Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Science ; 255(5048): 1137-41, 1992 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-1372131

RESUMO

Severe combined immunodeficient (SCID) mice transplanted with human bone marrow were treated with human mast cell growth factor, a fusion of interleukin-3 and granulocyte-macrophage colony-stimulating factor (PIXY321), or both, starting immediately or 1 month later. Immature human cells repopulated the mouse bone marrow with differentiated human cells of multiple myeloid and lymphoid lineages; inclusion of erythropoietin resulted in human red cells in the peripheral blood. The bone marrow of growth factor-treated mice contained both multipotential and committed myeloid and erythroid progenitors, whereas mice not given growth factors had few human cells and only granulocyte-macrophage progenitors. Thus, this system allows the detection of immature human cells, identification of the growth factors that regulate them, and the establishment of animal models of human hematopoietic diseases.


Assuntos
Transplante de Medula Óssea , Citocinas/farmacologia , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Animais , Células da Medula Óssea , Eritropoetina/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Humanos , Interleucina-3/farmacologia , Camundongos , Camundongos SCID , Proteínas Recombinantes de Fusão/farmacologia , Fator de Células-Tronco
2.
Science ; 246(4937): 1597-600, 1989 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-2595371

RESUMO

A human acute lymphoblastic leukemia (ALL) cell line that was transplanted into immune-deficient SCID mice proliferated in the hematopoietic tissues, invaded various organs, and led to the death of the mice. The distribution of leukemic cells in SCID mice was similar to the course of the disease in children. A-1 cells marked with a retrovirus vector showed clonal evolution after the transplant. SCID mice that were injected with bone marrow from three patients with non-T ALL had leukemic cells in their bone marrow and spleen. This in vivo model of human leukemia is an approach to understanding leukemic growth and progression and is a novel system for testing new treatment strategies.


Assuntos
Síndromes de Imunodeficiência/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Animais , Encéfalo/patologia , Linhagem Celular , Células Clonais , DNA de Neoplasias/isolamento & purificação , Humanos , Rim/patologia , Fígado/patologia , Camundongos , Camundongos Mutantes , Transplante de Neoplasias , Transplante Heterólogo
3.
Leukemia ; 6(1): 8-17, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1531243

RESUMO

The sensitivity of the scid mouse model was assessed by comparing the growth of two pre-B acute lymphoblastic leukemia (ALL) cell lines, A1 and G2, established from patients at relapse. When cell numbers varying from 10(4) to 10(7) were injected intravenously into scid mice, advanced growth and dissemination of leukemia was observed at 10-12 weeks with the G2 cells. Bone marrow, spleen and thymus contained high levels of human leukemic cells and infiltration into lung, kidney, liver, and brain was observed. Two of three mice grafted with only 100 cells showed high levels of infiltration at 15 weeks, suggesting that 100 G2 cells was near the limiting cell number that could produce disseminated leukemia. With the A1 line, a minimum of 10(5) cells was needed to obtain dissemination to liver, lung, brain, and kidney; a low level of spleen infiltration occurred and thymus invasion was not observed. In vitro, both lines showed a density dependent growth in clonogenic assays but the cloning efficiency of the A1 line was 10-fold higher than for G2 cells. These results indicate that G2 and A1 lines have a dissimilar aggressiveness in vivo which does not correlate with clonogenic assay in vitro. Neither G2 nor A1 lines, growing in vitro, expressed CD10/CALLA on their surface, despite low levels of antigen on the freshly obtained relapse samples. Although A1 cells remained CD10-negative in the scid mice, G2 cells showed detectable levels of CD10, particularly on those cells found in the thymus. Several subclones of the G2 line were derived from isolated colonies in vitro; they were found to be CD10- in vitro, but to become CD10+ when proliferating into scid mouse thymus, suggesting the induction of CD10 by the murine microenvironment.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Animais , Antígenos de Diferenciação/metabolismo , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/análise , Criança , DNA de Neoplasias/análise , Citometria de Fluxo , Humanos , Cinética , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias/patologia , Neprilisina , Fenótipo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Receptores de Retorno de Linfócitos/análise , Células Tumorais Cultivadas/patologia , Ensaio Tumoral de Célula-Tronco
4.
Exp Hematol ; 27(11): 1667-74, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10560914

RESUMO

Fanconi anemia (FA) is a complex recessive genetic disease that causes bone marrow failure in children. The mechanism by which the gene for FA group C (Fancc) impinges on the normal hematopoietic program is unknown. Here we demonstrate that the bone marrow from Fancc-/- mice have reduced ability for primary and secondary long-term reconstitution of myeloablated recipients compared to wild-type or heterozygous mice, indicating that the Fancc gene product is required for the maintenance of normal numbers of hematopoietic stem cells. Long-term and secondary transplant studies suggested that there also were qualitative changes in their developmental potential. Consistent with the reduction in reconstitution, flow cytometric analysis of the primitive subfractions of hematopoietic cells obtained from the bone marrow of Fancc -/- mice demonstrated that they contained 40 to 70% fewer lineage-negative (Lin-)Thy1.2-/lowScal(+) c-Kit(+)CD34+ cells compared to controls. In contrast, the number of Lin Thy1.2-/ lowScal(+)c-Kit CD34(-)cells was comparable to that of wild-type mice. The differential behavior of Lin(-)Thy1.2-/lowScal+c-Kit+CD34+ and Lin(-)Thy1.2-/lowScal(+)c-Kit CD34 subfractions also was observed in mice treated with the DNA cross-linking agent mitomycin C(MMC). Fancc-/- mice treated with MMC had an 92% reduction of CD34 cells as compared to Fancc+/+ mice. The number of CD34 cells only was reduced about 20%. These results suggest that the Fancc gene may act at a stage of primitive hematopoietic cell development identified by CD34 expression.


Assuntos
Antígenos CD34/sangue , Anemia de Fanconi/imunologia , Células-Tronco Hematopoéticas/imunologia , Animais , Contagem de Células , Linhagem da Célula , Corantes , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Leukemia ; 24(5): 1012-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20220775

RESUMO

Children with constitutional trisomy 21 or Down's syndrome (DS) are predisposed to develop myeloid leukemia (ML) at a young age. DS-ML is frequently preceded by transient leukemia (TL), a spontaneously resolving accumulation of blasts during the newborn period. Somatic mutations of GATA1 in the blasts of TL and DS-ML likely function as an initiating event. We hypothesized that the phenotypic difference between TL and DS-ML is due to a divergent functional repertoire of the leukemia-initiating cells. Using an NOD/SCID model, we found that cells initiating DS-ML engrafted, disseminated to distant bone marrow sites, and propagated the leukemic clone in secondary recipients. In contrast, TL cells lacked the ability to expand and to migrate, but were able to persist in the recipient bone marrow. We found some evidence of genomic progression with 1 of 9 DS-ML samples and none of 11 TL samples harboring a mutation of N-RAS. The findings of this pilot study provide evidence for the functional impact of second events underlying the transformation of TL into DS-ML and a needed experimental tool for the functional testing of these promoting events.


Assuntos
Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Síndrome de Down/patologia , Leucemia Mieloide/patologia , Animais , Síndrome de Down/complicações , Citometria de Fluxo , Genes ras/genética , Humanos , Hibridização in Situ Fluorescente , Recém-Nascido , Leucemia Mieloide/etiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação/genética , Fenótipo , Projetos Piloto
6.
Blood ; 89(11): 3919-24, 1997 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9166828

RESUMO

We have previously reported the development of in vivo functional assays for primitive human hematopoietic cells based on their ability to repopulate the bone marrow (BM) of severe combined immunodeficient (SCID) and nonobese diabetic/SCID (NOD/SCID) mice following intravenous transplantation. Accumulated data from gene marking and cell purification experiments indicate that the engrafting cells (defined as SCID-repopulating cells or SRC) are biologically distinct from and more primitive than most cells that can be assayed in vitro. Here we demonstrate through limiting dilution analysis that the NOD/SCID xenotransplant model provides a quantitative assay for SRC. Using this assay, the frequency of SRC in cord blood (CB) was found to be 1 in 9.3 x 10(5) cells. This was significantly higher than the frequency of 1 SRC in 3.0 x 10(6) adult BM cells or 1 in 6.0 x 10(6) mobilized peripheral blood (PB) cells from normal donors. Mice transplanted with limiting numbers of SRC were engrafted with both lymphoid and multilineage myeloid human cells. This functional assay is currently the only available method for quantitative analysis of human hematopoietic cells with repopulating capacity. Both CB and mobilized PB are increasingly being used as alternative sources of hematopoietic stem cells in allogeneic transplantation. Thus, the findings reported here will have important clinical as well as biologic implications.


Assuntos
Bioensaio/métodos , Células da Medula Óssea , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Adulto , Animais , Contagem de Células Sanguíneas , Diferenciação Celular , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos SCID
7.
Blood ; 78(3): 624-34, 1991 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-1859880

RESUMO

The ability to transfer new genetic material into human hematopoietic cells provides the foundation for characterizing the organization and developmental program of human hematopoietic stem cells. It also provides a valuable model in which to test gene transfer and long-term expression in human hematopoietic cells as a prelude to human gene therapy. At the present time such studies are limited by the absence of in vivo assays for human stem cells, although recent descriptions of the engraftment of human hematopoietic cells in immune-deficient mice may provide the basis for such an assay. This study focuses on the establishment of conditions required for high efficiency retrovirus-mediated gene transfer into human hematopoietic progenitors that can be assayed in vitro in short-term colony assays and in vivo in immune-deficient mice. Here we report that a 24-hour preincubation of human bone marrow in 5637-conditioned medium, before infection, increases gene transfer efficiency into in vitro colony-forming cells by sixfold; interleukin-6 (IL-6) and leukemia inhibitory factor (LIF) provide the same magnitude increase as 5637-conditioned medium. In contrast, incubation in recombinant growth factors IL-1, IL-3, and granulocyte-macrophage colony-stimulating factor increases gene transfer efficiency by 1.5- to 3-fold. Furthermore, preselection in high concentrations of G418 results in a population of cells significantly enriched for G418-resistant progenitors (up to 100%). These results, obtained using detailed survival curves based on colony formation in G418, have been substantiated by directly detecting the neo gene in individual colonies using the polymerase chain reaction. Using these optimized protocols, human bone marrow cells were genetically manipulated with a neo retrovirus vector and transplanted into immune-deficient bg/nu/xid mice. At 1 month and 4 months after the transplant, the hematopoietic tissues of these animals remained engrafted with genetically manipulated human cells. More importantly, G418-resistant progenitors that contained the neo gene were recovered from the bone marrow and spleen of engrafted animals after 4 months. These experiments establish the feasibility of characterizing human stem cells using the unique retrovirus integration site as a clonal marker, similar to techniques developed to elucidate the murine stem cell hierarchy.


Assuntos
Substâncias de Crescimento/farmacologia , Células-Tronco Hematopoéticas/fisiologia , Transfecção , Adulto , Animais , Sequência de Bases , Células da Medula Óssea , Transplante de Medula Óssea , Sobrevivência Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Resistência Microbiana a Medicamentos/genética , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Sondas de Oligonucleotídeos , Reação em Cadeia da Polimerase , Proteínas Recombinantes/farmacologia , Valores de Referência , Transfecção/efeitos dos fármacos , Transplante Heterólogo
8.
Blood ; 91(8): 2737-44, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9531583

RESUMO

Fanconi anemia (FA) is a pleiotropic inherited disease that causes bone marrow failure in children. However, the specific involvement of FA genes in hematopoiesis and their relation to bone marrow (BM) failure is still unclear. The increased sensitivity of FA cells to DNA cross-linking agents such as mitomycin C (MMC) and diepoxybutane (DEB), including the induction of chromosomal aberrations and delay in the G2 phase of the cell cycle, have suggested a role for the FA genes in DNA repair, cell cycle regulation, and apoptosis. We previously reported the cloning of the FA group C gene (FAC) and the generation of a Fac mouse model. Surprisingly, the Fac -/- mice did not show any of the hematologic defects found in FA patients. To better understand the relationship of FA gene functions to BM failure, we have analyzed the in vivo effect of an FA-specific DNA damaging agent in Fac -/- mice. The mice were found to be highly sensitive to DNA cross-linking agents; acute exposure to MMC produced a marked BM hypoplasia and degeneration of proliferative tissues and caused death within a few days of treatment. However, sequential, nonlethal doses of MMC caused a progressive decrease in all peripheral blood parameters of Fac -/- mice. This treatment targeted specifically the BM compartment, with no effect on other proliferative tissues. The progressive pancytopenia resulted from a reduction in the number of early and committed hematopoietic progenitors. These results indicate that the FA genes are involved in the physiologic response of hematopoietic progenitor cells to DNA damage.


Assuntos
Medula Óssea/patologia , Proteínas de Ciclo Celular , Dano ao DNA , Proteínas de Ligação a DNA , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Regulação da Expressão Gênica , Proteínas Nucleares , Proteínas/genética , Animais , Medula Óssea/efeitos dos fármacos , Reagentes de Ligações Cruzadas/administração & dosagem , Dano ao DNA/efeitos dos fármacos , Modelos Animais de Doenças , Proteína do Grupo de Complementação C da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Regulação da Expressão Gênica/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitomicina/administração & dosagem , Inibidores da Síntese de Ácido Nucleico/administração & dosagem
9.
Blood ; 78(11): 2973-81, 1991 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-1835412

RESUMO

Bone marrow samples from patients with pre-B acute lymphoblastic leukemia (pre-B ALL), either at diagnosis or at relapse, were transplanted into scid mice to determine whether these freshly obtained leukemic cells could proliferate in vivo and whether there were any differences in their in vivo growth characteristics. Cells from three patients who relapsed within 13 months of diagnosis proliferated rapidly in the murine bone marrow, spleen, and thymus, invaded peripheral organs, and resulted in morbidity and mortality of the animals within 4 to 16 weeks. Cells from two patients who relapsed 3.5 years after diagnosis grew much slower than the early relapse samples, taking up to 30 weeks to infiltrate the bone marrow of recipient mice. In contrast, leukemic cells were absent or were detected at low numbers in scid mice transplanted with cells obtained at diagnosis from three patients who have not yet relapsed. These results show an increased ability of leukemic cells from patients with aggressive lymphoblastic leukemia of poor prognosis to proliferate in scid mice.


Assuntos
Medula Óssea/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Adolescente , Animais , Antígenos de Diferenciação/análise , Antígenos de Neoplasias/análise , Encéfalo/patologia , Divisão Celular , Criança , Pré-Escolar , Feminino , Antígenos HLA-DR/análise , Humanos , Lactente , Rim/patologia , Fígado/patologia , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neprilisina
10.
Blood ; 89(12): 4307-16, 1997 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9192753

RESUMO

Based on initial observations of human CD34+ Thy-1+ cells and long-term culture-initiating cells (LTC-IC) in the bone marrow of some sublethally irradiated severe combined immunodeficient (SCID) mice transplanted intravenously with normal human marrow cells, and the subsequent finding that the NOD/LtSz-scid/scid (NOD/SCID) mouse supports higher levels of human cell engraftment, we undertook a series of time course experiments to examine posttransplant changes in the number, tissue distribution, cycling activity, and in vivo differentiation pattern of various human hematopoietic progenitor cell populations in this latter mouse model. These studies showed typical rapid posttransplant recovery curves for human CD34- CD19+ (B-lineage) cells, CD34+ granulopoietic, erythroid, and multilineage colony-forming cells (CFC), LTC-IC, and CD34+ Thy-1+ cells from a small initial population representing <0.1% of the original transplant. The most primitive human cell populations reached maximum values at 5 weeks posttransplant, after which they declined. More mature cell types peaked after another 5 weeks and then declined. A 2-week course of thrice weekly injections of human Steel factor, interleukin (IL)-3, granulocyte-macrophage colony-stimulating factor (GM-CSF), and erythropoietin (administered just before the mice were killed for analysis) did not alter the pace of regeneration of either primitive or mature human hematopoietic cells, or their predominantly granulopoietic and B-lymphoid pattern of differentiation, although a significant enhancing effect on the level of human cell engraftment sustained after 3 months was noted. Cycling studies showed the human CFC present at 4 to 5 weeks posttransplant to be rapidly proliferating even in mice not given human growth factors. However, by 10 weeks and thereafter, only quiescent human CFC were detected; interestingly, even in mice that were given the 2-week course of growth factor injections. These studies indicate the use of this model for future analysis of the properties and in vivo regulation of primitive human hematopoietic cells that possess in vivo repopulating ability.


Assuntos
Transplante de Medula Óssea/patologia , Linhagem da Célula , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Imunodeficiência Combinada Severa/patologia , Transplante Heterólogo/patologia , Animais , Medula Óssea/patologia , Ciclo Celular , Divisão Celular , Células Cultivadas , Feminino , Sobrevivência de Enxerto , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos SCID , Quimera por Radiação , Especificidade da Espécie , Baço/patologia
11.
Blood ; 91(7): 2406-14, 1998 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9516140

RESUMO

We have previously shown that intravenously injected peripheral blood (PB) or bone marrow (BM) cells from newly diagnosed chronic myeloid leukemia (CML) patients can engraft the BM of sublethally irradiated severe combined immunodeficient (SCID) mice. We now report engraftment results for chronic phase CML cells in nonobese diabetic (NOD)/SCID recipients which show the superiority of this latter model. Transplantation of NOD/SCID mice with 7 to 10 x 10(7) patient PB or BM cells resulted in the continuing presence of human cells in the BM of the mice for up to 7 months, and primitive human CD34+ cells, including those detectable as colony-forming cells (CFC), as long-term culture-initiating cells, or by their coexpression of Thy-1, were found in a higher proportion of the NOD/SCID recipients analyzed, and at higher levels than were seen previously in SCID recipients. The human CFC and total human cells present in the BM of the NOD/SCID mice transplanted with CML cells also contained higher proportions of leukemic cells than were obtained in the SCID model, and NOD/SCID mice could be repopulated with transplants of enriched CD34+ cells from patients with CML. These results suggest that the NOD/SCID mouse may allow greater engraftment and amplification of both normal and leukemic (Ph+) cells sufficient for the quantitation and characterization of the normal and leukemic stem cells present in patients with CML. In addition, this model should make practical the investigation of mechanisms underlying progression of the disease and the development of more effective in vivo therapies.


Assuntos
Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Adulto , Idoso , Animais , Contagem de Células Sanguíneas , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Transplante de Neoplasias , Transplante Homólogo
12.
Blood ; 87(4): 1539-48, 1996 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8608245

RESUMO

Progress in understanding the abnormal regulation of hematopoiesis in chronic myelogenous leukemia (CML) would be facilitated if neoplastic cells, at all stages of the disease, could be studied in an animal model. In this report, we show that irradiated severe combined immunodeficient (SCID) mice can be transplanted with both normal (Philadelphia chromosome [Ph]-negative) and neoplastic (Ph+) cells from CML patients with either chronic or blast phase disease. Mice transplanted with peripheral blood (PB) or bone marrow (BM) cells from 9 of 12 chronic phase CML patients were well engrafted with human cells including multilineage colony-forming progenitors and CD34+ cells for at least 90 days posttransplantation. Repeated posttransplant injections of cytokines did not enhance the number of engrafted human cells. Interestingly, approximately 70% of the human progenitors found in the engrafted SCID BM were Ph-, suggesting that the growth of primitive normal cells is favored in this in vivo transplant model. A similar number of normal cells were found in mice transplanted with either PB or BM cells, suggesting that elevated numbers of primitive normal cells are present in CML PB. When cells from patients with CML in either myeloid or lymphoid blast crisis were transplanted into SCID mice, the BM of these mice was more rapidly repopulated and to a higher level than that observed with transplants of chronic phase cells. Moreover, all human colony-forming progenitors present in the BM of mice transplanted with blast crisis cells were Ph+, and the majority of cells showed the same morphological features of the blast crisis cells originally transplanted. These experiments provide a starting point for the creation of an animal model of CML and establish the feasibility of using this model for the future characterization of transplantable CML stem cells during disease progression.


Assuntos
Medula Óssea/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células-Tronco Neoplásicas/patologia , Animais , Antígenos CD19/análise , Antígenos CD34/análise , Sequência de Bases , Crise Blástica , Doença Crônica , Primers do DNA/química , Proteínas de Fusão bcr-abl/genética , Genes abl , Humanos , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Transplante de Neoplasias , Quimera por Radiação
13.
Proc Natl Acad Sci U S A ; 92(24): 11024-8, 1995 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-7479929

RESUMO

Many cancers overexpress a member of the bcl-2 family of inhibitors of apoptosis. To determine the role of these proteins in maintaining cancer cell viability, an adenovirus vector that expresses bcl-xs, a functional inhibitor of these proteins, was constructed. Even in the absence of an exogenous apoptotic signal such as x-irradiation, this virus specifically and efficiently kills carcinoma cells arising from multiple organs including breast, colon, stomach, and neuroblasts. In contrast, normal hematopoietic progenitor cells and primitive cells capable of repopulating severe combined immunodeficient mice were refractory to killing by the bcl-xs adenovirus. These results suggest that Bcl-2 family members are required for survival of cancer cells derived from solid tissues. The bcl-xs adenovirus vector may prove useful in killing cancer cells contaminating the bone marrow of patients undergoing autologous bone marrow transplantation.


Assuntos
Apoptose , Células-Tronco Hematopoéticas/citologia , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/fisiologia , Adenovírus Humanos/genética , Animais , Células da Medula Óssea , Terapia Genética , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos SCID , Neoplasias/genética , Neoplasias/terapia , Proteínas Recombinantes , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína bcl-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA