Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Cell ; 135(6): 1118-29, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-19062086

RESUMO

Bone marrow hematopoietic stem cells (HSCs) are crucial to maintain lifelong production of all blood cells. Although HSCs divide infrequently, it is thought that the entire HSC pool turns over every few weeks, suggesting that HSCs regularly enter and exit cell cycle. Here, we combine flow cytometry with label-retaining assays (BrdU and histone H2B-GFP) to identify a population of dormant mouse HSCs (d-HSCs) within the lin(-)Sca1+cKit+CD150+CD48(-)CD34(-) population. Computational modeling suggests that d-HSCs divide about every 145 days, or five times per lifetime. d-HSCs harbor the vast majority of multilineage long-term self-renewal activity. While they form a silent reservoir of the most potent HSCs during homeostasis, they are efficiently activated to self-renew in response to bone marrow injury or G-CSF stimulation. After re-establishment of homeostasis, activated HSCs return to dormancy, suggesting that HSCs are not stochastically entering the cell cycle but reversibly switch from dormancy to self-renewal under conditions of hematopoietic stress.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Adultas/fisiologia , Animais , Antígenos de Diferenciação/metabolismo , Medula Óssea/fisiologia , Bromouracila/análogos & derivados , Fluoruracila/metabolismo , Proteínas de Fluorescência Verde , Células-Tronco Hematopoéticas/fisiologia , Homeostase , Camundongos , Camundongos Transgênicos , Uridina/análogos & derivados , Uridina/metabolismo
2.
J Immunol ; 183(10): 6422-31, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19880447

RESUMO

The pool of hematopoietic stem/progenitor cells, which provide life-long reconstitution of all hematopoietic lineages, is tightly controlled and regulated by self-renewal and apoptosis. Histone modifiers and chromatin states are believed to govern establishment, maintenance, and propagation of distinct patterns of gene expression in stem cells, however the underlying mechanism remains poorly understood. In this study, we identified a role for the histone acetytransferase cofactor Trrap in the maintenance of hematopietic stem/progenitor cells. Conditional deletion of the Trrap gene in mice resulted in ablation of bone marrow and increased lethality. This was due to the depletion of early hematopoietic progenitors, including hematopoietic stem cells, via a cell-autonomous mechanism. Analysis of purified bone marrow progenitors revealed that these defects are associated with induction of p53-independent apoptosis and deregulation of Myc transcription factors. Together, this study has identified a critical role for Trrap in the mechanism that maintains hematopoietic stem cells and hematopoietic system, and underscores the importance of Trrap and histone modifications in tissue homeostasis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Coenzimas/imunologia , Células-Tronco Hematopoéticas/imunologia , Histona Acetiltransferases/imunologia , Proteínas Nucleares/imunologia , Proteína Supressora de Tumor p53/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/imunologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Coenzimas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Histona Acetiltransferases/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
Ann N Y Acad Sci ; 1106: 64-75, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17442778

RESUMO

In the mouse, over the last 20 years, a set of cell-surface markers and activities have been identified, enabling the isolation of bone marrow (BM) populations highly enriched in hematopoietic stem cells (HSCs). These HSCs have the ability to generate multiple lineages and are capable of long-term self-renewal activity such that they are able to reconstitute and maintain a functional hematopoietic system after transplantation into lethally irradiated recipients. Using single-cell reconstitution assays, various marker combinations can be used to achieve a functional HSC purity of almost 50%. Here we have used the differential expression of six of these markers (Sca1, c-Kit, CD135, CD48, CD150, and CD34) on lineage-depleted BM to refine cell hierarchies within the HSC population. At the top of the hierarchy, we propose a dormant HSC population (Lin(-)Sca1(+)c-Kit(+) CD48(-)CD150(+)CD34(-)) that gives rise to an active self-renewing CD34(+) HSC population. HSC dormancy, as well as the balance between self-renewal and differentiation activity, is at least, in part, controlled by the stem cell niches individual HSCs are attached to. Here we review the current knowledge about HSC niches and propose that dormant HSCs are located in niches at the endosteum, whereas activated HSCs are in close contact to sinusoids of the BM microvasculature.


Assuntos
Células-Tronco Hematopoéticas/citologia , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Regulação da Expressão Gênica , Camundongos , Modelos Biológicos , Modelos Genéticos , Osteoblastos/metabolismo , Fenótipo
4.
J Exp Med ; 210(11): 2337-49, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-24127490

RESUMO

The phosphatase and tumor suppressor PTEN inhibits the phosphoinositol-3-kinase (PI3K) signaling pathway and plays a key role in cell growth, proliferation, survival, and migration. Pten conditional deletion using MxCre or Scl-CreER(T) leads to splenomegaly and leukemia formation, which occurs after the relocation of normal hematopoietic stem cells (HSCs) from the bone marrow to the spleen. Unexpectedly, dormant HSCs in the bone marrow do not enter the cell cycle upon Pten loss, they do not lose self-renewal activity, and they are not exhausted. Instead, Pten deficiency causes an up-regulation of the PI3K pathway in myeloid cells, but not in HSCs. Strikingly, myeloid cells secrete high levels of G-CSF upon Pten loss, leading to the mobilization of HSCs from the bone marrow and accumulation in the spleen. After deletion of Pten in mice lacking G-CSF, the splenomegaly, myeloproliferative disease, and splenic HSC accumulation are rescued. Our data show that although PTEN has little if any role in normal HSCs, it is essential to prevent overt G-CSF production by myeloid and stromal cells which otherwise causes HSCs to relocate to the spleen followed by lethal leukemia initiation.


Assuntos
Medula Óssea/enzimologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , PTEN Fosfo-Hidrolase/deficiência , Alelos , Animais , Citocinas/metabolismo , Deleção de Genes , Fator Estimulador de Colônias de Granulócitos/deficiência , Integrases/metabolismo , Camundongos , Transtornos Mieloproliferativos/enzimologia , Transtornos Mieloproliferativos/patologia , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/enzimologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Baço/metabolismo , Baço/patologia
5.
Genes Dev ; 18(22): 2747-63, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15545632

RESUMO

The activity of adult stem cells is essential to replenish mature cells constantly lost due to normal tissue turnover. By a poorly understood mechanism, stem cells are maintained through self-renewal while concomitantly producing differentiated progeny. Here, we provide genetic evidence for an unexpected function of the c-Myc protein in the homeostasis of hematopoietic stem cells (HSCs). Conditional elimination of c-Myc activity in the bone marrow (BM) results in severe cytopenia and accumulation of HSCs in situ. Mutant HSCs self-renew and accumulate due to their failure to initiate normal stem cell differentiation. Impaired differentiation of c-Myc-deficient HSCs is linked to their localization in the differentiation preventative BM niche environment, and correlates with up-regulation of N-cadherin and a number of adhesion receptors, suggesting that release of HSCs from the stem cell niche requires c-Myc activity. Accordingly, enforced c-Myc expression in HSCs represses N-cadherin and integrins leading to loss of self-renewal activity at the expense of differentiation. Endogenous c-Myc is differentially expressed and induced upon differentiation of long-term HSCs. Collectively, our data indicate that c-Myc controls the balance between stem cell self-renewal and differentiation, presumably by regulating the interaction between HSCs and their niche.


Assuntos
Diferenciação Celular , Sobrevivência Celular , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Anemia/etiologia , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Caderinas/metabolismo , Adesão Celular , Feminino , Humanos , Integrases/metabolismo , Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa