Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
BMC Biol ; 20(1): 104, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35550087

RESUMEN

BACKGROUND: Lifelong production of the many types of mature blood cells from less differentiated progenitors is a hierarchically ordered process that spans multiple cell divisions. The nature and timing of the molecular events required to integrate the environmental signals, transcription factor activity, epigenetic modifications, and changes in gene expression involved are thus complex and still poorly understood. To address this gap, we generated comprehensive reference epigenomes of 8 phenotypically defined subsets of normal human cord blood. RESULTS: We describe a striking contraction of H3K27me3 density in differentiated myelo-erythroid cells that resembles a punctate pattern previously ascribed to pluripotent embryonic stem cells. Phenotypically distinct progenitor cell types display a nearly identical repressive H3K27me3 signature characterized by large organized chromatin K27-modification domains that are retained by mature lymphoid cells but lost in terminally differentiated monocytes and erythroblasts. We demonstrate that inhibition of polycomb group members predicted to control large organized chromatin K27-modification domains influences lymphoid and myeloid fate decisions of primary neonatal hematopoietic progenitors in vitro. We further show that a majority of active enhancers appear in early progenitors, a subset of which are DNA hypermethylated and become hypomethylated and induced during terminal differentiation. CONCLUSION: Primitive human hematopoietic cells display a unique repressive H3K27me3 signature that is retained by mature lymphoid cells but is lost in monocytes and erythroblasts. Intervention data implicate that control of this chromatin state change is a requisite part of the process whereby normal human hematopoietic progenitor cells make lymphoid and myeloid fate decisions.


Asunto(s)
Histonas , Células Madre Pluripotentes , Diferenciación Celular , Cromatina/genética , Cromatina/metabolismo , Células Madre Hematopoyéticas/metabolismo , Histonas/genética , Humanos , Recién Nacido , Células Madre Pluripotentes/metabolismo
2.
Oncogene ; 34(23): 3000-10, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25088197

RESUMEN

TWIST2 has a dual function in tumors. Its implication in the initiation and metastasis of various solid tumors is well established, and its tumor-suppressor role in murine osteosarcoma cells has been reported recently. However, the function of TWIST2 and its underlying mechanisms in human normal and malignant hematopoiesis remain unclear. In the present study, we found that TWIST2 directly regulated p21 in human hematopoietic cells and whose silence promoted cell proliferation and cell cycle progression. Hypermethylation of TWIST2 occurred to 23 out of the 75 adult acute myeloid leukemia (AML) patients and resulted in the impaired expression of both TWIST2 and p21. Conversely, TWIST2 overexpression inhibited the growth of AML cells partially through its direct activation of p21 with intact HLH (helix-loop-helix) domain. The microarray data and gene expression validation showed that TWIST2 was sufficient to activate known tumor-suppressor genes, whereas suppress known oncogenes, which further supported its inhibitory effect against AML cells. Taken together, our data have identified a novel TWIST2-p21 axis that modulates the cell cycle of both normal and leukemic cells and demonstrated that the direct regulation of p21 by TWIST2 has a role in its tumor-suppressor function in AML.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas Represoras/genética , Proteína 1 Relacionada con Twist/genética , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Metilación de ADN , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Represoras/metabolismo , Proteína 1 Relacionada con Twist/metabolismo
3.
Leukemia ; 27(1): 159-69, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22868969

RESUMEN

HOX genes have been implicated as regulators of normal and leukemic stem cell functionality, but the extent to which these activities are linked is poorly understood. Previous studies revealed that transduction of primitive mouse hematopoietic cells with a NUP98HOXA10homeodomain (NA10HD) fusion gene enables a subsequent rapid and marked expansion in vitro of hematopoietic stem cell numbers without causing their transformation or deregulated expansion in vivo. To determine whether forced expression of NA10HD in primitive human cells would have a similar effect, we compared the number of long-term culture-initiating cells (LTC-ICs) present in cultures of lenti-NA10HD versus control virus-transduced CD34(+) cells originally isolated from human cord blood and chronic phase (CP) chronic myeloid leukemia (CML) patients. We found that NA10HD greatly increases outputs of both normal and Ph(+)/BCR-ABL(+) LTC-ICs, and this effect is particularly pronounced in cultures containing growth factor-producing feeders. Interestingly, NA10HD did not affect the initial cell cycle kinetics of the transduced cells nor their subsequent differentiation. Moreover, immunodeficient mice repopulated with NA10HD-transduced CP-CML cells for more than 8 months showed no evidence of altered behavior. Thus, NA10HD provides a novel tool to enhance both normal and CP-CML stem cell expansion in vitro, without apparently altering other properties.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Leucemia Mieloide de Fase Crónica/patología , Células Madre Neoplásicas/citología , Proteínas de Complejo Poro Nuclear/genética , Animales , Antígenos CD34/metabolismo , Western Blotting , Ciclo Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Sangre Fetal/citología , Sangre Fetal/metabolismo , Proteínas de Fusión bcr-abl/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Homeobox A10 , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia Mieloide de Fase Crónica/genética , Leucemia Mieloide de Fase Crónica/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Fusión Oncogénica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-2 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
4.
Leukemia ; 24(11): 1823-33, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20861912

RESUMEN

Chronic myeloid leukemia (CML) has long served as a paradigm for generating new insights into the cellular origin, pathogenesis and improved approaches to treating many types of human cancer. Early studies of the cellular phenotypes and genotypes represented in leukemic populations obtained from CML patients established the concept of an evolving clonal disorder originating in and initially sustained by a rare, multipotent, self-maintaining hematopoietic stem cell (HSC). More recent investigations continue to support this model, while also revealing new insights into the cellular and molecular mechanisms that explain how knowledge of CML stem cells and their early differentiating progeny can predict the differing and variable features of chronic phase and blast crisis. In particular, these emphasize the need for new agents that effectively and specifically target CML stem cells to produce non-toxic, but curative therapies that do not require lifelong treatments.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre/patología , Apoptosis , Crisis Blástica/genética , Crisis Blástica/patología , Diferenciación Celular , División Celular , Proteínas de Fusión bcr-abl/genética , Genotipo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Mielopoyesis , Fenotipo , Transcripción Genética
6.
Oncogene ; 28(11): 1421-31, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19151767

RESUMEN

Basal-like breast cancers (BLBCs) are aggressive tumors with high relapse rates and poor survival. We recently reported that >70% of primary BLBCs express the oncogenic transcription/translation factor Y-box binding protein-1 (YB-1) and silencing it with small interfering RNAs (siRNAs) attenuates the growth of BLBC cell lines. To understand the basis of these earlier findings, we profiled YB-1:DNA complexes by chromatin immunoprecipitation (ChIP)-on-chip. Several tumor growth-promoting genes such as MET, CD44, CD49f, WNT and NOTCH family members were identified. In addition, YB-1 and MET are coordinately expressed in BLBC cell lines, as well as in normal human mammary progenitor cells. MET was confirmed to be a YB-1 target through traditional ChIP and gel-shift assays. More specifically, YB-1 binds to -1018 bp on the MET promoter. Silencing YB-1 with siRNA decreased MET promoter activity, transcripts, as well as protein levels and signaling. Conversely, expressing wild-type YB-1 or a constitutively active mutant YB-1 (D102) increased MET expression. Finally, silencing YB-1 or MET attenuated anchorage-independent growth of BLBC cell lines. Together, these findings implicate MET as a target of YB-1 that work in concert to promote BLBC growth.


Asunto(s)
Neoplasias de la Mama/patología , Mama/química , Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Factores de Crecimiento/fisiología , Neoplasias de la Mama/química , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Femenino , Perfilación de la Expresión Génica , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-met , ARN Interferente Pequeño/genética , Receptores de Factores de Crecimiento/análisis , Receptores de Factores de Crecimiento/genética , Células Madre/química , Proteína 1 de Unión a la Caja Y
7.
Curr Oncol ; 15(2): 90-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18454182

RESUMEN

Chronic myeloid leukemia (cml) is a myeloproliferative disorder whose therapy has changed dramatically since the late 1990s. With the introduction of the tyrosine kinase inhibitor (tki) imatinib mesylate, the treatment outcomes for patients with cml have improved markedly, and hematopoietic stem-cell transplantation is no longer routinely offered as first-line therapy for most patients in chronic phase.However, resistance to tki therapy is increasingly being recognized, and alternative therapy is needed for this group of patients. In addition, the development of models predicting response to tki therapy is desired, so that appropriate treatment strategies can be used for individual patients. The present report serves to outline the approach to the treatment of cml in British Columbia and to highlight areas of ongoing research.

8.
Leukemia ; 19(7): 1184-91, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15889158

RESUMEN

Recent studies indicate that a rare population of primitive quiescent BCR-ABL(+) cells are innately insensitive to imatinib mesylate (IM) and persist after IM therapy of patients with chronic myeloid leukemia (CML). New approaches to the eradication of these cells are therefore likely to be crucial to the development of curative therapies for CML. We have now found that Ara-C, LY294002 (a PI-3 (phosphatidylinositol-3' kinase) kinase inhibitor), 17AAG (a heat-shock protein (HSP)-90 antagonist) and lonafarnib (a farnesyltransfease inhibitor) all enhance the toxicity of IM on K562 cells and on the total CD34(+) leukemic cell population from chronic phase CML patients. However, for quiescent CD34(+) leukemic cells, this was achieved only by concomitant exposure of the cells to lonafarnib. Ara-C or LY294002 alone blocked the proliferation of these cells but did not kill them, and Ara-C, LY294002 or 17AAG in combination with IM enhanced the cytostatic effect of IM but did not prevent the subsequent regrowth of the surviving leukemic cells. These studies demonstrate the importance of in vitro testing of novel agents on the subset of primary leukemic cells most likely to determine long-term treatment outcomes in vivo.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proliferación Celular/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piperazinas/farmacología , Piperidinas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Antígenos CD34/efectos de los fármacos , Benzamidas , Benzoquinonas , Línea Celular Tumoral , Cromonas/farmacología , Citarabina/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Humanos , Mesilato de Imatinib , Lactamas Macrocíclicas , Masculino , Morfolinas/farmacología , Rifabutina/análogos & derivados , Rifabutina/farmacología
9.
Leukemia ; 19(3): 435-41, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15674418

RESUMEN

Xenograft models of chronic phase human chronic myeloid leukemia (CML) have been difficult to develop because of the persistence of normal hematopoietic stem cells in most chronic phase CML patients and the lack of methods to selectively isolate the rarer CML stem cells. To circumvent this problem, we first identified nine patients' samples in which the long-term culture-initiating cells were predominantly leukemic and then transplanted cells from these samples into sublethally irradiated NOD/SCID and NOD/SCID-beta2microglobulin-/- mice. This resulted in the consistent and durable (>5 months) repopulation of both host genotypes with similar numbers of BCR-ABL+/Ph+ cells. The regenerated leukemic cells included an initial, transient population derived from CD34+CD38+ cells as well as more sustained populations derived from CD34+CD38- progenitors, indicative of a hierarchy of transplantable leukemic cells. Analysis of the phenotypes produced revealed a reduced output of B-lineage cells, enhanced myelopoiesis with excessive production of erythroid and megakaropoietic cells and the generation of primitive (CD34+) leukemic cells displaying an autocrine IL-3 and G-CSF phenotype, all characteristics of primary CML cells. These findings demonstrate the validity of this xenograft model of chronic phase human CML, which should enable future investigation of disease pathogenesis and new approaches to therapy.


Asunto(s)
Modelos Animales de Enfermedad , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Leucemia Experimental/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/trasplante , Animales , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Interleucina-3/genética , Interleucina-3/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Fenotipo , Quimera por Radiación , Factores de Tiempo , Trasplante Heterólogo/métodos
10.
Leukemia ; 18(5): 1006-12, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15014528

RESUMEN

The tyrosine kinase activity of p210BCR-ABL is essential to its leukemogenic potential, but the role of other functional domains in primary human hematopoietic cells has not been previously investigated. Here we show that infection of normal human CD34+ cord blood (CB) cells with a retroviral vector encoding p210BCR-ABL rapidly activates a factor-independent phenotype and autocrine interleukin-3/granulocyte colony-stimulating factor/erythropoietin production in the transduced cells. These changes are characteristic of primitive chronic myeloid leukemic (CML) cells and are important to the leukemogenicity of BCR-ABL-transduced murine hematopoietic stem cells. When BCR-ABL-transduced human CB cells were incubated with imatinib mesylate, an inhibitor of the p210BCR-ABL kinase, or when human CB cells were transduced with a BCR-ABL cDNA lacking the SH2 domain (p210DeltaSH2), factor independence was significantly reduced. In contrast, deletion of the SH2 domain had little impact on the p210BCR-ABL kinase-dependent promotion of erythropoietic differentiation also seen immediately following the BCR-ABL transduction of primitive human CB cells, but not in naturally occurring CML. Thus, p210BCR-ABL has distinct biological effects in primary human hematopoietic cells, which variably mimic features of human CML, and activation of these changes can show different dependencies on the integrity of the SH1 and SH2 domains of p210BCR-ABL.


Asunto(s)
Sangre Fetal/citología , Proteínas de Fusión bcr-abl/fisiología , Células Madre Hematopoyéticas/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Antígenos CD34/análisis , División Celular , Linaje de la Célula , Eritropoyetina/biosíntesis , Proteínas de Fusión bcr-abl/química , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Interleucina-3/farmacología , Transducción Genética , Dominios Homologos src
11.
Leukemia ; 18(2): 341-7, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14628073

RESUMEN

Transplantation of immunodeficient mice with human hematopoietic cells has greatly facilitated studies of the earliest stages of human hematopoiesis. These include demonstration of the ability of injected 'human-specific' hematopoietic growth factors to enhance the production of human cells at multiple levels of differentiation. In contrast, the effects of continuous exposure to such molecules have not been well investigated. Here, we show that nonobese diabetic severe combined immunodeficiency mice genetically engineered to produce ng/ml serum levels of human interleukin-3 (IL-3), granulocyte/macrophage-stimulating factor (GM-CSF) and Steel factor (SF) display a complex phenotype when transplanted with primitive human bone marrow (BM) or fetal liver cells. This phenotype is characterized by an enhancement of terminal human myelopoiesis and a matched suppression of terminal human erythropoiesis, with a slight reduction in human B-lymphopoiesis in the BM of the engrafted mice. Human clonogenic progenitors are more prevalent in the blood of the transplanted growth factor-producing mice and this is accompanied by a very marked reduction of more primitive human cells in the BM. Our findings suggest that long-term exposure of primitive human hematopoietic cells to elevated levels of human IL-3, GM-CSF and SF in vivo may deleteriously affect the stem cell compartment, while expanding terminal myelopoiesis.


Asunto(s)
Factores de Crecimiento de Célula Hematopoyética/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Hematopoyesis/efectos de los fármacos , Factores de Crecimiento de Célula Hematopoyética/genética , Células Madre Hematopoyéticas/fisiología , Humanos , Interleucina-3/genética , Interleucina-3/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Regeneración , Factor de Células Madre/genética , Factor de Células Madre/farmacología , Trasplante Heterólogo
13.
Leukemia ; 17(4): 760-3, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12682634

RESUMEN

Primitive malignant progenitors defined as nonobese diabetic/severe combined immunodeficient (NOD/SCID) leukemia-initiating cells or NOD/SL-IC from patients with acute myeloid leukemia (AML) can be detected and quantitated in sublethally irradiated NOD/SCID mice. However, there is variability in the levels of bone marrow (BM) engraftment obtained after intravenous injection of cells from different AML samples. In the current study, AML cell engraftment in standard NOD/SCID mice was compared to that obtained with NOD/SCID mice transgenic for the human growth factor genes Steel factor (SF), interleukin-3 (IL-3) and granulocyte macrophage-colony-stimulating factor (GM-CSF) (N/S-S/GM/3) as well as beta 2 microglobulin-null NOD/SCID (N/S-beta 2m(-/-)) mice. Three of the eight AML samples that failed to engraft in standard NOD/SCID animals showed easily detectable and up to 70-fold increased in the number of leukemic cells in BM 8-12 weeks post-transplantation in each of the N/S-beta 2m(-/-) and N/S-S/GM/3 mouse strains. In two of the four AML samples studied at limiting dilution, the frequency of NOD/SL-IC detected was increased six- and seven-fold. Thus, in these novel mouse strains a broader spectrum of AML patient samples can be evaluated for their progenitor content and potentially studied for their response to innovative therapeutics in vivo.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Interleucina-3/fisiología , Leucemia Mieloide/patología , Trasplante de Neoplasias , Factor de Células Madre/fisiología , Trasplante Heterólogo , Microglobulina beta-2/deficiencia , Enfermedad Aguda , Animales , Médula Ósea/patología , Citometría de Flujo , Supervivencia de Injerto , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Hibridación Fluorescente in Situ , Interleucina-3/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Quimera por Radiación , Proteínas Recombinantes de Fusión/fisiología , Factor de Células Madre/genética , Microglobulina beta-2/genética
14.
Cytotherapy ; 4(5): 407-13, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12473207

RESUMEN

BACKGROUND: CML progenitor-cell studies would be greatly facilitated if samples could be repeatedly accessed from a source of well-characterized cells. The present study was designed to develop a simple, inexpensive Ab cocktail that would provide subpopulations of cells enriched for CD34+ cells and simultaneously depleted of CD15+ mature myeloid cells. METHODS: Cells from leukapheresis products from CML patients at diagnosis were incubated with each of two Ab cocktails. The standard cocktail (debulking, DB), containing 11 Abs, is recommended for obtaining a highly enriched population of CD34+ cells. The efficacy of an alternative, simpler cocktail (CML custom, CC), containing only four Abs was tested. The recoveries of CD34+ cells, CD15+ cells, colony-forming unit granulocyte-macrophage, and LTCIC were monitored. The samples were then cryopreserved, thawed, and the recoveries remeasured. RESULTS: The purity of CD34+ cells was significantly superior using the DB cocktail than with the CC cocktail. Conversely, using the CC cocktail, the yield of CD34+ cells was significantly higher compared to the DB cocktail. These results were maintained even when the amount of Ab was reduced 10-fold. Both Ab cocktails consistently removed > 99% of the CD15+ cells. Consistent with the CD34+ cell-enrichment data, higher colony-forming cell (CFC) frequencies were obtained with the DB cocktail, although superior yields of CFC were obtained with the CC cocktail. After cryopreservation and thawing the yield of CD34+ cells remained high, and a further reduction in the number of CD15+ cells was obtained. DISCUSSION: A method is described that allows the rapid and efficient debulking of large CML samples. This strategy will provide a source of well-characterized CML stem/progenitor cells that can be repeatedly accessed.


Asunto(s)
Antígenos CD34/biosíntesis , Separación Celular/métodos , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Antígeno Lewis X/biosíntesis , Linfocitos T/inmunología , Criopreservación , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Separación Inmunomagnética/métodos , Células Mieloides/inmunología , Trasplante de Células Madre
15.
Leukemia ; 16(4): 549-58, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11960331

RESUMEN

Chronic myeloid leukemia (CML) has been studied intensively for many years; yet its treatment remains problematic and its biology remains elusive. In chronic phase, the leukemic clone appears to be maintained by a small number of BCR-ABL-positive hematopoietic stem cells that differentiate normally and amplify slowly. In contrast, as these cells enter the intermediate stages of lineage restriction, their progeny are selectively expanded and generate an enlarged pool of neoplastic progenitors. Recent analyses of purified subsets of primitive CML cells have provided a coherent explanation for this dichotomous behavior of BCR-ABL-positive stem and progenitor cells based on the discovery of an unusual autocrine IL-3/G-CSF mechanism activated in them. This only partially counteracts in vivosignals that maintain normal stem cells in a quiescent state but, when active in CML stem cells, promotes their differentiation in favor of their self-renewal. In more differentiated CML progenitors, the same mechanism has a more potent mitogenic effect which is then extinguished when the cells enter the terminal stages of differentiation. Thus, further expansion of the clone is limited until inevitably additional mutations are acquired that further distort or override the regulatory mechanisms still operative in the chronic phase.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , División Celular , Aberraciones Cromosómicas , Proteínas de Fusión bcr-abl/metabolismo , Hematopoyesis , Humanos , Persona de Mediana Edad , Telómero/fisiología
16.
Science ; 294(5550): 2368-71, 2001 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-11743206

RESUMEN

Sickle cell disease (SCD) is caused by a single point mutation in the human betaA globin gene that results in the formation of an abnormal hemoglobin [HbS (alpha2betaS2)]. We designed a betaA globin gene variant that prevents HbS polymerization and introduced it into a lentiviral vector we optimized for transfer to hematopoietic stem cells and gene expression in the adult red blood cell lineage. Long-term expression (up to 10 months) was achieved, without preselection, in all transplanted mice with erythroid-specific accumulation of the antisickling protein in up to 52% of total hemoglobin and 99% of circulating red blood cells. In two mouse SCD models, Berkeley and SAD, inhibition of red blood cell dehydration and sickling was achieved with correction of hematological parameters, splenomegaly, and prevention of the characteristic urine concentration defect.


Asunto(s)
Anemia de Células Falciformes/terapia , Terapia Genética , Vectores Genéticos , Globinas/genética , VIH-1/genética , Anemia de Células Falciformes/genética , Animales , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Expresión Génica , Globinas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Hemoglobina Falciforme/metabolismo , Humanos , Lentivirus/genética , Región de Control de Posición , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxihemoglobinas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Talasemia/genética , Talasemia/terapia , Transducción Genética , Transgenes , Globinas beta
17.
J Clin Invest ; 108(7): 1071-7, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11581308

RESUMEN

Cells with a verapamil-sensitive ability to efflux Hoechst 33342 (termed side population [SP] cells) have been identified in adult marrow from several species including humans and in several tissues from adult mice. In mice, the SP phenotype appears to be a common feature of stem cells, but human SP cells have been less well characterized. We show here, for the first time to our knowledge, that SP cells are present in the second-trimester human fetal liver. They include all of the transplantable human hematopoietic stem cell activity detectable in NOD/SCID mice and also certain other, more differentiated hematopoietic cell types. Notably, the stem cell activity was confined to the CD34(+)CD38(-) SP(+) population, and isolation of these cells gave an approximately tenfold enrichment of transplantable stem cells. This subset was not, however, coenriched in hematopoietic progenitors detectable by either short- or long-term in vitro assays, indicating most of these to be distinct from transplantable stem cells. These findings suggest that the SP phenotype is an important and distinguishing property of human hematopoietic stem cells and that early in ontogeny they express CD34.


Asunto(s)
Antígenos CD34 , Antígenos CD , Células Madre Hematopoyéticas/inmunología , Hígado/citología , Hígado/embriología , ADP-Ribosil Ciclasa , ADP-Ribosil Ciclasa 1 , Animales , Antígenos de Diferenciación , Biomarcadores , Separación Celular , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/clasificación , Hepatocitos/clasificación , Hepatocitos/citología , Hepatocitos/inmunología , Humanos , Inmunofenotipificación , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos NOD , Ratones SCID , NAD+ Nucleosidasa
18.
Breast Cancer Res Treat ; 67(2): 93-109, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11519870

RESUMEN

The purpose of the present study was to characterize primitive epithelial progenitor populations present in adult normal human mammary tissue using a combination of flow cytometry and in vitro colony assay procedures. Three types of human breast epithelial cell (HBEC) progenitors were identified: luminal-restricted, myoepithelial-restricted and bipotent progenitors. The first type expressed epithelial cell adhesion molecule (EpCAM), alpha6 integrin and MUC1 and generated colonies composed exclusively of cells positive for the luminal-associated markers keratin 8/18, keratin 19, EpCAM and MUC1. Bipotent progenitors produced colonies containing a central core of cells expressing luminal markers surrounded by keratin 14+ myoepithelial-like cells. Single cell cultures confirmed the bipotentiality of these progenitors. Their high expression of alpha6 integrin and low expression of MUC1 suggests a basal position of these cells in the mammary epithelium in vivo. Serial passage in vitro of an enriched population of bipotent progenitors demonstrated that only myoepithelial-restricted progenitors could be readily generated under the culture conditions used. These results support a hierarchical branching model of HBEC progenitor differentiation from a primitive uncommitted cell to luminal- and myoepithelial-restricted progenitors.


Asunto(s)
Mama/citología , Diferenciación Celular , Células Epiteliales/fisiología , Células Madre/fisiología , Antígenos de Neoplasias/biosíntesis , Biomarcadores/análisis , Moléculas de Adhesión Celular/biosíntesis , Técnicas de Cultivo de Célula , Molécula de Adhesión Celular Epitelial , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Integrinas/biosíntesis , Mucina-1/biosíntesis
19.
Ann N Y Acad Sci ; 938: 293-303; discussion 303-4, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11458518

RESUMEN

The telomere length in nucleated peripheral blood (PB) cells indirectly reflects the mitotic history of their precursors: the hematopoietic stem cells (HSCs). The average length of telomeres in PB leukocytes can be measured using fluorescence in situ hybridization and flow cytometry (flow FISH). We previously used flow FISH to characterize the age-related turnover of HSCs in healthy individuals. In this review, we describe results of recent flow FISH studies in patients with selected hematopoietic stem cell-associated disorders: chronic myelogenous leukemia (CML) and several bone marrow failure syndromes. CML is characterized by a marked expansion of myeloid Philadelphia chromosome positive (Ph+) cells. Nevertheless, nonmalignant (Ph-) HSCs typically coexist in the bone marrow of CML patients. We analyzed the telomere length in > 150 peripheral blood leukocytes (PBLs) and bone marrow samples of patients with CML as well as samples of Ph- T-lymphocytes. Compared to normal controls, the overall telomere fluorescence in PBLs of patients with CML was significantly reduced. However, no telomere shortening was observed in Ph- T-lymphocytes. Patients in late chronic phase (CP) had significantly shorter telomeres than those assessed earlier in CP. Our data suggest that progressive telomere shortening is correlated with disease progression in CML. Within the group of patients with bone marrow failure syndromes, we only found significantly shortened telomeres (compared to age-adjusted controls) in granulocytes from patients with aplastic anemia (AA). Strikingly, the telomere length in granulocytes from AA patients who had recovered after immunosuppressive therapy (recAA) did not differ significantly from controls, whereas untreated patients and nonresponders with persistent severe pancytopenia (sAANR) showed marked and significant telomere shortening compared to healthy donors and patients with recAA. Furthermore, an inverse correlation between age-adjusted telomere length and peripheral blood counts was found in support of a model in which the degree of cytopenia and the amount of telomere shortening are correlated. These results support the concept of extensive proliferation of HSCs in subgroups of AA patients and suggest a potential use of telomere-length measurements as a prognostic tool in this group of disorders as well.


Asunto(s)
Anemia Aplásica/patología , Células Sanguíneas/ultraestructura , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/ultraestructura , Telómero/ultraestructura , Anemia Aplásica/sangre , Animales , División Celular , Senescencia Celular , Anemia de Fanconi/sangre , Anemia de Fanconi/patología , Citometría de Flujo , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/patología , Humanos , Hibridación Fluorescente in Situ , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/patología
20.
Blood ; 97(8): 2286-92, 2001 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11290589

RESUMEN

Several studies point to multiple members of the Hox transcription factor family as playing key roles in normal hematopoietic development, and they link the imbalanced expression of these transcription factors, in particular of the Abd-like A cluster HOX genes HOXA9 and HOXA10, to leukemogenesis. To test directly the hypothesis that HOXA10 is involved in human hematopoietic development, the gene was retrovirally overexpressed in human highly purified CD34(+)/GFP(+) hematopoietic progenitor cells derived from cord blood or fetal liver sources, and the impact of aberrant gene expression was analyzed on differentiation and proliferation in vitro and in vivo. HOXA10 misexpression profoundly impaired myeloid differentiation with a higher yield of blast cells in liquid culture and a greater than 100-fold increased generation of blast colonies after in vitro expansion or after replating of primary colonies first plated in methylcellulose directly after transduction (P < .01). Furthermore, aberrant HOXA10 expression almost completely blocked erythroid differentiation in methylcellulose (P < .02). HOXA10 deregulation also severely perturbed the differentiation of human progenitors in vivo, reducing B-cell development by 70% in repopulated NOD/SCID mice and enhancing myelopoiesis in the transduced compartment. The data provide evidence that the balanced expression of HOXA10 is pivotal for normal human hematopoietic development and that aberrant expression of the gene contributes to impaired differentiation and increased proliferation of human hematopoietic progenitor cells. These results also provide a framework to initiate more detailed analyses of HOX regulatory domains and HOX cofactors in the human system in vitro and in vivo.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio , Células Mieloides/citología , Animales , Diferenciación Celular , División Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Células Precursoras Eritroides/citología , Sangre Fetal/citología , Regulación de la Expresión Génica , Genes Homeobox , Vectores Genéticos/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Proteínas Homeobox A10 , Humanos , Hígado/citología , Hígado/embriología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Quimera por Radiación , Proteínas Recombinantes de Fusión/fisiología , Retroviridae/genética , Transfección , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...