Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Leukemia ; 31(4): 872-881, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27740633

RESUMO

Traditional response criteria in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are based on bone marrow morphology and may not accurately reflect clonal tumor burden in patients treated with non-cytotoxic chemotherapy. We used next-generation sequencing of serial bone marrow samples to monitor MDS and AML tumor burden during treatment with epigenetic therapy (decitabine and panobinostat). Serial bone marrow samples (and skin as a source of normal DNA) from 25 MDS and AML patients were sequenced (exome or 285 gene panel). We observed that responders, including those in complete remission (CR), can have persistent measurable tumor burden (that is, mutations) for at least 1 year without disease progression. Using an ultrasensitive sequencing approach, we detected extremely rare mutations (equivalent to 1 heterozygous mutant cell in 2000 non-mutant cells) months to years before their expansion at disease relapse. While patients can live with persistent clonal hematopoiesis in a CR or stable disease, ultimately we find evidence that expansion of a rare subclone occurs at relapse or progression. Here we demonstrate that sequencing of serial samples provides an alternative measure of tumor burden in MDS or AML patients and augments traditional response criteria that rely on bone marrow blast percentage.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Evolução Clonal/genética , Epigênese Genética/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Idoso , Idoso de 80 Anos ou mais , Medula Óssea/patologia , Exoma , Feminino , Genes p53 , Sequenciamento de Nucleotídeos em Larga Escala , Inibidores de Histona Desacetilases/administração & dosagem , Humanos , Leucemia Mieloide Aguda/diagnóstico , Masculino , Pessoa de Meia-Idade , Mutação , Síndromes Mielodisplásicas/diagnóstico , Polimorfismo de Nucleotídeo Único , Indução de Remissão , Resultado do Tratamento , Carga Tumoral
4.
Leukemia ; 29(6): 1279-89, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25600023

RESUMO

HOX genes are highly expressed in many acute myeloid leukemia (AML) samples, but the patterns of expression and associated regulatory mechanisms are not clearly understood. We analyzed RNA sequencing data from 179 primary AML samples and normal hematopoietic cells to understand the range of expression patterns in normal versus leukemic cells. HOX expression in AML was restricted to specific genes in the HOXA or HOXB loci, and was highly correlated with recurrent cytogenetic abnormalities. However, the majority of samples expressed a canonical set of HOXA and HOXB genes that was nearly identical to the expression signature of normal hematopoietic stem/progenitor cells. Transcriptional profiles at the HOX loci were similar between normal cells and AML samples, and involved bidirectional transcription at the center of each gene cluster. Epigenetic analysis of a subset of AML samples also identified common regions of chromatin accessibility in AML samples and normal CD34(+) cells that displayed differences in methylation depending on HOX expression patterns. These data provide an integrated epigenetic view of the HOX gene loci in primary AML samples, and suggest that HOX expression in most AML samples represents a normal stem cell program that is controlled by epigenetic mechanisms at specific regulatory elements.


Assuntos
Biomarcadores Tumorais/genética , Metilação de DNA , Epigenômica , Regulação Leucêmica da Expressão Gênica , Genes Homeobox/genética , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/genética , Estudos de Casos e Controles , Aberrações Cromossômicas , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Leucemia Mieloide Aguda/mortalidade , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida
5.
Leukemia ; 29(4): 909-17, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25311244

RESUMO

We previously identified missense mutations in the U2AF1 splicing factor affecting codons S34 (S34F and S34Y) or Q157 (Q157R and Q157P) in 11% of the patients with de novo myelodysplastic syndrome (MDS). Although the role of U2AF1 as an accessory factor in the U2 snRNP is well established, it is not yet clear how these mutations affect splicing or contribute to MDS pathophysiology. We analyzed splice junctions in RNA-seq data generated from transfected CD34+ hematopoietic cells and found significant differences in the abundance of known and novel junctions in samples expressing mutant U2AF1 (S34F). For selected transcripts, splicing alterations detected by RNA-seq were confirmed by analysis of primary de novo MDS patient samples. These effects were not due to impaired U2AF1 (S34F) localization as it co-localized normally with U2AF2 within nuclear speckles. We further found evidence in the RNA-seq data for decreased affinity of U2AF1 (S34F) for uridine (relative to cytidine) at the e-3 position immediately upstream of the splice acceptor site and corroborated this finding using affinity-binding assays. These data suggest that the S34F mutation alters U2AF1 function in the context of specific RNA sequences, leading to aberrant alternative splicing of target genes, some of which may be relevant for MDS pathogenesis.


Assuntos
Processamento Alternativo , Leucócitos Mononucleares/metabolismo , Proteínas Nucleares/genética , Precursores de RNA/genética , Ribonucleoproteínas/genética , Spliceossomos/metabolismo , Antígenos CD34/genética , Antígenos CD34/metabolismo , Sequência de Bases , Sítios de Ligação , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Humanos , Leucócitos Mononucleares/citologia , Dados de Sequência Molecular , Mutação , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/patologia , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Plasmídeos , Cultura Primária de Células , Ligação Proteica , Precursores de RNA/química , Precursores de RNA/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Transdução de Sinais , Spliceossomos/genética , Fator de Processamento U2AF , Transfecção
6.
Leukemia ; 29(4): 869-76, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25252869

RESUMO

Clonal architecture in myeloproliferative neoplasms (MPNs) is poorly understood. Here we report genomic analyses of a patient with primary myelofibrosis (PMF) transformed to secondary acute myeloid leukemia (sAML). Whole genome sequencing (WGS) was performed on PMF and sAML diagnosis samples, with skin included as a germline surrogate. Deep sequencing validation was performed on the WGS samples and an additional sample obtained during sAML remission/relapsed PMF. Clustering analysis of 649 validated somatic single-nucleotide variants revealed four distinct clonal groups, each including putative driver mutations. The first group (including JAK2 and U2AF1), representing the founding clone, included mutations with high frequency at all three disease stages. The second clonal group (including MYB) was present only in PMF, suggesting the presence of a clone that was dispensable for transformation. The third group (including ASXL1) contained mutations with low frequency in PMF and high frequency in subsequent samples, indicating evolution of the dominant clone with disease progression. The fourth clonal group (including IDH1 and RUNX1) was acquired at sAML transformation and was predominantly absent at sAML remission/relapsed PMF. Taken together, these findings illustrate the complex clonal dynamics associated with disease evolution in MPNs and sAML.


Assuntos
Transformação Celular Neoplásica/genética , Evolução Clonal/genética , Genoma Humano , Leucemia Mieloide Aguda/genética , Mielofibrose Primária/genética , Transformação Celular Neoplásica/patologia , Células Clonais , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Progressão da Doença , Feminino , Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Isocitrato Desidrogenase/genética , Janus Quinase 2/genética , Leucemia Mieloide Aguda/patologia , Pessoa de Meia-Idade , Taxa de Mutação , Proteínas Nucleares/genética , Polimorfismo de Nucleotídeo Único , Mielofibrose Primária/patologia , Proteínas Proto-Oncogênicas c-myb/genética , Proteínas Repressoras/genética , Ribonucleoproteínas/genética , Fator de Processamento U2AF
8.
Leukemia ; 27(6): 1275-82, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23443460

RESUMO

Recent studies suggest that most cases of myelodysplastic syndrome (MDS) are clonally heterogeneous, with a founding clone and multiple subclones. It is not known whether specific gene mutations typically occur in founding clones or subclones. We screened a panel of 94 candidate genes in a cohort of 157 patients with MDS or secondary acute myeloid leukemia (sAML). This included 150 cases with samples obtained at MDS diagnosis and 15 cases with samples obtained at sAML transformation (8 were also analyzed at the MDS stage). We performed whole-genome sequencing (WGS) to define the clonal architecture in eight sAML genomes and identified the range of variant allele frequencies (VAFs) for founding clone mutations. At least one mutation or cytogenetic abnormality was detected in 83% of the 150 MDS patients and 17 genes were significantly mutated (false discovery rate ≤0.05). Individual genes and patient samples displayed a wide range of VAFs for recurrently mutated genes, indicating that no single gene is exclusively mutated in the founding clone. The VAFs of recurrently mutated genes did not fully recapitulate the clonal architecture defined by WGS, suggesting that comprehensive sequencing may be required to accurately assess the clonal status of recurrently mutated genes in MDS.


Assuntos
Mutação , Síndromes Mielodisplásicas/genética , Feminino , Frequência do Gene , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Pessoa de Meia-Idade , Recidiva
9.
Leukemia ; 25(7): 1153-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21415852

RESUMO

Alterations in DNA methylation have been implicated in the pathogenesis of myelodysplastic syndromes (MDS), although the underlying mechanism remains largely unknown. Methylation of CpG dinucleotides is mediated by DNA methyltransferases, including DNMT1, DNMT3A and DNMT3B. DNMT3A mutations have recently been reported in patients with de novo acute myeloid leukemia (AML), providing a rationale for examining the status of DNMT3A in MDS samples. In this study, we report the frequency of DNMT3A mutations in patients with de novo MDS, and their association with secondary AML. We sequenced all coding exons of DNMT3A using DNA from bone marrow and paired normal cells from 150 patients with MDS and identified 13 heterozygous mutations with predicted translational consequences in 12/150 patients (8.0%). Amino acid R882, located in the methyltransferase domain of DNMT3A, was the most common mutation site, accounting for 4/13 mutations. DNMT3A mutations were expressed in the majority of cells in all tested mutant samples regardless of myeloblast counts, suggesting that DNMT3A mutations occur early in the course of MDS. Patients with DNMT3A mutations had worse overall survival compared with patients without DNMT3A mutations (P=0.005) and more rapid progression to AML (P=0.007), suggesting that DNMT3A mutation status may have prognostic value in de novo MDS.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Mutação , Síndromes Mielodisplásicas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Códon/genética , Ilhas de CpG/genética , Metilação de DNA/genética , DNA Metiltransferase 3A , DNA de Neoplasias/genética , Progressão da Doença , Éxons/genética , Feminino , Células Precursoras de Granulócitos/enzimologia , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidade , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/enzimologia , Síndromes Mielodisplásicas/mortalidade , Prognóstico , Análise de Sequência de DNA , Adulto Jovem
10.
Leukemia ; 24(5): 950-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20376082

RESUMO

The t(8;21)(q22;q22) translocation, present in approximately 5% of adult acute myeloid leukemia (AML) cases, produces the AML1/ETO (AE) fusion protein. Dysregulation of the Pit/Oct/Unc (POU) domain-containing transcription factor POU4F1 is a recurring abnormality in t(8;21) AML. In this study, we showed that POU4F1 overexpression is highly correlated with, but not caused by, AE. We observed that AE markedly increases the self-renewal capacity of myeloid progenitors from murine bone marrow or fetal liver and drives the expansion of these cells in liquid culture. POU4F1 is neither necessary nor sufficient for these AE-dependent properties, suggesting that it contributes to leukemia through novel mechanisms. To identify targets of POU4F1, we performed gene expression profiling in primary mouse cells with genetically defined levels of POU4F1 and identified 140 differentially expressed genes. This expression signature was significantly enriched in human t(8;21) AML samples and was sufficient to cluster t(8;21) AML samples in an unsupervised hierarchical analysis. Among the most highly differentially expressed genes, half are known AML1/ETO targets, implying that the unique transcriptional signature of t(8;21) AML is, in part, attributable to POU4F1 and not AML1/ETO itself. These genes provide novel candidates for understanding the biology and developing therapeutic approaches for t(8;21) AML.


Assuntos
Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 8/genética , Perfilação da Expressão Gênica , Leucemia Mieloide Aguda/genética , Fator de Transcrição Brn-3A/genética , Fator de Transcrição Brn-3A/fisiologia , Translocação Genética/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Feto/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Fusão Oncogênica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína 1 Parceira de Translocação de RUNX1 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Brn-3A/metabolismo
12.
Proc Natl Acad Sci U S A ; 98(26): 14985-90, 2001 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-11752447

RESUMO

Granzyme B (GzmB) is a serine protease that is used by activated cytotoxic T lymphocytes to induce target cell apoptosis. Although GzmB directly cleaves the Bcl2 family member BID on target cell entry, Bid-deficient (and Bax, Bak doubly deficient) cells are susceptible to GzmB-induced death, even though they fail to release cytochrome c from mitochondria. GzmB still induces mitochondrial depolarization in Bax, Bak double knockout cells without cytochrome c release or opening of the permeability transition pore. Because GzmB cannot directly cause depolarization of isolated mitochondria, novel intracellular factor(s) may be required for GzmB to depolarize mitochondria in situ. GzmB therefore utilizes two distinct mitochondrial pathways to amplify the proapoptotic signal that it delivers to target cells.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Transporte/fisiologia , Proteínas de Membrana/fisiologia , Mitocôndrias Hepáticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/fisiologia , Serina Endopeptidases/farmacologia , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Permeabilidade da Membrana Celular , Grupo dos Citocromos c/metabolismo , Imunofluorescência , Granzimas , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias Hepáticas/enzimologia , Mitocôndrias Hepáticas/fisiologia , Frações Subcelulares/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína X Associada a bcl-2
13.
Curr Opin Hematol ; 8(4): 206-11, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11561157

RESUMO

Translocations involving a variety of fusion partners, such as promyelocytic leukemia gene, promyelocytic leukemia zinc finger, nucleophosmin, nuclear matrix protein, and signal transducer and activator of transcription protein 5B, with the retinoic acid receptor alpha gene are commonly associated with development of acute promyelocytic leukemia. Through the development of transgenic mouse models, some retinoic acid receptor alpha translocation fusion proteins have been shown to be capable of initiating acute promyelocytic leukemia development, and dictate the leukemias' responsiveness to retinoic acid. Transgenic mouse models also have identified the influence of reciprocal translocation fusion proteins on acute promyelocytic leukemia development, and have demonstrated that additional mutations can contribute to the development of acute promyelocytic leukemia. In this review, the authors summarize current mouse models of acute promyelocytic leukemia and describe current knowledge about additional genetic alterations that occur during development of acute promyelocytic leukemia in the mouse.


Assuntos
Modelos Animais de Doenças , Leucemia Promielocítica Aguda/genética , Animais , Antineoplásicos/uso terapêutico , Arsênio/uso terapêutico , Deleção Cromossômica , Leucemia Promielocítica Aguda/tratamento farmacológico , Camundongos , Modelos Biológicos , Mutação , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Tretinoína/uso terapêutico
14.
Blood ; 98(2): 266-71, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11435292

RESUMO

Arsenic trioxide has been shown to be effective in treating acute promyelocytic leukemia (APL), with minimal overall toxicity reported to date. A phase I/II study was initiated in June 1998 using arsenic trioxide for relapsed APL to determine the maximum tolerated or minimal effective dose and to determine the efficacy of treatment at that dose. Ten patients received 1 to 4 monthly cycles of treatment with 0.1 mg/kg per day intravenous arsenic trioxide. Six of 7 patients evaluable for response achieved cytogenetic or molecular complete remission. However, 3 patients died suddenly during the first cycle of treatment. Autopsies obtained on 2 of these failed to identify a cause of sudden death, despite evidence of pulmonary hemorrhage in one. A third patient, for whom an autopsy was not performed, became asystolic and died while on continuous cardiac telemetry. These observations suggest that arsenic trioxide may be significantly or even fatally toxic at doses currently used and that caution is warranted in its use.


Assuntos
Antineoplásicos/efeitos adversos , Arsenicais/efeitos adversos , Morte Súbita , Leucemia Promielocítica Aguda/tratamento farmacológico , Óxidos/efeitos adversos , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Trióxido de Arsênio , Arsenicais/administração & dosagem , Arsenicais/farmacocinética , Criança , Feminino , Humanos , Cinética , Masculino , Pessoa de Meia-Idade , Óxidos/administração & dosagem , Óxidos/farmacocinética , Recidiva
15.
Blood ; 98(1): 65-73, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11418464

RESUMO

For the treatment of beta-globin gene defects, a homologous recombination-mediated gene correction approach would provide advantages over random integration-based gene therapy strategies. However, "neighborhood effects" from retained selectable marker genes in the targeted locus are among the key issues that must be taken into consideration for any attempt to use this strategy for gene correction. An Ala-to-Ile mutation was created in the beta6 position of the mouse beta-major globin gene (beta(6I)) as a step toward the development of a murine model system that could serve as a platform for therapeutic gene correction studies. The marked beta-major gene can be tracked at the level of DNA, RNA, and protein, allowing investigation of the impact of a retained phosphoglycerate kinase (PGK)-neo cassette located between the mutant beta-major and beta-minor globin genes on expression of these 2 neighboring genes. Although the PGK-neo cassette was expressed at high levels in adult erythroid cells, the abundance of the beta(6I) mRNA was indistinguishable from that of the wild-type counterpart in bone marrow cells. Similarly, the output from the beta-minor globin gene was also normal. Therefore, in this specific location, the retained, transcriptionally active PGK-neo cassette does not disrupt the regulated expression of the adult beta-globin genes. (Blood. 2001;98:65-73)


Assuntos
Globinas/genética , Fosfoglicerato Quinase/genética , Transcrição Gênica/genética , Animais , Medula Óssea/metabolismo , Eritrócitos/metabolismo , Regulação da Expressão Gênica , Marcação de Genes , Hemoglobinas/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Mutantes , Mutagênese Insercional , Mutagênese Sítio-Dirigida , Oxigênio/metabolismo , RNA/metabolismo , Recombinação Genética
16.
Proc Natl Acad Sci U S A ; 97(24): 13306-11, 2000 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-11087871

RESUMO

We previously generated a transgenic mouse model for acute promyelocytic leukemia (APL) by expressing the promyelocytic leukemia (PML)-retinoic acid receptor (RARalpha) cDNA in early myeloid cells. This fusion protein causes a myeloproliferative disease in 100% of animals, but only 15-20% of the animals develop acute leukemia after a long latency period (6-13 months). PML-RARalpha is therefore necessary, but not sufficient, for APL development. The coexpression of a reciprocal form of the fusion, RARalpha-PML, increased the likelihood of APL development (55-60%), but did not shorten latency. Together, these results suggested that additional genetic events are required for the development of APL. We therefore evaluated the splenic tumor cells from 18 transgenic mice with APL for evidence of secondary genetic events, by using spectral karyotyping analysis. Interstitial or terminal deletions of the distal region of one copy of chromosome 2 [del(2)] were found in 1/5 tumors expressing PML-RARalpha, but in 11/13 tumors expressing both PML-RARalpha and RARalpha-PML (P < 0.05). Leukemic cells that contained a deletion on chromosome 2 often contained additional chromosomal gains (especially of 15), chromosomal losses (especially of 11 or X/Y), or were tetraploid (P

Assuntos
Aberrações Cromossômicas , Transtornos Cromossômicos , Mapeamento Cromossômico , Leucemia Promielocítica Aguda/genética , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/genética , Animais , Aberrações Cromossômicas/genética , Deleção Cromossômica , Cruzamentos Genéticos , Feminino , Humanos , Cariotipagem , Leucemia Promielocítica Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estadiamento de Neoplasias
17.
Immunity ; 12(6): 621-32, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10894162

RESUMO

Granzyme B (GzmB) is a component of cytotoxic lymphocyte granules that can rapidly initiate apoptosis in target cells. While several procaspases are cleaved and activated by GzmB, the absolute requirement of caspase activation for GzmB-induced apoptosis is controversial. In this report, we demonstrate that GzmB can initiate apoptosis in the absence of caspase-3 activity by directly cleaving DFF45/ICAD to liberate activated DFF40/CAD. DFF45/ICAD cleavage occurs less efficiently in cells that lack caspase-3 activity, suggesting that the caspases normally amplify the GzmB death signal. DFF45/ICAD-deficient mouse embryo fibroblasts are partially resistant to GzmB-induced death, demonstrating the biological importance of DFF45/ICAD for GzmB-mediated apoptosis.


Assuntos
Apoptose/imunologia , Fragmentação do DNA/imunologia , Desoxirribonucleases/antagonistas & inibidores , Inibidores Enzimáticos/metabolismo , Processamento de Proteína Pós-Traducional/imunologia , Proteínas/metabolismo , Serina Endopeptidases/metabolismo , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Citotoxicidade Imunológica , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Granzimas , Imunidade Inata , Células Matadoras Ativadas por Linfocina/citologia , Células Matadoras Ativadas por Linfocina/enzimologia , Células Matadoras Ativadas por Linfocina/imunologia , Camundongos , Proteínas/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/fisiologia , Especificidade por Substrato , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/enzimologia , Linfócitos T Citotóxicos/imunologia
18.
Proc Natl Acad Sci U S A ; 96(26): 15103-8, 1999 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-10611345

RESUMO

Acute promyelocytic leukemia (APML) most often is associated with the balanced reciprocal translocation t(15;17) (q22;q11.2) and the expression of both the PML-RARalpha and RARalpha-PML fusion cDNAs that are formed by this translocation. In this report, we investigated the biological role of a bcr-3 isoform of RARalpha-PML for the development of APML in a transgenic mouse model. Expression of RARalpha-PML alone in the early myeloid cells of transgenic mice did not alter myeloid development or cause APML, but its expression significantly increased the penetrance of APML in mice expressing a bcr-1 isoform of PML-RARalpha (15% of animals developed APML with PML-RARalpha alone vs. 57% with both transgenes, P < 0.001). The latency of APML development was not altered substantially by the expression of RARalpha-PML, suggesting that it does not behave as a classical "second hit" for development of the disease. Leukemias that arose from doubly transgenic mice were less mature than those from PML-RARalpha transgenic mice, but they both responded to all-trans retinoic acid in vitro. These findings suggest that PML-RARalpha drives the development of APML and defines its basic phenotype, whereas RARalpha-PML potentiates this phenotype via mechanisms that are not yet understood.


Assuntos
Leucemia Promielocítica Aguda/etiologia , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Translocação Genética , Animais , Células da Medula Óssea , Catepsinas/genética , Cruzamentos Genéticos , Expressão Gênica , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/mortalidade , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/genética , Penetrância , Fenótipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Baço/patologia , Tretinoína/farmacologia
19.
Proc Natl Acad Sci U S A ; 96(15): 8627-32, 1999 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-10411926

RESUMO

Dipeptidyl peptidase I (DPPI) is a lysosomal cysteine protease that has been implicated in the processing of granzymes, which are neutral serine proteases exclusively expressed in the granules of activated cytotoxic lymphocytes. In this report, we show that cytotoxic lymphocytes derived from DPPI-/- mice contain normal amounts of granzymes A and B, but these molecules retain their prodipeptide domains and are inactive. Cytotoxic assays with DPPI-/- effector cells reveal severe defects in the induction of target cell apoptosis (as measured by [(125)I]UdR release) at both early and late time points; this defect is comparable to that detected in perforin-/- or granzyme A-/- x B-/- cytotoxic lymphocytes. DPPI therefore plays an essential role in the in vivo processing and activation of granzymes A and B, which are required for cytotoxic lymphocyte granule-mediated apoptosis.


Assuntos
Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Serina Endopeptidases/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Apoptose/imunologia , Catepsina C , Grânulos Citoplasmáticos/enzimologia , Grânulos Citoplasmáticos/imunologia , Testes Imunológicos de Citotoxicidade , Desoxiuridina/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/deficiência , Ativação Enzimática , Citometria de Fluxo , Marcação de Genes/métodos , Granzimas , Células Matadoras Ativadas por Linfocina , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Perforina , Proteínas Citotóxicas Formadoras de Poros , Precursores de Proteínas/metabolismo , Linfócitos T Citotóxicos/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA