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











Base de dados
Intervalo de ano de publicação
1.
Blood ; 129(18): 2493-2506, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28232582

RESUMO

RNA-binding proteins (RBPs) have emerged as important regulators of invertebrate adult stem cells, but their activities remain poorly appreciated in mammals. Using a short hairpin RNA strategy, we demonstrate here that the 2 mammalian RBPs, PUMILIO (PUM)1 and PUM2, members of the PUF family of posttranscriptional regulators, are essential for hematopoietic stem/progenitor cell (HSPC) proliferation and survival in vitro and in vivo upon reconstitution assays. Moreover, we found that PUM1/2 sustain myeloid leukemic cell growth. Through a proteomic approach, we identified the FOXP1 transcription factor as a new target of PUM1/2. Contrary to its canonical repressive activity, PUM1/2 rather promote FOXP1 expression by a direct binding to 2 canonical PUM responsive elements present in the FOXP1-3' untranslated region (UTR). Expression of FOXP1 strongly correlates with PUM1 and PUM2 levels in primary HSPCs and myeloid leukemia cells. We demonstrate that FOXP1 by itself supports HSPC and leukemic cell growth, thus mimicking PUM activities. Mechanistically, FOXP1 represses the expression of the p21-CIP1 and p27-KIP1 cell cycle inhibitors. Enforced FOXP1 expression reverses shPUM antiproliferative and proapoptotic activities. Altogether, our results reveal a novel regulatory pathway, underscoring a previously unknown and interconnected key role of PUM1/2 and FOXP1 in regulating normal HSPC and leukemic cell growth.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Animais , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fatores de Transcrição Forkhead/genética , Humanos , Leucemia Mieloide/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Proteínas de Neoplasias/genética , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/genética
2.
Eur Cytokine Netw ; 27(2): 16-26, 2016 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-27478075

RESUMO

In spite of recent key improvements, in vitro mass production of erythrocytes from human stem cells is still limited by difficulties in obtaining sufficient numbers of erythroid progenitors. In fact, such progenitors are as scarce in the bone marrow as in peripheral blood. We used a two-step culture model of human cord blood-derived erythroid progenitors in the presence or absence of high-purity neuraminidase, in a serum-free, defined culture medium. Granulocytic and megakaryocytic progenitor cell expansions were also studied. We show that significant enhancement of erythroid cell generation is obtained when CD34(+) human hematopoietic progenitors are cultured in the presence of neuraminidase. Interestingly, in so doing, expanded red cell progenitors remained erythropoietin-dependent for further expansion and survival, and cells thus generated displayed a normal phenotype. Moreover, the activity of neuraminidase on these cells can be reversed by simple cell washing. Finally, growth of cells of the other myeloid lineages (granulocytes and megakaryocytes) is either decreased or unchanged in the presence of neuraminidase. This specific feature of neuraminidase, that of stimulation of human red cell progenitor proliferation, provides a safe technique for producing greater numbers of in vitro-generated red blood cells for both basic research and transfusion use.


Assuntos
Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Neuraminidase/metabolismo , Antígenos CD34/metabolismo , Antígenos Glicosídicos Associados a Tumores/genética , Antígenos Glicosídicos Associados a Tumores/metabolismo , Apoptose/efeitos dos fármacos , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Células Precursoras Eritroides/efeitos dos fármacos , Expressão Gênica , Granulócitos/efeitos dos fármacos , Granulócitos/metabolismo , Humanos , Imunofenotipagem , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Neuraminidase/farmacologia , Fenótipo
3.
Haematologica ; 97(2): 168-78, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22298821

RESUMO

BACKGROUND: Expansion of hematopoietic stem cells represents an important objective for improving cell and gene therapy protocols. Retroviral transduction of the HoxB4 homeogene in mouse and human hematopoietic stem cells and hematopoietic progenitors is known to promote the cells' expansion. A safer approach consists in transferring homeobox proteins into hematopoietic stem cells taking advantage of the natural ability of homeoproteins to cross cell membranes. Thus, HOXB4 protein transfer is operative for expanding human hematopoietic cells, but such expansion needs to be improved. DESIGN AND METHODS: To that aim, we evaluated the effects of HOXC4, a protein encoded by a HOXB4 paralog gene, by co-culturing HOXC4-producing stromal cells with human CD34(+) hematopoietic cells. Numbers of progenitors and stem cells were assessed by in vitro cloning assays and injection into immuno-deficient mice, respectively. We also looked for activation or inhibition of target downstream gene expression. RESULTS: We show that the HOXC4 homeoprotein expands human hematopoietic immature cells by 3 to 6 times ex vivo and significantly improves the level of in vivo engraftment. Comparative transcriptome analysis of CD34(+) cells subjected or not to HOXB4 or HOXC4 demonstrated that both homeoproteins regulate the same set of genes, some of which encode key hematopoietic factors and signaling molecules. Certain molecules identified herein are factors reported to be involved in stem cell fate or expansion in other models, such as MEF2C, EZH2, DBF4, DHX9, YPEL5 and Pumilio. CONCLUSIONS: The present study may help to identify new HOX downstream key factors potentially involved in hematopoietic stem cell expansion or in leukemogenesis.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
4.
Haematologica ; 97(4): 491-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22180429

RESUMO

BACKGROUND: Although mobilization of hematopoietic stem cells and hematopoietic progenitor cells can be achieved with a combination of granulocyte colony-stimulating factor and plerixafor (AMD3100), improving approaches for hematopoietic progenitor cell mobilization is clinically important. DESIGN AND METHODS: Heparan sulfate proteoglycans are ubiquitous macromolecules associated with the extracellular matrix that regulates biology of hematopoietic stem cells. We studied the effects of a new family of synthetic oligosaccharides mimicking heparan sulfate on hematopoietic stem cell mobilization. These oligosaccharides were administered intravenously alone or in combination with granulocyte colony-stimulating factor and/or AMD3100 in mice. Mobilized hematopoietic cells were counted and phenotyped at different times and the ability of mobilized hematopoietic stem cells to reconstitute long-term hematopoiesis was determined by competitive transplantation into syngenic lethally irradiated mice followed by secondary transplantation. RESULTS: Mimetics of heparan sulfate induced rapid mobilization of B-lymphocytes, T-lymphocytes, hematopoietic stem cells and hematopoietic progenitor cells. They increased the mobilization of hematopoietic stem cells and hematopoietic progenitor cells more than 3-fold when added to the granulocyte colony-stimulating factor/AMD3100 association. Hematopoietic stem cells mobilized by mimetics of heparan sulfate or by the granulocyte colony-stimulating factor/AMD3100/mimetics association were as effective as hematopoietic stem cells mobilized by the granulocyte colony-stimulating factor/AMD3100 association for primary and secondary hematopoietic reconstitution of lethally irradiated mice. CONCLUSIONS: This new family of mobilizing agents could alone or in combination with granulocyte colony-stimulating factor and/or AMD3100 mobilize a high number of hematopoietic stem cells that were able to maintain long-term hematopoiesis. These results strengthen the role of heparan sulfates in the retention of hematopoietic stem cells in bone marrow and support the use of small glyco-drugs based on heparan sulfate in combination with granulocyte colony-stimulating factor and AMD3100 to improve high stem cell mobilization, particularly in a prospect of use in human therapeutics.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Heparitina Sulfato/farmacologia , Compostos Heterocíclicos/farmacologia , Animais , Benzilaminas , Ciclamos , Sinergismo Farmacológico , Hematopoese/efeitos dos fármacos , Hematopoese/fisiologia , Transplante de Células-Tronco Hematopoéticas , Heparitina Sulfato/síntese química , Cinética , Camundongos , Camundongos Endogâmicos C57BL
5.
Biol Cell ; 103(11): 531-42, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21810080

RESUMO

BACKGROUND INFORMATION: The identification of a source of stem cells able to regenerate skeletal muscle was the goal of numerous studies with the aim to develop new therapeutic approaches for genetic muscle diseases or muscle injuries. A series of studies have demonstrated that stem cells derived from various tissues may have a role in the regeneration of damaged muscles, but this contribution is always very weak. Thus we established a project aiming to reprogramme non-muscle cells into the skeletal striated differentiation pathway. RESULTS: We transduced several human primary adult stem or progenitor cells using a recombinant lentivirus containing the coding sequence of the Myf5 gene considered as a master gene for the determination of skeletal striated muscle. These original results are the first demonstration of a myogenic conversion of human mesenchymal and endothelial cells by Myf5. CONCLUSIONS: The procedure described in the present paper could be used to develop new research protocols with the prospect of using these cells as therapeutic agents.


Assuntos
Reprogramação Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Mioblastos/citologia , Fator Regulador Miogênico 5/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Mioblastos/metabolismo , Fator Regulador Miogênico 5/genética
6.
Blood ; 118(13): 3657-60, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21791426

RESUMO

Sustained undetectable molecular residual disease (UMRD) is obtained in a minority of patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. It remains unclear whether these patients are definitively cured of their leukemia or whether leukemic stem cells (LSCs) persist in their BM. We have evaluated the presence of BCR-ABL-expressing marrow LSCs in 6 patients with chronic myeloid leukemia with sustained UMRD induced by IFN-α (n = 3), imatinib mesylate after IFN-α failure (n = 2), and dasatinib after imatinib intolerance (n = 1). Purified CD34(+) cells were used for clonogenic and long-term culture-initiating cell assays performed on classic or HOXB4-expressing MS-5 feeders. Using this strategy, we identified BCR-ABL-expressing LSCs in all patients. Interestingly, long-term culture-initiating cell assays with MS-5/HOXB4 stromal feeders increased detected numbers of LSCs in 3 patients. The relation between LSC persistency and a potential risk of disease relapse for patients with durable UMRD (on or off tyrosine kinase inhibitor therapy) warrants further investigation.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/diagnóstico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Neoplásicas/fisiologia , Adulto , Idoso , Algoritmos , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Sobrevivência Celular , Feminino , Humanos , Limite de Detecção , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Técnicas de Diagnóstico Molecular/normas , Técnicas de Diagnóstico Molecular/estatística & dados numéricos , Neoplasia Residual , Células-Tronco Neoplásicas/patologia , Indução de Remissão , Células Tumorais Cultivadas
7.
Blood ; 114(9): 1875-83, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19525480

RESUMO

Platelets originate from megakaryocytes (MKs) by cytoplasmic elongation into proplatelets. Direct platelet release is not seen in bone marrow hematopoietic islands. It was suggested that proplatelet fragmentation into platelets can occur intravascularly, yet evidence of its dependence on hydrodynamic forces is missing. Therefore, we investigated whether platelet production from MKs could be up-regulated by circulatory forces. Human mature MKs were perfused at a high shear rate on von Willebrand factor. Cells were observed in real time by videomicroscopy, and by confocal and electron microscopy after fixation. Dramatic cellular modifications followed exposure to high shear rates: 30% to 45% adherent MKs were converted into proplatelets and released platelets within 20 minutes, contrary to static conditions that required several hours, often without platelet release. Tubulin was present in elongated proplatelets and platelets, thus ruling out membrane tethers. By using inhibitors, we demonstrated the fundamental roles of microtubule assembly and MK receptor GPIb. Secretory granules were present along the proplatelet shafts and in shed platelets, as shown by P-selectin labeling. Platelets generated in vitro were functional since they responded to thrombin by P-selectin expression and cytoskeletal reorganization. In conclusion, MK exposure to high shear rates promotes platelet production via GPIb, depending on microtubule assembly and elongation.


Assuntos
Plaquetas/citologia , Plaquetas/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Citometria de Fluxo/métodos , Humanos , Microscopia Confocal/métodos , Microscopia Eletrônica/métodos , Microscopia de Vídeo/métodos , Modelos Biológicos , Perfusão , Estresse Mecânico , Trombopoese/imunologia , Tubulina (Proteína)/metabolismo , Fator de von Willebrand/metabolismo
8.
Stem Cells ; 27(1): 108-15, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18974210

RESUMO

To further clarify the contribution of nuclear architecture in the regulation of gene expression patterns during differentiation of human multipotent cells, we analyzed expression status, histone modifications, and subnuclear positioning relative to repressive compartments, of hematopoietic loci in multipotent and lineage-committed primary human hematopoietic progenitors. We report here that positioning of lineage-affiliated loci relative to pericentromeric heterochromatin compartments (PCH) is identical in multipotent cells from various origins and is unchanged between multipotent and lineage-committed hematopoietic progenitors. However, during differentiation of multipotent hematopoietic progenitors, changes in gene expression and histone modifications at these loci occur in committed progenitors, prior to changes in gene positioning relative to pericentromeric heterochromatin compartments, detected at later stages in precursor and mature cells. Therefore, during normal human hematopoietic differentiation, changes in gene subnuclear location relative to pericentromeric heterochromatin appear to be dictated by whether the gene will be permanently silenced or activated, rather than being predictive of commitment toward a given lineage.


Assuntos
Compartimento Celular , Cromatina/metabolismo , Ordem dos Genes , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Multipotentes/metabolismo , Acetilação , Animais , Diferenciação Celular/genética , Linhagem da Célula , Células Eritroides/metabolismo , Globinas/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Histonas/metabolismo , Humanos , Cadeias kappa de Imunoglobulina/genética , Lactente , Camundongos , Células-Tronco Multipotentes/citologia
10.
Stem Cells ; 26(2): 312-22, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17962697

RESUMO

The HOXB4 homeoprotein is known to promote the expansion of mouse and human hematopoietic stem cells (HSCs) and progenitors of the myeloid lineages. However, the putative involvement of HOXB4 in lymphopoiesis and particularly in the expansion of early lymphoid progenitor cells has remained elusive. Based on the ability of the HOXB4 protein to passively enter hematopoietic cells, our group previously designed a long-term culture procedure of human HSCs that allows ex vivo expansion of these cells. Here, this method has been further used to investigate whether HOXB4 could cause similar expansion on cells originating from CD34(+) hematopoietic progenitor cells (HPCs) committed at various levels toward the lymphoid lineages. We provide evidence that HOXB4 protein delivery promotes the expansion of primitive HPCs that generate lymphoid progenitors. Moreover, HOXB4 acts on lymphomyeloid HPCs and committed T/natural killer HPCs but not on primary B-cell progenitors. Our results clarify the effect of HOXB4 in the early stages of human lymphopoiesis, emphasizing the contribution of this homeoprotein in the maintenance of the intrinsic lymphomyeloid differentiation potential of defined HPC subsets. Finally, this study supports the potential use of HOXB4 protein for HSC and HPC expansion in a therapeutic setting and furthers our understanding of the mechanisms of the molecular regulation of hematopoiesis.


Assuntos
Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/fisiologia , Linfopoese/fisiologia , Fatores de Transcrição/fisiologia , Animais , Antígenos CD/metabolismo , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/fisiologia , Diferenciação Celular , Linhagem Celular , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/farmacologia , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/fisiologia , Linfopoese/efeitos dos fármacos , Camundongos , Fenótipo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/farmacologia , Transdução Genética
11.
J Mol Biol ; 355(1): 9-19, 2006 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-16298389

RESUMO

The TAL-1/SCL gene encodes a basic helix-loop-helix (bHLH) transcription factor essential for primitive hematopoiesis and for adult erythroid and megakaryocytic development. Activated transcription of TAL-1 as a consequence of chromosomal rearrangements is associated with a high proportion of human T cell acute leukemias, showing that appropriate control of TAL-1 is crucial for the formation and subsequent fate of hematopoietic cells. Hence, the knowledge of the mechanisms, which govern the pattern of TAL-1 expression in hematopoiesis, is of great interest. We previously described a silencer in the 3'-untranslated region of human TAL-1, the activity of which is mediated through binding of a tissue-specific 40 kDa nuclear protein to a new DNA recognition motif, named tal-RE. Here, we show that tal-RE-binding activity, high in immature human hematopoietic progenitors is down regulated upon erythroid and megakaryocytic differentiation. This expression profile helped us to identify that PU.1/Spi-1 binds to the tal-RE sequences in vitro and occupies the TAL-1 silencer in vivo. By expressing a mutant protein containing only the ETS domain of PU.1 in human erythroleukemic HEL cells, we demonstrated that PU.1 mediates the transcriptional repression activity of the silencer. We found that ectopic PU.1 is not able to induce silencing activity in PU.1-negative Jurkat T cells, indicating that PU.1 activity, although necessary, is not sufficient to confer transcriptional repression activity to the TAL-1 silencer. Finally, we showed that the silencer is also active in TAL-1-negative myeloid HL60 cells that express PU.1 at high levels. In summary, our study shows that PU.1, in addition to its positive role in TAL-1 expression in early hematopoietic progenitors, may also act as a mediator of TAL-1 silencing in some hematopoietic lineages.


Assuntos
Inativação Gênica , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Ubiquitina-Proteína Ligases/genética , Linhagem Celular Tumoral , Regulação para Baixo , Hematopoese/genética , Humanos , Leucemia/patologia , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Elementos de Resposta , Transativadores/metabolismo , Fatores de Transcrição , Ativação Transcricional , Transfecção
12.
J Soc Biol ; 200(3): 235-41, 2006.
Artigo em Francês | MEDLINE | ID: mdl-17417138

RESUMO

Expansion of human hematopoietic stem cells (HSCs) is a challenge for cellular therapy. It currently relies on either the use of recombinant cytokines or transfer of transcription factor genes. Among these, the HOXB4 homeoprotein is of particular interest since it promotes the expansion of mouse HSCs without inducing leukemia. To prevent potential deleterious side effects associated with stable HOXB4 gene transfer into the cells, we took advantage of the ability of homeoproteins to passively pass through cell membranes. We have shown that, when co-cultured with stromal cells engineered to secrete HOXB4, human stem cells and immature progenitors clearly were expanded. This expansion was associated with enhanced stem cell repopulating capacity in vivo and maintenance of pluripotentiality. The role that HOXB4 plays on stem cell expansion has also been tested on human lymphoid progenitors. We found that our model of protein transfer was also able to induce the expansion of the immature lympho-myeloid and pro-T/NK progenitors as well as of more mature NK progenitors. We then looked for synergistic activities between HOXB4 and other homeoproteins such as HOXC4. We found that HOXC4 was able to promote the expansion of human hematopoietic cells in vitro roughly as HOXB4 did and that the presence of both HOXB4 and HOXC4 molecules induced even higher expansion levels of these cells. Our method provides a basis for developing cell therapy strategies using expanded HSCs that are not genetically modified.


Assuntos
Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição/fisiologia , Animais , Divisão Celular , Linhagem Celular/metabolismo , Linhagem Celular/virologia , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Técnicas de Cocultura , Vetores Genéticos/genética , Hematopoese , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/farmacologia , Humanos , Lentivirus/genética , Linfócitos/citologia , Camundongos , Modelos Biológicos , Transporte Proteico , Proteínas Recombinantes de Fusão/fisiologia , Fatores de Transcrição/química , Transdução Genética
13.
J Cell Sci ; 118(Pt 8): 1595-605, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15784678

RESUMO

We have previously identified the transcript encoding NACA (the alpha chain of the nascent-polypeptide-associated complex) as a cytokine-modulated specific transcript in the human TF-1 erythroleukemic cell line. This protein was already known to be a transcriptional co-activator that acts by potentiating AP-1 activity in osteoblasts, and is known to be involved in the targeting of nascent polypeptides. In this study, we investigate the role of NACA in human hematopoiesis. Protein distribution analyses indicate that NACA is expressed in undifferentiated TF-1 cells and in human-cord-blood-derived CD34(+) progenitor cells. Its expression is maintained during in vitro erythroid differentiation but, in marked contrast, its expression is suppressed during their megakaryocytic or granulocytic differentiation. Ectopic expression of NACA in CD34(+) cells under culture conditions that induce erythroid-lineage differentiation leads to a marked acceleration of erythroid-cell differentiation. Moreover, ectopic expression of NACA induces erythropoietin-independent differentiation of TF-1 cells, whereas downregulation of NACA by RNA interference abolishes the induction of hemoglobin production in these cells and diminishes glycophorin-A (GPA) expression by CD34(+) progenitors cultured under erythroid differentiation conditions. Altogether, these results characterize NACA as a new factor involved in the positive regulation of human erythroid-cell differentiation.


Assuntos
Diferenciação Celular/fisiologia , Eritrócitos/metabolismo , Células Precursoras Eritroides/metabolismo , Hematopoese/fisiologia , Transativadores/metabolismo , Antígenos CD34/metabolismo , Linhagem Celular Tumoral , Linhagem da Célula/fisiologia , Regulação para Baixo/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicoforinas/metabolismo , Granulócitos/metabolismo , Hemoglobinas/biossíntese , Humanos , Megacariócitos/metabolismo , Chaperonas Moleculares , Interferência de RNA/fisiologia , Transativadores/genética , Regulação para Cima/fisiologia
14.
J Immunol ; 174(7): 3948-58, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15778351

RESUMO

Class I MHC complexes (MHC(I)) are essential in mediating immune response. The transport of antigenic peptides (TAP) to MHC(I) and the stable expression of MHC(I) on the cell surface require the presence of a dedicated TAP. In this study we report that IFN-gamma and thrombopoietin (TPO) strongly increase TAP1 protein expression in megakaryocytes, followed by an enhanced expression of MHC(I) on the cell surface. This expression parallels the enhanced TAP1 promoter activity and TAP1 mRNA expression, which are independent of protein synthesis. We also show that this cytokine-dependent expression of TAP1 transcripts depends on STAT1 and IFN regulatory factor-2 (IRF-2), but not on IRF-1, and provide evidence that IRF-2 constitutively binds to the TAP1 gene promoter and enhances TAP1 promoter activity. We show that IRF-2 forms a complex with STAT1 and the cytokine-responsive region of the TAP1 promoter in any TPO or IFN-gamma target cells tested. Interaction of IRF-2 and STAT1 on the promoter depends on the DNA-binding domain of IRF-2. Overall, our data indicate that TPO and IFN-gamma activate the expression of TAP1 via a new mechanism that involves functional cooperation between STAT1 and IRF-2 on the TAP1 promoter.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Antígenos de Histocompatibilidade Classe I/genética , Megacariócitos/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras/fisiologia , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Transportadores de Cassetes de Ligação de ATP , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fator Regulador 2 de Interferon , Interferon gama/farmacologia , Ligação Proteica , RNA Mensageiro/biossíntese , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT1 , Trombopoetina/farmacologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
16.
Blood ; 103(9): 3326-35, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-14715640

RESUMO

The transcription factor TAL1 has major functions during embryonic hematopoiesis and in adult erythropoiesis and megakaryocytopoiesis. These functions rely on different TAL1 structural domains that are responsible for dimerization, transactivation, and DNA binding. Previous work, most often done in mice, has shown that some TAL1 functions do not require DNA binding. To study the role of TAL1 and the relevance of the TAL1 DNA-binding domain in human erythropoiesis, we developed an approach that allows an efficient enforced wild-type or mutant TAL1 protein expression in human hematopoietic CD34(+) cells using a lentiviral vector. Differentiation capacities of the transduced cells were studied in a culture system that distinguishes early and late erythroid development. Results indicate that enforced TAL1 expression enhances long-term culture initiating cell (LTC-IC) potential and erythroid differentiation of human CD34(+) cells as shown by increased beta globin and porphobilinogen deaminase (PBGD) gene expressions and erythroid colony-forming units (CFU-Es), erythroid burst-forming units (BFU-Es), and glycophorin A-positive (GPA(+)) cell productions. Enforced expression of a TAL1 protein deleted of its DNA-binding domain (named Delta bTAL1) mimicked most TAL1 effects except for the LTC-IC enhancement, the down-regulation of the CD34 surface marker, and the GPA(+) cell production. These results provide the first functional indications of DNA-binding-dependent and -independent roles of TAL1 in human erythropoiesis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Eritropoese , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição/fisiologia , Antígenos CD34/análise , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Sítios de Ligação , Técnicas de Cultura de Células/métodos , Diferenciação Celular , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Células Precursoras Eritroides/citologia , Sangue Fetal/citologia , Regulação da Expressão Gênica/fisiologia , Glicoforinas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Glicoproteínas de Membrana/análise , Mutação , Proteínas Proto-Oncogênicas/genética , Sialoglicoproteínas/análise , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição/genética , Transfecção
17.
Thromb Haemost ; 90(5): 844-52, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14597980

RESUMO

alpha-Granule protein storage is important for producing platelets with normal haemostatic function. The low to undetectable levels of several megakaryocyte-synthesized alpha-granule proteins in normal plasma suggest megakaryocytes are important to sequester these proteins in vivo. alpha-Granule protein storage in vitro has been studied using other cell types, with differences observed in how some proteins are processed compared to platelets. Human megakaryocytes, cultured from cord blood CD34(+) cells and grown in serum-free media containing thrombopoietin, were investigated to determine if they could be used as a model for studying normal alpha-granule protein processing and storage. ELISA indicated that cultured megakaryocytes contained the alpha-granule proteins multimerin, von Willebrand factor, thrombospondin-1, beta-thromboglobulin and platelet factor 4, but no detectable fibrinogen and factor V. A significant proportion of the alpha-granule protein in megakaryocyte cultures was contained within the cells (averages: 41-71 %), consistent with storage. Detailed analyses of multimerin and von Willebrand factor confirmed that alpha-granule proteins were processed to mature forms and were predominantly located in the alpha-granules of cultured megakaryocytes.Thrombopoietin-stimulated cultured megakaryocytes provide a useful model for studying alpha-granule protein processing and storage.


Assuntos
Grânulos Citoplasmáticos/química , Megacariócitos/química , Proteínas/metabolismo , Proteínas Sanguíneas/análise , Proteínas Sanguíneas/metabolismo , Células Cultivadas , Humanos , Megacariócitos/citologia , Megacariócitos/ultraestrutura , Fator Plaquetário 4/análise , Fator Plaquetário 4/metabolismo , Proteínas/análise , Trombopoetina/farmacologia , Trombospondina 1/análise , Trombospondina 1/metabolismo , beta-Tromboglobulina/análise , beta-Tromboglobulina/metabolismo , Fator de von Willebrand/análise , Fator de von Willebrand/metabolismo
18.
Nat Med ; 9(11): 1423-7, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14578882

RESUMO

Expansion of human hematopoietic stem cells (HSCs) is a major challenge in cellular therapy, and currently relies on the use of recombinant cytokines or on gene transfer of transcription factors. Of these, the HOXB4 homeoprotein protein is of particular interests as it promotes the expansion of mouse HSCs without inducing the development of leukemia. To eliminate any deleterious effects that might be associated with stable HOXB4 gene transfer into human cells, we took advantage of the ability of HOX proteins to passively translocate through cell membranes. Here we show that when cultured on stromal cells genetically engineered to secrete HOXB4, human long-term culture-initiating cells (LTC-ICs) and nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mouse repopulating cells (SRCs) were expanded by more than 20- and 2.5-fold, respectively, over their input numbers. This expansion was associated with enhanced stem cell repopulating capacity in vivo and maintenance of pluripotentiality. This method provides a basis for developing cell therapy strategies using expanded HSCs that are not genetically modified.


Assuntos
Divisão Celular/fisiologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Técnicas de Cocultura , Humanos , Camundongos
19.
Mol Ther ; 7(3): 317-24, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12668127

RESUMO

X-linked adrenoleukodystrophy (ALD), an inherited demyelinating disorder of the central nervous system, can be corrected by allogeneic bone marrow transplantation, likely due to the turnover of brain macrophages that are bone marrow derived. ALD is characterized by an accumulation of very long chain fatty acids (VLCFA) due to the deficiency of an ATP binding cassette transporter that imports these fatty acids in peroxisomes. Murine retroviral transduction results in metabolic correction of ALD CD34(+) cells in vitro but reinfusion of these cells into ALD patients would not provide clinical benefit owing to the absence of selective advantage conferred by transgene expression. High-efficiency transduction of ALD CD34(+) peripheral blood mobilized cells was achieved using an HIV-based vector driving ALD gene expression under the elongation factor 1 alpha promoter and a protocol without prestimulation of CD34(+) cells with cytokines prior to transduction to preserve their stem cell properties. Efficient expression of the ALD gene was demonstrated in monocytes/macrophages derived from cultures of transduced ALD CD34(+) cells and in long-term culture initiating cells. VLCFA metabolism was corrected in transduced CD34(+), CFU-derived, and LTC-derived cells, indicating that the vector-encoded ALD protein was fully functional. Transplantation of transduced ALD CD34(+) cells into NOD/SCID mice resulted in long-term expression of ALD protein in monocytes/macrophages derived from engrafted stem cells.


Assuntos
Adrenoleucodistrofia/terapia , Antígenos CD34/imunologia , Terapia Genética , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Lentivirus/genética , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adrenoleucodistrofia/sangue , Adrenoleucodistrofia/genética , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Sobrevivência Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Citometria de Fluxo , Expressão Gênica , Técnicas de Transferência de Genes , HIV/genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fator 1 de Elongação de Peptídeos/genética , Transfecção
20.
Mol Ther ; 6(5): 673-7, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12436963

RESUMO

Gene therapy using permanent modifications of hematopoietic stem cells (HSC) has increasing potential applications for both genetic and acquired diseases. Considerable progress has been made recently in gene transfer to HSC by the use of lentivirus-derived vectors, which have the capacity to transduce noncycling cells. However, overall efficiency of HSC transduction reported so far is still not sufficient for numerous applications. We describe here an improved HSC transduction protocol, using the previously described lentiviral vector, that leads to more than 90% transduction of human CD34+ cells from cord blood, including NOD-LtSz-scid/scid repopulating cells. Moreover, under the same conditions, we transduce more than 75% and 80% of CD34+ cells mobilized in peripheral blood and from bone marrow, respectively. We further show that transgene expression is stable through time and hematopoietic cell differentiation in vitro as well as in vivo. Such a high HSC transduction efficiency opens new opportunities for both gene therapy applications and functional studies of regulator proteins of hematopoiesis.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Células-Tronco Hematopoéticas/metabolismo , Lentivirus/genética , Transgenes , Animais , Antígenos CD34/biossíntese , Antígenos CD34/genética , Células da Medula Óssea/metabolismo , Separação Celular , Sangue Fetal/citologia , Citometria de Fluxo , Proteínas de Fluorescência Verde , Hematopoese , Humanos , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos SCID , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA