Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 124(12): 5263-74, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25384216

RESUMO

Multiple myeloma (MM) is a plasma cell neoplasm that results from clonal expansion of an Ig-secreting terminally differentiated B cell. Advanced MM is characterized by tissue damage that involves bone, kidney, and other organs and is typically associated with recurrent genetic abnormalities. IL-6 signaling via the IL-6 signal transducer GP130 has been implicated as an important driver of MM pathogenesis. Here, we demonstrated that ectopic expression of constitutively active GP130 (L-GP130) in a murine retroviral transduction-transplantation model induces rapid MM development of high penetrance. L-GP130-expressing mice recapitulated all of the characteristics of human disease, including monoclonal gammopathy, BM infiltration with lytic bone lesions, and protein deposition in the kidney. Moreover, the disease was easily transplantable and allowed different therapeutic options to be evaluated in vitro and in vivo. Using this model, we determined that GP130 signaling collaborated with MYC to induce MM and was responsible and sufficient for directing the plasma cell phenotype. Accordingly, we identified Myc aberrations in the L-GP130 MM model. Evaluation of human MM samples revealed recurrent activation of STAT3, a downstream target of GP130 signaling. Together, our results indicate that deregulated GP130 activity contributes to MM pathogenesis and that pathways downstream of GP130 activity have potential as therapeutic targets in MM.


Assuntos
Receptor gp130 de Citocina/metabolismo , Mieloma Múltiplo/metabolismo , Neoplasias Experimentais/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Linhagem Celular Tumoral , Receptor gp130 de Citocina/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Plasmócitos/metabolismo , Plasmócitos/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética
2.
Blood ; 119(17): 4026-33, 2012 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-22411868

RESUMO

Mutations of Fms-like tyrosine kinase 3 (FLT3) are among the most frequently detected molecular abnormalities in AML patients. Internal tandem duplications (ITDs) are found in approximately 25% and point mutations within the second tyrosine kinase domain (TKD) in approximately 7% of AML patients. Patients carrying the FLT3-ITD but not the FLT3-TKD mutation have a significantly worse prognosis. Therefore, both FLT3 mutations seem to exert different biologic functions. FLT3-ITD but not FLT3-TKD has been shown to induce robust activation of the STAT5 signaling pathway. In the present study, we investigated the mechanisms leading to differential STAT5 activation and show that FLT3-ITD but not FLT3-TKD uses SRC to activate STAT5. Coimmunoprecipitation and pull-down experiments revealed an exclusive interaction between SRC but not other Src family kinases and FLT3-ITD, which is mediated by the SRC SH2 domain. We identified tyrosines 589 and 591 of FLT3-ITD to be essential for SRC binding and subsequent STAT5 activation. Using site-specific Abs, we found that both residues were significantly more strongly phosphorylated in FLT3-ITD compared with FLT3-TKD. SRC inhibition and knock-down blocked STAT5 activation and proliferation induced by FLT3-ITD but not by FLT3-TKD. We conclude that SRC might be a therapeutic target in FLT3-ITD(+) AML.


Assuntos
Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteínas Tirosina Quinases/metabolismo , Sequências de Repetição em Tandem/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo , Quinases da Família src/metabolismo , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C3H , Células NIH 3T3 , Fosforilação , RNA Interferente Pequeno/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Domínios de Homologia de src , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
3.
Blood ; 116(22): 4600-11, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20696946

RESUMO

The oncogenic JAK2V617F mutation is found in myeloproliferative neoplasms (MPNs) and is believed to be critical for leukemogenesis. Here we show that JAK2V617F requires an intact SH2 domain for constitutive activation of downstream signaling pathways. In addition, there is a strict requirement of cytokine receptor expression for the activation of this oncogene. Further analysis showed that the SH2 domain mutation did not interfere with JAK2 membrane distribution. However, coimmunoprecipitated experiments revealed a role for the SH2 domain in the aggregation and cross-phosphorylation of JAK2V617F at the cell membrane. Forced overexpression of cytokine receptors could rescue the JAK2V617F SH2 mutant supporting a critical role of JAK2V617F abundance for constitutive activation. However, under physiologic cytokine receptor expression the SH2 domain is absolutely necessary for oncogenic JAK2V617F activation. This is demonstrated in a bone marrow transplantation model, in which an intact SH2 domain in JAK2V617F is required for the induction of an MPN-like disease. Thus, our results points to an indispensable role of the SH2 domain in JAK2V617F-induced MPNs.


Assuntos
Janus Quinase 2/genética , Mutação , Transtornos Mieloproliferativos/enzimologia , Domínios de Homologia de src , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Fosforilação
4.
Cancer Res ; 70(13): 5368-78, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20551051

RESUMO

Epidermal growth factor receptor (EGFR) overexpression and activation are hallmarks of non-small cell lung carcinoma (NSCLC). Although EGFR-targeted therapies are used, the prognosis of NSCLC remains poor. ADAM17 induces activation of the EGFR through ligand cleavage. However, we show that inhibition or knockdown of ADAM17 markedly reduces tumorigenesis and survival to a large part independently from EGFR ligand shedding in NSCLC cells. These findings strongly indicate additional oncogenic mechanisms regulated by ADAM17. We identified Notch1 signaling as an ADAM17-controlled pathway and a critical regulator of anchorage-independent growth by using both Notch1 shRNA and ectopic expression of the active intracellular Notch1 fragment. Strikingly, Notch1 knockdown led to a strong reduction of EGFR expression in all analyzed cell lines. Proliferation, survival, and colony formation of Notch1-deficient cells were insensitive to EGF stimulation. Moreover, targeting Notch1 or ADAM17 resulted in substantial cell death, whereas EGFR inhibition predominantly induced cell cycle arrest. Immunohistochemical analysis of primary human tissue revealed a significant correlation between ADAM17, Notch1 signaling, and high EGFR expression levels. In conclusion, this article describes a novel molecular circuitry in NSCLC, incorporating ADAM17 as a regulator of EGFR expression through the activation of Notch1. Due to their central role in tumorigenesis and survival of NSCLC cells, both ADAM17 and Notch1 constitute promising targets for the treatment of NSCLC.


Assuntos
Proteínas ADAM/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Receptores ErbB/biossíntese , Neoplasias Pulmonares/metabolismo , Receptor Notch1/metabolismo , Proteínas ADAM/biossíntese , Proteína ADAM17 , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Carcinoma Pulmonar de Células não Pequenas/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Fator de Crescimento Epidérmico/farmacologia , Proteínas de Homeodomínio/biossíntese , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Receptor Notch1/biossíntese , Transdução de Sinais , Fatores de Transcrição HES-1 , Transplante Heterólogo
5.
Blood ; 114(19): 4197-208, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19734451

RESUMO

Somatic mutations of Kit have been found in leukemias and gastrointestinal stromal tumors. The proto-oncogene c-Cbl negatively regulates Kit and Flt3 by its E3 ligase activity and acts as a scaffold. We recently identified the first c-Cbl mutation in human disease in an acute myeloid leukemia patient, called Cbl-R420Q. Here we analyzed the role of Cbl mutants on Kit-mediated transformation. Coexpression of Cbl-R420Q or Cbl-70Z with Kit induced cytokine-independent proliferation, survival, and clonogenic growth. Primary murine bone marrow retrovirally transduced with c-Cbl mutants and transplanted into mice led to a generalized mastocytosis, a myeloproliferative disease, and myeloid leukemia. Overexpression of these Cbl mutants inhibited stem cell factor (SCF)-induced ubiquitination and internalization of Kit. Both Cbl mutants enhanced the basal activation of Akt and prolonged the ligand-dependent activation. Importantly, transformation was observed also with kinase-dead forms of Kit and Flt3 in the presence of Cbl-70Z, but not in the absence of Kit or Flt3, suggesting a mechanism dependent on receptor tyrosine kinases, but independent of their kinase activity. Instead, transformation depends on the Src family kinase Fyn, as c-Cbl coimmunoprecipitated with Fyn and inhibition abolished transformation. These findings may explain primary resistance to tyrosine kinase inhibitors targeted at receptor tyrosine kinases.


Assuntos
Mastocitose/genética , Mutação , Transtornos Mieloproliferativos/genética , Proteínas Proto-Oncogênicas c-cbl/genética , Animais , Transplante de Medula Óssea , Células COS , Transformação Celular Neoplásica , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Humanos , Ligantes , Mastocitose/etiologia , Mastocitose/metabolismo , Mastocitose/patologia , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Transtornos Mieloproliferativos/etiologia , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais , Ubiquitinação
6.
J Exp Med ; 206(9): 1957-70, 2009 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-19687226

RESUMO

FLT3-ITD-mediated leukemogenesis is associated with increased expression of oncogenic PIM serine/threonine kinases. To dissect their role in FLT3-ITD-mediated transformation, we performed bone marrow reconstitution assays. Unexpectedly, FLT3-ITD cells deficient for PIM1 failed to reconstitute lethally irradiated recipients, whereas lack of PIM2 induction did not interfere with FLT3-ITD-induced disease. PIM1-deficient bone marrow showed defects in homing and migration and displayed decreased surface CXCR4 expression and impaired CXCL12-CXCR4 signaling. Through small interfering RNA-mediated knockdown, chemical inhibition, expression of a dominant-negative mutant, and/or reexpression in knockout cells, we found PIM1 activity to be essential for proper CXCR4 surface expression and migration of cells toward a CXCL12 gradient. Purified PIM1 led to the phosphorylation of serine 339 in the CXCR4 intracellular domain in vitro, a site known to be essential for normal receptor recycling. In primary leukemic blasts, high levels of surface CXCR4 were associated with increased PIM1 expression, and this could be significantly reduced by a small molecule PIM inhibitor in some patients. Our data suggest that PIM1 activity is important for homing and migration of hematopoietic cells through modification of CXCR4. Because CXCR4 also regulates homing and maintenance of cancer stem cells, PIM1 inhibitors may exert their antitumor effects in part by interfering with interactions with the microenvironment.


Assuntos
Células da Medula Óssea/citologia , Movimento Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Animais , Células da Medula Óssea/metabolismo , Quimiocina CXCL12/metabolismo , Primers do DNA/genética , Citometria de Fluxo , Humanos , Immunoblotting , Imunofenotipagem , Células Jurkat , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-pim-1/genética , Interferência de RNA , Receptores CXCR4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tirosina Quinase 3 Semelhante a fms
7.
Br J Haematol ; 144(6): 865-74, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19183186

RESUMO

Inhibition of the mutated fms-like tyrosine kinase 3 (FLT3) receptor tyrosine kinase is a promising therapeutic strategy in acute myeloid leukaemia (AML). However, development of resistance to FLT3 tyrosine kinase inhibitors (TKI), such as PKC412A, has been described recently. This observation may have an increasing impact on the duration of response and relapse rates in upcoming clinical trials employing FLT3-TKI. Herein we investigated two representatives of a novel class of FLT3-TKI: Bis(1H-indol-2-yl)methanones. Both compounds effectively induced apoptosis in FLT3-internal tandem duplicate (ITD)-transfected murine myeloid cells and in primary FLT3-ITD positive blasts. Combination of both compounds with chemotherapy revealed synergistic effects in apoptosis assays. The compounds did not show significant toxicity in human bone marrow cells derived from healthy donors. Compound102 overcame resistance to PKC412 within a non-myelotoxic dose-range. Western Blotting experiments of 32D-FLT3-ITD cells showed dose-dependent dephosphorylation of FLT3-ITD and of its downstream targets STAT5, AKT and ERK upon incubation with either compound. In conclusion, bis(1H-indol-2-yl)methanones overcome resistance mediated by FLT3-ITD mutations at position N676 and show strong efficacy in FLT3-ITD-positive cells alone as well as in combination with chemotherapy. We propose that further development of methanone compounds overcoming resistance to currently established FLT3-TKIs is an important step forward to an anticipated need within our future therapeutic algorithm in FLT3-ITD-positive AML.


Assuntos
Antineoplásicos/uso terapêutico , Indóis/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Estaurosporina/análogos & derivados , Sequências de Repetição em Tandem , Tirosina Quinase 3 Semelhante a fms/genética , Apoptose/efeitos dos fármacos , Linhagem Celular , Resistencia a Medicamentos Antineoplásicos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Estaurosporina/uso terapêutico , Transfecção/métodos , Células Tumorais Cultivadas
8.
Blood ; 113(17): 4074-7, 2009 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-18483393

RESUMO

In acute myeloid leukemia (AML), internal tandem duplications (ITDs) of the juxtamembrane (JM) of FLT3 have been shown to play a crucial role in driving proliferation and survival of the leukemic clone. Here, we report the identification of FLT3_ITD mutations located in non-JM domains of the FLT3-receptor. This novel type of FLT3_ITD mutation was found in 216 of 753 (28.7%) of unselected FLT3_ITD-positive AML cases. An FLT3 receptor harbouring a prototypic non-JM ITD (FLT3_ITD627E) mediated constitutive phosphorylation of FLT3 and of STAT5, suggesting that non-JM ITDs confer constitutive activation of the receptor. FLT3_ITD627E induced transformation of hematopoietic 32D cells and led to a lethal myeloproliferative disease in a syngeneic mouse model. Our results indicate that a significant proportion of activating FLT3_ITD mutations is not confined to the JM domain of FLT3. Further studies are warranted to define the biologic and clinical characteristics of non-JM ITDs.


Assuntos
Membrana Celular/enzimologia , Tirosina Quinase 3 Semelhante a fms/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Estrutura Terciária de Proteína , Tirosina Quinase 3 Semelhante a fms/química , Tirosina Quinase 3 Semelhante a fms/genética
9.
Exp Hematol ; 35(10): 1522-6, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17889720

RESUMO

OBJECTIVE: Activating mutations in FLT3 are known to be a frequent transforming event in acute myeloid leukemia. Small molecule-inhibitor therapy targeting the FLT3 kinase is, therefore, an attractive strategy. FLT3 kinase inhibitors, such as PKC412, have already entered clinical trials. Even though results are encouraging, emergence of primary and secondary resistance does occur in the majority of patients. Thus, it will be crucial to carefully characterize the activity of every single compound against different activating and resistance FLT3-internal tandem duplication (ITD) mutations. Here we tested the efficacy of sunitinib and sorafenib to inhibit primary FLT3 activating mutations (ITD and D835Y) and of secondary resistance mutations. METHODS: Ba/F3 cell lines stably expressing oncogenic FLT3 mutations were used to calculate cellular IC(50) values for sunitinib and sorafenib using cell proliferation assays. Differential IC(50) values for sorafenib toward FLT3-ITD and FLT3-D835Y were confirmed by Western blotting. Cell death was measured by propidium-iodide staining and flow cytometry. RESULTS: Sorafenib inhibits FLT3-ITD more potent than FLT3-D835Y, while sunitinib is equally effective against both mutant forms of FLT3. Importantly, sensitivity toward sorafenib and sunitinib varies between the different secondary FLT3-ITD resistance mutations. CONCLUSIONS: These results establish sensitivity profiles for the FLT3 inhibitors sunitinib and sorafenib. This may help to develop rational treatment strategies for acute myeloid leukemia with these compounds.


Assuntos
Antineoplásicos/farmacologia , Benzenossulfonatos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Indóis/farmacologia , Leucemia Mieloide Aguda/dietoterapia , Mutação , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Benzenossulfonatos/uso terapêutico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Indóis/uso terapêutico , Leucemia Mieloide Aguda/enzimologia , Niacinamida/análogos & derivados , Compostos de Fenilureia , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Pirróis/uso terapêutico , Sorafenibe , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia , Estaurosporina/uso terapêutico , Sunitinibe , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
10.
Blood ; 110(3): 1004-12, 2007 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-17446348

RESUMO

In acute myeloid leukemia (AML), mutational activation of the receptor tyrosine kinase (RTK) Flt3 is frequently involved in leukemic transformation. However, little is known about a possible role of highly expressed wild-type Flt3 in AML. The proto-oncogene c-Cbl is an important regulator of RTK signaling, acting through its ubiquitin ligase activity and as a platform for several signaling adaptor molecules. Here, we analyzed the role of c-Cbl in Flt3 signal transduction and myeloid transformation. C-Cbl physically interacted with Flt3 and was tyrosine phosphorylated in the presence of Flt3-ligand (FL). Overexpression of a dominant-negative form of c-Cbl (Cbl-70Z) inhibited FL-induced Flt3 ubiquitylation and internalization, indicating involvement of c-Cbl in Flt3 signaling. DNA sequencing of AML bone marrow revealed a case with a c-Cbl point mutation (Cbl-R420Q). Cbl-R420Q inhibited Flt3 internalization and ubiquitylation. Coexpression of Cbl-R420Q or Cbl-70Z with Flt3 induced cytokine-independent growth and survival of 32Dcl3 cells in the absence of FL. Also, the mutant Cbl proteins altered the amplitude and duration of Flt3-dependent signaling events. Our results indicate an important role of Cbl proteins in Flt3 signal modulation. Also, the data suggest a novel mechanism of leukemic transformation in AML by mutational inactivation of negative RTK regulators.


Assuntos
Transformação Celular Neoplásica/metabolismo , Leucemia Mieloide Aguda/metabolismo , Mutação Puntual , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Células COS , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Chlorocebus aethiops , Regulação Leucêmica da Expressão Gênica , Genes Dominantes , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-cbl/genética , Transdução de Sinais , Tirosina Quinase 3 Semelhante a fms/genética
11.
Proc Natl Acad Sci U S A ; 104(11): 4594-9, 2007 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-17360569

RESUMO

The kinase inhibitor imatinib mesylate targeting the oncoprotein Bcr-Abl has revolutionized the treatment of chronic myeloid leukemia (CML). However, even though imatinib successfully controls the leukemia in chronic phase, it seems not to be able to cure the disease, potentially necessitating lifelong treatment with the inhibitor under constant risk of relapse. On a molecular level, the cause of disease persistence is not well understood. Initial studies implied that innate features of primitive progenitor cancer stem cells may be responsible for the phenomenon. Here, we describe an assay using retroviral insertional mutagenesis (RIM) to identify genes contributing to disease persistence in vivo. We transplanted mice with bone marrow cells retrovirally infected with the Bcr-Abl oncogene and subsequently treated the animals with imatinib to select for leukemic cells in which the proviral integration had affected genes modulating the imatinib response. Southern blot analysis demonstrated clonal outgrowth of cells carrying similar integration sites. Candidate genes located near the proviral insertion sites were identified, among them the transcription factor RUNX3. Proviral integration near the RUNX3 promoter induced RUNX3 expression, and Bcr-Abl-positive cell lines with stable or inducible expression of RUNX1 or RUNX3 were protected from imatinib-induced apoptosis. Furthermore, imatinib treatment selected for RUNX1-expressing cells in vitro and in vivo after infection of primary bone marrow cells with Bcr-Abl and RUNX1. Our results demonstrate the utility of RIM for probing molecular modulators of targeted therapies and suggest a role for members of the RUNX transcription factor family in disease persistence in CML patients.


Assuntos
Antineoplásicos/farmacologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Resistencia a Medicamentos Antineoplásicos , Técnicas Genéticas , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Mutagênese , Piperazinas/farmacologia , Pirimidinas/farmacologia , Retroviridae/genética , Animais , Benzamidas , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subunidade alfa 3 de Fator de Ligação ao Core/fisiologia , Mesilato de Imatinib , Hibridização in Situ Fluorescente , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Masculino , Camundongos , Mutagênese Sítio-Dirigida
12.
Blood ; 105(12): 4792-9, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15718420

RESUMO

Activating mutations of the Fms-like tyrosine kinase 3 (FLT3) receptor are the most common genetic alteration in acute myeloid leukemia (AML). Two distinct groups of FLT3 mutations are found: internal tandem duplications (ITDs) of the juxtamembrane region and point mutations within the tyrosine kinase domain (TKD). Recently, point mutations within the activation loop of FLT3 have also been described in childhood acute lymphoblastic leukemia (ALL). FLT3-ITD has been shown to induce a myeloproliferative syndrome in a murine bone marrow transplantation model. The phenotype of FLT3-TKD in mice has not yet been investigated. We transduced murine bone marrow with retrovirus-expressing FLT3-TKD mutants or FLT3-ITD and transplanted these cells into lethally irradiated mice. Mice that received a transplant of FLT3-ITD developed an oligoclonal myeloproliferative disease as previously described. In contrast, FLT3-TKD mutants induced an oligoclonal lymphoid disorder with longer latency and distinct hematologic manifestations: importantly, induction of the lymphoid phenotype was not due to a low number of transplanted cells. The lymphoid manifestation and longer latency of FLT3-TKD compared with FLT3-ITD mutants together with the lack of influence of FLT3-TKD mutations on the clinical outcome of patients with AML suggest differences in cell signaling between FLT3-TKD mutants and FLT3-ITDs. Indeed strong signal transducers and activators of transcription 5 (STAT5) activation could only be demonstrated for FLT3-ITDs.


Assuntos
Transplante de Medula Óssea , Leucemia Mieloide Aguda/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Tirosina Quinases/química , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/genética , Animais , Southern Blotting , Células da Medula Óssea/citologia , Separação Celular , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Immunoblotting , Imunofenotipagem , Imunoprecipitação , Camundongos , Proteínas do Leite/metabolismo , Mutação , Transtornos Mieloproliferativos/genética , Fenótipo , Mutação Puntual , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Retroviridae/genética , Fator de Transcrição STAT5 , Transdução de Sinais , Esplenomegalia/patologia , Transativadores/metabolismo , Resultado do Tratamento , Tirosina Quinase 3 Semelhante a fms
13.
Oncogene ; 22(30): 4642-7, 2003 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-12879008

RESUMO

A t(2;5) (p23;q35) chromosomal translocation can be found in a high percentage of anaplastic large-cell lymphomas (ALCL). This genetic abnormality leads to the expression of the NPM-ALK fusion protein, which encodes a constitutively active tyrosine kinase that plays a causative role in lymphomagenesis. Employing a modified infection/transplantation protocol utilizing an MSCV-based vector, we were able to reproducibly induce two phenotypically different lymphoma-like diseases dependent on the retroviral titers used. The first phenotype presented as a polyclonal histiocytic malignancy of myeloid/macrophage origin with a short latency period of 3-4 weeks. Clinically, the diseased mice showed rapidly progressive wasting, lymphadenopathy and pancytopenia. Mice displaying the second phenotype developed monoclonal B-lymphoid tumors with a longer latency of approximately 12-16 weeks, primarily involving the spleen and the bone marrow, with less extensive lymph node but also histologically evident extranodal organ infiltration by large immature plasmoblastic cells. The described retroviral mouse model will be useful to analyse the role of NPM-ALK in lymphomagenesis in vivo and may contribute to the development of new treatment options for NPM-ALK induced malignancies.


Assuntos
Proteínas Tirosina Quinases/fisiologia , Retroviridae/genética , Animais , Separação Celular , DNA Complementar/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Camundongos , Modelos Genéticos , Transplante de Neoplasias , Neoplasias Experimentais , Fenótipo , Proteínas Tirosina Quinases/metabolismo , Fatores de Tempo
14.
Blood ; 102(2): 646-51, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12663439

RESUMO

Activating mutations of FLT3 have been detected in patients with acute myeloid leukemia (AML). Two distinct types of FLT3 mutations are most common: internal tandem duplication (ITD) of sequences coding for the juxtamembrane domain and point mutations at codon 835 (Asp835) within the kinase domain. Both types of mutations constitutively activate the tyrosine kinase activity of FLT3 in experimental systems and result in factor-independent proliferation of Ba/F3 and 32D cells. Recently, novel mutations within the activation loop were identified in patients with AML: deletion of isoleucine 836 (Ile836del) and an exchange of isoleucine 836 to methionine plus an arginine insertion (Ile836Met+Arg). To examine whether the Ile836 mutations result in constitutive activation of the FLT3 receptor, we introduced both mutant FLT3 cDNAs transiently into HEK 293 cells. Both mutant FLT3 receptors were constitutively autophosphorylated in the absence of ligand and kinase activity led to constitutive activation of downstream signaling cascades as determined by activation of the STAT5 (signal transducer and activator of transcription 5) pathway. When stably expressed in the growth factor-dependent cell lines Ba/F3 and 32D, both deletion and insertion mutants led to factor-independent proliferation, indicating that both mutants have transforming capabilities. We then examined the sensitivity of the FLT3 ITD, FLT3 Asp835Tyr, and the novel FLT3 receptor mutants toward the kinase inhibitors AG1296, PKC412, and SU5614. We show that these FLT3 kinase inhibitors have distinct inhibitory potencies against different activating FLT3 receptor mutants. These results suggest that it may be useful to determine the exact kind of FLT3 mutation when applying receptor kinase inhibitors in clinical trials.


Assuntos
Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Proteínas do Leite , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia , Tirfostinas/farmacologia , Doença Aguda , Substituição de Aminoácidos , Animais , Divisão Celular , Linhagem Celular/citologia , Linhagem Celular/enzimologia , Códon/genética , Proteínas de Ligação a DNA/metabolismo , Resistência a Medicamentos , Ativação Enzimática , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , Humanos , Leucemia Mieloide/enzimologia , Leucemia Mieloide/genética , Proteínas de Membrana/farmacologia , Camundongos , Mutagênese Insercional , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT5 , Deleção de Sequência , Relação Estrutura-Atividade , Transativadores/metabolismo , Transfecção , Tirosina Quinase 3 Semelhante a fms
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA