Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Leukemia ; 31(6): 1269-1277, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27840424

RESUMO

Chromosomal rearrangements of the mixed lineage leukemia (MLL/KMT2A) gene leading to oncogenic MLL-fusion proteins occur in ~10% of acute leukemias and are associated with poor clinical outcomes, emphasizing the need for new treatment modalities. Inhibition of the DOT1-like histone H3K79 methyltransferase (DOT1L) is a specific therapeutic approach for such leukemias that is currently being tested in clinical trials. However, in most MLL-rearranged leukemia models responses to DOT1L inhibitors are limited. Here, we performed deep-coverage short hairpin RNA sensitizer screens in DOT1L inhibitor-treated MLL-rearranged leukemia cell lines and discovered that targeting additional nodes of MLL complexes concomitantly with DOT1L inhibition bears great potential for superior therapeutic results. Most notably, combination of a DOT1L inhibitor with an inhibitor of the MLL-Menin interaction markedly enhanced induction of differentiation and cell killing in various MLL disease models including primary leukemia cells, while sparing normal hematopoiesis and leukemias without MLL rearrangements. Gene expression analysis on human and murine leukemic cells revealed that target genes of MLL-fusion proteins and MYC were suppressed more profoundly upon combination treatment. Our findings provide a strong rationale for a novel targeted combination therapy that is expected to improve therapeutic outcomes in patients with MLL-rearranged leukemia.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/farmacologia , Rearranjo Gênico , Histona-Lisina N-Metiltransferase/genética , Leucemia/tratamento farmacológico , Metiltransferases/metabolismo , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , RNA Interferente Pequeno/genética , Animais , Apoptose , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Regulação Leucêmica da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Leucemia/genética , Leucemia/patologia , Metiltransferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteína de Leucina Linfoide-Mieloide/metabolismo , Células Tumorais Cultivadas
2.
Leukemia ; 29(5): 1153-62, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25510432

RESUMO

While the nucleoporin 98-retinoic acid receptor gamma (NUP98-RARG) is the first RARG fusion protein found in acute leukemia, its roles and the molecular basis in oncogenic transformation are currently unknown. Here, we showed that homodimeric NUP98-RARG not only acquired unique nuclear localization pattern and ability of recruiting both RXRA and wild-type NUP98, but also exhibited similar transcriptional properties as RARA fusions found in acute promyelocytic leukemia (APL). Using murine bone marrow retroviral transduction/transformation assay, we further demonstrated that NUP98-RARG fusion protein had gained transformation ability of primary hematopoietic stem/progenitor cells, which was critically dependent on the C-terminal GLFG domain of NUP98 and the DNA binding domain (DBD) of RARG. In contrast to other NUP98 fusions, cells transformed by the NUP98-RARG fusion were extremely sensitive to all-trans retinoic acid (ATRA) treatment. Interestingly, while pan-RXR agonists, SR11237 and LGD1069 could specifically inhibit NUP98-RARG transformed cells, mutation of the RXR interaction domain in NUP98-RARG had little effect on its transformation, revealing that therapeutic functions of rexinoid can be independent of the direct biochemical interaction between RXR and the fusion. Together, these results indicate that deregulation of the retinoid/rexinoid signaling pathway has a major role and may represent a potential therapeutic target for NUP98-RARG-mediated transformation.


Assuntos
Regulação Leucêmica da Expressão Gênica , Leucemia/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Animais , Núcleo Celular/metabolismo , Transformação Celular Neoplásica , Células HEK293 , Células HeLa , Humanos , Camundongos , Microscopia de Fluorescência , Ligação Proteica , Mapeamento de Interação de Proteínas , Multimerização Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Transdução de Sinais , Tretinoína/química , Receptor gama de Ácido Retinoico
3.
Leukemia ; 28(3): 566-76, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23817178

RESUMO

The CXCR4 receptor is a major regulator of hematopoietic cell migration. Overexpression of CXCR4 has been associated with poor prognosis in acute myelogenous leukemia (AML). We have previously shown that ligand-mediated phosphorylation of the Serine339 (CXCR4-S339) residue of the intracellular domain by PIM1 is implicated in surface re-expression of this receptor. Here, we report that phosphorylation of CXCR4-S339 in bone marrow (BM) biopsies correlated with poor prognosis in a cohort of AML patients. To functionally address the impact of CXCR4-S339 phosphorylation, we generated cell lines-expressing CXCR4 mutants that mimic constitutive phosphorylation (S339E) or abrogate phosphorylation (S339A). Whereas the expression of CXCR4 significantly increased, both CXCR4-S339E and the CXCR4-S339A mutants significantly reduced the BM homing and engraftment of Kasumi-1 AML cells in immunodeficient mice. In contrast, only expression of the CXCR4-S339E mutant increased the BM retention of the cells and resistance to cytarabine treatment, and impaired detachment capacity and AMD3100-induced mobilization of engrafted leukemic cells. These observations suggest that the poor prognosis in AML patients displaying CXCR4-S339 phosphorylation can be the consequence of an increased retention to the BM associated with an enhanced chemoresistance of leukemic cells. Therefore, CXCR4-S339 phosphorylation could serve as a novel prognostic marker in human AML.


Assuntos
Biomarcadores/metabolismo , Adesão Celular/fisiologia , Leucemia Mieloide Aguda/patologia , Receptores CXCR4/fisiologia , Serina/química , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Receptores CXCR4/química , Receptores CXCR4/genética
4.
Leukemia ; 27(6): 1245-53, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23318960

RESUMO

The lens epithelium-derived growth factor (LEDGF/p75) tethers the mixed-lineage leukemia (MLL1) protein complex to chromatin. Likewise, LEDGF/p75 tethers the HIV-1 pre-integration complex to chromatin. We previously demonstrated that expression of the C-terminal fragment fused to enhanced green fluorescent protein (eGFP) (eGFP-LEDGF(325-530)) impaired HIV-1 replication. Here, we explored this strategy to selectively interfere with the leukemogenic activity of MLL-fusion proteins. We found that expression of LEDGF(325-530) impaired the clonogenic growth of MLL-fusion gene transformed human and mouse hematopoietic cells, without affecting the growth of control cells immortalized by the FLT3-ITD mutant or normal lineage-marker-depleted murine bone marrow cells. Expression of LEDGF(325-530) was associated with downregulation of the MLL target Hoxa9 and impaired cell cycle progression. Structure-function analysis revealed two small eGFP-fused LEDGF/p75 peptides, LEDGF(424-435) and LEDGF(375-386) phenocopying these effects. Both LEDGF(325-530) and the smaller active peptides were able to disrupt the LEDGF/p75-MLL interaction. Expression of LEDGF(325-530) or LEDGF(375-386) fragments increased the latency period to disease development in vivo in a mouse bone marrow transplant model of MLL-AF9-induced AML. We conclude that small peptides disrupting the LEDGF/p75-MLL interface have selective anti-leukemic activity providing a direct rationale for the design of small molecule inhibitors targeting this interaction.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas Recombinantes de Fusão/genética , Animais , Transformação Celular Neoplásica , Histona-Lisina N-Metiltransferase/metabolismo , Leucemia Experimental/genética , Camundongos , Proteína de Leucina Linfoide-Mieloide/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Br J Cancer ; 107(3): 491-500, 2012 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-22722314

RESUMO

BACKGROUND: PIM serine/threonine kinases are often highly expressed in haematological malignancies. We have shown that PIM inhibitors reduced the survival and migration of leukaemic cells. Here, we investigated PIM kinases in diffuse large B-cell lymphoma (DLBCL) biopsy samples and DLBCL cell lines. METHODS: Immunohistochemical staining for PIM kinases and CXCR4 was performed on tissue microarrays from a cohort of 101 DLBCL cases, and the effects of PIM inhibitors on the survival and migration of DLBCL cell lines were determined. RESULTS: PIM1 expression significantly correlated with the activation of signal transducer and activator of transcription (STAT) 3 and 5, P-glycoprotein expression, CXCR4-S339 phosphorylation, and cell proliferation. Whereas most cases exhibited cytoplasmic or cytoplasmic and nuclear PIM1 and PIM2 expression, 12 cases (10 of the non-germinal centre DLBCL type) expressed PIM1 predominately in the nucleus. Interestingly, nuclear expression of PIM1 significantly correlated with disease stage. Exposure of DLBCL cell lines to PIM inhibitors modestly impaired cellular proliferation and CXCR4-mediated migration. CONCLUSION: This work demonstrates that PIM expression in DLBCL is associated with activation of the JAK/STAT signalling pathway and with the proliferative activity. The correlation of nuclear PIM1 expression with disease stage and the modest response to small-molecule inhibitors suggests that PIM kinases are progression markers rather than primary therapeutic targets in DLBCL.


Assuntos
Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/enzimologia , Linfoma Difuso de Grandes Células B/genética , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Estudos de Coortes , Citoplasma/efeitos dos fármacos , Citoplasma/genética , Citoplasma/metabolismo , Progressão da Doença , Feminino , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , Linfoma Difuso de Grandes Células B/metabolismo , Masculino , Terapia de Alvo Molecular , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Taxa de Sobrevida
6.
Leukemia ; 24(3): 601-12, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20072157

RESUMO

Retroviral expression of leukemogenic oncogenes in the murine hematopoietic system is essential but not sufficient to induce acute leukemia. Proviral integration-mediated elevated expression of the meningioma 1 (MN1) oncogene suggested MN1 acting as cooperating event in mixed-lineage leukemia 1 (MLL) and eleven nineteen leukemia (ENL)-induced murine leukemia. Indeed, co-expression of MN1 with MLL-ENL enhanced transformation in vivo, and resulted in a significantly reduced latency for induction of an aggressive acute leukemia when compared with MN1 or MLL-ENL alone. In addition, co-expression of MN1 increased the granulocyte macrophage progenitor cell population with leukemia-initiating properties as shown in secondary transplantation experiments. Gene expression profiling experiments identified putative downstream MN1 targets, of which FMS-like tyrosine kinase 3 (FLT3) and CD34 were upregulated in both MN1-overexpressing murine leukemias and in pediatric acute leukemias with high MN1 levels. Interestingly, small interfering RNA (siRNA)-mediated MN1 knockdown resulted in cell cycle arrest and impaired clonogenic growth of human leukemia cell lines with high MN1 levels. Our work shows for the first time that high MN1 levels are important for the growth of leukemic cells, and that increased MN1 expression can synergize with MLL-ENL and probably other transforming fusion genes in leukemia induction through a distinct gene expression program that is able to expand the leukemia-initiating cell population.


Assuntos
Leucemia/genética , Oncogenes , Proteínas Supressoras de Tumor/fisiologia , Doença Aguda , Animais , Linhagem Celular Tumoral , Humanos , Leucemia/etiologia , Camundongos , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Transativadores , Proteínas Supressoras de Tumor/genética
8.
J Leukoc Biol ; 80(4): 697-704, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16793914

RESUMO

Abundant mRNA expression for a proliferation-inducing ligand (APRIL) from tumor necrosis factor (TNF) family is observed in many solid tumors. Here, we analyzed in situ the cellular source of APRIL in human solid tumors with anti-APRIL antibodies. In most cases, neutrophils present in the tumor stroma constituted the main source of APRIL. In cutaneous lesions such as melanoma or basal cell carcinoma, tumor-adjacent keratinocytes also produced APRIL. APRIL production by tumor cells themselves was a rare event, only observed in urothelial bladder cancer and squamous cell carcinoma. Detailed analysis revealed that APRIL dissociated from producing cells, and secreted APRIL was retained in the tumor lesions. A direct binding onto tumor cells via heparan sulfate proteoglycans (HSPG) was observed in in vitro experiments and confirmed in situ. Taken together, our analysis indicates a potential role for HSPG/APRIL interactions in the development of solid tumors.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/biossíntese , Regulação para Cima , Linhagem Celular Tumoral , Granulócitos/metabolismo , Granulócitos/patologia , Células HeLa , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Imuno-Histoquímica , Queratinócitos/metabolismo , Queratinócitos/patologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
9.
Eur J Haematol ; 71(4): 243-9, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12950232

RESUMO

The anaplastic large cell lymphoma kinase (ALK)-positive anaplastic large cell lymphoma (ALCL) is a rare type of non-Hodgkin lymphoma which occurs in children mostly. The ALK protein is highly immunogenic and elicits both humoral and cellular immune responses. A 15-yr-old child presented with fever and adenopathy and did not respond to antibiotics. Biopsy of the enlarged lymph node contained almost no lymphoid element except for a few CD8-positive T cells, plasma cells and isolated CD30-positive blasts. The patient's condition improved following lymphadenectomy but relapse occurred 3 months later with multiple nodes, high fever and an abdominal mass. This time an ALK-positive ALCL was diagnosed and the retrospective analysis of the initial biopsy revealed rare, isolated ALK+ cells. Molecular analysis showed T-cell clones and oligoclonal B cells in both biopsies and peripheral blood of the patient. The tumour cells harbour a t(2;5) translocation, revealing a null phenotype by immunohistochemistry and no evidence for T-cell clonality by Southern blotting. The patient's serum contained anti-ALK antibodies. Our findings suggest that the T-cell clones and anti-ALK antibodies in this patient constitute an anti-tumour response that caused the hypocellularity of the initial lymph node. Hypocellular and oedematous lymph nodes occurring in a child with evocative symptoms should be tested for the presence of ALK.


Assuntos
Linfócitos B/imunologia , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Linfócitos T/imunologia , Adolescente , Linfócitos B/metabolismo , Southern Blotting , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Inflamação , Antígeno Ki-1/biossíntese , Linfonodos/citologia , Linfonodos/patologia , Metástase Linfática , Linfoma não Hodgkin/genética , Modelos Genéticos , RNA Mensageiro/metabolismo , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Translocação Genética
10.
Pathobiology ; 69(2): 59-66, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11752899

RESUMO

OBJECTIVE: TCL1, MTCP1 and TCL1b are three members of a new family of oncogenes that are expressed in T cell leukemias of ataxia telangiectasia patients (T-PLL, T-CLL). TCL1 is located at 14q32.1 and activated by juxtaposition to the alpha/delta-locus at 14q11 or beta-locus at 7q35 of the T cell receptor during the reciprocal translocations t(14;14)(q11;q32), t(7;14)(q35;q32), or inversion inv(14)(q11;q32). TCL1 encodes a predominantly cytoplasmic protein of 114 aa (14 kD) of unknown function. Recent studies suggest that TCL1 promotes cell survival rather than stimulating cell proliferation, as previously proposed. METHODS: In an attempt to clarify the contexts in which TCL1 is expressed, we investigated TCL1 expression in 114 lymphoma and leukemia patients by Northern blot, RT-PCR and immunohistochemistry. RESULTS: TCL1 expression is restricted to lymphoid cells, and is found in neoplastic (T and B cell neoplasms, and Hodgkin's disease) and nonneoplastic proliferations (reactive lesions). Out of 114 cases, 18 neoplasms of myeloid and 4 cases of epithelial origin were TCL1-negative. In lesions of the lymphoid system, both low- and high-grade lymphomas were found to express TCL1. CONCLUSIONS: We propose that TCL1 expression especially in high-grade B cell non-Hodgkin's lymphomas might interfere with B cell differentiation and promote the transition from low- to high-grade lymphoma.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Leucemia/metabolismo , Linfoma/metabolismo , Proteínas Proto-Oncogênicas , Fatores de Transcrição/biossíntese , Northern Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Proteínas de Ligação a DNA/sangue , Humanos , Imuno-Histoquímica , Leucemia/sangue , Leucemia/patologia , Linfoma/sangue , Linfoma/patologia , RNA Neoplásico/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/sangue , Células Tumorais Cultivadas
11.
Cancer Genet Cytogenet ; 130(2): 93-104, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11675129

RESUMO

ETV6/CBFA2 (TEL/AML1) is the most frequent genetic abnormality associated with acute lymphoblastic leukemias in children, and is associated with a favorable prognosis. To investigate the influence of ETV6/CBFA2 on cellular transformation, the fusion gene was cloned into a murine ecotropic retroviral vector and transduced into IL-3-dependent Ba/F3 and 32Dcl.3 and IL-7-dependent IxN/2b murine hematopoietic cell lines. Different variants of ETV6/CBFA2, corresponding to CBFA2 alternatively spliced variants, and the reciprocal product CBFA2/ETV6, were stably expressed in each of these cell lines. However, although Western blot analysis demonstrated expression of each variant, none of the stable cell lines expressing CBFA2/ETV6 or the variants conferred factor-independent growth. We further investigated the effect of ETV6/CBFA2 expression in vivo by generating transgenic mice in which expression of the fusion was directed to lymphoid cells using the immunoglobulin heavy chain enhancer/promoter. Four founder mice were identified showing transmission and expression of the chimeric product. The mice were bred for five generations and followed for more than 24 months. The mice did not develop a malignant hematologic disorder, nor did they display histopathologic, morphologic, or immunophenotypic abnormalities, although ETV6/CBFA2 expression was confirmed in each line. We conclude that the expression of ETV6/CBFA2 alone is not sufficient for induction of growth factor independence in hematopoietic cell lines or hematologic disease in transgenic mice.


Assuntos
Transformação Celular Neoplásica , Neoplasias Hematológicas/etiologia , Neoplasias Hematológicas/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Fusão Oncogênica/genética , Animais , Western Blotting , Diferenciação Celular , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core , Eletroporação , Elementos Facilitadores Genéticos , Citometria de Fluxo , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Leucemia/etiologia , Leucemia/genética , Camundongos , Camundongos Transgênicos , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Ativação Transcricional , Transdução Genética
12.
Blood ; 97(12): 3910-8, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11389034

RESUMO

The molecular cloning of the t(5;10)(q33;q22) associated with atypical chronic myeloid leukemia (CML) is reported. Fluorescence in situ hybridization (FISH), Southern blot, and reverse transcriptase- polymerase chain reaction analysis demonstrated that the translocation resulted in an H4/platelet-derived growth factor receptor betaR (PDGFbetaR) fusion transcript that incorporated 5' sequences from H4 fused in frame to 3' PDGFbetaR sequences encoding the transmembrane, WW-like, and tyrosine kinase domains. FISH combined with immunophenotype analysis showed that t(5;10)(q33;q22) was present in CD13(+) and CD14(+) cells but was not observed in CD3(+) or CD19(+) cells. H4 has previously been implicated in pathogenesis of papillary thyroid carcinoma as a fusion partner of RET. The H4/RET fusion incorporates 101 amino acids of H4, predicted to encode a leucine zipper dimerization domain, whereas the H4/PDGFbetaR fusion incorporated an additional 267 amino acids of H4. Retroviral transduction of H4/PDGFbetaR, but not a kinase-inactive mutant, conferred factor-independent growth to Ba/F3 cells and caused a T-cell lymphoblastic lymphoma in a murine bone marrow transplantation assay of transformation. Mutational analysis showed that the amino-terminal H4 leucine zipper domain (amino acids 55-93), as well as H4 amino acids 101 to 386, was required for efficient induction of factor-independent growth of Ba/F3 cells. Tryptophan-to-alanine substitutions in the PDGFbetaR WW-like domain at positions 566/593, or tyrosine-to-phenylalanine substitutions at PDGFbetaR positions 579/581 impaired factor-independent growth of Ba/F3 cells. H4/PDGFbetaR is an oncoprotein expressed in t(5;10)(q33;q22) atypical CML and requires dimerization motifs in the H4 moiety, as well as residues implicated in signal transduction by PDGFbetaR, for efficient induction of factor-independent growth of Ba/F3 cells. (Blood. 2001;97:3910-3918)


Assuntos
Carcinoma Papilar/genética , Cromossomos Humanos Par 10 , Cromossomos Humanos Par 5 , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Proteínas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Neoplasias da Glândula Tireoide/genética , Translocação Genética , Transformação Celular Neoplásica/genética , Cromossomos Humanos Par 10/genética , Cromossomos Humanos Par 5/genética , Clonagem Molecular , Análise Citogenética , Proteínas do Citoesqueleto , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , Rearranjo Gênico , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/etiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Mutagênese , Células Mieloides/metabolismo , Células Mieloides/patologia , Proteínas de Fusão Oncogênica , Estrutura Terciária de Proteína , Proteínas/metabolismo , Transfecção
13.
Mol Cell Biol ; 21(10): 3547-57, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11313480

RESUMO

TEL-JAK2 fusion proteins, which are a result of t(9;12)(p24;p13) translocations associated with human leukemia, activate Stat5 in vitro and in vivo and cause a myelo- and lymphoproliferative disease in a murine bone marrow transplant model. We report that Socs-1, a member of the SOCS family of endogenous inhibitors of JAKs and STATs, inhibits transformation of Ba/F3 cells by TEL-JAK2 but has no effect on Ba/F3 cells transformed by BCR-ABL, TEL-ABL, or TEL-platelet-derived growth factor receptor beta. TEL-JAK2, in addition to activating Stat5, associates with Shc and Grb2 and induces activation of Erk2, and expression of Socs-1 inhibits engagement of each of these signaling molecules. TEL-JAK2 kinase activity is inhibited by Socs-1, as assessed by in vitro kinase assays. In addition, Socs-1 induces proteasomal degradation of TEL-JAK2. Mutational analysis indicates that the SOCS box of Socs-1 is required for proteasomal degradation and for abrogation of growth of TEL-JAK2-transformed cells. Furthermore, murine bone marrow transplant assays demonstrate that expression of Socs-1 prolongs latency of TEL-JAK2-mediated disease in vivo. Collectively, these data indicate that Socs-1 inhibits TEL-JAK2 in vitro and in vivo through inhibition of kinase activity and induction of TEL-JAK2 protein degradation.


Assuntos
Proteínas de Transporte/fisiologia , Cisteína Endopeptidases/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Complexos Multienzimáticos/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas , Proteínas Repressoras , Animais , Linhagem Celular , Transformação Celular Neoplásica , Ativação Enzimática , Células-Tronco Hematopoéticas/patologia , Janus Quinase 2 , Camundongos , Complexo de Endopeptidases do Proteassoma , Transdução de Sinais/fisiologia , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina
14.
Mol Cell ; 6(3): 693-704, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11030348

RESUMO

STAT5 is activated in a broad spectrum of human hematologic malignancies. We addressed whether STAT5 activation is necessary for the myelo- and lymphoproliferative disease induced by TEL/JAK2 using a genetic approach. Whereas mice transplanted with bone marrow transduced with retrovirus expressing TEL/JAK2 develop a rapidly fatal myelo- and lymphoproliferative syndrome, reconstitution with bone marrow derived from Stat5ab-deficient mice expressing TEL/JAK2 did not induce disease. Disease induction in the Stat5a/b-deficient background was rescued with a bicistronic retrovirus encoding TEL/JAK2 and Stat5a. Furthermore, myeloproliferative disease was induced by reconstitution with bone marrow cells expressing a constitutively active mutant, Stat5a, or a single Stat5a target, murine oncostatin M (mOSM). These data define a critical role for Stat5a/b and mOSM in the pathogenesis of TEL/JAK2 disease.


Assuntos
Proteínas de Ligação a DNA/genética , Transtornos Linfoproliferativos/fisiopatologia , Proteínas do Leite , Transtornos Mieloproliferativos/fisiopatologia , Proteínas de Fusão Oncogênica/genética , Transativadores/genética , Animais , Southern Blotting , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Ensaio de Unidades Formadoras de Colônias , DNA de Neoplasias/análise , Fibrose , Citometria de Fluxo , Técnicas de Transferência de Genes , Transtornos Linfoproliferativos/genética , Camundongos , Camundongos Mutantes , Mutagênese/fisiologia , Transtornos Mieloproliferativos/genética , Transplante de Neoplasias , Oncostatina M , Peptídeos/genética , Fenótipo , Retroviridae/genética , Fator de Transcrição STAT5
15.
Leuk Lymphoma ; 37(5-6): 639-48, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-11042528

RESUMO

Chronic B-cell lymphocytic leukaemia (CLL) and low-grade B-cell Non Hodgkin's lymphomas (Lg-NHL) are characterized by slow accumulation of neoplastic cells arrested in the G0/G1 phase of the cell cycle. In contrast, proliferation rates are high in aggressive B-cell lymphomas (Hg-NHL). Divergent expression of cyclin-dependent kinase inhibitors (CKI) in the cell cycle may contribute to these differences. We analysed CLL as well as low and high grade B-cell NHL for expression of G1-specific and universal CKI by competitive RT-PCR and immunostaining. p16(INK4A) expression was low in all types of neoplasms. Highest p14(ARF) /p16 beta expression levels were found in normal lymphocytes. Expression of this CKI was significantly lower in CLL, but still higher in CLL than in the lymphomas (median 27 vs. 3 mRNA transcripts x 10(3), p = 0.0001). p14(ARF) /p16 beta immunostaining correlated with mRNA expression. Highest p21 mRNA levels were found in CLL, but three of four CLL with abundant p21 mRNA production were negative on immunostaining. High grade lymphomas showed markedly decreased p21 expression (3.9 in Hg-NHL vs. 12 in Lg-NHL and 29 in CLL; values expressed as mRNA transcripts x 10(3), p < 0.009). mRNA and protein expression of p27 was considerably higher in CLL than in the lymphomas. Differential CKI expression in various B-cell neoplasias may provide important biological markers, if not the molecular underpinning of their different cell cycle kinetics. Targeted interference with such genes governing cell cycle control in lymphoid neoplasia may pave the way towards new treatment strategies.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Regulação Neoplásica da Expressão Gênica , Genes p16 , Leucemia Linfocítica Crônica de Células B/genética , Linfoma não Hodgkin/genética , Proteínas de Neoplasias/biossíntese , Proteína Supressora de Tumor p14ARF/biossíntese , Proteínas Supressoras de Tumor/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Ciclo Celular/genética , Divisão Celular , Inibidor de Quinase Dependente de Ciclina p15 , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/biossíntese , Ciclinas/genética , Feminino , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia Linfocítica Crônica de Células B/metabolismo , Linfoma não Hodgkin/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Supressoras de Tumor/genética
16.
Pediatr Res ; 48(3): 299-303, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10960493

RESUMO

IL-8, a chemokine with striking neutrophil-activating properties, is important in the pathogenesis of various disorders of the adult lung. Little is known about its production and possible role in fetal and neonatal lung disorders. We therefore examined IL-8 expression by immunohistochemistry in lung tissue from neonates with hyaline membrane disease, from fetuses with amniotic infection, and from a fetal control group with noninflammatory diseases. In the majority of cases with hyaline membrane disease, intense IL-8 immunoreaction was seen in fetal and neonatal neutrophils and in almost half of these cases, in epithelial cells of the terminal airways as well as in the connective tissue cell compartment. In contrast, in the amniotic infection group, strong IL-8 immunostaining was almost exclusively seen in maternal aspirated neutrophils. Little or no IL-8 signal was seen in the control cases in all cell types examined. Also, no IL-8 production by fetal lung cells was detected in fetuses <18 wk of gestation. The marked presence of IL-8 in all cell types of the lung in hyaline membrane disease cases indicates a role for IL-8 in the pathobiology of hyaline membrane disease possibly similar to that in adult respiratory distress syndrome. It further suggests that the cytokine network of the fetal lung is already well developed by the second trimester of pregnancy.


Assuntos
Âmnio/imunologia , Doença da Membrana Hialina/imunologia , Interleucina-8/imunologia , Pulmão/imunologia , Adulto , Âmnio/microbiologia , Âmnio/patologia , Infecções Bacterianas/imunologia , Infecções Bacterianas/patologia , Feminino , Humanos , Doença da Membrana Hialina/patologia , Imuno-Histoquímica , Recém-Nascido , Inflamação/imunologia , Inflamação/patologia , Pulmão/patologia , Gravidez
17.
EMBO J ; 19(8): 1827-38, 2000 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-10775267

RESUMO

The TEL-TRKC fusion is expressed as a consequence of t(12;15)(p13;q25), and is associated with two human cancers: congenital fibrosarcoma and acute myelogenous leukemia (AML). We report that the T/T(F) and T/T(L) fusion variants associated with congenital fibrosarcoma and AML, respectively, are constitutively tyrosine phosphorylated, and confer factor-independent growth to the murine hematopoietic cell line Ba/F3. Retroviral transduction of T/T(L) causes a rapidly fatal myeloproliferative disease in a murine bone marrow transplant (BMT) model, whereas T/T(F) causes a long-latency, pre-B-cell lymphoblastic lymphoma. TEL-TRKC variants are potent activators of the MAP kinase pathway, but neither variant activates Stat5 or other Stat family members. T/T(L), but not T/T(F), induces tyrosine phosphorylation of phospholipase Cgamma (PLCgamma), phosphoinositol-3 kinase and SHC. However, mutation analysis demonstrates that PLCgamma tyrosine phos phorylation by T/T(L) is dispensable for induction of the myeloproliferative phenotype by T/T(L). Collectively, these data demonstrate that the TEL-TRKC fusion variants are oncoproteins that activate the MAP kinase pathway, and do not require activation of either PLCgamma or Stat5 for efficient induction of a myeloproliferative phenotype in the murine BMT model.


Assuntos
Cromossomos Humanos Par 12 , Cromossomos Humanos Par 15 , Proteínas de Ligação a DNA/metabolismo , Fibrossarcoma/genética , Leucemia Mieloide Aguda/genética , Proteínas do Leite , Proteínas de Fusão Oncogênica/metabolismo , Receptor trkC/metabolismo , Proteínas Repressoras , Fatores de Transcrição/metabolismo , Translocação Genética , Células 3T3 , Animais , Medula Óssea/metabolismo , Transformação Celular Neoplásica , Fibrossarcoma/congênito , Células-Tronco Hematopoéticas/metabolismo , Humanos , Isoenzimas/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama , Fosforilação , Proteínas Proto-Oncogênicas c-ets , Fator de Transcrição STAT5 , Transdução de Sinais , Distribuição Tecidual , Transativadores/metabolismo , Células Tumorais Cultivadas , Fosfolipases Tipo C/metabolismo , Tirosina/metabolismo , Variante 6 da Proteína do Fator de Translocação ETS
18.
Blood ; 93(5): 1707-14, 1999 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10029600

RESUMO

The TEL/PDGFbetaR fusion protein is expressed as the consequence of a recurring t(5;12) translocation associated with chronic myelomonocytic leukemia (CMML). Unlike other activated protein tyrosine kinases associated with hematopoietic malignancies, TEL/PDGFbetaR is invariably associated with a myeloid leukemia phenotype in humans. To test the transforming properties of TEL/PDGFbetaR in vivo, and to analyze the basis for myeloid lineage specificity in humans, we constructed transgenic mice with TEL/PDGFbetaR expression driven by a lymphoid-specific immunoglobulin enhancer-promoter cassette. These mice developed lymphoblastic lymphomas of both T and B lineage, demonstrating that TEL/PDGFbetaR is a transforming protein in vivo, and that the transforming ability of this fusion is not inherently restricted to the myeloid lineage. Treatment of TEL/PDGFbetaR transgenic animals with a protein tyrosine kinase inhibitor with in vitro activity against PDGFbetaR (CGP57148) resulted in suppression of disease and a prolongation of survival. A therapeutic benefit was apparent both in animals treated before the development of overt clonal disease and in animals transplanted with clonal tumor cells. These results suggest that small-molecule tyrosine kinase inhibitors may be effective treatment for activated tyrosine kinase-mediated malignancies both early in the course of disease and after the development of additional transforming mutations.


Assuntos
Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/uso terapêutico , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Repressoras , Fatores de Transcrição/genética , Translocação Genética , Animais , Antineoplásicos/uso terapêutico , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-ets , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Variante 6 da Proteína do Fator de Translocação ETS
19.
EMBO J ; 17(18): 5321-33, 1998 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9736611

RESUMO

Recent reports have demonstrated fusion of the TEL gene on 12p13 to the JAK2 gene on 9p24 in human leukemias. Three variants have been identified that fuse the TEL pointed (PNT) domain to (i) the JAK2 JH1-kinase domain, (ii) part of and (iii) all of the JH2 pseudokinase domain. We report that all of the human TEL/JAK2 variants, and a human/mouse chimeric hTEL/mJAK2(JH1) fusion gene, transform the interleukin-3 (IL-3)-dependent murine hematopoietic cell line Ba/F3 to IL-3-independent growth. Transformation requires both the TEL PNT domain and JAK2 kinase activity. Furthermore, all TEL/JAK2 variants strongly activated STAT 5 by phosphotyrosine Western blots and by electrophoretic mobility shift assays (EMSA). Mice (n = 40) transplanted with bone marrow infected with the MSCV retrovirus containing either the hTEL/mJAK2(JH1) fusion or its human counterpart developed a fatal mixed myeloproliferative and T-cell lymphoproliferative disorder with a latency of 2-10 weeks. In contrast, mice transplanted with a TEL/JAK2 mutant lacking the TEL PNT domain (n = 10) or a kinase-inactive TEL/JAK2(JH1) mutant (n = 10) did not develop the disease. We conclude that all human TEL/JAK2 fusion variants are oncoproteins in vitro that strongly activate STAT 5, and cause lethal myelo- and lymphoproliferative syndromes in murine bone marrow transplant models of leukemia.


Assuntos
Células-Tronco Hematopoéticas , Transtornos Linfoproliferativos/genética , Proteínas do Leite , Transtornos Mieloproliferativos/genética , Proteínas de Fusão Oncogênica/genética , Animais , Transplante de Medula Óssea , Divisão Celular , Linhagem Celular Transformada , DNA/metabolismo , DNA Recombinante , DNA Viral/análise , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/citologia , Humanos , Interleucina-3/fisiologia , Camundongos , Camundongos Transgênicos , Transtornos Mieloproliferativos/patologia , Proteínas de Fusão Oncogênica/fisiologia , Retroviridae/genética , Fator de Transcrição STAT5 , Transativadores/metabolismo , Transformação Genética , Integração Viral
20.
Leukemia ; 11(1): 54-63, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9001419

RESUMO

In hematological malignancies, structural alterations of genes for G1-specific cyclin-dependent kinases inhibitors (CKIs) have been extensively investigated. G1-CKIs might play an important role not only as tumor suppressor genes but also in cellular differentiation. We examined constitutive and differentiation-induced expression and regulation of the four members of the G1-CKI family p16INK4A, p15INK4B, p18INK4C and p19INK4D in acute myeloid leukemia as well as their expression in normal granulocytes and monocytes. p18INK4C and p19INK4D mRNA were expressed constitutively at high levels in seven myeloid cell lines and 16 AML patient samples, whereas expression of p15INK4B mRNA was very low and only detectable by nested RT-PCR analysis. During phorbol ester-induced monocytic differentiation of leukemic HL-60 cells expression of particular G1-CKIs was disparately regulated. This process was associated with growth arrest of the majority of the cells (> or = 80%) in G1/G0, and in parallel p15INK4B were upregulated whereas p18INK4C and p19INK4D expression was downregulated. In contrast, granulocytic differentiation induced by DMSO was accompanied by an increase of p18INK4C and p19INK4D expression only. PMA treatment of blast cells from two AML patients confirmed these cell line results. Disparate regulation of p15INK4B and p18INK4C mRNA was dependent on intermediary protein synthesis and occurred at the post-transcriptional level as shown by nuclear run-on analysis and mRNA half-life studies. In normal granulocytes and monocytes low constitutive p15INK4B and p18INK4C mRNA expression was detectable by RT-PCR only, but p19INK4D transcripts were noted by Northern blotting in both cell types. Disparate expression of G1-specific cell cycle inhibitors indicates complex and divergent roles of particular CKIs during normal and leukemic myeloid hematopoiesis.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Inibidores Enzimáticos , Leucemia Mieloide/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Supressoras de Tumor , Doença Aguda , Proteínas de Transporte/genética , Diferenciação Celular , Inibidor de Quinase Dependente de Ciclina p15 , Inibidor p16 de Quinase Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p18 , Inibidor de Quinase Dependente de Ciclina p19 , Fase G1 , Regulação Leucêmica da Expressão Gênica , Granulócitos/metabolismo , Células HL-60/metabolismo , Humanos , Leucemia Mieloide/genética , Monócitos/metabolismo , Proteínas de Neoplasias/genética , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...