Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Science ; 373(6551)2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34244384

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/genética , Síndrome de Down/genética , Fator de Transcrição GATA1/genética , Células-Tronco Hematopoéticas/fisiologia , Leucemia Mieloide/genética , Pré-Leucemia/genética , Animais , Antígenos CD34/análise , Proteínas de Ciclo Celular/metabolismo , Linhagem da Célula , Proliferação de Células , Transformação Celular Neoplásica , Proteínas Cromossômicas não Histona/genética , Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 21/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Síndrome de Down/complicações , Feminino , Fator de Transcrição GATA1/metabolismo , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Xenoenxertos , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Fígado/embriologia , Masculino , Megacariócitos/fisiologia , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Mutação , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Inibidores de Proteínas Quinases/farmacologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-kit/análise , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Coesinas
2.
J Intern Med ; 289(5): 636-649, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33511694

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/patologia , Leucemia/patologia , Pré-Leucemia/patologia , Biomarcadores Tumorais , Medula Óssea/patologia , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Variação Genética , Hematopoese , Humanos , Leucemia/genética , Leucemia/metabolismo , Mutação , Fenótipo , Pré-Leucemia/genética , Pré-Leucemia/metabolismo , Microambiente Tumoral
3.
Blood ; 137(7): 945-958, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33254233

RESUMO

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.


Assuntos
Eritropoese/genética , Células-Tronco Hematopoéticas/metabolismo , Heme/biossíntese , Isocitrato Desidrogenase/genética , Mutação de Sentido Incorreto , Mutação Puntual , Pré-Leucemia/genética , Acil Coenzima A/biossíntese , Acil Coenzima A/deficiência , Anemia/genética , Animais , Medula Óssea/patologia , Eritroblastos/metabolismo , Técnicas de Introdução de Genes , Glutaratos/metabolismo , Heme/deficiência , Heme Oxigenase-1/metabolismo , Isocitrato Desidrogenase/fisiologia , Complexo Cetoglutarato Desidrogenase/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/patologia , Mielopoese/genética , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/metabolismo , Esplenomegalia/etiologia , Trombocitopenia/genética
4.
Leukemia ; 31(2): 423-433, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27568523

RESUMO

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.


Assuntos
Dano ao DNA , Glicoproteínas de Membrana/metabolismo , Células-Tronco Neoplásicas/metabolismo , Pré-Leucemia/genética , Pré-Leucemia/metabolismo , Receptores Imunológicos/metabolismo , Animais , Biomarcadores , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Análise por Conglomerados , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patologia , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Expressão Gênica , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Genótipo , Imunomodulação/genética , Masculino , Camundongos , Camundongos Knockout , Células-Tronco Neoplásicas/patologia , Pré-Leucemia/mortalidade , Pré-Leucemia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides
5.
Leukemia ; 30(3): 728-39, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26449661

RESUMO

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.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , Proteínas de Fusão Oncogênica/genética , Pré-Leucemia/genética , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas c-cbl/genética , Animais , Proliferação de Células , Cromossomos Humanos Par 21 , Cromossomos Humanos Par 8 , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sangue Fetal/citologia , Sangue Fetal/efeitos dos fármacos , Sangue Fetal/metabolismo , Xenoenxertos , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos SCID , MicroRNAs/genética , MicroRNAs/metabolismo , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-cbl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-cbl/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína 1 Parceira de Translocação de RUNX1 , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Trombopoetina/farmacologia , Transgenes , Translocação Genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
6.
Nat Med ; 21(10): 1172-81, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26343801

RESUMO

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.


Assuntos
Leucemia Mieloide Aguda/metabolismo , Pré-Leucemia/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Progressão da Doença , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Pré-Leucemia/patologia , Proteínas Proto-Oncogênicas/genética , Transativadores/genética
7.
Br J Haematol ; 167(5): 587-99, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25155832

RESUMO

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.


Assuntos
Síndrome de Down , Células-Tronco Hematopoéticas , Leucemia de Células B , Leucemia Mieloide , Mielopoese , Pré-Leucemia , Adolescente , Animais , Criança , Pré-Escolar , Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 21/metabolismo , Síndrome de Down/genética , Síndrome de Down/metabolismo , Síndrome de Down/patologia , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Regulação Leucêmica da Expressão Gênica/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia de Células B/genética , Leucemia de Células B/metabolismo , Leucemia de Células B/patologia , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Pré-Leucemia/genética , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Nicho de Células-Tronco/genética
8.
Blood ; 124(3): 426-36, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24894773

RESUMO

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.


Assuntos
Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Oncogênicas/metabolismo , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Sobrevivência Celular , Técnicas de Introdução de Genes , Leucemia Experimental/etiologia , Leucemia Experimental/metabolismo , Leucemia Experimental/patologia , Leucemia Mieloide Aguda/etiologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Monoméricas de Ligação ao GTP/genética , Mutação de Sentido Incorreto , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Oncogênicas/genética , Proteínas de Fusão Oncogênica/genética , Pré-Leucemia/genética , Proteínas Proto-Oncogênicas/metabolismo
10.
J Biol Chem ; 288(25): 18219-27, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23673656

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Transformação Celular Neoplásica/genética , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptor Notch1/genética , Adulto , Animais , Transplante de Medula Óssea , Transformação Celular Neoplásica/metabolismo , Citometria de Fluxo , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos 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/cirurgia , Pré-Leucemia/genética , Pré-Leucemia/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
11.
Ann Hematol ; 91(8): 1221-33, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22488406

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Síndromes Mielodisplásicas/diagnóstico , Proteínas de Neoplasias/genética , Proto-Oncogenes/genética , Transativadores/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Estudos de Coortes , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteína do Locus do Complexo MDS1 e EVI1 , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/mortalidade , Proteínas de Neoplasias/metabolismo , Valor Preditivo dos Testes , Pré-Leucemia/diagnóstico , Pré-Leucemia/genética , Pré-Leucemia/metabolismo , Prognóstico , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Regulador Transcricional ERG , Proteínas Supressoras de Tumor/metabolismo , Estudos de Validação como Assunto
12.
Blood ; 120(4): 709-19, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22337712

RESUMO

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.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Leucemia Mieloide Aguda/patologia , Proteínas de Fusão Oncogênica/metabolismo , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Receptores de Trombopoetina/metabolismo , Trombopoetina/metabolismo , Proteína bcl-X/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Feto/citologia , Feto/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Fusão Oncogênica/genética , RNA Mensageiro/genética , Proteína 1 Parceira de Translocação de RUNX1 , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Trombopoetina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Trombopoetina/genética , Proteína bcl-X/genética
14.
Science ; 333(6043): 765-9, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21764752

RESUMO

The chromosomal translocations found in acute myelogenous leukemia (AML) generate oncogenic fusion transcription factors with aberrant transcriptional regulatory properties. Although therapeutic targeting of most leukemia fusion proteins remains elusive, the posttranslational modifications that control their function could be targetable. We found that AML1-ETO, the fusion protein generated by the t(8;21) translocation, is acetylated by the transcriptional coactivator p300 in leukemia cells isolated from t(8;21) AML patients, and that this acetylation is essential for its self-renewal-promoting effects in human cord blood CD34(+) cells and its leukemogenicity in mouse models. Inhibition of p300 abrogates the acetylation of AML1-ETO and impairs its ability to promote leukemic transformation. Thus, lysine acetyltransferases represent a potential therapeutic target in AML.


Assuntos
Transformação Celular Neoplásica , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteína p300 Associada a E1A/metabolismo , Células-Tronco Hematopoéticas/citologia , Leucemia Mieloide Aguda/metabolismo , Lisina/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Acetilação , Animais , Linhagem Celular , Linhagem Celular Tumoral , Subunidade alfa 2 de Fator de Ligação ao Core/química , Proteína p300 Associada a E1A/antagonistas & inibidores , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Proteínas de Fusão Oncogênica/química , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Proteína 1 Parceira de Translocação de RUNX1 , Ativação Transcricional , Células Tumorais Cultivadas
15.
Science ; 327(5967): 879-83, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20093438

RESUMO

The LMO2 oncogene causes a subset of human T cell acute lymphoblastic leukemias (T-ALL), including four cases that arose as adverse events in gene therapy trials. To investigate the cellular origin of LMO2-induced leukemia, we used cell fate mapping to study mice in which the Lmo2 gene was constitutively expressed in the thymus. Lmo2 induced self-renewal of committed T cells in the mice more than 8 months before the development of overt T-ALL. These self-renewing cells retained the capacity for T cell differentiation but expressed several genes typical of hematopoietic stem cells (HSCs), suggesting that Lmo2 might reactivate an HSC-specific transcriptional program. Forced expression of one such gene, Hhex, was sufficient to initiate self-renewal of thymocytes in vivo. Thus, Lmo2 promotes the self-renewal of preleukemic thymocytes, providing a mechanism by which committed T cells can then accumulate additional genetic mutations required for leukemic transformation.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/genética , Metaloproteínas/genética , Oncogenes , Células Precursoras de Linfócitos T/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Linfócitos T/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Proteínas com Domínio LIM , Metaloproteínas/metabolismo , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Células Precursoras de Linfócitos T/transplante , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Pré-Leucemia/genética , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Proteínas Proto-Oncogênicas , Subpopulações de Linfócitos T , Linfócitos T/transplante , Timo/metabolismo , Timo/patologia , Fatores de Transcrição/genética , Transcrição Gênica , Regulação para Cima
16.
Science ; 319(5861): 336-9, 2008 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-18202291

RESUMO

Understanding cancer pathogenesis requires knowledge of not only the specific contributory genetic mutations but also the cellular framework in which they arise and function. Here we explore the clonal evolution of a form of childhood precursor-B cell acute lymphoblastic leukemia that is characterized by a chromosomal translocation generating a TEL-AML1 fusion gene. We identify a cell compartment in leukemic children that can propagate leukemia when transplanted in mice. By studying a monochorionic twin pair, one preleukemic and one with frank leukemia, we establish the lineal relationship between these "cancer-propagating" cells and the preleukemic cell in which the TEL-AML1 fusion first arises or has functional impact. Analysis of TEL-AML1-transduced cord blood cells suggests that TEL-AML1 functions as a first-hit mutation by endowing this preleukemic cell with altered self-renewal and survival properties.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Doenças em Gêmeos , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Pré-Leucemia/patologia , ADP-Ribosil Ciclase 1/análise , Doença Aguda , Animais , Antígenos CD19/análise , Antígenos CD34/análise , Apoptose , Transplante de Medula Óssea , Pré-Escolar , Subunidade alfa 2 de Fator de Ligação ao Core/análise , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Doenças em Gêmeos/genética , Doenças em Gêmeos/metabolismo , Doenças em Gêmeos/patologia , Feminino , Sangue Fetal/transplante , Rearranjo Gênico de Cadeia Pesada de Linfócito B , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Células-Tronco Neoplásicas/patologia , Proteínas de Fusão Oncogênica/análise , Proteínas de Fusão Oncogênica/fisiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Células Precursoras de Linfócitos B/química , Células Precursoras de Linfócitos B/fisiologia , Pré-Leucemia/genética , Pré-Leucemia/metabolismo , Recombinação Genética , Transplante Heterólogo , Gêmeos Monozigóticos
17.
Proc Natl Acad Sci U S A ; 104(50): 20013-8, 2007 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-18056627

RESUMO

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.


Assuntos
Medula Óssea/metabolismo , Proteínas de Homeodomínio/metabolismo , Pré-Leucemia/metabolismo , Pré-Leucemia/patologia , Animais , Transplante de Medula Óssea , Diferenciação Celular , Células Cultivadas , Proteínas de Homeodomínio/genética , Interleucina-3/biossíntese , Mastócitos/citologia , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pré-Leucemia/genética
18.
J Virol ; 81(8): 4374-7, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17287277

RESUMO

Infection of thymic lymphocytes by a mink cell focus-forming murine leukemia virus induces apoptosis during the preleukemic period of lymphomagenesis. In this study, we observed that during this period, the viral envelope precursor polyprotein accumulated to high levels in thymic lymphocytes from mice inoculated with virus. Envelope accumulation occurred with the same kinetics as the induction of endoplasmic reticulum (ER) stress, which resulted in the upregulation of the 78-kDa glucose-regulated protein (GRP78). In thymic lymphomas, GRP78 levels were higher than those in virus-infected preleukemic cells, and GRP58 was upregulated. These results suggest that Env precursor accumulation induces ER stress, which participates in thymic lymphocyte apoptosis. The subsequent upregulation of ER chaperone proteins GRP78 and GRP58 may contribute to rescuing cells from virus-induced apoptosis.


Assuntos
Retículo Endoplasmático/metabolismo , Produtos do Gene env/metabolismo , Vírus da Leucemia Murina/fisiologia , Pré-Leucemia/metabolismo , Infecções por Retroviridae/metabolismo , Linfócitos T/virologia , Infecções Tumorais por Vírus/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Retículo Endoplasmático/química , Chaperona BiP do Retículo Endoplasmático , Regulação da Expressão Gênica , Proteínas de Choque Térmico/biossíntese , Linfoma/metabolismo , Linfoma/patologia , Linfoma/virologia , Camundongos , Chaperonas Moleculares/biossíntese , Pré-Leucemia/patologia , Pré-Leucemia/virologia , Isomerases de Dissulfetos de Proteínas/biossíntese , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Linfócitos T/metabolismo , Neoplasias do Timo/metabolismo , Neoplasias do Timo/patologia , Neoplasias do Timo/virologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
19.
Blood ; 108(8): 2703-11, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16794253

RESUMO

The pre-B-cell receptor (pre-BCR) is thought to signal transcriptional activation of the immunoglobulin light (L) chain gene locus, proceeding to its V-J rearrangement. The pre-BCR signaling pathway for this process is largely unknown but may involve the adaptor protein BASH (BLNK/SLP-65). Here we report that the pre-B leukemia cell lines established from affected BASH-deficient mice rearrange kappaL-chain gene locus and down-regulate pre-BCR upon PMA treatment or BASH reconstitution. Analyses with specific inhibitors revealed that activation of novel PKC (nPKC) and MEK, but not Ras, is necessary for the rearrangement. Accordingly, retroviral transduction of active PKCeta, PKCepsilon, or Raf-1, but not Ras, induced the kappa gene rearrangement and expression in the pre-B-cell line. Tamoxifen-mediated BASH reconstitution resulted in the translocation of PKCeta to the plasma membrane and kappa chain expression. These data make evident that the Ras-independent BASH-nPKC-Raf-1 pathway of pre-BCR signaling induces the L-chain gene rearrangement and expression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Rearranjo Gênico de Cadeia Leve de Linfócito B , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Diferenciação Celular , Linhagem Celular Tumoral , DNA de Neoplasias/genética , Leucemia de Células B/genética , Leucemia de Células B/imunologia , Leucemia de Células B/metabolismo , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Pré-Leucemia/genética , Pré-Leucemia/imunologia , Pré-Leucemia/metabolismo , Transdução de Sinais , Proteínas ras/metabolismo
20.
J Clin Invest ; 116(4): 974-83, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16585963

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/virologia , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Adulto , Apoptose , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Portador Sadio/virologia , Ciclo Celular , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pré-Leucemia/metabolismo , Pré-Leucemia/virologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA