Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 34(4): 1068-82, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27095138

RESUMO

Folliculin (FLCN) is an autosomal dominant tumor suppressor gene that modulates diverse signaling pathways required for growth, proliferation, metabolism, survival, motility, and adhesion. FLCN is an essential protein required for murine embryonic development, embryonic stem cell (ESC) commitment, and Drosophila germline stem cell maintenance, suggesting that Flcn may be required for adult stem cell homeostasis. Conditional inactivation of Flcn in adult hematopoietic stem/progenitor cells (HSPCs) drives hematopoietic stem cells (HSC) into proliferative exhaustion resulting in the rapid depletion of HSPC, loss of all hematopoietic cell lineages, acute bone marrow (BM) failure, and mortality after 40 days. HSC that lack Flcn fail to reconstitute the hematopoietic compartment in recipient mice, demonstrating a cell-autonomous requirement for Flcn in HSC maintenance. BM cells showed increased phosphorylation of Akt and mTorc1, and extramedullary hematopoiesis was significantly reduced by treating mice with rapamycin in vivo, suggesting that the mTorc1 pathway was activated by loss of Flcn expression in hematopoietic cells in vivo. Tfe3 was activated and preferentially localized to the nucleus of Flcn knockout (KO) HSPCs. Tfe3 overexpression in HSPCs impaired long-term hematopoietic reconstitution in vivo, recapitulating the Flcn KO phenotype, and supporting the notion that abnormal activation of Tfe3 contributes to the Flcn KO phenotype. Flcn KO mice develop an acute histiocytic hyperplasia in multiple organs, suggesting a novel function for Flcn in macrophage development. Thus, Flcn is intrinsically required to maintain adult HSC quiescence and homeostasis, and Flcn loss leads to BM failure and mortality in mice.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Diferenciação Celular/genética , Estrona/genética , Células-Tronco Hematopoéticas/patologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Células da Medula Óssea/patologia , Linhagem da Célula/genética , Proliferação de Células/genética , Desenvolvimento Embrionário/genética , Células-Tronco Hematopoéticas/metabolismo , Homeostase/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout
2.
Blood ; 120(6): 1254-61, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22709692

RESUMO

Birt-Hogg-Dubé (BHD) syndrome is an autosomal dominant disorder characterized by cutaneous fibrofolliculomas, pulmonary cysts, and kidney malignancies. Affected individuals carry germ line mutations in folliculin (FLCN), a tumor suppressor gene that becomes biallelically inactivated in kidney tumors by second-hit mutations. Similar to other factors implicated in kidney cancer, FLCN has been shown to modulate activation of mammalian target of rapamycin (mTOR). However, its precise in vivo function is largely unknown because germ line deletion of Flcn results in early embryonic lethality in animal models. Here, we describe mice deficient in the newly characterized folliculin-interacting protein 1 (Fnip1). In contrast to Flcn, Fnip1(-/-) mice develop normally, are not susceptible to kidney neoplasia, but display a striking pro-B cell block that is entirely independent of mTOR activity. We show that this developmental arrest results from rapid caspase-induced pre-B cell death, and that a Bcl2 transgene reconstitutes mature B-cell populations, respectively. We also demonstrate that conditional deletion of Flcn recapitulates the pro-B cell arrest of Fnip1(-/-) mice. Our studies thus demonstrate that the FLCN-FNIP complex deregulated in BHD syndrome is absolutely required for B-cell differentiation, and that it functions through both mTOR-dependent and independent pathways.


Assuntos
Linfócitos B/fisiologia , Síndrome de Birt-Hogg-Dubé/genética , Proteínas de Transporte/genética , Diferenciação Celular/genética , Deleção de Genes , Proteínas Proto-Oncogênicas/genética , Proteínas Supressoras de Tumor/genética , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Diferenciação Celular/imunologia , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Especificidade da Espécie , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia
3.
Blood ; 114(6): 1186-95, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19478045

RESUMO

Development of hematopoietic stem cells (HSCs) and their immediate progeny is maintained by the interaction with cells in the microenvironment. We found that hematopoiesis was dysregulated in Id1(-/-) mice. Although the frequency of HSCs in Id1(-/-) bone marrow was increased, their total numbers remained unchanged as the result of decreased bone marrow cellularity. In addition, the ability of Id1(-/-) HSCs to self-renew was normal, suggesting Id1 does not affect HSC function. Id1(-/-) progenitors showed increased cycling in vivo but not in vitro, suggesting cell nonautonomous mechanisms for the increased cycling. Id1(-/-) HSCs developed normally when transplanted into Id1(+/+) mice, whereas the development of Id1(+/+) HSCs was impaired in Id1(-/-) recipients undergoing transplantation and reproduced the hematologic features of Id1(-/-) mice, indicating that the Id1(-/-) microenvironment cannot support normal hematopoietic development. Id1(-/-) stromal cells showed altered production of cytokines in vitro, and cytokine levels were deregulated in vivo, which could account for the Id1(-/-) hematopoietic phenotypes. Thus, Id1 is required for regulating the hematopoietic progenitor cell niche but is dispensable for maintaining HSCs.


Assuntos
Medula Óssea/metabolismo , Ciclo Celular/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteína 1 Inibidora de Diferenciação/metabolismo , Animais , Citocinas/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Knockout , Células Estromais/citologia , Células Estromais/metabolismo
4.
Blood ; 112(4): 1068-77, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18523151

RESUMO

Inhibitors of DNA binding (Id) family members are key regulators of cellular differentiation and proliferation. These activities are related to the ability of Id proteins to antagonize E proteins and other transcription factors. As negative regulators of E proteins, Id proteins have been implicated in lymphocyte development. Overexpression of Id1, Id2, or Id3 has similar effects on lymphocyte development. However, which Id protein plays a physiologic role during lymphocyte development is not clear. By analyzing Id2 knock-out mice and retroviral transduced hematopoietic progenitors, we demonstrated that Id2 is an intrinsic negative regulator of B-cell development. Hematopoietic progenitor cells overexpressing Id2 did not reconstitute B-cell development in vivo, which resembled the phenotype of E2A null mice. The B-cell population in bone marrow was significantly expanded in Id2 knock-out mice compared with their wild-type littermates. Knock-down of Id2 by shRNA in hematopoietic progenitor cells promoted B-cell differentiation and induced the expression of B-cell lineage-specific genes. These data identified Id2 as a physiologically relevant regulator of E2A during B lymphopoiesis. Furthermore, we identified a novel Id2 function in erythroid development. Overexpression of Id2 enhanced erythroid development, and decreased level of Id2 impaired normal erythroid development. Id2 regulation of erythroid development is mediated via interacting with transcription factor PU.1 and modulating PU.1 and GATA-1 activities. We conclude that Id2 regulates lymphoid and erythroid development via interaction with different target proteins.


Assuntos
Linfócitos B/citologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Linhagem da Célula , Células Eritroides/citologia , Fator de Transcrição GATA1/fisiologia , Proteína 2 Inibidora de Diferenciação/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Animais , Medula Óssea , Eritropoese , Células-Tronco Hematopoéticas , Proteína 2 Inibidora de Diferenciação/genética , Linfócitos/citologia , Linfopoese , Camundongos , Camundongos Knockout , Ligação Proteica/fisiologia
5.
Epigenetics ; 3(3): 134-42, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18487951

RESUMO

Hematopoietic malignancies are frequently associated with DNA hypomethylation but the molecular mechanisms involved in tumor formation remain poorly understood. Here we report that mice lacking Lsh develop leukemia associated with DNA hypomethylation and oncogene activation. Lsh is a member of the SNF2 chromatin remodeling family and is required for de novo methylation of genomic DNA. Mice that received Lsh deficient hematopoietic progenitors showed severe impairment of hematopoiesis, suggesting that Lsh is necessary for normal hematopoiesis. A subset of mice developed erythroleukemia, a tumor that does not spontaneously occur in mice. Tumor tissues were CpG hypomethylated and showed a modest elevation of the transcription factor PU.1, an oncogene that is crucial for Friend virus induced erythroleukemia. Analysis of Lsh(-/-) hematopoietic progenitors revealed widespread DNA hypomethylation at repetitive sequences and hypomethylation at specific retroviral elements within the PU.1 gene. Wild type cells showed Lsh and Dnmt3b binding at the retroviral elements located within the PU.1 gene. On the other hand, Lsh deficient cells had no detectable Dnmt3b association suggesting that Lsh is necessary for recruitment of Dnmt3b to its target. Furthermore, Lsh(-/-) hematopoietic precursors showed impaired suppression of retroviral elements in the PU.1 gene, an increase of PU.1 transcripts and protein levels. Thus DNA hypomethylation caused by Lsh depletion is linked to transcriptional upregulation of retroviral elements and oncogenes such as PU.1 which in turn may promote the development of erythroleukemia in mice.


Assuntos
DNA Helicases/deficiência , Metilação de DNA , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Animais , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Helicases/genética , Regulação da Expressão Gênica , Hematopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Retroviridae , Transativadores/genética , DNA Metiltransferase 3B
6.
Dev Cell ; 10(5): 563-73, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16678773

RESUMO

Mouse knockouts of Cdk2 and Cdk4 have demonstrated that, individually, these genes are not essential for viability. To investigate whether there is functional redundancy, we have generated double knockout (DKO) mice. Cdk2-/- Cdk4-/- DKOs die during embryogenesis around E15 as a result of heart defects. We observed a gradual decrease of Retinoblastoma protein (Rb) phosphorylation and reduced expression of E2F-target genes, like Cdc2 and cyclin A2, during embryogenesis and in embryonic fibroblasts (MEFs). DKO MEFs are characterized by a decreased proliferation rate, impaired S phase entry, and premature senescence. HPV-E7-mediated inactivation of Rb restored normal expression of E2F-inducible genes, senescence, and proliferation in DKO MEFs. In contrast, loss of p27 did not rescue Cdk2-/- Cdk4-/- phenotypes. Our results demonstrate that Cdk2 and Cdk4 cooperate to phosphorylate Rb in vivo and to couple the G1/S phase transition to mitosis via E2F-dependent regulation of gene expression.


Assuntos
Quinase 2 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/deficiência , Embrião de Mamíferos/anormalidades , Proteína do Retinoblastoma/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Quinase 2 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/genética , Fatores de Transcrição E2F/antagonistas & inibidores , Fibroblastos/citologia , Inativação Gênica , Hematopoese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus , Fenótipo , Fosforilação , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/química
7.
Blood ; 107(11): 4308-16, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16469877

RESUMO

C/EBPalpha is an essential transcription factor required for myeloid differentiation. While C/EBPalpha can act as a cell fate switch to promote granulocyte differentiation in bipotential granulocyte-macrophage progenitors (GMPs), its role in regulating cell fate decisions in more primitive progenitors is not known. We found increased numbers of erythroid progenitors and erythroid cells in C/EBPalpha(-/-) fetal liver (FL). Also, enforced expression of C/EBPalpha in hematopoietic stem cells resulted in a loss of erythroid progenitors and an increase in myeloid cells by inhibition of erythroid development and inducing myeloid differentiation. Conditional expression of C/EBPalpha in murine erythroleukemia (MEL) cells induced myeloid-specific genes, while inhibiting erythroid-specific gene expression including erythropoietin receptor (EpoR), which suggests a novel mechanism to determine hematopoietic cell fate. Thus, C/EBPalpha functions in hematopoietic cell fate decisions by the dual actions of inhibiting erythroid and inducing myeloid gene expression in multipotential progenitors.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/fisiologia , Diferenciação Celular , Células Eritroides/citologia , Hematopoese , Células-Tronco Multipotentes/citologia , Células Mieloides/citologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Contagem de Células , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Fígado/citologia , Fígado/embriologia , Camundongos , Camundongos Knockout , Receptores da Eritropoetina/genética , Transfecção
8.
Blood ; 104(6): 1639-47, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15073037

RESUMO

CCAAT enhancer binding protein-alpha (C/EBPalpha) inhibits proliferation in multiple cell types; therefore, we evaluated whether C/EBPalpha-deficient hematopoietic progenitor cells (HPCs) have an increased proliferative potential in vitro and in vivo. In this study we demonstrate that C/EBPalpha(-/-) fetal liver (FL) progenitors are hyperproliferative, show decreased differentiation potential, and show increased self-renewal capacity in response to hematopoietic growth factors (HGFs). There are fewer committed bipotential progenitors in C/EBPalpha(-/-) FL, whereas multipotential progenitors are unaffected. HGF-dependent progenitor cell lines can be derived by directly culturing C/EBPalpha(-/-) FL cells in vitro Hyperproliferative spleen colonies and myelodysplastic syndrome (MDS) are observed in mice reconstituted with C/EBPalpha(-/-) FL cells, indicating progenitor hyperproliferation in vitro and in vivo. C/EBPalpha(-/-) FL lacked macrophage progenitors in vitro and had impaired ability to generate macrophages in vivo. These findings show that C/EBPalpha deficiency results in hyperproliferation of HPCs and a block in the ability of multipotential progenitors to differentiate into bipotential granulocyte/macrophage progenitors and their progeny.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/deficiência , Diferenciação Celular , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Transplante de Células , Células Cultivadas , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Interleucina-3/metabolismo , Fígado/embriologia , Fígado/metabolismo , Fígado/patologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Taxa de Sobrevida
9.
Leuk Res ; 26(4): 369-76, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11839380

RESUMO

As a part of our continuing efforts to develop gene therapy for acute myelogenous leukemia (AML), this study was undertaken to evaluate the possibility of using autologous bone marrow stromal fibroblasts (BMSFs) as a target cell population. Autologous BMSFs in AML were isolated from the stromal layers of long-term bone marrow culture (LTBMC) using immunomagnetic beads. BMSFs exhibited rapid proliferation even in the absence of growth factors. Cultures stimulated with bFGF produced significantly increased numbers of BMSFs than cultures without added growth factors. Using LNC/LacZ retroviral vector, the transduction efficiency of BMSFs was 13+/-4% at a 5 multiplicity of infection (MOI). LNC/interleukin-2 (IL-2)-transduced BMSFs produced between 1200 and 4800pg of IL-2/10(6) cells per 24h. Using adenoviral vector AdV/LacZ, the transduction efficiency was 84+/-10% at 100, and 92+/-8% at a MOI of 1000. Although the addition of basic fibroblast growth factor, epidermal growth factor, or platelet-derived growth factor did not affect the transduction efficiency, they increased the numbers of transduced cells significantly (P<0.01). AdV/IL-2-treated BMSFs produced high levels of IL-2 over the course of 7 days between 9820 and 22,700pg of IL-2/10(6) cells per 24h. Our finding that the genetically engineered autologous BMSFs of AML could be successfully established in vitro implies that BMSFs obtained from LTBMC might be considered as a target cell population for certain types of clinical gene therapy in AML.


Assuntos
Células da Medula Óssea/patologia , Terapia Genética , Leucemia Mieloide Aguda/patologia , Células Estromais/patologia , Transdução Genética , Células Tumorais Cultivadas , Adenoviridae , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Engenharia Genética , Terapia Genética/métodos , Vetores Genéticos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Células Estromais/fisiologia , Fatores de Tempo , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...