Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Leukemia ; 32(2): 419-428, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28720764

RESUMO

Both proto-oncogenic and tumor-suppressive functions have been reported for enhancer of zeste homolog 2 (EZH2). To investigate the effects of its inactivation, a mutant EZH2 lacking its catalytic domain was prepared (EZH2-dSET). In a mouse bone marrow transplant model, EZH2-dSET expression in bone marrow cells induced a myelodysplastic syndrome (MDS)-like disease in transplanted mice. Analysis of these mice identified Abcg2 as a direct target of EZH2. Intriguingly, Abcg2 expression alone induced the same disease in the transplanted mice, where stemness genes were enriched. Interestingly, ABCG2 expression is specifically high in MDS patients. The present results indicate that ABCG2 de-repression induced by EZH2 mutations have crucial roles in MDS pathogenesis.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Animais , Modelos Animais de Doenças , Camundongos , Mutação/genética
2.
Oncogene ; 36(18): 2589-2598, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27819671

RESUMO

Eradication of leukemia stem cells (LSCs) is the ultimate goal of treating acute myeloid leukemia (AML). We recently showed that the combined loss of Runx1/Cbfb inhibited the development of MLL-AF9-induced AML. However, c-Kit+/Gr-1- cells remained viable in Runx1/Cbfb-deleted cells, indicating that suppressing RUNX activity may not eradicate the most immature LSCs. In this study, we found upregulation of several hemostasis-related genes, including the thrombin-activatable receptor PAR-1 (protease-activated receptor-1), in Runx1/Cbfb-deleted MLL-AF9 cells. Similar to the effect of Runx1/Cbfb deletion, PAR-1 overexpression induced CDKN1A/p21 expression and attenuated proliferation in MLL-AF9 cells. To our surprise, PAR-1 deficiency also prevented leukemia development induced by a small number of MLL-AF9 leukemia stem cells (LSCs) in vivo. PAR-1 deficiency also reduced leukemogenicity of AML1-ETO-induced leukemia. Re-expression of PAR-1 in PAR-1-deficient cells combined with a limiting-dilution transplantation assay demonstrated the cell-dose-dependent role of PAR-1 in MLL-AF9 leukemia: PAR-1 inhibited rapid leukemic proliferation when there were a large number of LSCs, while a small number of LSCs required PAR-1 for their efficient growth. Mechanistically, PAR-1 increased the adherence properties of MLL-AF9 cells and promoted their engraftment to bone marrow. Taken together, these data revealed a multifaceted role for PAR-1 in leukemogenesis, and highlight this receptor as a potential target to eradicate primitive LSCs in AML.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Leucemia Mieloide Aguda/genética , Receptor PAR-1/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Células-Tronco Neoplásicas/patologia , Receptor PAR-1/biossíntese
3.
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
4.
Oncogene ; 34(27): 3483-92, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25263451

RESUMO

The transcription factor RUNX1 is a master regulator of hematopoiesis. Disruption of RUNX1 activity has been implicated in the development of hematopoietic neoplasms. Recent studies also highlight the importance of RUNX1 in solid tumors both as a tumor promoter and a suppressor. Given its central role in cancer development, RUNX1 is an excellent candidate for targeted therapy. A potential strategy to target RUNX1 is through modulation of its posttranslational modifications (PTMs). Numerous studies have shown that RUNX1 activity is regulated by PTMs, including phosphorylation, acetylation, methylation and ubiquitination. These PTMs regulate RUNX1 activity either positively or negatively by altering RUNX1-mediated transcription, promoting protein degradation and affecting protein interactions. In this review, we first summarize the available data on the context- and dosage-dependent roles of RUNX1 in various types of neoplasms. We then provide a comprehensive overview of RUNX1 PTMs from biochemical and biologic perspectives. Finally, we discuss how aberrant PTMs of RUNX1 might contribute to tumorigenesis and also strategies to develop anticancer therapies targeting RUNX1 PTMs.


Assuntos
Antineoplásicos/uso terapêutico , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Terapia de Alvo Molecular/métodos , Processamento de Proteína Pós-Traducional , Animais , Sequência de Bases , Carcinogênese/genética , Carcinogênese/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/química , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/genética , Humanos , Lisina/genética , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , Tirosina/genética
5.
Bone Marrow Transplant ; 32(12): 1175-9, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14647272

RESUMO

Late-onset hemorrhagic cystitis (LHC) after hematopoietic stem cell transplantation (HSCT) is mainly caused by viral infections. We retrospectively analyzed the records of 141 Japanese adult patients who underwent a first allogeneic HSCT from 1995 to 2002. In all, 19 patients developed LHC a median of 51 days after HSCT. Adenovirus (AdV) was detected in the urine of 10 LHC patients, of whom eight had AdV type 11. Five of the six available serum samples from these patients were also positive for AdV type 11, but the detection of AdV in serum was not associated with a worse outcome. Male sex and the development of grade II-IV acute graft-versus-host disease were identified as independent significant risk factors for LHC. Male predominance was detected in LHC after HSCT, as has been previously shown in children with AdV-induced acute HC. The detection of AdV DNA in serum did not predict a poor outcome.


Assuntos
Cistite/epidemiologia , Transplante de Células-Tronco Hematopoéticas , Hemorragia/epidemiologia , Infecções por Adenoviridae/complicações , Infecções por Adenoviridae/epidemiologia , Adenovírus Humanos/isolamento & purificação , Adolescente , Adulto , Idoso , Anemia Aplástica/complicações , Anemia Aplástica/terapia , Vírus BK/isolamento & purificação , Bussulfano/efeitos adversos , Ciclofosfamida/efeitos adversos , Cistite/etiologia , Cistite/virologia , Feminino , Doença Enxerto-Hospedeiro/epidemiologia , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/terapia , Hemorragia/etiologia , Hemorragia/virologia , Humanos , Terapia de Imunossupressão/efeitos adversos , Imunossupressores/efeitos adversos , Japão/epidemiologia , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/complicações , Síndromes Mielodisplásicas/terapia , Infecções por Polyomavirus/complicações , Infecções por Polyomavirus/epidemiologia , Estudos Retrospectivos , Fatores de Risco , Distribuição por Sexo , Condicionamento Pré-Transplante/efeitos adversos , Transplante Homólogo , Irradiação Corporal Total/efeitos adversos
6.
Leuk Lymphoma ; 43(11): 2159-63, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12533042

RESUMO

Hepatitis B virus (HBV) reactivation in patients previously positive for hepatitis B surface antibody (HBsAb), so-called reverse seroconversion, has been considered to be a rare complication after hematopoietic stem cell transplantation (HSCT). We experienced two patients who developed reverse seroconversion among nine who were HBsAb positive and Hepatitis B core antibody (HBcAb) positive before HSCT; one after autologous bone marrow transplantation (BMT) and another after allogeneic peripheral blood stem cell transplantation (PBSCT). We reviewed the literature and considered that reverse seroconversion of HBV after HSCT is not uncommon among HBsAb positive recipients. The use of corticosteroids, the lack of HBsAb in donor, and a decrease in serum HBsAb and HBcAb levels may predict reverse seroconversion after HSCT.


Assuntos
Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Vírus da Hepatite B/fisiologia , Ativação Viral/efeitos dos fármacos , Corticosteroides/efeitos adversos , Corticosteroides/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/métodos , Hepatite B/induzido quimicamente , Antígenos do Núcleo do Vírus da Hepatite B/sangue , Antígenos de Superfície da Hepatite B/sangue , Vírus da Hepatite B/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Transplante Autólogo , Transplante Homólogo
7.
Oncogene ; 33(42): 5028-38, 2014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24747972

RESUMO

Relapse of chronic myeloid leukemia (CML) is triggered by stem cells with a reconstituting capacity similar to that of hematopoietic stem cells (HSCs) and CML stem cells are a source of resistance in drug therapy with tyrosine kinase inhibitors (TKIs). Ecotropic viral integration site 1 (EVI1), a key transcription factor in HSC regulation, is known to predict poor outcomes in myeloid malignancies, however, incapability of prospective isolation of EVI1-high leukemic cells precludes the functional evaluation of intraindividual EVI1-high cells. Introduction of CML into Evi1-internal ribosomal entry site (IRES)-green fluorescent protein (GFP) knock-in mice, a versatile HSC-reporter strain, enables us to separate Evi1-high CML cells from the individual. Evi1-IRES-GFP allele models of CML in chronic phase (CML-CP), by retroviral overexpression of BCR-ABL and by crossing BCR-ABL transgenic mice, revealed that Evi1 is predominantly enriched in the stem cell fraction and associated with an enhanced proliferative as well as a leukemia-initiating capacity and that Evi1-high CML-CP cells exhibit resistance to TKIs. Overexpressing BCR-ABL and NUP98-HOXA9 in Evi1-IRES-GFP knock-in mice to model CML in blast crisis (CML-BC), in which Evi1-high cells turned to be a major population as opposed to a minor population in CML-CP models, showed that Evi1-high CML-BC cells have a greater potential to recapitulate the disease and appear resistant to TKIs. Furthermore, given that Evi1 heterozygosity ameliorates CML-CP and CML-BC development and that the combination of Evi1 and BCR-ABL causes acute myeloid leukemia resembling CML-BC, Evi1 could regulate CML development as a potent driver. In addition, in human CML-CP cases, we show that EVI1 is highly expressed in stem cell-enriched CD34+CD38-CD90+ fraction at single-cell level. This is the first report to clarify directly that Evi1-high leukemic cells themselves possess the superior potential to Evi1-low cells in oncogenic self-renewal, which highlights the role of Evi1 as a valuable and a functional marker of CML stem cells.


Assuntos
Antineoplásicos/farmacologia , Crise Blástica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Células-Tronco Neoplásicas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Fatores de Transcrição/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Carcinogênese/metabolismo , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Proteínas de Fusão bcr-abl/fisiologia , Regulação Leucêmica da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteína do Locus do Complexo MDS1 e EVI1 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Fenótipo , Proto-Oncogenes , Regulação para Cima
8.
Leukemia ; 24(1): 81-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19776757

RESUMO

The ecotropic viral integration site-1 (EVI-1) is a nuclear transcription factor and has an essential function in the proliferation/maintenance of haematopoietic stem cells. Aberrant expression of EVI-1 has been frequently found in myeloid leukaemia as well as in several solid tumours, and is associated with a poor patient survival. It was recently shown that EVI-1 associates with two different histone methyltransferases (HMTs), SUV39H1 and G9a. However, the functional roles of these HMTs in EVI-1-mediated leukemogenesis remain unclear. In this study, we showed that EVI-1 physically interacts with SUV39H1 and G9a, but not with Set9. Immunofluorescence analysis revealed that EVI-1 colocalizes with these HMTs in nuclei. We also found that the catalytically inactive form of SUV39H1 abrogates the transcriptional repression mediated by EVI-1, suggesting that SUV39H1 is actively involved in EVI-1-mediated transcriptional repression. Furthermore, RNAi-based knockdown of SUV39H1 or G9a in Evi-1-expressing progenitors significantly reduced their colony-forming activity. In contrast, knockdown of these HMTs did not impair bone marrow immortalization by E2A/HLF. These results indicate that EVI-1 forms higher-order complexes with HMTs, and this association has a role in the transcription repression and bone marrow immortalization. Targeting these HMTs may be of therapeutic benefit in the treatment for EVI-1-related haematological malignancies.


Assuntos
Medula Óssea/metabolismo , Proteínas de Ligação a DNA/fisiologia , Antígenos de Histocompatibilidade/fisiologia , Histona-Lisina N-Metiltransferase/fisiologia , Metiltransferases/fisiologia , Proto-Oncogenes/fisiologia , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Animais , Células COS , Chlorocebus aethiops , Proteínas de Ligação a DNA/análise , Antígenos de Histocompatibilidade/análise , Histona-Lisina N-Metiltransferase/análise , Humanos , Proteína do Locus do Complexo MDS1 e EVI1 , Metilação , Metiltransferases/análise , Proteínas Repressoras/análise , Fatores de Transcrição/análise
9.
Oncogene ; 28(49): 4364-74, 2009 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-19767769

RESUMO

Ecotropic viral integration site-1 (Evi-1) is a nuclear transcription factor, which is essential for the proliferation/maintenance of hematopoietic stem cells (HSCs). Aberrant expression of Evi-1 has been frequently found in myeloid leukemia, and is associated with a poor patient survival. Recently, we reported candidate target genes of Evi-1 shared in HSCs and leukemic cells using gene expression profiling analysis. In this study, we identified Pbx1, a proto-oncogene in hematopoietic malignancy, as a target gene of Evi-1. Overexpression of Evi-1 increased Pbx1 expression in hematopoietic stem/progenitor cells. An analysis of the Pbx1 promoter region revealed that Evi-1 upregulates Pbx1 transcription. Furthermore, reduction of Pbx1 levels through RNAi-mediated knockdown significantly inhibited Evi-1-induced transformation. In contrast, knockdown of Pbx1 did not impair bone marrow transformation by E2A/HLF or AML1/ETO, suggesting that Pbx1 is specifically required for the maintenance of bone marrow transformation mediated by Evi-1. These results indicate that Pbx1 is a target gene of Evi-1 involved in Evi-1-mediated leukemogenesis.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Leucemia/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes/fisiologia , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Células COS , Transformação Celular Neoplásica/genética , Células Cultivadas , Chlorocebus aethiops , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia/metabolismo , Leucemia/patologia , Proteína do Locus do Complexo MDS1 e EVI1 , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Dados de Sequência Molecular , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/metabolismo
10.
Leukemia ; 22(6): 1241-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18337762

RESUMO

The t(3;21) chromosomal translocation seen in blastic crisis of chronic myeloid leukemia and secondary leukemias results in a formation of a chimeric protein AML1-Evi-1, which suppresses wild-type AML1 function. Loss of AML1 function causes expansion of hematopoietic progenitor cells, whereas it is not sufficient for the development of leukemia. To identify essential mechanisms through which AML1-Evi-1 exerts full leukemogenic potential, we introduced AML1-Evi-1 and its mutants in murine bone marrow cells, and evaluated their transforming activities by colony replating assays. The transforming activity of AML1-Evi-1 was lost when any of the known functional domains of Evi-1 was deleted from the chimeric protein, and forced expression of Evi-1 did not transform the AML1-deleted bone marrow cells. Unlike the MLL-ENL and AML1-ETO leukemia-related chimeric proteins, AML1-Evi-1 could transform only the hematopoietic stem cell fraction. Moreover, AML1-Evi-1-transformed cells show a cell-marker profile distinct from that of the cells transformed by AML1-ETO, which also suppresses AML1 function. Thus, leukemogenic activity of AML1-Evi-1 may be due to activation of molecular mechanisms distinct from those activated by MLL-ENL or AML1-ETO in the hematopoietic stem cell fractions.


Assuntos
Células da Medula Óssea/fisiologia , Transformação Celular Neoplásica , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Proteínas de Ligação a DNA/fisiologia , Células-Tronco Hematopoéticas , Proteínas de Fusão Oncogênica/fisiologia , Proto-Oncogenes/fisiologia , Fatores de Transcrição/fisiologia , Animais , Western Blotting , Citometria de Fluxo , Integrases/metabolismo , Proteína do Locus do Complexo MDS1 e EVI1 , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Células Progenitoras Mieloides/fisiologia , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-kit/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa