Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Leukemia ; 21(4): 764-71, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17268528

RESUMEN

Evidence is continuing to accumulate that the FMS-like tyrosine kinase 3 (FLT3) receptor plays an important role in acute leukemias. Acute myeloid leukemia patients often express constitutive active mutant forms of the receptor in their leukemic cells. A t(12;13)(p13;q12) translocation between Tel and the FLT3 receptor was recently described in a patient with myeloproliferative disease (MPD). Here a Tel-FLT3 construct mimicking this fusion protein was used to generate transgenic mice. The fusion protein was previously found to constitutively activate FLT3 signaling and transform Ba/F3 cells. Expression of the fusion protein in the transgenic mice was found in all tissues assayed including spleen, bone marrow (BM), thymus and liver. These mice developed splenomegaly and had a high incidence of MPD with extramedullary hematopoiesis in the liver and lymph nodes. Spleens also had increased dendritic and natural killer cell populations. In vitro analysis of the hematopoietic progenitor cells derived from Tel-FLT3 transgenic mice showed a significant increase in the number of CFU-GM in the BM, and CFU-GM, BFU-E and CFU-GEMM in the spleen. BM also showed significant increases of in vivo CFU-S colonies. Thus, transgenic mice expressing constitutively activated Tel-FLT3 develop MPD with a long latency and also result in the expansion of the hematopoietic stem/progenitor cells.


Asunto(s)
Trastornos Mieloproliferativos/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Médula Ósea/patología , Activación Enzimática , Humanos , Hígado/patología , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/metabolismo , Bazo/patología , Timo/patología
3.
Ann N Y Acad Sci ; 938: 191-4; discussion 194-5, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11458508

RESUMEN

Using an in vivo selection technique, we can isolate individual cells that can repopulate the hematopoietic system of a lethally irradiated murine recipient. These cells rapidly acquire a CD34 phenotype in the animal. Progenitors in our long-term chimeras are of donor type. We also have evidence that transplantation of limiting numbers (as low as a single cell) that have this long-term repopulating ability (LTRA) can self-renew. This is demonstrated by serial transplantation of marrow from engrafted recipients 11 months post transplant into new hosts for four additional months.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Compuestos Orgánicos , Animales , Antígenos CD34/análisis , Médula Ósea/patología , División Celular , Movimiento Celular , Femenino , Colorantes Fluorescentes , Supervivencia de Injerto , Células Madre Hematopoyéticas/clasificación , Células Madre Hematopoyéticas/citología , Inmunofenotipificación , Hibridación Fluorescente in Situ , Masculino , Ratones , Quimera por Radiación
4.
Cell ; 105(3): 369-77, 2001 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-11348593

RESUMEN

Purification of rare hematopoietic stem cell(s) (HSC) to homogeneity is required to study their self-renewal, differentiation, phenotype, and homing. Long-term repopulation (LTR) of irradiated hosts and serial transplantation to secondary hosts represent the gold standard for demonstrating self-renewal and differentiation, the defining properties of HSC. We show that rare cells that home to bone marrow can LTR primary and secondary recipients. During the homing, CD34 and SCA-1 expression increases uniquely on cells that home to marrow. These adult bone marrow cells have tremendous differentiative capacity as they can also differentiate into epithelial cells of the liver, lung, GI tract, and skin. This finding may contribute to clinical treatment of genetic disease or tissue repair.


Asunto(s)
Antígenos CD34/metabolismo , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Proteínas de la Membrana/metabolismo , Compuestos Orgánicos , Células Madre/citología , Animales , Linaje de la Célula , Movimiento Celular , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , Colorantes Fluorescentes/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Intestino Delgado/citología , Queratinas/metabolismo , Pulmón/citología , Masculino , Ratones , Ratones Noqueados , Surfactantes Pulmonares/genética , Surfactantes Pulmonares/metabolismo , Células Madre/fisiología , Cromosoma Y/genética , Cromosoma Y/metabolismo
5.
Ann N Y Acad Sci ; 872: 48-54; discussion 54-6, 1999 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-10372110

RESUMEN

Long-term repopulating hematopoietic stem cells can be separated from cells which provided radioprotection (short-term repopulating cells) on the basis of size. This might be a result of the quiescent nature of long-term repopulating cells. To define the activity of these populations we utilized a dye, PKH26, which incorporates into the membrane of cells and is equally distributed to daughter cells when they divide. We were able to retrieve PKH26(+)-labeled cells posttransplant in the hematopoietic tissues of the recipients. We could also assess their cell cycle status and their ability, short- and long-term, to reconstitute secondary lethally irradiated hosts in limiting dilution. The results suggest that long-term repopulating cells remain quiescent in the bone marrow shortly after engraftment, whereas cells which radioprotect are more rapidly dividing. We could not detect labeled cells in the peripheral blood posttransplant, and even though cells homed to both the spleen and bone marrow the cells in the bone marrow were significantly more competent at reconstituting lethally irradiated secondary hosts.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Compuestos Orgánicos , Animales , Ciclo Celular , Separación Celular/métodos , Femenino , Colorantes Fluorescentes , Supervivencia de Injerto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Bazo/citología , Factores de Tiempo
6.
Blood ; 93(6): 1916-21, 1999 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10068664

RESUMEN

We have previously demonstrated that we could separate long-term repopulating stem cells from cells that provided radioprotection (short-term repopulating cells) on the basis of size and suggested that this might be due to the quiescent nature of long-term repopulating cells. To further define the activity of these populations, we used a dye (PKH26), which incorporates into the membrane of cells and is equally distributed to daughter cells when they divide. We developed an assay, which allowed us to retrieve PKH26(+) long-term and short-term repopulating cells in the hematopoietic tissues of the recipients posttransplant. We were able to recover the labeled cells and determine their cell cycle activity, as well as their ability to reconstitute secondary lethally irradiated hosts in limiting dilution. The results of our assay suggest that long-term repopulating cells are quiescent in the bone marrow (BM) 48 hours after transplant. We were able to detect only a few labeled cells in the peripheral blood posttransplant and even though cells homed to both the spleen and BM, more long-term repopulating cells homed to the marrow and only these cells, which homed to the marrow, were capable of reconstituting lethally irradiated secondary hosts long-term.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Compuestos Orgánicos , Animales , Ciclo Celular , División Celular , Membrana Celular/química , Separación Celular , Femenino , Colorantes Fluorescentes/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Bazo/citología , Irradiación Corporal Total
8.
J Hematother ; 7(2): 151-7, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9597572

RESUMEN

T cell depletion (TCD) performed by elutriation has decreased the incidence of acute and chronic graft-versus-host disease (GvHD) following bone marrow transplantation (BMT). However, as with all forms of TCD, patients may experience graft failure (10%), delayed engraftment, and mixed chimerism. Because 66%-75% of the CD34+ cells coseparate with the small lymphocytes, which are removed by elutriation, we designed a phase I trial in HLA-identical siblings to determine if the readdition of these previously lost small CD34+ cells would improve elutriation's engraftment kinetics. CD34+ cells were isolated from the small cell fraction of 10 consecutive donor grafts and infused into the recipients along with the TCD graft. The positively selected product had a mean T cell content of 1.2 x 10(5)/kg and was 80% CD34+, doubling the CD34+ content of the graft. All patients engrafted promptly with a median time to 500 neutrophils/mm3, untransfused 50,000 platelets/mm3, and discharge from the hospital of 19 (range 10-25), 24 (14-52), and 24 (18-29) days, respectively. Acute GvHD occurred in 2 patients, and no patient had chronic GvHD. Augmenting stem cell dose may be an efficient and safe alternative for overcoming TCD-associated delayed engraftment and graft failure, rather than increasing immunosuppression.


Asunto(s)
Antígenos CD34/inmunología , Trasplante de Médula Ósea/efectos adversos , Linfocitos T/inmunología , Adulto , Trasplante de Médula Ósea/inmunología , Separación Celular , Femenino , Humanos , Depleción Linfocítica , Masculino , Persona de Mediana Edad , Morbilidad , Proyectos Piloto , Sobrevivientes , Trasplante Homólogo
9.
Cancer Res ; 57(5): 837-41, 1997 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-9041182

RESUMEN

Inactivation of the cyclin-dependent kinase inhibitors p16INK4A and p15INK4B are frequent alterations in neoplasia, often resulting from homozygous deletion or promoter region hypermethylation. We have analyzed both modes of inactivation of p15INK4B and p16INK4A in the major types of adult and pediatric hematological malignancies. Hypermethylation of p15INK4B, without alteration of p16INK4A, was an almost universal finding in adult acute myelogenous leukemia, and occurred very frequently in adult acute lymphocytic leukemia and pediatric acute myelogenous leukemia and acute lymphocytic leukemia. In contrast, neither p15INK4B nor p16INK4A were inactivated in any stage of chronic myelogenous leukemia. Hypermethylation of p16INK4A, often without alterations of p15INK4B, was found in non-Hodgkin's lymphoma and was much more frequent in cases with high-grade than low-grade histology. Enriched normal bone marrow stem cells had no detectable promoter region methylation of these genes, as analyzed by a newly developed PCR method. Remarkably distinct patterns of inactivation of p15INK4B and p16INK4A characterize different types of hematological malignancy, and alterations in these tumor suppressor genes are one of the most common alterations in hematological malignancies.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Leucemia/genética , Linfoma/genética , Proteínas Supresoras de Tumor , Adulto , Biomarcadores de Tumor , Niño , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Metilación de ADN , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico , Eliminación de Secuencia
10.
Stem Cells ; 15 Suppl 1: 41-4; discussion 44-5, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9368323

RESUMEN

The purpose of this report is to demonstrate the phenotypic and functional characteristics of a primitive hematopoietic stem cell (HSC). We present evidence that an isolated murine HSC can repopulate the hematopoietic tissues of lethally irradiated recipient animals long term. By limiting dilution, as few as ten isolated stem cells can reconstitute mice for their lifetime. The stem cell which we have isolated does not copurify with colony forming unit-spleen or radioprotect recipients from lethal radiation. The phenotypic characterization of this rare cell, which represents 0.005% of total bone marrow, includes either the absence or very low expression of markers associated with long-term repopulating cells described by other groups. We believe this stem cell represents a very early self-renewing stem cell in the mouse.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Antígenos de Diferenciación/análisis , Trasplante de Médula Ósea , Femenino , Células Madre Hematopoyéticas/química , Masculino , Ratones , Fenotipo , Tolerancia a Radiación , Bazo/citología
11.
Cancer Res ; 56(20): 4551-5, 1996 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8840958

RESUMEN

The induction of apoptosis by the Fas/APO-1 receptor is important for T-cell-mediated cytotoxicity and down-regulation of immune responses. Binding of Fas ligand to the Fas/APO-1 receptor transduces an apoptotic signal that requires activation of interleukin 1beta-converting enzyme (ICE) and CPP32beta, members of a family of cysteine proteases that are evolutionarily conserved determinants of cell death. We report here that Fas/APO-1-triggered apoptosis involves ICE-mediated activation of p34cdc2 kinase. Ligation of the Fas receptor resulted in the rapid stimulation of ICE proteolytic activity and activation of p34cdc2 kinase. Specific tetrapeptide inhibitors of ICE (Acetyl-Tyr-Val-Ala-Asp-chloromethylketone) or CPP32beta (Acetyl-Asp-Glu-Val-Asp-aldehyde) prevented the anti-Fas antibody-mediated activation of p34cdc2 and inhibited apoptosis. Inhibition of p34cdc2 activity by transient overexpression of a dominant-negative cdc2 construct or human WEE1 kinase inhibited Fas-mediated apoptosis. These results suggest that activation of p34cdc2 kinase is a critical determinant of cell death mediated by Fas and ICE family proteases.


Asunto(s)
Apoptosis/fisiología , Proteína Quinasa CDC2/metabolismo , Cisteína Endopeptidasas/biosíntesis , Proteínas Quinasas/biosíntesis , Receptor fas/inmunología , Apoptosis/genética , Caspasa 1 , Activación Enzimática/efectos de los fármacos , Vectores Genéticos , Humanos , Células Jurkat/metabolismo , Células Jurkat/patología , Receptores de Lipoproteína/fisiología , Transfección
12.
Nat Med ; 2(10): 1140-3, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8837615

RESUMEN

The induction of tumor cell death by anticancer therapy results from a genetic program of autonomous cell death termed apoptosis. Because the p53 tumor suppressor gene is a critical component for induction of apoptosis in response to DNA damage, its inactivation in cancers may be responsible for their resistance to genotoxic anticancer agents. The cellular response to DNA damage involves a cell-cycle arrest at both the G1/S and G2/M transitions; these checkpoints maintain viability by preventing the replication or segregation of damaged DNA. The arrest at the G1 checkpoint is mediated by p53-dependent induction of p21WAF1/CIP1, whereas the G2 arrest involves inactivation of p34cdc2 kinase. Following DNA damage, p53-deficient cells fail to arrest at G1 and accumulate at the G2/M transition. We demonstrate that abrogation of G2 arrest by caffeine-mediated activation of p34cdc2 kinase results in the selective sensitization of p53-deficient primary and tumor cells to irradiation-induced apoptosis. These data suggest that pharmacologic activation of p34cdc2 kinase may be a useful therapeutic strategy for circumventing the resistance of p53-deficient cancers to genotoxic anticancer agents.


Asunto(s)
Apoptosis/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Cafeína/farmacología , Tolerancia a Radiación/efectos de los fármacos , Proteína p53 Supresora de Tumor/deficiencia , Animales , Apoptosis/efectos de la radiación , Médula Ósea/efectos de la radiación , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Daño del ADN , Activación Enzimática/efectos de los fármacos , Femenino , Fase G2/efectos de los fármacos , Genes p53 , Masculino , Ratones , Ratones Noqueados , Proteína p53 Supresora de Tumor/fisiología
13.
Blood ; 88(2): 487-91, 1996 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-8695796

RESUMEN

The classical definition of lymphohematopoietic stem cells (LHSC), the most primitive progenitors of all blood cells, requires that they have the capacity for self-renewal and for the long-term production of all blood cell lineages. However, other characteristics of LHSC have been debated. Our previous data suggested that mouse LHSC are very slowly proliferating cells that generate delayed multilineage engraftment, while "radioprotection" (rapid engraftment that will prevent early death from radiation-induced marrow aplasia) results from more committed progenitors. Alternatively, some groups have reported that mouse LHSC are responsible for both radioprotection and long-term repopulation of all blood cell lineages. A possible explanation for this difference is that cells with the capacity for long-term production of all blood cell lineages are biologically heterogeneous. We now show that 10 LHSC can generate all blood cell lineages for the lifetime of the animal. However, these cells lacked radioprotection and spleen colony-forming activity. LHSC were identified and isolated by their small size, their lack of expression of antigens characteristic of mature blood cell lineages, and their high expression of aldehyde dehydrogenase. In addition, these cells were found to express undetectable or low levels of many antigens presumed to mark LHSC, including Thy-1, Ly-6A/E (Sca-1), c-kit, and CD34. There appears to be at least two classes of LHSC with the capacity for long-term production of all blood cell lineages: one that generates both radioprotection and long-term engraftment and one that produces delayed but durable engraftment. Our data suggest that this latter class may represent a very primitive class of LHSC.


Asunto(s)
Células de la Médula Ósea , Células Madre Hematopoyéticas/clasificación , Bazo/citología , Aldehído Deshidrogenasa/análisis , Animales , Antígenos Ly/análisis , Secuencia de Bases , Biomarcadores , Trasplante de Médula Ósea , Linaje de la Célula , Tamaño de la Célula , Ensayo de Unidades Formadoras de Colonias , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Inmunofenotipificación , Masculino , Proteínas de la Membrana/análisis , Ratones , Datos de Secuencia Molecular , Quimera por Radiación , Tolerancia a Radiación , Antígenos Thy-1/análisis
14.
Cancer Res ; 56(5): 973-77, 1996 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-8640788

RESUMEN

Estrogen appears to be a negative regulator of normal hematopoiesis. Chromosome 6q, which contains the estrogen receptor (ER) gene, is frequently altered in human hematopoietic neoplasms. The ER gene, which has growth and metastasis suppressor activity in many different cell types, is inactivated by promoter methylation in some ER-negative breast tumors and 100% of colorectal tumors. We now report that the promoter region of the ER gene is aberrantly methylated in 86% of human hematopoietic tumors, including 8 of 9 pediatric acute lymphocytic leukemia, 17 of 18 adult acute lymphocytic leukemia, 21 of 23 adult acute myelogenous leukemia, 3 of 6 chronic phase chronic myelogenous leukemia, 9 of 9 blast crisis chronic myelogenous leukemia and 5 of 8 lymphomas. This methylation event was also present in all nine leukemia cell lines examined, where it was associated with very low or absent ER expression. In addition, rat and mouse leukemia cell line also exhibited this change, indicating that ER CpG island methylation in leukemias is conserved among species. Our results suggest that ER CpG island methylation could be an important step in the genesis of human hematopoietic neoplasms and might be a useful molecular marker for monitoring the clinical status of these diseases.


Asunto(s)
Cromosomas Humanos Par 6 , Leucemia/genética , Linfoma/genética , Receptores de Estrógenos/genética , Adulto , Animales , Biomarcadores de Tumor , Niño , Preescolar , Islas de CpG , Humanos , Lactante , Recién Nacido , Leucemia/metabolismo , Linfoma/metabolismo , Metilación , Ratones , Ratas , Células Tumorales Cultivadas
15.
Curr Opin Hematol ; 2(1): 12-21, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9371967

RESUMEN

The hematopoietic developmental hierarchy originates with a rare population of lymphohematopoietic stem cells that are capable of extensive self-renewal as well as the continuous generation of more developmentally restricted progeny. The generation of mature blood cells from these pluripotent hematopoietic stem cells involves the highly regulated progression through successive stages involving commitment to a specific cell lineage, terminal differentiation of lineage-restricted progenitors, and growth arrest. Although the differentiation commitment of stem cells may be intrinsically determined, it is apparent that a wide variety of external and internal stimuli can influence and modulate lineage choice and differentiation. These factors cooperate with cellular transcription factors to activate or repress the expression of genes responsible for lineage choice, diverse mature phenotypes, and cell cycle progression. The extrinsic and genetic mechanisms that orchestrate the differentiation commitment and myeloid lineage restriction of pluripotent stem cells are of fundamental importance in the regulation of hematopoiesis. The elucidation of these mechanisms of normal myeloid differentiation has provided instrumental insights into the biology of leukemia and other hematopoietic disorders.


Asunto(s)
Células Madre Hematopoyéticas/citología , Leucemia Mieloide/patología , Animales , Blastocisto/citología , Diferenciación Celular/fisiología , Hematopoyesis/fisiología , Factores de Crecimiento de Célula Hematopoyética/fisiología , Humanos , Interferones/fisiología , Tretinoina/fisiología
16.
Cancer Res ; 54(21): 5535-8, 1994 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7923192

RESUMEN

Expression of the BCR-ABL chimeric gene in chronic myeloid leukemia results in the inhibition of apoptosis, a genetically programmed process of autonomous cell death. BCR-ABL and other genetic factors that suppress apoptosis confer cross-resistance to cytotoxic agents with diverse mechanisms of action. Eradication of the chronic myeloid leukemia clone requires strategies that circumvent this inherent resistance to cytotoxic therapy. We have determined that BCR-ABL expression augments the sensitivity of hematopoietic cells to growth factor-mediated signals of differentiation; hematopoietic growth factors induce the selective terminal differentiation of chronic myeloid leukemia progenitors at concentrations that allow optimal growth of normal progenitors. Hematopoietic growth factors may be an effective strategy for the elimination of cytotoxic therapy-resistant leukemic cells by inducing their terminal differentiation while allowing concomitant expansion of coexistent normal hematopoietic progenitors.


Asunto(s)
Diferenciación Celular/genética , Regulación Leucémica de la Expresión Génica/genética , Interleucina-3/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proto-Oncogenes/fisiología , Animales , Secuencia de Bases , Genes abl/genética , Granulocitos/patología , Humanos , Macrófagos/patología , Ratones , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/farmacología , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
17.
Blood ; 84(3): 691-701, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-7519070

RESUMEN

CD34 is expressed on human hematopoietic stem and progenitor cells, and its clinical usefulness for the purification of stem cells has been well established. However, a similar pattern of expression for murine CD34 (mCD34) has not yet been determined. Two polyclonal anti-mCD34 antibodies that specifically recognize both endogenous and recombinant murine CD34 were developed to characterize the mCD34 protein and to determine its pattern of expression on murine cell lines and hematopoietic progenitor cells. Fluorescence-activated cell sorter analysis showed that mCD34 is expressed on NIH/3T3 embryonic fibroblasts, PA6 stromal cells, embryonic stem cells, M1 leukemia cells, and a subpopulation of normal bone marrow cells. Murine CD34 was found to be a glycoprotein expressed on the cell surface as either a full-length (approximately 100 kD) or truncated (approximately 90 kD) protein in NIH/3T3 and PA6 cells. Recombinant full-length CD34, when expressed in the CHO-K1 cell line, had a molecular weight of approximately 105 kD. Full-length CD34 expressed on M1 leukemia cells, had a higher apparent molecular weight (110 kD). These results suggest that there are glycosylation differences between CD34 expressed by different cell types. The full-length form, but not the truncated form, is a phosphoprotein that is hyperphosphorylated in response to 12-0-Tetradecanoyl phorbol 13-acetate treatment, suggesting potential functional differences between the two forms. Selection of the 3% highest-expressing CD34+ bone marrow cells enriched for the hematopoietic precursors that form colony-forming unit-spleen (CFU-S), CFU-granulocyte-macrophage, and burst-forming unit-erythroid. Transplantation of lethally irradiated mice with these cells demonstrated both short- and long-term repopulating ability, indicating that this population contains both functional hematopoietic progenitors and the putative stem cell. These antibodies should be useful to select for murine hematopoietic stem cells.


Asunto(s)
Antígenos CD/metabolismo , Células Madre Hematopoyéticas/inmunología , Células 3T3/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD34 , Antígenos de Superficie/metabolismo , Secuencia de Bases , Células de la Médula Ósea , Citoplasma/inmunología , Cartilla de ADN/química , Leucemia Mieloide Aguda/inmunología , Ratones , Datos de Secuencia Molecular , Fosforilación , Proteínas Recombinantes de Fusión/inmunología , Células Madre/inmunología
18.
Blood ; 83(8): 2038-44, 1994 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-8161775

RESUMEN

BCR-ABL expression is presumed to effect clonal expansion in chronic myeloid leukemia (CML) by deregulation of cell proliferation. However, most studies have found that relative rates of cell proliferation are not increased in CML. Moreover, we found that CML progenitors display a normal proliferative response to growth factors and do not manifest greater proliferative potential than normal progenitors. Growth of malignancies depends on an imbalance between the rate of cell production and the rate of cell death. We found that BCR-ABL expression inappropriately prolongs the growth factor-independent survival of CML myeloid progenitors and granulocytes by inhibiting apoptosis, a genetically programmed process of active cell death; inhibition of BCR-ABL expression by antisense oligonucleotides reversed the suppression of apoptosis as well as the enhancement of survival. The decreased rate of programmed cell death appears to be the primary mechanism by which BCR-ABL effects expansion of the leukemic clone in CML.


Asunto(s)
Apoptosis , Proteínas de Fusión bcr-abl/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Animales , Secuencia de Bases , División Celular , Línea Celular , Supervivencia Celular , Células Madre Hematopoyéticas/patología , Humanos , Interleucina-3/farmacología , Linfoma Folicular/patología , Ratones , Datos de Secuencia Molecular
19.
Blood ; 81(11): 2898-902, 1993 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-8499629

RESUMEN

Chronic myeloid leukemia (CML) is characterized by an initial chronic phase of expanded yet orderly clonal hematopoiesis that is distinguished by the BCR-ABL gene rearrangement. We found that although the mature myeloid compartment in patients with CML was expanded and entirely derived from the dominant leukemic clone, the primitive hematopoietic progenitor compartment did not show a corresponding expansion and was substantially enriched for cells without the BCR-ABL gene rearrangement. More importantly, primitive progenitors exhibiting the BCR-ABL gene rearrangement did not express either the BCR-ABL hybrid mRNA or fusion protein (P210). Expression of P210 protein and BCR-ABL mRNA increased with myeloid commitment in vivo as well as with growth factor-induced proliferation and differentiation of the primitive CML progenitors in vitro. This differential expression of BCR-ABL between primitive and mature CML progenitors may explain the expansion of the leukemic clone at the level of mature myeloid progenitors and granulocytes without a concomitant expansion of primitive CML progenitors. Because BCR-ABL mRNA is minimally expressed or may be absent in primitive CML progenitors, these cells may escape detection by reverse transcriptase-polymerase chain reaction and eradication by antisense oligonucleotides targeted against BCR-ABL mRNA.


Asunto(s)
Proteínas de Fusión bcr-abl/metabolismo , Células Madre Hematopoyéticas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Secuencia de Bases , Aberraciones Cromosómicas/diagnóstico , Trastornos de los Cromosomas , Expresión Génica , Reordenamiento Génico , Genes abl , Células Madre Hematopoyéticas/citología , Humanos , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/química , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Neoplásico/genética , Translocación Genética
20.
Cancer Genet Cytogenet ; 64(2): 145-8, 1992 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1486564

RESUMEN

The mouse Y-specific DNA sequence pY2 was used as a probe for fluorescence in situ hybridization (FISH) to evaluate murine hematopoietic tissues after sex-mismatched bone marrow transplant (BMT). The pY2 probe was localized to the long arm of the Y chromosome on BM metaphases. Hybridization of pY2 in FISH of interphase cells from BM, spleen, and thymus after BMT was compared with Southern blot analysis; both methods gave comparable results. Only FISH was able to analyze post-BMT peripheral blood (PB) samples successfully, and provides a useful method for following engraftment status in the mouse on an ongoing basis.


Asunto(s)
Células de la Médula Ósea , Trasplante de Médula Ósea , Hibridación Fluorescente in Situ , Animales , Southern Blotting , ADN/análisis , Sondas de ADN , Femenino , Marcadores Genéticos , Cariotipificación , Masculino , Ratones , Trasplante Isogénico , Cromosoma Y
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...