Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 19(3): 295-304, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23377280

RESUMO

The mechanisms that regulate hematopoietic stem cell (HSC) regeneration after myelosuppressive injury are not well understood. We identified epidermal growth factor (EGF) to be highly enriched in the bone marrow serum of mice bearing deletion of Bak and Bax in TIE2-expressing cells in Tie2Cre; Bak1(-/-); Bax(flox/-) mice. These mice showed radioprotection of the HSC pool and 100% survival after a lethal dose of total-body irradiation (TBI). Bone marrow HSCs from wild-type mice expressed functional EGF receptor (EGFR), and systemic administration of EGF promoted the recovery of the HSC pool in vivo and improved the survival of mice after TBI. Conversely, administration of erlotinib, an EGFR antagonist, decreased both HSC regeneration and the survival of mice after TBI. Mice with EGFR deficiency in VAV-expressing hematopoietic cells also had delayed recovery of bone marrow stem and progenitor cells after TBI. Mechanistically, EGF reduced radiation-induced apoptosis of HSCs and mediated this effect through repression of the proapoptotic protein PUMA. Our findings show that EGFR signaling regulates HSC regeneration after myelosuppressive injury.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/efeitos da radiação , Lesões Experimentais por Radiação/tratamento farmacológico , Regeneração , Animais , Apoptose/efeitos da radiação , Proteínas Reguladoras de Apoptose/biossíntese , Medula Óssea/efeitos da radiação , Células da Medula Óssea/efeitos da radiação , Células Cultivadas , Receptores ErbB/antagonistas & inibidores , Cloridrato de Erlotinib , Feminino , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Transdução de Sinais/efeitos da radiação , Proteínas Supressoras de Tumor/biossíntese , Irradiação Corporal Total , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína X Associada a bcl-2/genética
2.
Stem Cells ; 31(2): 327-37, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23132593

RESUMO

Hematopoietic stem cells (HSCs) reside in proximity to bone marrow endothelial cells (BM ECs) and maintenance of the HSC pool is dependent upon EC-mediated c-kit signaling. Here, we used genetic models to determine whether radioprotection of BM ECs could facilitate hematopoietic regeneration following radiation-induced myelosuppression. We developed mice bearing deletion of the proapoptotic proteins, BAK and BAX, in Tie2(+) ECs and HSCs (Tie2Bak/Bax(Fl/-) mice) and compared their hematopoietic recovery following total body irradiation (TBI) with mice which retained Bax in Tie2(+) cells. Mice bearing deletion of Bak and Bax in Tie2(+) cells demonstrated protection of BM HSCs, preserved BM vasculature, and 100% survival following lethal dose TBI. In contrast, mice that retained Bax expression in Tie2(+) cells demonstrated depletion of BM HSCs, disrupted BM vasculature, and 10% survival post-TBI. In a complementary study, VEcadherinBak/Bax(Fl/-) mice, which lack Bak and Bax in VEcadherin(+) ECs, also demonstrated increased recovery of BM stem/progenitor cells following TBI compared to mice which retained Bax in VEcadherin(+) ECs. Importantly, chimeric mice that lacked Bak and Bax in HSCs but retained Bak and Bax in BM ECs displayed significantly decreased HSC content and survival following TBI compared to mice lacking Bak and Bax in both HSCs and BM ECs. These data suggest that the hematopoietic response to ionizing radiation is dependent upon HSC-autonomous responses but is regulated by BM EC-mediated mechanisms. Therefore, BM ECs may be therapeutically targeted as a means to augment hematopoietic reconstitution following myelosuppression.


Assuntos
Células da Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Lesões Experimentais por Radiação/metabolismo , Receptores Proteína Tirosina Quinases/genética , Irradiação Corporal Total , Animais , Células da Medula Óssea/patologia , Células da Medula Óssea/efeitos da radiação , Caderinas/genética , Caderinas/metabolismo , Células Endoteliais/patologia , Células Endoteliais/efeitos da radiação , Expressão Gênica/efeitos da radiação , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/efeitos da radiação , Camundongos , Camundongos Transgênicos , Lesões Experimentais por Radiação/genética , Lesões Experimentais por Radiação/mortalidade , Lesões Experimentais por Radiação/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2 , Regeneração/efeitos da radiação , Transdução de Sinais/efeitos da radiação , Análise de Sobrevida , Proteína Killer-Antagonista Homóloga a bcl-2/deficiência , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína X Associada a bcl-2/deficiência , Proteína X Associada a bcl-2/genética
3.
Cell Rep ; 2(4): 964-75, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23084748

RESUMO

The mechanisms through which the bone marrow (BM) microenvironment regulates hematopoietic stem cell (HSC) fate remain incompletely understood. We examined the role of the heparin-binding growth factor pleiotrophin (PTN) in regulating HSC function in the niche. PTN(-/-) mice displayed significantly decreased BM HSC content and impaired hematopoietic regeneration following myelosuppression. Conversely, mice lacking protein tyrosine phosphatase receptor zeta, which is inactivated by PTN, displayed significantly increased BM HSC content. Transplant studies revealed that PTN action was not HSC autonomous, but rather was mediated by the BM microenvironment. Interestingly, PTN was differentially expressed and secreted by BM sinusoidal endothelial cells within the vascular niche. Furthermore, systemic administration of anti-PTN antibody in mice substantially impaired both the homing of hematopoietic progenitor cells to the niche and the retention of BM HSCs in the niche. PTN is a secreted component of the BM vascular niche that regulates HSC self-renewal and retention in vivo.


Assuntos
Células da Medula Óssea/citologia , Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Células-Tronco Hematopoéticas/citologia , Animais , Anticorpos/imunologia , Proteínas de Transporte/genética , Células Cultivadas , Quimiocina CXCL12/metabolismo , Citocinas/deficiência , Citocinas/genética , Transplante de Células-Tronco Hematopoéticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/metabolismo , Nicho de Células-Tronco
4.
PLoS One ; 5(7): e11535, 2010 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-20634956

RESUMO

In the event of a terrorist-mediated attack in the United States using radiological or improvised nuclear weapons, it is expected that hundreds of thousands of people could be exposed to life-threatening levels of ionizing radiation. We have recently shown that genome-wide expression analysis of the peripheral blood (PB) can generate gene expression profiles that can predict radiation exposure and distinguish the dose level of exposure following total body irradiation (TBI). However, in the event a radiation-mass casualty scenario, many victims will have heterogeneous exposure due to partial shielding and it is unknown whether PB gene expression profiles would be useful in predicting the status of partially irradiated individuals. Here, we identified gene expression profiles in the PB that were characteristic of anterior hemibody-, posterior hemibody- and single limb-irradiation at 0.5 Gy, 2 Gy and 10 Gy in C57Bl6 mice. These PB signatures predicted the radiation status of partially irradiated mice with a high level of accuracy (range 79-100%) compared to non-irradiated mice. Interestingly, PB signatures of partial body irradiation were poorly predictive of radiation status by site of injury (range 16-43%), suggesting that the PB molecular response to partial body irradiation was anatomic site specific. Importantly, PB gene signatures generated from TBI-treated mice failed completely to predict the radiation status of partially irradiated animals or non-irradiated controls. These data demonstrate that partial body irradiation, even to a single limb, generates a characteristic PB signature of radiation injury and thus may necessitate the use of multiple signatures, both partial body and total body, to accurately assess the status of an individual exposed to radiation.


Assuntos
Perfilação da Expressão Gênica/métodos , Radiação Ionizante , Animais , Feminino , Expressão Gênica/efeitos da radiação , Leucócitos Mononucleares/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Lesões Experimentais por Radiação , Irradiação Corporal Total/efeitos adversos
5.
Nat Med ; 16(4): 475-82, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20305662

RESUMO

Hematopoietic stem cell (HSC) self-renewal is regulated by both intrinsic and extrinsic signals. Although some of the pathways that regulate HSC self-renewal have been uncovered, it remains largely unknown whether these pathways can be triggered by deliverable growth factors to induce HSC growth or regeneration. Here we show that pleiotrophin, a neurite outgrowth factor with no known function in hematopoiesis, efficiently promotes HSC expansion in vitro and HSC regeneration in vivo. Treatment of mouse bone marrow HSCs with pleiotrophin caused a marked increase in long-term repopulating HSC numbers in culture, as measured in competitive repopulating assays. Treatment of human cord blood CD34(+)CDCD38(-)Lin(-) cells with pleiotrophin also substantially increased severe combined immunodeficient (SCID)-repopulating cell counts in culture, compared to input and cytokine-treated cultures. Systemic administration of pleiotrophin to irradiated mice caused a pronounced expansion of bone marrow stem and progenitor cells in vivo, indicating that pleiotrophin is a regenerative growth factor for HSCs. Mechanistically, pleiotrophin activated phosphoinositide 3-kinase (PI3K) signaling in HSCs; antagonism of PI3K or Notch signaling inhibited pleiotrophin-mediated expansion of HSCs in culture. We identify the secreted growth factor pleiotrophin as a new regulator of both HSC expansion and regeneration.


Assuntos
Proteínas de Transporte/fisiologia , Citocinas/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Proteínas de Transporte/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/farmacologia , Relação Dose-Resposta a Droga , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Receptores Notch/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Células-Tronco/farmacologia , Trombopoetina/farmacologia
6.
Stem Cells ; 28(3): 523-34, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20054864

RESUMO

Hematopoietic stem cells (HSCs) are enriched for aldehyde dehydrogenase (ALDH) activity and ALDH is a selectable marker for human HSCs. However, the function of ALDH in HSC biology is not well understood. We sought to determine the function of ALDH in regulating HSC fate. Pharmacologic inhibition of ALDH with diethylaminobenzaldehyde (DEAB) impeded the differentiation of murine CD34(-)c-kit(+)Sca-1(+)lineage(-) (34(-)KSL) HSCs in culture and facilitated a ninefold expansion of cells capable of radioprotecting lethally irradiated mice compared to input 34(-)KSL cells. Treatment of bone marrow (BM) 34(-)KSL cells with DEAB caused a fourfold increase in 4-week competitive repopulating units, verifying the amplification of short-term HSCs (ST-HSCs) in response to ALDH inhibition. Targeted siRNA of ALDH1a1 in BM HSCs caused a comparable expansion of radioprotective progenitor cells in culture compared to DEAB treatment, confirming that ALDH1a1 was the target of DEAB inhibition. The addition of all trans retinoic acid blocked DEAB-mediated expansion of ST-HSCs in culture, suggesting that ALDH1a1 regulates HSC differentiation via augmentation of retinoid signaling. Pharmacologic inhibition of ALDH has therapeutic potential as a means to amplify ST-HSCs for transplantation purposes.


Assuntos
Aldeído Desidrogenase/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Citoproteção/fisiologia , Inibidores Enzimáticos/farmacologia , Células-Tronco Hematopoéticas/enzimologia , Transplante de Células-Tronco/métodos , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Citoproteção/efeitos dos fármacos , Inibidores Enzimáticos/uso terapêutico , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos da radiação , Humanos , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/genética , Radiação Ionizante , Retinal Desidrogenase , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tretinoína/metabolismo , Tretinoína/farmacologia , p-Aminoazobenzeno/análogos & derivados , p-Aminoazobenzeno/farmacologia , p-Aminoazobenzeno/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...