Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 137(7): 945-958, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33254233

RESUMEN

Isocitrate dehydrogenase (IDH) mutations are common genetic alterations in myeloid disorders, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Epigenetic changes, including abnormal histone and DNA methylation, have been implicated in the pathogenic build-up of hematopoietic progenitors, but it is still unclear whether and how IDH mutations themselves affect hematopoiesis. Here, we show that IDH1-mutant mice develop myeloid dysplasia in that these animals exhibit anemia, ineffective erythropoiesis, and increased immature progenitors and erythroblasts. In erythroid cells of these mice, D-2-hydroxyglutarate, an aberrant metabolite produced by the mutant IDH1 enzyme, inhibits oxoglutarate dehydrogenase activity and diminishes succinyl-coenzyme A (CoA) production. This succinyl-CoA deficiency attenuates heme biosynthesis in IDH1-mutant hematopoietic cells, thus blocking erythroid differentiation at the late erythroblast stage and the erythroid commitment of hematopoietic stem cells, while the exogenous succinyl-CoA or 5-ALA rescues erythropoiesis in IDH1-mutant erythroid cells. Heme deficiency also impairs heme oxygenase-1 expression, which reduces levels of important heme catabolites such as biliverdin and bilirubin. These deficits result in accumulation of excessive reactive oxygen species that induce the cell death of IDH1-mutant erythroid cells. Our results clearly show the essential role of IDH1 in normal erythropoiesis and describe how its mutation leads to myeloid disorders. These data thus have important implications for the devising of new treatments for IDH-mutant tumors.


Asunto(s)
Eritropoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Hemo/biosíntesis , Isocitrato Deshidrogenasa/genética , Mutación Missense , Mutación Puntual , Preleucemia/genética , Acilcoenzima A/biosíntesis , Acilcoenzima A/deficiencia , Anemia/genética , Animales , Médula Ósea/patología , Eritroblastos/metabolismo , Técnicas de Sustitución del Gen , Glutaratos/metabolismo , Hemo/deficiencia , Hemo-Oxigenasa 1/metabolismo , Isocitrato Deshidrogenasa/fisiología , Complejo Cetoglutarato Deshidrogenasa/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Mielopoyesis/genética , Preleucemia/metabolismo , Preleucemia/patología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Esplenomegalia/etiología , Trombocitopenia/genética
2.
J Intern Med ; 289(5): 636-649, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33511694

RESUMEN

Haematopoietic stem and progenitor cells (HSPCs) are defined as unspecialized cells that give rise to more differentiated cells. In a similar way, leukaemic stem and progenitor cells (LSPCs) are defined as unspecialized leukaemic cells, which can give rise to more differentiated cells. Leukaemic cells carry leukaemic mutations/variants and have clear differentiation abnormalities. Pre-leukaemic HSPCs (PreL-HSPCs) carry pre-leukaemic mutations/variants (pLMs) and are capable of producing mature functional cells, which will carry the same variants. Under the roof of LSPCs, one can find a broad range of cell types genetic and disease phenotypes. Present-day knowledge suggests that this phenotypic heterogeneity is the result of interactions between the cell of origin, the genetic background and the microenvironment background. The combination of these attributes will define the LSPC phenotype, frequency, differentiation capacity and evolutionary trajectory. Importantly, as LSPCs are leukaemia-initiating cells that sustain clinical remission and are the source of relapse, an improved understanding of LSPCs phenotype would offer better clinical opportunities for the treatment and hopefully prevention of human leukaemia. The current review will focus on LSPCs attributes in the context of human haematologic malignancies.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia/patología , Preleucemia/patología , Biomarcadores de Tumor , Médula Ósea/patología , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Variación Genética , Hematopoyesis , Humanos , Leucemia/genética , Leucemia/metabolismo , Mutación , Fenotipo , Preleucemia/genética , Preleucemia/metabolismo , Microambiente Tumoral
3.
Blood ; 124(3): 426-36, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24894773

RESUMEN

Acute myeloid leukemia (AML) results from the activity of driver mutations that deregulate proliferation and survival of hematopoietic stem cells (HSCs). The fusion protein CBFß-SMMHC impairs differentiation in hematopoietic stem and progenitor cells and induces AML in cooperation with other mutations. However, the combined function of CBFß-SMMHC and cooperating mutations in preleukemic expansion is not known. Here, we used Nras(LSL-G12D); Cbfb(56M) knock-in mice to show that allelic expression of oncogenic Nras(G12D) and Cbfß-SMMHC increases survival of preleukemic short-term HSCs and myeloid progenitor cells and maintains the differentiation block induced by the fusion protein. Nras(G12D) and Cbfß-SMMHC synergize to induce leukemia in mice in a cell-autonomous manner, with a shorter median latency and higher leukemia-initiating cell activity than that of mice expressing Cbfß-SMMHC. Furthermore, Nras(LSL-G12D); Cbfb(56M) leukemic cells were sensitive to pharmacologic inhibition of the MEK/ERK signaling pathway, increasing apoptosis and Bim protein levels. These studies demonstrate that Cbfß-SMMHC and Nras(G12D) promote the survival of preleukemic myeloid progenitors primed for leukemia by activation of the MEK/ERK/Bim axis, and define Nras(LSL-G12D); Cbfb(56M) mice as a valuable genetic model for the study of inversion(16) AML-targeted therapies.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Oncogénicas/metabolismo , Preleucemia/metabolismo , Preleucemia/patología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Supervivencia Celular , Técnicas de Sustitución del Gen , Leucemia Experimental/etiología , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/genética , Mutación Missense , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Oncogénicas/genética , Proteínas de Fusión Oncogénica/genética , Preleucemia/genética , Proteínas Proto-Oncogénicas/metabolismo
4.
J Biol Chem ; 288(25): 18219-27, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23673656

RESUMEN

Acute T-cell lymphoblastic leukemia/lymphoma (T-ALL) is an aggressive hematopoietic malignancy affecting both children and adults. Previous studies of T-ALL mouse models induced by different genetic mutations have provided highly diverse results on the issues of T-cell leukemia/lymphoma-initiating cells (T-LICs) and potential mechanisms contributing to T-LIC transformation. Here, we show that oncogenic Kras (Kras G12D) expressed from its endogenous locus is a potent inducer of T-ALL even in a less sensitized BALB/c background. Notch1 mutations, including exon 34 mutations and recently characterized type 1 and 2 deletions, are detected in 100% of Kras G12D-induced T-ALL tumors. Although these mutations are not detected at the pre-leukemia stage, incremental up-regulation of NOTCH1 surface expression is observed at the pre-leukemia and leukemia stages. As secondary genetic hits in the Kras G12D model, Notch1 mutations target CD8(+) T-cells but not hematopoietic stem cells to further promote T-ALL progression. Pre-leukemia T-cells without detectable Notch1 mutations do not induce T-ALL in secondary recipient mice compared with T-ALL tumor cells with Notch1 mutations. We found huge variations in T-LIC frequency and immunophenotypes of cells enriched for T-LICs. Unlike Pten deficiency-induced T-ALL, oncogenic Kras-initiated T-ALL is not associated with up-regulation of the Wnt/ß-catenin pathway. Our results suggest that up-regulation of NOTCH1 signaling, through either overexpression of surface NOTCH1 or acquired gain-of-function mutations, is involved in both T-ALL initiation and progression. Notch1 mutations and Kras G12D contribute cooperatively to leukemogenic transformation of normal T-cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Transformación Celular Neoplásica/genética , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptor Notch1/genética , Adulto , Animales , Trasplante de Médula Ósea , Transformación Celular Neoplásica/metabolismo , Citometría de Flujo , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/cirugía , Preleucemia/genética , Preleucemia/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
5.
Br J Haematol ; 167(5): 587-99, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25155832

RESUMEN

Children with constitutional trisomy 21 (cT21, Down Syndrome, DS) are at a higher risk for both myeloid and B-lymphoid leukaemias. The myeloid leukaemias are often preceded by a transient neonatal pre-leukaemic syndrome, Transient Abnormal Myelopoiesis (TAM). TAM is caused by cooperation between cT21 and acquired somatic N-terminal truncating mutations in the key haematopoietic transcription factor GATA1. These mutations, which are not leukaemogenic in the absence of cT21, are found in almost one-third of neonates with DS. Analysis of primary human fetal liver haematopoietic cells and of human embryonic stem cells demonstrates that cT21 itself substantially alters human fetal haematopoietic development. Consequently, many haematopoietic developmental defects are observed in neonates with DS even in the absence of TAM. Although studies in mouse models have suggested a pathogenic role of deregulated expression of several chromosome 21-encoded genes, their role in human leukaemogenesis remains unclear. As cT21 exists in all embryonic cells, the molecular basis of cT21-associated leukaemias probably reflects a complex interaction between deregulated gene expression in haematopoietic cells and the fetal haematopoietic microenvironment in DS.


Asunto(s)
Síndrome de Down , Células Madre Hematopoyéticas , Leucemia de Células B , Leucemia Mieloide , Mielopoyesis , Preleucemia , Adolescente , Animales , Niño , Preescolar , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/metabolismo , Síndrome de Down/genética , Síndrome de Down/metabolismo , Síndrome de Down/patología , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Regulación Leucémica de la Expresión Génica/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Leucemia de Células B/genética , Leucemia de Células B/metabolismo , Leucemia de Células B/patología , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Preleucemia/genética , Preleucemia/metabolismo , Preleucemia/patología , Nicho de Células Madre/genética
6.
Blood ; 120(4): 709-19, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22337712

RESUMEN

AML1-ETO (AE) is a fusion product of translocation (8;21) that accounts for 40% of M2 type acute myeloid leukemia (AML). In addition to its role in promoting preleukemic hematopoietic cell self-renewal, AE represses DNA repair genes, which leads to DNA damage and increased mutation frequency. Although this latter function may promote leukemogenesis, concurrent p53 activation also leads to an increased baseline apoptotic rate. It is unclear how AE expression is able to counterbalance this intrinsic apoptotic conditioning by p53 to promote survival and self-renewal. In this report, we show that Bcl-xL is up-regulated in AE cells and plays an essential role in their survival and self-renewal. Further investigation revealed that Bcl-xL expression is regulated by thrombopoietin (THPO)/MPL-signaling induced by AE expression. THPO/MPL-signaling also controls cell cycle reentry and mediates AE-induced self-renewal. Analysis of primary AML patient samples revealed a correlation between MPL and Bcl-xL expression specifically in t(8;21) blasts. Taken together, we propose that survival signaling through Bcl-xL is a critical and intrinsic component of a broader self-renewal signaling pathway downstream of AML1-ETO-induced MPL.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Preleucemia/metabolismo , Preleucemia/patología , Receptores de Trombopoyetina/metabolismo , Trombopoyetina/metabolismo , Proteína bcl-X/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Sangre Fetal/citología , Sangre Fetal/metabolismo , Feto/citología , Feto/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Hígado/citología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Fusión Oncogénica/genética , ARN Mensajero/genética , Proteína 1 Compañera de Translocación de RUNX1 , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Trombopoyetina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Trombopoyetina/genética , Proteína bcl-X/genética
7.
Cancer Cell ; 9(1): 57-68, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16413472

RESUMEN

The acute myeloid leukemia (AML)-associated CBF beta-SMMHC fusion protein impairs hematopoietic differentiation and predisposes to leukemic transformation. The mechanism of leukemia progression, however, is poorly understood. In this study, we report a conditional Cbfb-MYH11 knockin mouse model that develops AML with a median latency of 5 months. Cbf beta-SMMHC expression reduced the multilineage repopulation capacity of hematopoietic stem cells (HSCs) while maintaining their numbers under competitive conditions. The fusion protein induced abnormal myeloid progenitors (AMPs) with limited proliferative potential but leukemic predisposition similar to that of HSCs in transplanted mice. In addition, Cbf beta-SMMHC blocked megakaryocytic maturation at the CFU-Meg to megakaryocyte transition. These data show that a leukemia oncoprotein can inhibit differentiation and proliferation while not affecting the maintenance of long-term HSCs.


Asunto(s)
Leucemia Mieloide/patología , Células Progenitoras Mieloides/patología , Proteínas de Fusión Oncogénica/metabolismo , Preleucemia/patología , Enfermedad Aguda , Animales , Linfocitos B/patología , Plaquetas/patología , Proliferación Celular , Hematopoyesis , Leucemia Mieloide/metabolismo , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células Progenitoras Mieloides/metabolismo , Proteínas de Fusión Oncogénica/genética , Preleucemia/metabolismo
8.
Ann Hematol ; 91(8): 1221-33, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22488406

RESUMEN

Overexpression of MN1, ERG, BAALC, and EVI1 (MEBE) genes in cytogenetically normal acute myeloid leukemia (AML) patients is associated with poor prognosis, but their prognostic effect in patients with myelodysplastic syndromes (MDS) has not been studied systematically. Expression data of the four genes from 140 MDS patients were combined in an additive score, which was validated in an independent patient cohort of 110 MDS patients. A high MEBE score, defined as high expression of at least two of the four genes, predicted a significantly shorter overall survival (OS) (HR 2.29, 95 % CI 1.3-4.09, P= .005) and time to AML progression (HR 4.83, 95 % CI 2.01-11.57, P< .001) compared to a low MEBE score in multivariate analysis independent of karyotype, percentage of bone marrow blasts, transfusion dependence, ASXL1, and IDH1 mutation status. In a validation cohort of 110 MDS patients, a high MEBE score predicted shorter OS (HR 1.77; 95 % CI 1.04-3.0, P= .034) and time to AML progression (HR 3.0, 95 % CI 1.17-7.65, P= .022). A high MEBE expression score is an unfavorable prognostic marker in MDS and is associated with an increased risk for progression to AML. Expression of the MEBE genes is regulated by FLI1 and c-MYC, which are potential upstream targets of the MEBE signature.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndromes Mielodisplásicos/diagnóstico , Proteínas de Neoplasias/genética , Proto-Oncogenes/genética , Transactivadores/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Estudios de Cohortes , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteína del Locus del Complejo MDS1 y EV11 , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/mortalidad , Proteínas de Neoplasias/metabolismo , Valor Predictivo de las Pruebas , Preleucemia/diagnóstico , Preleucemia/genética , Preleucemia/metabolismo , Pronóstico , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Regulador Transcripcional ERG , Proteínas Supresoras de Tumor/metabolismo , Estudios de Validación como Asunto
9.
Science ; 373(6551)2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244384

RESUMEN

Children with Down syndrome have a 150-fold increased risk of developing myeloid leukemia, but the mechanism of predisposition is unclear. Because Down syndrome leukemogenesis initiates during fetal development, we characterized the cellular and developmental context of preleukemic initiation and leukemic progression using gene editing in human disomic and trisomic fetal hematopoietic cells and xenotransplantation. GATA binding protein 1 (GATA1) mutations caused transient preleukemia when introduced into trisomy 21 long-term hematopoietic stem cells, where a subset of chromosome 21 microRNAs affected predisposition to preleukemia. By contrast, progression to leukemia was independent of trisomy 21 and originated in various stem and progenitor cells through additional mutations in cohesin genes. CD117+/KIT proto-oncogene (KIT) cells mediated the propagation of preleukemia and leukemia, and KIT inhibition targeted preleukemic stem cells.


Asunto(s)
Proteínas de Ciclo Celular/genética , Síndrome de Down/genética , Factor de Transcripción GATA1/genética , Células Madre Hematopoyéticas/fisiología , Leucemia Mieloide/genética , Preleucemia/genética , Animales , Antígenos CD34/análisis , Proteínas de Ciclo Celular/metabolismo , Linaje de la Célula , Proliferación Celular , Transformación Celular Neoplásica , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Síndrome de Down/complicaciones , Femenino , Factor de Transcripción GATA1/metabolismo , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Hígado/embriología , Masculino , Megacariocitos/fisiología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Mutación , Preleucemia/metabolismo , Preleucemia/patología , Inhibidores de Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-kit/análisis , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Cohesinas
10.
Proc Natl Acad Sci U S A ; 104(50): 20013-8, 2007 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-18056627

RESUMEN

Murine granulocytic cells, in becoming leukemic, need to acquire enhanced self-generation and a capacity for autocrine growth stimulation. Mice transplanted with bone marrow cells transduced with the Mixl1 homeobox gene develop a very high frequency of myeloid leukemia derived from the transduced cells. Preleukemic mice contained a high frequency of transduced clonogenic granulocytic cells. They exhibited an abnormally high capacity for self-replication and could generate immortalized granulocytic cell lines that remained absolutely dependent on either GM-CSF or IL-3 and were not leukemic. Organs from mice repopulated by marrow cells transduced either with Mixl1 or the control murine stem cell virus vector exhibited a capacity to produce IL-3 in vitro, activity being highest with the lungs, marrow, bladder, and thymus. Supporting evidence for the in vivo production of IL-3 was the frequent development of mast cells in the marrow. Overexpression of Mixl1 appears capable of inducing an abnormal self-renewal capacity in granulocytic precursors. Aberrant production of IL-3 was not present in the continuous Mixl cell lines and was therefore not in itself likely to be a leukemogenic change but it could support the enhanced survival and proliferation of the Mixl1 granulocytic populations until a final leukemogenic mutation occurs in them.


Asunto(s)
Médula Ósea/metabolismo , Proteínas de Homeodominio/metabolismo , Preleucemia/metabolismo , Preleucemia/patología , Animales , Trasplante de Médula Ósea , Diferenciación Celular , Células Cultivadas , Proteínas de Homeodominio/genética , Interleucina-3/biosíntesis , Mastocitos/citología , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Preleucemia/genética
11.
J Clin Invest ; 116(4): 974-83, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16585963

RESUMEN

Human T cell leukemia virus type 1 (HTLV-1) infects both CD4+ and CD8+ lymphocytes, yet it induces adult T cell leukemia/lymphoma (ATLL) that is regularly of the CD4+ phenotype. Here we show that in vivo infected CD4+ and CD8+ T cells displayed similar patterns of clonal expansion in carriers without malignancy. Cloned infected cells from individuals without malignancy had a dramatic increase in spontaneous proliferation, which predominated in CD8+ lymphocytes and depended on the amount of tax mRNA. In fact, the clonal expansion of HTLV-1-positive CD8+ and CD4+ lymphocytes relied on 2 distinct mechanisms--infection prevented cell death in the former while recruiting the latter into the cell cycle. Cell cycling, but not apoptosis, depended on the level of viral-encoded tax expression. Infected tax-expressing CD4+ lymphocytes accumulated cellular defects characteristic of genetic instability. Therefore, HTLV-1 infection establishes a preleukemic phenotype that is restricted to CD4+ infected clones.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Adulto , Apoptosis , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Portador Sano/virología , Ciclo Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Preleucemia/metabolismo , Preleucemia/virología , Factores de Tiempo
12.
Oncogene ; 25(21): 3023-31, 2006 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-16407836

RESUMEN

Analysis of the INK4A/ARF locus in human T-ALL patients revealed frequent deletions in exon 2, the exon common to both p16(INK4A) and p14(ARF). Other studies have described selective deletion of exon 1beta of p14(ARF) or methylation of the p16(INK4A) promoter. Therefore, it is unclear from these studies whether loss of p16(INK4A) and/or p14(ARF) contributes to the development of T-ALL. To elucidate the relative contribution of the ink4a/arf locus to T-cell leukemogenesis, we mated our tal1 transgenic mice to ink4a/arf-/-, p16(ink4a)-/-, and p19(arf)-/- mice and generated tal1/ink4a/arf+/-, tal1/p16(ink4a)+/-, and tal1/p19(arf)+/- mice. Each of these mice developed T-cell leukemia rapidly, indicating that loss of either p16(ink4a) or p19(arf) cooperates with Tal1 to induce leukemia in mice. Preleukemic studies reveal that Tal1 expression stimulates entry into the cell cycle and thymocyte apoptosis in vivo. Interestingly, mice expressing a DNA-binding mutant of Tal1 do not exhibit increases in S phase cells. The S phase induction is accompanied by an increase in thymocyte apoptosis in tal1 transgenic mice. Whereas apoptosis is reduced to wild-type levels in tal1/ink4a/arf-/- mice, S phase induction remains unaffected. Thus, Tal1 stimulates cell cycle entry independent of the ink4a/arf locus, but its ability to induce apoptosis is Ink4a/Arf-dependent.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Transformación Celular Neoplásica/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Eliminación de Gen , Leucemia-Linfoma de Células T del Adulto/genética , Proteínas Proto-Oncogénicas/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Animales , Antígenos CD/análisis , Antígenos de Neoplasias/análisis , Apoptosis/genética , Ciclo Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , ADN/metabolismo , Exones/genética , Regulación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Genes p16 , Glicoproteínas/fisiología , Inmunofenotipificación , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Preleucemia/genética , Preleucemia/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptor Notch1/fisiología , Fase S/genética , Proteína 1 de la Leucemia Linfocítica T Aguda , Subgrupos de Linfocitos T/patología , Factores de Transcripción/fisiología , Proteína p14ARF Supresora de Tumor/deficiencia , Proteína p14ARF Supresora de Tumor/genética , Proteínas Wnt/fisiología
13.
Leukemia ; 31(2): 423-433, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27568523

RESUMEN

Fanconi anemia (FA) is an inherited bone marrow failure syndrome with extremely high risk of leukemic transformation. Here we investigate the relationship between DNA damage response (DDR) and leukemogenesis using the Fanca knockout mouse model. We found that chronic exposure of the Fanca-/- hematopoietic stem cells to DNA crosslinking agent mitomycin C in vivo leads to diminished DDR, and the emergence/expansion of pre-leukemia stem cells (pre-LSCs). Surprisingly, although genetic correction of Fanca deficiency in the pre-LSCs restores DDR and reduces genomic instability, but fails to prevent pre-LSC expansion or delay leukemia development in irradiated recipients. Furthermore, we identified transcription program underlying dysregulated DDR and cell migration, myeloid proliferation, and immune response in the Fanca-/- pre-LSCs. Forced expression of the downregulated DNA repair genes, Rad51c or Trp53i13, in the Fanca-/- pre-LSCs partially rescues DDR but has no effect on leukemia, whereas shRNA knockdown of the upregulated immune receptor genes Trem1 or Pilrb improves leukemia-related survival, but not DDR or genomic instability. Furthermore, Trem1 cooperates with diminished DDR in vivo to promote Fanca-/- pre-LSC expansion and leukemia development. Our study implicates diminishing DDR as a root cause of FA leukemogenesis, which subsequently collaborates with other signaling pathways for leukemogenic transformation.


Asunto(s)
Daño del ADN , Glicoproteínas de Membrana/metabolismo , Células Madre Neoplásicas/metabolismo , Preleucemia/genética , Preleucemia/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Biomarcadores , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Expresión Génica , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Inestabilidad Genómica , Genotipo , Inmunomodulación/genética , Masculino , Ratones , Ratones Noqueados , Células Madre Neoplásicas/patología , Preleucemia/mortalidad , Preleucemia/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Activador Expresado en Células Mieloides 1
14.
Leukemia ; 30(3): 728-39, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26449661

RESUMEN

The t(8;21) rearrangement, which creates the AML1-ETO fusion protein, represents the most common chromosomal translocation in acute myeloid leukemia (AML). Clinical data suggest that CBL mutations are a frequent event in t(8;21) AML, but the role of CBL in AML1-ETO-induced leukemia has not been investigated. In this study, we demonstrate that CBL mutations collaborate with AML1-ETO to expand human CD34+ cells both in vitro and in a xenograft model. CBL depletion by shRNA also promotes the growth of AML1-ETO cells, demonstrating the inhibitory function of endogenous CBL in t(8;21) AML. Mechanistically, loss of CBL function confers hyper-responsiveness to thrombopoietin and enhances STAT5/AKT/ERK/Src signaling in AML1-ETO cells. Interestingly, we found the protein tyrosine phosphatase UBASH3B/Sts-1, which is known to inhibit CBL function, is upregulated by AML1-ETO through transcriptional and miR-9-mediated regulation. UBASH3B/Sts-1 depletion induces an aberrant pattern of CBL phosphorylation and impairs proliferation in AML1-ETO cells. The growth inhibition caused by UBASH3B/Sts-1 depletion can be rescued by ectopic expression of CBL mutants, suggesting that UBASH3B/Sts-1 supports the growth of AML1-ETO cells partly through modulation of CBL function. Our study reveals a role of CBL in restricting myeloid proliferation of human AML1-ETO-induced leukemia, and identifies UBASH3B/Sts-1 as a potential target for pharmaceutical intervention.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/genética , Preleucemia/genética , Proteínas Tirosina Fosfatasas/genética , Proteínas Proto-Oncogénicas c-cbl/genética , Animales , Proliferación Celular , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Sangre Fetal/metabolismo , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones SCID , MicroARNs/genética , MicroARNs/metabolismo , Células Mieloides/citología , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Preleucemia/metabolismo , Preleucemia/patología , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-cbl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-cbl/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Trombopoyetina/farmacología , Transgenes , Translocación Genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
15.
Nat Med ; 21(10): 1172-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26343801

RESUMEN

Modest transcriptional changes caused by genetic or epigenetic mechanisms are frequent in human cancer. Although loss or near-complete loss of the hematopoietic transcription factor PU.1 induces acute myeloid leukemia (AML) in mice, a similar degree of PU.1 impairment is exceedingly rare in human AML; yet, moderate PU.1 inhibition is common in AML patients. We assessed functional consequences of modest reductions in PU.1 expression on leukemia development in mice harboring DNA lesions resembling those acquired during human stem cell aging. Heterozygous deletion of an enhancer of PU.1, which resulted in a 35% reduction of PU.1 expression, was sufficient to induce myeloid-biased preleukemic stem cells and their subsequent transformation to AML in a DNA mismatch repair-deficient background. AML progression was mediated by inhibition of expression of a PU.1-cooperating transcription factor, Irf8. Notably, we found marked molecular similarities between the disease in these mice and human myelodysplastic syndrome and AML. This study demonstrates that minimal reduction of a key lineage-specific transcription factor, which commonly occurs in human disease, is sufficient to initiate cancer development, and it provides mechanistic insight into the formation and progression of preleukemic stem cells in AML.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Preleucemia/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Progresión de la Enfermedad , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Preleucemia/patología , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética
16.
Free Radic Biol Med ; 33(1): 76-82, 2002 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12086685

RESUMEN

The evolution of mitochondrial oxidative phosphorylation was studied during cancer induction in a model of thymic radiolymphomagenesis in C57BL/Ka mice. During the preneoplastic period, thymuses displayed an increase of the cytochrome c oxidase activity and oxygen consumption together with oxidative DNA damage assessed by the presence of the 8-hydroxydeoxyguanine DNA base modification. These transient changes in mitochondrial functional activity were not observed in thymuses of mice rescued from lymphoma development by a bone marrow graft, suggesting an important role of mitochondria for neoplastic transformation in this model, which might therefore be of interest to test the utilization of antioxidants for the prevention of radiation-induced malignancies.


Asunto(s)
Desoxiguanosina/análogos & derivados , Leucemia Inducida por Radiación/metabolismo , Linfoma/metabolismo , Neoplasias del Timo/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Trasplante de Médula Ósea , Respiración de la Célula , Transformación Celular Neoplásica , Desoxiguanosina/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Citometría de Flujo , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Estrés Oxidativo/fisiología , Consumo de Oxígeno , Preleucemia/metabolismo , Timo/efectos de la radiación , Regulación hacia Arriba , Irradiación Corporal Total
17.
Semin Oncol ; 12(2 Suppl 3): 200-7, 1985 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3925558

RESUMEN

Although the mechanism of action responsible for the effects of low-dose ara-C remains unclear, certain insights are available concerning the interaction of this agent with DNA. Ara-C incorporates into DNA, and the extent of (ara-C)DNA formation correlates significantly with loss of clonogenic survival. The inhibition of DNA replication by ara-C also results in DNA fragmentation and terminal differentiation of leukemic cells. Other studies have demonstrated that inhibition of DNA replication by ara-C results in an aberrant form of DNA synthesis with certain segments of DNA being replicated more than once within a single cell cycle. The additional copies of certain segments of DNA could lead to the accumulation of DNA fragments and alterations in gene expression. It is of interest that other inhibitors of S-phase DNA replication such as aphidicolin and hydroxyurea can also induce similar phenotypic changes in HL-60 and K562 leukemia cells. Although the in vitro data support the concept that ara-C is capable of inducing leukemic cell differentiation, there is no evidence to suggest that this agent induces differentiation of human leukemic cells in vivo. Drug levels achieved by administration of low-dose ara-C (42-64 nmol/L) result in the incorporation of ara-C into bone marrow mononuclear preparations from patients with preleukemia syndromes. This concentration of ara-C (5 X 10(-8) mol/L) slows DNA replication of human leukemic cells in vitro. Thus, the clinical use of low doses of ara-C that achieve plasma concentrations of 10(-8) to 10(-7) mol/L could theoretically induce maturational effects by partially inhibiting DNA synthesis. At the present time we have no available data to support this contention. On the basis of chromosomal analyses, low-dose ara-C apparently maintains sufficient drug levels to suppress more "malignant" clones, but even "clonal" selection may represent elimination of leukemic cells by either cytotoxicity or induction of terminal differentiation. Further studies will be necessary to define the mechanism of action of low-dose ara-C in preleukemia.


Asunto(s)
Citarabina/uso terapéutico , Leucemia/tratamiento farmacológico , Adulto , Anciano , Afidicolina , Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Citarabina/administración & dosificación , Citarabina/efectos adversos , Citarabina/metabolismo , Citarabina/farmacología , ADN/biosíntesis , Replicación del ADN/efectos de los fármacos , Diterpenos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Hemoglobinas/biosíntesis , Humanos , Cariotipificación , Cinética , Leucemia/patología , Masculino , Persona de Mediana Edad , Preleucemia/tratamiento farmacológico , Preleucemia/genética , Preleucemia/metabolismo , Preleucemia/patología
18.
Leuk Res ; 11(8): 731-7, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3498097

RESUMEN

The authors present a patient with the typical clinical picture of an acquired amegakaryocytic thrombocytopenic purpura. After 16 months of observation, the patient developed acute myelomonocytic leukemia. During the preleukemic phase and after progression to overt leukemia, serial in-vitro analyses of megakaryocytic, granulocytic, erythrocytic and T-lymphocytic colony growth were carried out in a microagar culture system. At presentation, a marked diminution of CFU-M was observed, whereas CFU-E, BFU-E, CFU-C and CFU-TL were in the normal range. The CFU-M number remained at its low level during the whole observation period. The CFU-C number declined steadily during the preleukemic period, while BFU-E, CFU-E and CFU-TL remained constant until January 1985 when the patient developed AML. After progression to overt leukemia, a distinct reduction became evident in all colony-forming cells. Cytogenetic studies performed during the preleukemic phase indicated the presence of a 5q- chromosome. The authors submit evidence here that the patient was not only characterized by defective megakaryocytic colony formation but also by a deficiency of functional megakaryocyte colony-stimulating activity. No humoral or cellular inhibitors of CFU-M colony formation were found. It is concluded that in preleukemia with a 5q- chromosome the megakaryocytic cell lineage may be involved in the process that precedes overt leukemia at an earlier time than cells of granulocytic and erythrocytic lineages. In addition, it is shown here that megakaryocytopoiesis during the preleukemic period can be characterized by two different defects: first, an intrinsic megakaryocytic stem cell defect and, second, a deficiency of functional megakaryocytic colony-stimulating activity.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 5 , Células Madre Hematopoyéticas/patología , Megacariocitos/patología , Preleucemia/diagnóstico , Púrpura Trombocitopénica Trombótica/diagnóstico , Adulto , Antígenos de Superficie/análisis , Médula Ósea/inmunología , Factores Estimulantes de Colonias/metabolismo , Diagnóstico Diferencial , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Megacariocitos/metabolismo , Preleucemia/genética , Preleucemia/metabolismo , Síndrome , Linfocitos T/patología
19.
Leuk Res ; 19(7): 457-61, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7637391

RESUMEN

The human pre-B acute lymphoblastic leukemia cell line REH6 was used to analyze the regulation of a ubiquitous intracellular phosphoprotein stathmin (Mr 19,000, pl = 5.6-6.2). We demonstrated by 32P-labeling that the short (1 h) treatment of the REH6 cells with the tumor promoting phorbol ester, 12-O-tetradecanoylphorbol-13-acetate (TPA), resulted in a rapid phosphorylation of at least three (P1, P2 and P3) stathmin isoforms without an alteration of stathmin isoform expression. Furthermore, Western blot analysis with specific antiserum showed that the prolonged period (48 h) of TPA treatment partially reduced protein levels particularly of two (N2 and P2) stathmin isoforms. The potent and relatively specific protein kinase C (PKC) inhibitor, 1,(5-isoquinolinesulphonyl)2methylpiperasine dihydrochloride (H7), partially inhibited these TPA effects, whereas the specific calmodulin inhibitor R24571 (calmidazolium) had no effect upon these events. Our findings suggest that stathmin phosphorylation in REH6 cells could be in part mediated by PKC activation.


Asunto(s)
Linfoma de Burkitt/patología , Isoquinolinas/farmacología , Proteínas de Microtúbulos , Fosfoproteínas/metabolismo , Piperazinas/farmacología , Preleucemia/patología , Proteína Quinasa C/antagonistas & inhibidores , Acetato de Tetradecanoilforbol/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina , Linfoma de Burkitt/metabolismo , Calmodulina/antagonistas & inhibidores , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Humanos , Imidazoles/farmacología , Fosforilación , Preleucemia/metabolismo , Estatmina , Células Tumorales Cultivadas
20.
Acta Med Okayama ; 42(1): 15-9, 1988 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2834918

RESUMEN

Superoxide anion (O2-) production by neutrophils from 14 untreated patients with acute nonlymphocytic leukemia (ANLL) was significantly less than that of healthy controls (4.93 +/- 1.99 vx 6.20 +/- 1.53 nmol/min/10(6) neutrophils, p less than 0.05). In 10 patients with myelodysplastic syndrome (MDS), however, it was not significantly different from the control level although 6 of the 10 patients had low levels, when individual patients were compared with the lower limit of the control range. An inverse correlation between the O2- production of neutrophils and the percentage of leukemic cells in the marrow existed in ANLL (r = -0.55, p less than 0.01), but not in MDS. Three of 4 MDS patients who died of pneumonia prior to leukemic conversion showed a low level of O2- production. The impaired O2- production by neutrophils from some MDS patients, probably due to the faulty differentiation from leukemic clones, may be one of the causes of enhanced susceptibility to infection.


Asunto(s)
Síndromes Mielodisplásicos/metabolismo , Neutrófilos/metabolismo , Preleucemia/metabolismo , Superóxidos/metabolismo , Anciano , Femenino , Humanos , Leucemia/metabolismo , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA