis (Bailey et al., 2017). While YTHDC2 protein is first expressed as male germ cells enter meiosis, when it is needed to shut down the mitotic program, YTHDC2 expression continues to increase and reaches its highest levels later in meiotic prophase, in pachytene spermatocytes. Here we show that YTHDC2 has an additional essential role regulating meiotic progression in late spermatocytes during mouse germ cell differentiation. Inducing conditional knockout of Ythdc2 during the first wave of spermatogenesis, after the germ cells have already initiated meiotic prophase, allowed Ythdc2-deficient germ cells to successfully reach the pachytene stage and properly express many meiotic markers. However, instead of continuing through meiotic prophase and initiating the meiotic divisions, late pachytene spermatocytes failed to transition to the diplotene stage and quickly died. Loss of function of Ythdc2 in spermatocytes resulted in changes in transcript levels for a number of genes, some up-regulated and some down-regulated, compared to control mid-stage spermatocytes. YTHDC2 interacts with different proteins in early and late spermatocytes, with many of the interacting proteins involved in post-transcriptional RNA regulation and present in RNA granules, similar to YTHDC2. Our findings suggest that YTHDC2 facilitates proper progression of germ cells through multiple steps of meiosis, potentially via several mechanisms of post-transcriptional RNA regulation.

2.
Bio Protoc ; 12(1): e4286, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-35118177

RESUMO

During development, cells must quickly switch from one cell state to the next to execute precise and timely differentiation. One method to ensure fast transitions in cell states is by controlling gene expression at the post-transcriptional level through action of RNA-binding proteins on mRNAs. The ability to accurately identify the RNA targets of RNA-binding proteins at specific stages is key to understanding the functional role of RNA-binding proteins during development. Here we describe an adapted formaldehyde RNA immunoprecipitation (fRIP) protocol to identify the in vivo RNA targets of a cytoplasmic RNA-binding protein, YTHDC2, from testis, during the first wave of spermatogenesis, at the stage when germ cells are shutting off the proliferative program and initiating terminal differentiation ( Bailey et al., 2017 ). This protocol enables quick and efficient identification of endogenous RNAs bound to an RNA-binding protein, and facilitates the monitoring of stage-specific changes during development.

3.
Elife ; 62017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29087293

RESUMO

The switch from mitosis to meiosis is the key event marking onset of differentiation in the germline stem cell lineage. In Drosophila, the translational repressor Bgcn is required for spermatogonia to stop mitosis and transition to meiotic prophase and the spermatocyte state. Here we show that the mammalian Bgcn homolog YTHDC2 facilitates a clean switch from mitosis to meiosis in mouse germ cells, revealing a conserved role for YTHDC2 in this critical cell fate transition. YTHDC2-deficient male germ cells enter meiosis but have a mixed identity, maintaining expression of Cyclin A2 and failing to properly express many meiotic markers. Instead of continuing through meiotic prophase, the cells attempt an abnormal mitotic-like division and die. YTHDC2 binds multiple transcripts including Ccna2 and other mitotic transcripts, binds specific piRNA precursors, and interacts with RNA granule components, suggesting that proper progression of germ cells through meiosis is licensed by YTHDC2 through post-transcriptional regulation.


Assuntos
Diferenciação Celular , Proliferação de Células , Células Germinativas/enzimologia , Células Germinativas/fisiologia , RNA Helicases/metabolismo , Animais , Regulação da Expressão Gênica , Meiose , Camundongos , Mitose , Ligação Proteica
4.
Cell Stem Cell ; 11(5): 689-700, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23122292

RESUMO

In adult stem cell lineages, progenitor cells commonly undergo mitotic transit amplifying (TA) divisions before terminal differentiation, allowing production of many differentiated progeny per stem cell division. Mechanisms that limit TA divisions and trigger the switch to differentiation may protect against cancer by preventing accumulation of oncogenic mutations in the proliferating population. Here we show that the switch from TA proliferation to differentiation in the Drosophila male germline stem cell lineage is mediated by translational control. The TRIM-NHL tumor suppressor homolog Mei-P26 facilitates accumulation of the differentiation regulator Bam in TA cells. In turn, Bam and its partner Bgcn bind the mei-P26 3' untranslated region and repress translation of mei-P26 in late TA cells. Thus, germ cells progress through distinct, sequential regulatory states, from Mei-P26 on/Bam off to Bam on/Mei-P26 off. TRIM-NHL homologs across species facilitate the switch from proliferation to differentiation, suggesting a conserved developmentally programmed tumor suppressor mechanism.


Assuntos
Células-Tronco Adultas/citologia , Diferenciação Celular , Linhagem da Célula , Drosophila/citologia , Regulação da Expressão Gênica , Células-Tronco Adultas/metabolismo , Animais , Divisão Celular , DNA Helicases/genética , DNA Helicases/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Masculino
5.
Cell Host Microbe ; 8(3): 284-91, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20833379

RESUMO

Human cytomegalovirus (HCMV) is a significant cause of morbidity and mortality in organ transplant recipients. The use of granulocyte-colony stimulating factor (G-CSF)-mobilized stem cells from HCMV seropositive donors is suggested to double the risk of late-onset HCMV disease and chronic graft-versus-host disease in recipients when compared to conventional bone marrow transplantation with HCMV seropositive donors, although the etiology of the increased risk is unknown. To understand mechanisms of HCMV transmission in patients receiving G-CSF-mobilized blood products, we generated a NOD-scid IL2Rγ(c)(null)-humanized mouse model in which HCMV establishes latent infection in human hematopoietic cells. In this model, G-CSF induces the reactivation of latent HCMV in monocytes/macrophages that have migrated into organ tissues. In addition to establishing a humanized mouse model for systemic and latent HCMV infection, these results suggest that the use of G-CSF mobilized blood products from seropositive donors pose an elevated risk for HCMV transmission to recipients.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Macrófagos/virologia , Ativação Viral , Latência Viral , Animais , Antígenos CD34/análise , Células da Medula Óssea/imunologia , Células da Medula Óssea/virologia , Citocinas/sangue , Citomegalovirus/genética , Citomegalovirus/imunologia , Citomegalovirus/isolamento & purificação , Modelos Animais de Doenças , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/virologia , Humanos , Camundongos , Camundongos SCID , Monócitos/virologia
6.
Stem Cell Res ; 4(1): 17-24, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19720572

RESUMO

Recent studies suggest that endothelial cells are a critical component of the normal hematopoietic microenvironment. Therefore, we sought to determine whether primary endothelial cells have the capacity to repair damaged hematopoietic stem cells. Highly purified populations of primary CD31(+) microvascular endothelial cells isolated from the brain or lung did not express the pan hematopoietic marker CD45, most hematopoietic lineage markers, or the progenitor marker c-kit and did not give rise to hematopoietic cells in vitro or in vivo. Remarkably, the transplantation of small numbers of these microvascular endothelial cells consistently restored hematopoiesis following bone marrow lethal doses of irradiation. Analysis of the peripheral blood of rescued recipients demonstrated that both short-term and long-term multilineage hematopoietic reconstitution was exclusively of host origin. Secondary transplantation studies revealed that microvascular endothelial cell-mediated hematopoietic regeneration also occurs at the level of the hematopoietic stem cell. These findings suggest a potential therapeutic role for microvascular endothelial cells in the self-renewal and repair of adult hematopoietic stem cells.


Assuntos
Diferenciação Celular , Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Animais , Biomarcadores , Linhagem da Célula , Separação Celular , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Células-Tronco Hematopoéticas/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo
7.
Blood ; 114(20): 4393-401, 2009 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19759357

RESUMO

Bone morphogenetic protein 4 (BMP4) is required for mesoderm commitment to the hematopoietic lineage during early embryogenesis. However, deletion of BMP4 is early embryonically lethal and its functional role in definitive hematopoiesis is unknown. Consequently, we used a BMP4 hypomorph to investigate the role of BMP4 in regulating hematopoietic stem cell (HSC) function and maintaining steady-state hematopoiesis in the adult. Reporter gene expression shows that Bmp4 is expressed in cells associated with the hematopoietic microenvironment including osteoblasts, endothelial cells, and megakaryocytes. Although resting hematopoiesis is normal in a BMP4-deficient background, the number of c-Kit+, Sca-1+, Lineage- cells is significantly reduced. Serial transplantation studies reveal that BMP4-deficient recipients have a microenvironmental defect that reduces the repopulating activity of wild-type HSCs. This defect is even more pronounced in a parabiosis model that demonstrates a profound reduction in wild-type hematopoietic cells within the bone marrow of BMP4-deficient recipients. Furthermore, wild-type HSCs that successfully engraft into the BMP4-deficient bone marrow show a marked decrease in functional stem cell activity when tested in a competitive repopulation assay. Taken together, these findings indicate BMP4 is a critical component of the hematopoietic microenvironment that regulates both HSC number and function.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Nicho de Células-Tronco/metabolismo , Animais , Apoptose/fisiologia , Western Blotting , Citometria de Fluxo , Expressão Gênica , Camundongos , Parabiose
8.
Proc Natl Acad Sci U S A ; 106(11): 4390-5, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19251665

RESUMO

The expression of ASPP2 (53BP2L), a proapoptotic member of a family of p53-binding proteins, is frequently suppressed in many human cancers. Accumulating evidence suggests that ASPP2 inhibits tumor growth; however, the mechanisms by which ASPP2 suppresses tumor formation remain to be clarified. To study this, we targeted the ASPP2 allele in a mouse by replacing exons 10-17 with a neoR gene. ASPP2(-/-) mice were not viable because of an early embryonic lethal event. Although ASPP2(+/-) mice appeared developmentally normal, they displayed an increased incidence of a variety of spontaneous tumors as they aged. Moreover, gamma-irradiated 6-week-old ASPP2(+/-) mice developed an increased incidence of high-grade T cell lymphomas of thymic origin compared with ASPP2(+/+) mice. Primary thymocytes derived from ASPP2(+/-) mice exhibited an attenuated apoptotic response to gamma-irradiation compared with ASPP2(+/+) thymocytes. Additionally, ASPP2(+/-) primary mouse embryonic fibroblasts demonstrated a defective G(0)/G(1) cell cycle checkpoint after gamma-irradiation. Our results demonstrate that ASPP2 is a haploinsufficient tumor suppressor and, importantly, open new avenues for investigation into the mechanisms by which disruption of ASPP2 pathways could play a role in tumorigenesis and response to therapy.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose/efeitos da radiação , Ciclo Celular/efeitos da radiação , Raios gama , Predisposição Genética para Doença , Heterozigoto , Linfoma de Células T/etiologia , Linfoma de Células T/genética , Camundongos , Camundongos Mutantes , Neoplasias/etiologia , Neoplasias/genética , Timo
9.
PLoS One ; 3(11): e3812, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19043576

RESUMO

BACKGROUND: Although the lymphatic system arises as an extension of venous vessels in the embryo, little is known about the role of circulating progenitors in the maintenance or development of lymphatic endothelium. Here, we investigated whether hematopoietic stem cells (HSCs) have the potential to give rise to lymphatic endothelial cells (LEC). METHODOLOGY/PRINCIPAL FINDINGS: Following the transfer of marked HSCs into irradiated recipients, donor-derived LEC that co-express the lymphatic endothelial markers Lyve-1 and VEGFR-3 were identified in several tissues. HSC-derived LEC persisted for more than 12 months and contributed to approximately 3-4% of lymphatic vessels. Donor-derived LECs were not detected in mice transplanted with common myeloid progenitors and granulocyte/macrophage progenitors, suggesting that myeloid lineage commitment is not a requisite step in HSC contribution to lymphatic endothelium. Analysis of parabiotic mice revealed direct evidence for the existence of functional, circulating lymphatic progenitors in the absence of acute injury. Furthermore, the transplantation of HSCs into Apc(Min/+) mice resulted in the incorporation of donor-derived LEC into the lymphatic vessels of spontaneously arising intestinal tumors. CONCLUSIONS/SIGNIFICANCE: Our results indicate that HSCs can contribute to normal and tumor associated lymphatic endothelium. These findings suggest that the modification of HSCs may be a novel approach for targeting tumor metastasis and attenuating diseases of the lymphatic system.


Assuntos
Endotélio Linfático/citologia , Células-Tronco Hematopoéticas/fisiologia , Transplante de Células-Tronco , Animais , Biomarcadores/análise , Granulócitos , Células-Tronco Hematopoéticas/citologia , Neoplasias Intestinais/patologia , Macrófagos , Camundongos , Células Mieloides
10.
J Exp Med ; 205(8): 1843-58, 2008 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-18625744

RESUMO

In mantle cell lymphoma (MCL) and some cases of multiple myeloma (MM), cyclin D1 expression is deregulated by chromosome translocations involving the immunoglobulin heavy chain (IgH) locus. To evaluate the mechanisms responsible, gene targeting was used to study long-distance gene regulation. Remarkably, these targeted cell lines lost the translocated chromosome (t(11;14)). In these MCL and MM cells, the nonrearranged cyclin D1 (CCND1) locus reverts from CpG hypomethylated to hypermethylated. Reintroduction of the translocated chromosome induced a loss of methylation at the unrearranged CCND1 locus, providing evidence of a transallelic regulatory effect. In these cell lines and primary MCL patient samples, the CCND1 loci are packaged in chromatin-containing CCCTC binding factor (CTCF) and nucleophosmin (NPM) at the nucleolus. We show that CTCF and NPM are bound at the IgH 3' regulatory elements only in the t(11;14) MCL cell lines. Furthermore, NPM short hairpin RNA produces a specific growth arrest in these cells. Our data demonstrate transvection in human cancer and suggest a functional role for CTCF and NPM.


Assuntos
Genes bcl-1 , Linfoma de Célula do Manto/genética , Translocação Genética , Fator de Ligação a CCCTC , Linhagem Celular Tumoral , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 14/genética , Ilhas de CpG , Ciclina D , Ciclinas/genética , Ciclinas/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Marcação de Genes , Genes de Imunoglobulinas , Humanos , Células Híbridas , Linfoma de Célula do Manto/imunologia , Linfoma de Célula do Manto/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/metabolismo , Proteínas Nucleares/metabolismo , Nucleofosmina , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo
11.
Proc Natl Acad Sci U S A ; 103(35): 13156-61, 2006 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-16920790

RESUMO

Despite an important role in vascular development and repair, the origin of endothelial progenitors remains unknown. Accumulating evidence indicates that cells derived from the hematopoietic system participate in angiogenesis. However, the identity and functional role of these cells remain controversial. Here we show that vascular endothelial cells can differentiate from common myeloid progenitors and granulocyte/macrophage progenitors. Endothelial cells derived from transplanted bone marrow-derived myeloid lineage progenitors expressed CD31, von Willebrand factor, and Tie2 but did not express the hematopoietic markers CD45 and F4/80 or the pericyte markers desmin and smooth muscle actin. Lineage tracing analysis in combination with a Tie2-driven Cre/lox reporter system revealed that, in contrast to bone marrow-derived hepatocytes, bone marrow-derived endothelial cells are not the products of cell fusion. The establishment of both hematopoietic and endothelial cell chimerism after parabiosis demonstrates that circulating cells can give rise to vascular endothelium in the absence of acute radiation injury. Our findings indicate that endothelial cells are an intrinsic component of myeloid lineage differentiation and underscore the close functional relationship between the hematopoietic and vascular systems.


Assuntos
Linhagem da Célula , Endotélio Vascular/citologia , Células Mieloides/citologia , Células-Tronco/citologia , Animais , Células da Medula Óssea/citologia , Fusão Celular , Células Endoteliais/citologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Parabiose
12.
Proc Natl Acad Sci U S A ; 103(16): 6321-5, 2006 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-16606845

RESUMO

Transplanted adult bone marrow-derived cells (BMDCs) have been shown to adopt the phenotype and function of several nonhematopoietic cell lineages and promote tumorigenesis. Beyond its cancer enhancing potential, cell fusion has recently emerged as an explanation of how BMDCs regenerate diseased heptocytes, contribute to Purkinje neurons and skeletal and cardiac muscle cells, and participate in skin and heart regeneration. Although bone marrow-derived epithelial cells also have been observed in the intestine, fusion as a mechanism has not been investigated. Here, we show that transplanted BMDCs fuse with both normal and neoplastic intestinal epithelium. Long-term repopulation by donor-derived cells was detected in all principal intestinal epithelial lineages including enterocytes, goblet cells, Paneth cells, and enteroendocrine cells, suggesting that the fusion partners of the BMDCs are long-lived intestinal progenitors or stem cells. Fusion of BMDCs with neoplastic epithelium did not result in tumor initiation. Our findings suggest an unexpected role for BMDCs in both regeneration and tumorigenesis of the intestine.


Assuntos
Células da Medula Óssea/fisiologia , Transformação Celular Neoplásica/patologia , Neoplasias Intestinais/patologia , Intestinos/citologia , Células-Tronco/citologia , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Fusão Celular , Feminino , Proteínas de Fluorescência Verde/análise , Intestinos/patologia , Masculino , Células-Tronco/patologia , Cromossomo Y/química , Cromossomo Y/genética
13.
Nat Med ; 10(7): 744-8, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15195088

RESUMO

Liver repopulation with bone marrow-derived hepatocytes (BMHs) can cure the genetic liver disease fumarylacetoacetate hydrolase (Fah) deficiency. BMHs emerge from fusion between donor bone marrow-derived cells and host hepatocytes. To use such in vivo cell fusion efficiently for therapy requires knowing the nature of the hematopoietic cells that fuse with hepatocytes. Here we show that the transplantation into Fah(-/-) mice of hematopoietic stem cells (HSCs) from lymphocyte-deficient Rag1(-/-) mice, lineage-committed granulocyte-macrophage progenitors (GMPs) or bone marrow-derived macrophages (BMMs) results in the robust production of BMHs. These results provide direct evidence that committed myelomonocytic cells such as macrophages can produce functional epithelial cells by in vivo fusion. Because stable bone marrow engraftment or HSCs are not required for this process, macrophages or their highly proliferative progenitors provide potential for targeted and well-tolerated cell therapy aimed at organ regeneration.


Assuntos
Fusão Celular , Transplante de Células-Tronco Hematopoéticas , Hepatócitos/citologia , Hidrolases/deficiência , Macrófagos/citologia , Animais , Feminino , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL
14.
Blood ; 104(4): 1010-6, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15126319

RESUMO

Platelet endothelial cell adhesion molecule-1 (PECAM-1) (CD31) is an adhesion molecule expressed on endothelial cells and subsets of leukocytes. Analysis of phenotypically defined hematopoietic stem cells (HSCs) from the yolk sac, fetal liver, and adult bone marrow demonstrates CD31 expression on these cells throughout development. CD31+ c-kit+ cells, but not CD31- c-kit+ cells, isolated from day-9.5 yolk sac give rise to multilineage hematopoiesis in vivo. Further evaluation of the CD31+ lineage marker-negative fraction of adult bone marrow reveals functionally distinct cell subsets. Transplantation of CD31+ Lin- c-kit- cells fails to protect lethally irradiated recipients, while CD31+ Lin- c-kit+ Sca-1- cells (CD31+ Sca-1-) provide radioprotection in the absence of long-term donor-derived hematopoiesis. Although donor-derived leukocytes were not detected in CD31+ Sca-1- recipients, donor-derived erythroid cells were transiently produced during the initial phases of bone marrow recovery. These results demonstrate CD31 expression on hematopoietic stem cells throughout ontogeny and identify a population of CD31+ short-term erythroid progenitors cells that confer protection from lethal doses of radiation.


Assuntos
Células Precursoras Eritroides/química , Células-Tronco Hematopoéticas/química , Molécula-1 de Adesão Celular Endotelial a Plaquetas , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Fatores Etários , Animais , Animais Recém-Nascidos , Células da Medula Óssea , Linhagem da Célula/efeitos da radiação , Células Precursoras Eritroides/citologia , Hematopoese/efeitos da radiação , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/efeitos da radiação , Leucócitos/citologia , Fígado/citologia , Fígado/embriologia , Camundongos , Camundongos Endogâmicos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Proteção Radiológica , Saco Vitelino/citologia
15.
Blood ; 103(1): 13-9, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12958072

RESUMO

During early embryogenesis, blood vessels and hematopoietic cells arise from a common precursor cell, the hemangioblast. Recent studies have identified endothelial progenitor cells in the peripheral blood, and there is accumulating evidence that a subset of these cells is derived from precursors in the bone marrow. Here we show that adult bone marrow-derived, phenotypically defined hematopoietic stem cells (c-kit+, Sca-1+, lineage-) give rise to functional endothelial cells. With the exception of the brain, donor-derived cells are rapidly integrated into blood vessels. Durably engrafted endothelial cells express CD31, produce von Willebrand factor, and take up low-density lipoprotein. Analysis of DNA content indicates that donor-derived endothelial cells are not the products of cell fusion. Self-renewal of stem cells with hematopoietic and endothelial cell potential was revealed by serial transplantation studies. The clonal origin of both hematopoietic and endothelial cell outcomes was established by the transfer of a single cell. These results suggest that adult bone marrow-derived hematopoietic stem cells may serve as a reservoir for endothelial cell progenitors.


Assuntos
Endotélio Vascular/citologia , Transplante de Células-Tronco Hematopoéticas , Animais , Diferenciação Celular , Células Clonais/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Feminino , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde , Lipoproteínas LDL/metabolismo , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Recombinantes/metabolismo
16.
Exp Hematol ; 31(11): 987-93, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14585360

RESUMO

Classical studies of the developing embryo first suggested the existence of the hemangioblast, a precursor cell with the potential to differentiate into both blood and blood vessels. Several lines of investigation demonstrated that many of the genes activated during early hematopoietic development are also expressed in the vascular endothelium. Gene-targeting studies using embryonic stem cells have identified Flk-1, SCL, and Runx-1 as important regulatory molecules that specify both hematopoietic and vascular outcomes. Although it was anticipated that the hemangioblast would be present only during the earliest stages of vascular development in the yolk sac, accumulating evidence now indicates that hematopoietic cells with hemangioblast activity persist into adulthood. In the adult, bone marrow-derived, circulating endothelial progenitors contribute to postnatal neovascularization and enhance vascular repair following ischemic injury. Highly purified populations of hematopoietic stem cells from humans and mice can differentiate into both blood cells and vascular tissue at the single cell level. These recent findings suggest that bone marrow-derived hematopoietic stem cells or their progeny may contribute to the maintenance and repair of both the hematopoietic and the vascular systems during adult life.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Animais , Diferenciação Celular , Embrião de Mamíferos/citologia , Endotélio Vascular/citologia , Humanos , Neovascularização Fisiológica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...