Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Cells ; 41(5): 444-453, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29477140

RESUMO

Aberrations in histone modifications are being studied in mixed-lineage leukemia (MLL)-AF9-driven acute myeloid leukemia (AML). In this study, we focused on the regulation of the differentiation of the MLL-AF9 type AML cell line THP-1. We observed that, upon phorbol 12-myristate 13-acetate (PMA) treatment, THP-1 cells differentiated into monocytes by down-regulating Aurora kinase A (AURKA), resulting in a reduction in H3S10 phosphorylation. We revealed that the AURKA inhibitor alisertib accelerates the expression of the H3K27 demethylase KDM6B, thereby dissociating AURKA and YY1 from the KDM6B promoter region. Using Flow cytometry, we found that alisertib induces THP-1 differentiation into monocytes. Furthermore, we found that treatment with the KDM6B inhibitor GSK-J4 perturbed the PMA-mediated differentiation of THP-1 cells. Thus, we discovered the mechanism of AURKA-KDM6B signaling that controls the differentiation of THP-1 cells, which has implications for biotherapy for leukemia.


Assuntos
Aurora Quinase A/fisiologia , Regulação Leucêmica da Expressão Gênica , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/fisiologia , Leucemia Monocítica Aguda/patologia , Proteínas de Neoplasias/fisiologia , Aurora Quinase A/antagonistas & inibidores , Azepinas/farmacologia , Benzazepinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Imunoprecipitação da Cromatina , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Genes Reporter , Células HEK293 , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Leucemia Monocítica Aguda/genética , Leucemia Monocítica Aguda/metabolismo , Monócitos/citologia , Proteína de Leucina Linfoide-Mieloide/fisiologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Fusão Oncogênica/fisiologia , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Células THP-1 , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição YY1/metabolismo
2.
Rinsho Byori ; 63(5): 631-42, 2015 May.
Artigo em Japonês | MEDLINE | ID: mdl-26524903

RESUMO

Acute promyelocytic leukemia (APL) is one of the well-characterized subtypes of acute myeloid leukemia (AML). The essential drugs used in the treatment strategy for APL include all-trans retinoic acid (ATRA) and arsenic trioxide (ATO), which are both pioneer molecular-targeting agents. They were initially administered to patients based on the therapeutic experience of traditional Chinese medicine, and their marked effectiveness has been demonstrated. Subsequently, the molecular mechanisms of these drugs, as well as the molecular pathogenesis of APL, have been elucidated, whereby the chimeric gene product PML-RARα induces epigenetic changes and transcription repression. This review summarizes the findings of previous studies related to the in vitro and in vivo function of PML-RARα and the effects of ATRA and ATO on PML-RARα and APL cells. These findings are very important, because the concept of epigenetic modulation in oncogenesis and their application as molecular targets in APL therapy have now been accepted in other types of leukemia, as well as for other malignancies.


Assuntos
Antineoplásicos/uso terapêutico , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/genética , Animais , Trióxido de Arsênio , Arsenicais/administração & dosagem , Arsenicais/farmacologia , Modelos Animais de Doenças , Epigenômica , Humanos , Medicina Tradicional Chinesa , Camundongos , Terapia de Alvo Molecular , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteínas de Fusão Oncogênica/fisiologia , Óxidos/administração & dosagem , Óxidos/farmacologia , Transcrição Gênica/genética , Tretinoína/administração & dosagem , Tretinoína/farmacologia
4.
Microbiol Mol Biol Rev ; 70(3): 646-59, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16959964

RESUMO

The positive transcription elongation factor b (P-TEFb) stimulates transcriptional elongation by phosphorylating the carboxy-terminal domain of RNA polymerase II and antagonizing the effects of negative elongation factors. Not only is P-TEFb essential for transcription of the vast majority of cellular genes, but it is also a critical host cellular cofactor for the expression of the human immunodeficiency virus (HIV) type 1 genome. Given its important role in globally affecting transcription, P-TEFb's activity is dynamically controlled by both positive and negative regulators in order to achieve a functional equilibrium in sync with the overall transcriptional demand as well as the proliferative state of cells. Notably, this equilibrium can be shifted toward either the active or inactive state in response to diverse physiological stimuli that can ultimately affect the cellular decision between growth and differentiation. In this review, we examine the mechanisms by which the recently identified positive (the bromodomain protein Brd4) and negative (the noncoding 7SK small nuclear RNA and the HEXIM1 protein) regulators of P-TEFb affect the P-TEFb-dependent transcriptional elongation. We also discuss the consequences of perturbations of the dynamic associations of these regulators with P-TEFb in relation to the pathogenesis and progression of several major human diseases, such as cardiac hypertrophy, breast cancer, and HIV infection.


Assuntos
Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , HIV/genética , Fator B de Elongação Transcricional Positiva/fisiologia , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/genética , Processos de Crescimento Celular/genética , Regulação Viral da Expressão Gênica , HIV/crescimento & desenvolvimento , Humanos , Modelos Biológicos , Proteínas Nucleares , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Fator B de Elongação Transcricional Positiva/genética , Fatores de Transcrição
5.
Proc Natl Acad Sci U S A ; 103(5): 1400-5, 2006 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-16432238

RESUMO

PML-RARalpha induces a block of hematopoietic differentiation and acute promyelocytic leukemia. This block is based on its capacity to inactivate target genes by recruiting histone deacetylase (HDAC) and DNA methyltransferase activities. Here we report that MBD1, a member of a conserved family of proteins able to bind methylated DNA, cooperates with PML-RARalpha in transcriptional repression and cellular transformation. PML-RARalpha recruits MBD1 to its target promoter through an HDAC3-mediated mechanism. Binding of HDAC3 and MBD1 is not confined to the promoter region but instead is spread over the locus. Knock-down of HDAC3 expression by RNA interference in acute promyelocytic leukemia cells alleviates PML-RAR-induced promoter silencing. We further demonstrate that retroviral expression of dominant-negative mutants of MBD1 in hematopoietic precursors compromises the ability of PML-RARalpha to block their differentiation and thus restored cell differentiation. Our results demonstrate that PML-RARalpha functions by recruiting an HDAC3-MBD1 complex that contributes to the establishment and maintenance of the silenced chromatin state.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/metabolismo , Proteínas de Fusão Oncogênica/fisiologia , Fatores de Transcrição/fisiologia , Western Blotting , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Cromatina/química , Cromatina/metabolismo , Imunoprecipitação da Cromatina , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/química , Epigênese Genética , Inativação Gênica , Genes Dominantes , Vetores Genéticos , Células HeLa , Células-Tronco Hematopoéticas/citologia , Histona Desacetilases/metabolismo , Humanos , Imunoprecipitação , Leucemia/metabolismo , Luciferases/metabolismo , Modelos Biológicos , Oligonucleotídeos/química , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Transcrição/química
6.
Oncogene ; 22(9): 1400-10, 2003 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-12618766

RESUMO

The mixed lineage leukemia gene (MLL, also known as HRX, ALL-1 and Htrx) located at 11q23 is involved in translocations with over 40 different chromosomal bands in a variety of leukemia subtypes. Here we report our analysis of a rare but recurring translocation, t(11;15)(q23;q14). This translocation has been described in a small subset of cases with both acute myeloblastic leukemia and ALL. Recent studies have shown that MLL is fused to AF15q14 in the t(11;15). Here we analyse a sample from another patient with this translocation and confirm the presence of an MLL-AF15q14 fusion. However, we have also identified and cloned another fusion transcript from the same patient sample. In this fusion transcript, MLL is fused to a novel gene, MLL partner containing FYVE domain (MPFYVE). Both MLL-AF15q14 and MLL-MPFYVE are in-frame fusion transcripts with the potential to code for novel fusion proteins. MPFYVE is also located on chromosome 15, approximately 170 kb telomeric to AF15q14. MPFYVE contains a highly conserved motif, the FYVE domain which, in other proteins, has been shown to bind to phosphotidyl-inositol-3 phosphate (PtdIns(3)P). The MLL-MPFYVE fusion may be functionally important in the leukemia process in at least some patients containing this translocation.


Assuntos
Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 15/genética , Leucemia Mieloide Aguda/genética , Proteínas de Fusão Oncogênica/genética , Proteínas/genética , Translocação Genética/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Sequência de Bases , Criança , Cromossomos Humanos Par 11/ultraestrutura , Cromossomos Humanos Par 15/ultraestrutura , DNA Complementar/genética , Etiquetas de Sequências Expressas , Humanos , Hibridização in Situ Fluorescente , Masculino , Dados de Sequência Molecular , Proteína de Leucina Linfoide-Mieloide , Proteínas Oncogênicas , Proteínas de Fusão Oncogênica/fisiologia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
7.
Blood ; 99(10): 3780-5, 2002 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11986236

RESUMO

The t(10;11)(p12;q23) chromosomal translocation in human acute myeloid leukemia results in the fusion of the MLL and AF10 genes. The latter codes for a novel leucine zipper protein, one of many MLL fusion partners of unknown function. In this report, we demonstrate that retroviral-mediated transduction of an MLL-AF10 complementary DNA into primary murine myeloid progenitors enhanced their clonogenic potential in serial replating assays and led to their efficient immortalization at a primitive stage of myeloid differentiation. Furthermore, MLL-AF10-transduced cells rapidly induced acute myeloid leukemia in syngeneic or severe combined immunodeficiency recipient mice. Structure/function analysis showed that a highly conserved 82-amino acid portion of AF10, comprising 2 adjacent alpha-helical domains, was sufficient for immortalizing activity when fused to MLL. Neither helical domain alone mediated immortalization, and deletion of the 29-amino acid leucine zipper within this region completely abrogated transforming activity. Similarly, the minimal oncogenic domain of AF10 exhibited transcriptional activation properties when fused to the MLL or GAL4 DNA-binding domains, while neither helical domain alone did. However, transcriptional activation per se was not sufficient because a second activation domain of AF10 was neither required nor competent for transformation. The requirement for alpha-helical transcriptional effector domains is similar to the oncogenic contributions of unrelated MLL partners ENL and ELL, suggesting a general mechanism of myeloid leukemogenesis by a subset of MLL fusion proteins, possibly through specific recruitment of the transcriptional machinery.


Assuntos
Transformação Celular Neoplásica , Leucemia Mieloide/etiologia , Células Progenitoras Mieloides/metabolismo , Proteínas de Fusão Oncogênica/fisiologia , Proto-Oncogenes , Fatores de Transcrição/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular , Células Cultivadas , Sequência Conservada , Proteínas de Ligação a DNA/genética , Histona-Lisina N-Metiltransferase , Zíper de Leucina , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Dados de Sequência Molecular , Proteína de Leucina Linfoide-Mieloide , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/genética , Estrutura Terciária de Proteína , Alinhamento de Sequência , Fatores de Transcrição/química , Ativação Transcricional , Transdução Genética
8.
Leukemia ; 15(11): 1743-9, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11681416

RESUMO

The MLL gene is frequently rearranged in leukemias, and MLL chimeric proteins generated by chromosomal translocations play crucial roles in leukemogenesis. Targets of murine Mll include HOX proteins that regulate body pattern formation and hematopoiesis. However, it is not known whether or not the MLL chimeric proteins regulate the HOX gene expression in human leukemia. To address this issue, THP-1 cells, a human leukemia cell line expressing MLL-AF9, were treated with antisense oligodeoxyribonucleotide (ODN) complementary to the coding sequence of the MLL-AF9 junction. Down-regulation of the MLL-AF9 transcript was accompanied by the reduced expression of the HOXA7 and -A10 genes, but not of the HOXA2, -A4, -A5, and -A9 genes. The number of viable cells cultured with 20 microM antisense ODN for 5 days was 10-fold lower than that of the sense ODN-treated control. And the number of the annexin V-/propidium iodide- apoptotic cells in the antisense ODN-treated cells after 3 days of culture was two-fold higher than that in the control. Staining of the antisense ODN-treated cells with Hoechst 33258 showed the morphology characteristic to apoptosis. These results indicate that MLL-AF9 regulates the expression of the selected HOX genes as well as prevents the leukemic cells from apoptosis.


Assuntos
Apoptose , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Leucemia/genética , Leucemia/patologia , Proteínas de Neoplasias , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Proteínas de Fusão Oncogênica/genética , Proto-Oncogenes , Fatores de Transcrição , Divisão Celular , Proteínas de Ligação a DNA/biossíntese , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Marcação de Genes , Células HL-60 , Histona-Lisina N-Metiltransferase , Proteínas Homeobox A10 , Proteínas de Homeodomínio/biossíntese , Humanos , Células K562 , Leucemia/metabolismo , Proteína de Leucina Linfoide-Mieloide , Proteínas Nucleares/genética , Proteínas de Fusão Oncogênica/fisiologia , RNA Neoplásico/biossíntese , Transcrição Gênica , Células Tumorais Cultivadas
9.
Blood ; 97(8): 2440-8, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11290609

RESUMO

The tyrosine kinase inhibitor STI571 inhibits BCR/ABL and induces hematologic remission in most patients with chronic myeloid leukemia. In addition to BCR/ABL, STI571 also inhibits v-Abl, TEL/ABL, the native platelet-derived growth factor (PDGF)beta receptor, and c-KIT, but it does not inhibit SRC family kinases, c-FMS, FLT3, the epidermal growth factor receptor, or multiple other tyrosine kinases. ARG is a widely expressed tyrosine kinase that shares substantial sequence identity with c-ABL in the kinase domain and cooperates with ABL to regulate neurulation in the developing mouse embryo. As described here, ARG has recently been implicated in the pathogenesis of leukemia as a fusion partner of TEL. A TEL/ARG fusion was constructed to determine whether ARG can be inhibited by STI571. When expressed in the factor-dependent murine hematopoietic cell line Ba/F3, the TEL/ARG protein was heavily phosphorylated on tyrosine, increased tyrosine phosphorylation of multiple cellular proteins, and induced factor-independent proliferation. The effects of STI571 on Ba/F3 cells transformed with BCR/ABL, TEL/ABL, TEL/PDGFbetaR, or TEL/ARG were then compared. STI571 inhibited tyrosine phosphorylation and cell growth of Ba/F3 cells expressing BCR/ABL, TEL/ABL, TEL/PDGFbetaR, and TEL/ARG with an IC(50) of approximately 0.5 microM in each case, but it had no effect on untransformed Ba/F3 cells growing in IL-3 or on Ba/F3 cells transformed by TEL/JAK2. Culture of TEL/ARG-transfected Ba/F3 cells with IL-3 completely prevented STI571-induced apoptosis in these cells, similar to what has been observed with BCR/ABL- or TEL/ABL-transformed cells. These results indicate that ARG is a target of the small molecule, tyrosine kinase inhibitor STI571.


Assuntos
Inibidores Enzimáticos/farmacologia , Piperazinas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Proteínas Recombinantes de Fusão , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Benzamidas , Sítios de Ligação , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/enzimologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , DNA Complementar/genética , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/fisiologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Mesilato de Imatinib , Camundongos , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Fosforilação/efeitos dos fármacos , Proteínas Quinases/química , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transfecção
10.
J Biol Chem ; 276(25): 22375-81, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11301322

RESUMO

The t(15;17) translocation, found in 95% of acute promyelocytic leukemia, encodes a promyelocytic leukemia (PML)-retinoic acid receptor alpha (RARalpha) fusion protein. Complete remission of acute promyelocytic leukemia can be obtained by treating patients with all-trans retinoic acid, and PML-RARalpha plays a major role in mediating retinoic acid effects in leukemia cells. A main model proposed for acute promyelocytic leukemia is that PML-RARalpha exerts its oncogenic effects by repressing the expression of retinoic acid-inducible genes critical to myeloid differentiation. By applying subtraction cloning to acute promyelocytic leukemia cells, we identified a retinoic acid-induced gene, PRAM-1 (PML-RARalpha target gene encoding an Adaptor Molecule-1), which encodes a novel adaptor protein sharing structural homologies with the SLAP-130/fyb adaptor. PRAM-1 is expressed and regulated during normal human myelopoiesis. In U937 myeloid precursor cells, PRAM-1 expression is inhibited by expression of PML-RARalpha in the absence of ligand and de novo superinduced by retinoic acid. PRAM-1 associates with other adaptors, SLP-76 and SKAP-55HOM, in myeloid cell lines and with protein tyrosine kinase lyn. By providing the first evidence that PML-RARalpha dysregulates expression of an adaptor protein, our data open new insights into signaling events that are disrupted during transformation by PML-RARalpha and induced by retinoic acid during de novo differentiation of acute promyelocytic leukemia cells.


Assuntos
Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Proteínas/metabolismo , Tretinoína/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Sequência de Bases , Diferenciação Celular , Clonagem Molecular , DNA Complementar , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Promielocítica Aguda/patologia , Dados de Sequência Molecular , Proteínas/química , Proteínas/genética , RNA Mensageiro/genética , Células Tumorais Cultivadas , Células U937
11.
Clin Cancer Res ; 6(9): 3522-9, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10999739

RESUMO

Desmoplastic small round cell tumor (DSRCT) is a primitive sarcoma with a consistent cytogenetic abnormality, t(11;22)(p13;q12). This chromosomal translocation generates a chimeric transcript that is formed by fusion of the 5' region of the Ewing's sarcoma gene, EWS, with the 3' DNA-binding segment of WT1, the Wilms' tumor suppressor gene. We collected 14 DSRCT tumor samples and examined the hybrid transcripts. We identified: (a) combinatorial heterogeneity of EWS exons fused to WT1 including use of EWS exons 7, 8, and 9; (b) subpopulations of variant transcripts in 6 of 14 tumors characterized by aberrant splicing resulting in loss of EWS exon 6 or WT1 exon 9; (c) multiple cDNA products with large internal deletions; and (d) insertion of small stretches of heterologous DNA at the fusion site or exon splice region in transcripts from two tumors. Most of the splice variants were in-frame, and in vitro translated fusion proteins with intact DNA-binding motifs formed complexes with a WT1 response element in gel mobility assays. Each of the chimeric proteins retains the ability to bind to the GC and TC elements of the early transcription factor EGR-1 as well as WT1 consensus sequences. We present evidence that various EWS-WT1 proteins up-regulated EGR-1 promoter activity and that this up-regulation is specifically dependent upon the absence of the exon 9 KTS domain of WT1. The molecular diversity and functionality exhibited by these fusion transcripts may have significant biological implications for their transactivating and tumorigenic potential.


Assuntos
Neoplasias Abdominais/genética , Carcinoma de Células Pequenas/genética , Proteínas Imediatamente Precoces , Proteínas de Fusão Oncogênica/fisiologia , Adolescente , Adulto , Sítios de Ligação , Criança , Proteínas Quimerinas/genética , DNA Complementar/genética , DNA Complementar/metabolismo , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Feminino , Genes do Tumor de Wilms/genética , Humanos , Masculino , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma de Ewing/genética , Fatores de Transcrição/genética , Ativação Transcricional/fisiologia , Proteínas WT1 , Dedos de Zinco/fisiologia
12.
Oncogene ; 18(29): 4275-81, 1999 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-10435641

RESUMO

TPR-MET, a transforming counterpart of the c-MET proto-oncogene detected in experimental and human cancer, results from fusion of the MET kinase domain with a dimerization motif encoded by TPR. In this rearrangement the exons encoding the Met extracellular, transmembrane and juxtamembrane domains are lost. The juxtamembrane domain has been suggested to be a regulatory region endowed with negative feedback control. To understand whether its absence is critical for the generation of the Tpr-Met transforming potential, we produced a chimeric molecule (Tpr-juxtaMet) with a conserved juxtamembrane domain. The presence of the domain (aa 962-1009) strongly inhibited Tpr-Met dependent cell transformation. Cell proliferation, anchorage-independent growth, motility and invasion were also impaired. The enzymatic behavior of Tpr-Met and Tpr-juxtaMet was the same, while Tpr-juxtaMet ability to associate cytoplasmic signal transducers and to elicit downstream signaling was severely impaired. These data indicate that the presence of the juxtamembrane domain counterbalances the Tpr-Met transforming potential and therefore the loss of the exon encoding the juxtamembrane domain is crucial in the generation of the active TPR-MET oncogene.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Transformação Celular Neoplásica/genética , Éxons/genética , Zíper de Leucina/fisiologia , Proteínas de Fusão Oncogênica/genética , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/fisiologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular Transformada , DNA Complementar/genética , Dimerização , Ativação Enzimática , Retroalimentação , Fibroblastos , Proteína Adaptadora GRB2 , Humanos , Zíper de Leucina/genética , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas/genética , Proteínas/metabolismo , Proto-Oncogene Mas , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência , Relação Estrutura-Atividade , Transfecção , Ensaio Tumoral de Célula-Tronco
13.
Leukemia ; 12(4): 563-70, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9557615

RESUMO

We have analyzed the differentiation program of growth factor-dependent TF-1 erythroleukemia cells as well as clones with inducible expression of the APL-specific PML/RARalpha protein. We have shown that TF-1 cells may be induced to megakaryocytic differentiation by phorbol ester (phorbol dibutyrate, PDB) addition, particularly when combined with thrombopoietin (Tpo). RT-PCR studies showed that Tpo induces Tpo receptor (TpoR or c-mpl), whose expression was further potentiated by PDB addition. When the cells are induced with both PDB and Tpo erythropoietin receptor (EpoR) expression was inhibited. In the absence of Zn2+-induced PML/RARalpha expression, PDB and Tpo induced megakaryocytic differentiation of TF-1 MTPR clones as observed in 'wild-type' TF-1 cells. Conversely, when PML/RARalpha expression was induced by Zn2+, PDB and Tpo treatment of these clones caused only a reduced level of megakaryocytic differentiation. These observations indicate that: (1) TF-1 cells as well as other erythroleukemic cells, possess the capacity to differentiate to megakaryocytic cells when grown in the presence of protein kinase (PKC) activators and more efficiently when combined with Tpo; (2) the PML/RARalpha gene has a wide capacity to interfere with the program of hematopoietic differentiation, including megakaryocytic differentiation. Finally, we also observed that PML/RARalpha expression in TF-1 cells induces an up-modulation of interleukin-3 receptor, c-kit and c-mpl, a phenomenon which may offer these cells a growth advantage.


Assuntos
Carcinógenos/farmacologia , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Dibutirato de 12,13-Forbol/farmacologia , Trombopoetina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , DNA Complementar/genética , DNA Complementar/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Megacariócitos/fisiologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/biossíntese , Proteínas de Fusão Oncogênica/genética , Receptores de Fatores de Crescimento/biossíntese , Receptores de Fatores de Crescimento/fisiologia , Células Tumorais Cultivadas
14.
Blood ; 87(2): 734-45, 1996 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-8555498

RESUMO

A novel human gene, LAF-4, was isolated from a subtracted cDNA library that showed strong sequence similarity to AF-4, a gene that is translocated in t(4;11)(q21;q23) acute lymphoblastic leukemias (ALLs). In t(4;11) ALL, the AF-4 gene at 4q21 is translocated into the MLL locus at 11q23, resulting in the expression of an MLL/AF-4 fusion protein that is the presumptive oncoprotein. AF-4 and LAF-4 are homologous throughout their coding regions, yet neither protein is related to previously cloned genes. Human LAF-4 readily hybridized with genes in mouse and chicken, thus showing that this gene family has been highly conserved during vertebrate evolution. In mouse tissues, LAF-4 mRNA was found to be present at highest levels in lymphoid tissues, present at lower levels in brain and lung, and absent from other tissues. In human and mouse lymphoid cell lines, LAF-4 expression was highest in pre-B cells, intermediate in mature B cells, and absent in plasma cells, thus pointing to a potential regulatory role for LAF-4 in lymphoid development. Antibodies to LAF-4 showed it to be a nuclear protein that showed an uneven, granular immunofluorescence pattern. In vitro-translated LAF-4 was able to bind strongly to double-stranded DNA cellulose. Furthermore, both LAF-4 and AF-4 had domains that activated transcription strongly when fused to the GAL4 DNA-binding domain. Interestingly, the AF-4 transactivation domain is retained in the MLL/AF-4 fusion protein; thus, it may contribute to the transforming potential of the oncoprotein. Therefore, the cloning of LAF-4 has defined a new family of potential regulatory proteins that may function in lymphoid development and oncogenesis.


Assuntos
Cromossomos Humanos Par 11/ultraestrutura , Cromossomos Humanos Par 4/ultraestrutura , Proteínas de Ligação a DNA/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/fisiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proto-Oncogenes , Fatores de Transcrição , Ativação Transcricional , Translocação Genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Galinhas/genética , DNA Complementar/genética , DNA de Neoplasias/genética , Proteínas de Ligação a DNA/genética , Drosophila melanogaster/genética , Histona-Lisina N-Metiltransferase , Humanos , Tecido Linfoide/metabolismo , Camundongos , Dados de Sequência Molecular , Proteína de Leucina Linfoide-Mieloide , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Especificidade de Órgãos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Frações Subcelulares/metabolismo , Fatores de Elongação da Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA