Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Cancer Cell ; 42(4): 568-582.e11, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38490213

RESUMO

Major histocompatibility complex (MHC) class I antigen presentation deficiency is a common cancer immune escape mechanism, but the mechanistic implications and potential strategies to address this challenge remain poorly understood. Studying ß2-microglobulin (B2M) deficient mouse tumor models, we find that MHC class I loss leads to a substantial immune desertification of the tumor microenvironment (TME) and broad resistance to immune-, chemo-, and radiotherapy. We show that treatment with long-lasting mRNA-encoded interleukin-2 (IL-2) restores an immune cell infiltrated, IFNγ-promoted, highly proinflammatory TME signature, and when combined with a tumor-targeting monoclonal antibody (mAB), can overcome therapeutic resistance. Unexpectedly, the effectiveness of this treatment is driven by IFNγ-releasing CD8+ T cells that recognize neoantigens cross-presented by TME-resident activated macrophages. These macrophages acquire augmented antigen presentation proficiency and other M1-phenotype-associated features under IL-2 treatment. Our findings highlight the importance of restoring neoantigen-specific immune responses in the treatment of cancers with MHC class I deficiencies.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Animais , Camundongos , Antígenos de Histocompatibilidade Classe I/genética , Interleucina-2/genética , Interleucina-2/imunologia , Neoplasias/genética , RNA Mensageiro , Microambiente Tumoral
3.
Oncotarget ; 11(20): 1862-1875, 2020 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-32499871

RESUMO

PLAC1 (placenta enriched 1) is a mammalian trophoblast-specific protein. Aberrant expression of PLAC1 is observed in various human cancers, where it is involved in the motility, migration, and invasion of tumor cells, which are associated with the phosphoinositide 3-kinase (PI3K)/AKT pathway. We previously demonstrated that AKT activation mediates the downstream effects of PLAC1; however, the molecular mechanisms of PLAC1-induced AKT-mediated tumor-related processes are unclear. We studied human choriocarcinoma and breast cancer cell lines to explore the localization and receptor-ligand interactions, as well as the downstream effects of PLAC1. We show secretion and adherence of PLAC1 to the extracellular matrix, where it forms a trimeric complex with fibroblast growth factor 7 (FGF7) and its receptor, FGF receptor 2 IIIb (FGFR2IIIb). We further show that PLAC1 signaling via FGFR2IIIb activates AKT phosphorylation in cancer cell lines. As the FGF pathway is of major interest in anticancer therapeutic strategies, these data further promote PLAC1 as a promising anticancer drug target.

4.
Mol Ther ; 27(4): 824-836, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30638957

RESUMO

Synthetic mRNA has emerged as a powerful tool for the transfer of genetic information, and it is being explored for a variety of therapeutic applications. Many of these applications require prolonged intracellular persistence of mRNA to improve bioavailability of the encoded protein. mRNA molecules are intrinsically unstable and their intracellular kinetics depend on the UTRs embracing the coding sequence, in particular the 3' UTR elements. We describe here a novel and generally applicable cell-based selection process for the identification of 3' UTRs that augment the expression of proteins encoded by synthetic mRNA. Moreover, we show, for two applications of mRNA therapeutics, namely, (1) the delivery of vaccine antigens in order to mount T cell immune responses and (2) the introduction of reprogramming factors into differentiated cells in order to induce pluripotency, that mRNAs tagged with the 3' UTR elements discovered in this study outperform those with commonly used 3' UTRs. This approach further leverages the utility of mRNA as a gene therapy drug format.


Assuntos
Regiões 3' não Traduzidas/genética , Biblioteca Gênica , Terapia Genética/métodos , Estabilidade de RNA , RNA Mensageiro/genética , Animais , Doadores de Sangue , Vacinas Anticâncer , Células Cultivadas , Reprogramação Celular/genética , Feminino , Fibroblastos , Técnicas de Transferência de Genes , Meia-Vida , Humanos , Células-Tronco Pluripotentes Induzidas , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/metabolismo , Vacinação
5.
Mol Pharm ; 15(9): 3909-3919, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30028629

RESUMO

Targeting mRNA to eukaryotic cells is an emerging technology for basic research and provides broad applications in cancer immunotherapy, vaccine development, protein replacement, and in vivo genome editing. Although a plethora of nanoparticles for efficient mRNA delivery exists, in vivo mRNA targeting to specific organs, tissue compartments, and cells remains a major challenge. For this reason, methods for reporting the in vivo targeting specificity of different mRNA nanoparticle formats will be crucial. Here, we describe a straightforward method for monitoring the in vivo targeting efficiency of mRNA-loaded nanoparticles in mice. To achieve accurate mRNA delivery readouts, we loaded lipoplex nanoparticles with Cre-recombinase-encoding mRNA and injected these into commonly used Cre reporter mouse strains. Our results show that this approach provides readouts that accurately report the targeting efficacy of mRNA into organs, tissue structures, and single cells as a function of the used mRNA delivery system. The method described here establishes a versatile basis for determining in vivo mRNA targeting profiles and can be systematically applied for testing and improving mRNA packaging formats.


Assuntos
Nanopartículas/química , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Animais , Cromatografia Líquida , Lipossomos/química , Espectrometria de Massas , Camundongos , Tamanho da Partícula
6.
Sci Rep ; 7(1): 16892, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203786

RESUMO

Recombinant vaccine strain-derived measles virus (MV) is clinically tested both as vaccine platform to protect against other pathogens and as oncolytic virus for tumor treatment. To investigate the potential synergism in anti-tumoral efficacy of oncolytic and vaccine properties, we chose Ovalbumin and an ideal tumor antigen, claudin-6, for pre-clinical proof of concept. To enhance immunogenicity, both antigens were presented by retroviral virus-like particle produced in situ during MV-infection. All recombinant MV revealed normal growths, genetic stability, and proper expression and presentation of both antigens. Potent antigen-specific humoral and cellular immunity were found in immunized MV-susceptible IFNAR-/--CD46Ge mice. These immune responses significantly inhibited metastasis formation or increased therapeutic efficacy compared to control MV in respective novel in vivo tumor models using syngeneic B16-hCD46/mCLDN6 murine melanoma cells. These data indicate the potential of MV to trigger selected tumor antigen-specific immune responses on top of direct tumor lysis for enhanced efficacy.


Assuntos
Antígenos de Neoplasias/genética , Vacinas Anticâncer/imunologia , Vírus do Sarampo/genética , Melanoma Experimental/terapia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Autoanticorpos/sangue , Autoanticorpos/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Chlorocebus aethiops , Claudinas/genética , Claudinas/imunologia , Claudinas/metabolismo , Imunidade Celular , Imunidade Humoral , Interferon gama/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Melanoma Experimental/imunologia , Camundongos , Camundongos Transgênicos , Terapia Viral Oncolítica , Ovalbumina/genética , Ovalbumina/imunologia , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/uso terapêutico , Células Vero
7.
Int J Cancer ; 122(12): 2744-52, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18366061

RESUMO

The BCR/ABL oncogene is responsible for the phenotype of Philadelphia chromosome-positive (Ph+) leukemia. BCR/ABL exhibits an aberrant ABL-tyrosine kinase activity. The treatment of advanced Ph+ leukemia with selective ABL-kinase inhibitors such as Imatinib, Nilotinib and Dasatinib is initially effective but rapidly followed by resistance mainly because of specific mutations in BCR/ABL. Tetramerization of ABL through the N-terminal coiled-coil region (CC) of BCR is essential for the ABL-kinase activation. Targeting the CC-domain forces BCR/ABL into a monomeric conformation reduces its kinase activity and increases the sensitivity for Imatinib. We show that (i) targeting the tetramerization by a peptide representing the Helix-2 of the CC efficiently reduced the autophosphorylation of both unmutated and mutated BCR/ABL; (ii) Helix-2 inhibited the transformation potential of BCR/ABL independently of the presence of mutations; and (iii) Helix-2 efficiently cooperated with Imatinib as revealed by their effects on the transformation potential and the factor-independence related to BCR/ABL with the exception of mutant T315I. These findings support earlier observations that BCR/ABL harboring the T315I mutation have a transformation potential that is at least partially independent of its kinase activity. These data provide evidence that the inhibition of tetramerization inhibits BCR/ABL-mediated transformation and can contribute to overcome Imatinib-resistance.


Assuntos
Antineoplásicos/farmacologia , Biopolímeros/química , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Mutação , Piperazinas/farmacologia , Pirimidinas/farmacologia , Sequência de Bases , Benzamidas , Linhagem Celular , Primers do DNA , Proteínas de Fusão bcr-abl/genética , Humanos , Mesilato de Imatinib , Mutagênese Sítio-Dirigida , Fosforilação
8.
Haematologica ; 92(3): 323-31, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17339181

RESUMO

BACKGROUND AND OBJECTIVES: Stem cells play an important role in the pathogenesis and maintenance of most malignant tumors. Acute myeloid leukemia (AML) is a stem cell disease. The inefficient targeting of the leukemic stem cells (LSC) is considered responsible for relapse after the induction of complete hematologic remission (CR) in AML. Acute promyelocytic leukemia (APL) is a subtype of AML characterized by the t(15;17) translocation and expression of the PML/RARalpha fusion protein. Treatment of APL with all-trans retinoic acid (ATRA) induces CR, but not molecular remission (CMR), because the fusion transcript remains detectable, followed by relapse within a few months. Arsenic induces high rates of CR and CMR followed by a long relapse-free survival (RFS). Here we compared the effects of ATRA and arsenic on PML/RARalpha-positive stem cell compartments. DESIGN AND METHODS: As models for the PML/RARalpha-positive LSC we used: (i) Sca1+/lin- murine HSC retrovirally transduced with PML/RARalpha; (ii) LSC from mice with PML/RARalpha-positive leukemia; (iii) the side population of the APL cell line NB4. RESULTS: In contrast to ATRA, arsenic abolishes the aberrant stem cell capacity of PML/RARalpha-positive stem cells. Arsenic had no apparent influence on the proliferation of PML/RARalpha-positive stem cells, whereas ATRA greatly increased the proliferation of these cells. Furthermore ATRA induces proliferation of APL-derived stem cells, whereas arsenic inhibits their growth. INTERPRETATIONS AND CONCLUSIONS: Taken together our data suggest a relationship between the capacity of a compound to target the leukemia-initiating cell and its ability to induce long relapse-free survival. These data strongly support the importance of efficient LSC-targeting for the outcome of patients with leukemia.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Inibidores do Crescimento/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucemia Promielocítica Aguda/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Óxidos/farmacologia , Animais , Trióxido de Arsênio , Ataxina-1 , Ataxinas , Biomarcadores Tumorais/análise , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Feminino , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/análise , Proteínas Nucleares/análise , Proteínas de Fusão Oncogênica/análise , Proteínas de Fusão Oncogênica/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transfecção , Tretinoína/administração & dosagem , Tretinoína/farmacologia , Ensaio Tumoral de Célula-Tronco
9.
BMC Cancer ; 6: 262, 2006 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-17090304

RESUMO

BACKGROUND: The reciprocal (9;22) translocation fuses the bcr (breakpoint cluster region) gene on chromosome 22 to the abl (Abelson-leukemia-virus) gene on chromosome 9. Depending on the breakpoint on chromosome 22 (the Philadelphia chromosome--Ph+) the derivative 9+ encodes either the p40(ABL/BCR) fusion transcript, detectable in about 65% patients suffering from chronic myeloid leukemia, or the p96(ABL/BCR) fusion transcript, detectable in 100% of Ph+ acute lymphatic leukemia patients. The ABL/BCRs are N-terminally truncated BCR mutants. The fact that BCR contains Rho-GEF and Rac-GAP functions strongly suggest an important role in cytoskeleton modeling by regulating the activity of Rho-like GTPases, such as Rho, Rac and cdc42. We, therefore, compared the function of the ABL/BCR proteins with that of wild-type BCR. METHODS: We investigated the effects of BCR and ABL/BCRs i.) on the activation status of Rho, Rac and cdc42 in GTPase-activation assays; ii.) on the actin cytoskeleton by direct immunofluorescence; and iii) on cell motility by studying migration into a three-dimensional stroma spheroid model, adhesion on an endothelial cell layer under shear stress in a flow chamber model, and chemotaxis and endothelial transmigration in a transwell model with an SDF-1alpha gradient. RESULTS: Here we show that both ABL/BCRs lost fundamental functional features of BCR regarding the regulation of small Rho-like GTPases with negative consequences on cell motility, in particular on the capacity to adhere to endothelial cells. CONCLUSION: Our data presented here describe for the first time an analysis of the biological function of the reciprocal t(9;22) ABL/BCR fusion proteins in comparison to their physiological counterpart BCR.


Assuntos
Movimento Celular/fisiologia , Citoesqueleto/fisiologia , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/fisiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Proteínas Proto-Oncogênicas c-bcr/genética , Proteínas Proto-Oncogênicas c-bcr/fisiologia , Humanos , Células Tumorais Cultivadas , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
10.
Cancer Res ; 65(14): 6080-8, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16024608

RESUMO

Acute myeloid leukemia is characterized by a differentiation block as well as by an increased self-renewal of hematopoietic precursors in the bone marrow. This phenotype is induced by specific acute myeloid leukemia-associated translocations, such as t(15;17) and t(11;17), which involve an identical portion of the retinoic acid receptor alpha (RARalpha) and either the promyelocytic leukemia (PML) or promyelocytic zinc finger (PLZF) genes, respectively. The resulting fusion proteins form high molecular weight complexes and aberrantly bind several histone deacetylase-recruiting nuclear corepressor complexes. The amino-terminal BTB/POZ domain is indispensable for the capacity of PLZF to form high molecular weight complexes. Here, we studied the role of dimerization and binding to histone deacetylase-recruiting nuclear corepressor complexes for the induction of the leukemic phenotype by PLZF/RARalpha and we show that (a) the BTB/POZ domain mediates the oligomerization of PLZF/RARalpha; (b) mutations that inhibit dimerization of PLZF do the same in PLZF/RARalpha; (c) the PLZF/RARalpha-related block of differentiation requires an intact BTB/POZ domain; (d) the mutations interfering with either folding of the BTB/POZ domain or with its charged pocket prevent the self-renewal of PLZF/RARalpha-positive hematopoietic stem cells. Taken together, these data provide evidence that the dimerization capacity and the formation of a functionally charged pocket are indispensable for the PLZF/RARalpha-induced leukemogenesis.


Assuntos
Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/metabolismo , Doença Aguda , Animais , Células COS , Dimerização , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Peso Molecular , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/genética , Mutação Puntual , Regiões Promotoras Genéticas , Ligação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Transcrição Gênica , Dedos de Zinco
11.
Cancer Res ; 65(7): 2537-41, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15805245

RESUMO

Histone deacetylase inhibitors have attracted considerable attention because of their ability to overcome the differentiation block in leukemic blasts, an effect achieved either alone or in combination with differentiating agents, such as all-trans retinoic acid. We have previously reported favorable effects of the potent histone deacetylase inhibitor valproic acid in combination with all-trans retinoic acid in patients with advanced acute myeloid leukemia leading to blast cell reduction and improvement of hemoglobin. These effects were accompanied by hypergranulocytosis most likely due to an enhancement of nonleukemic myelopoiesis and the suppression of malignant hematopoiesis rather than enforced differentiation of the leukemic cells. These data prompted us to investigate the effect of valproic acid on normal hematopoietic stem cells (HSC). Here we show that valproic acid increases both proliferation and self-renewal of HSC. It accelerates cell cycle progression of HSC accompanied by a down-regulation of p21(cip-1/waf-1). Furthermore, valproic acid inhibits GSK3beta by phosphorylation on Ser9 accompanied by an activation of the Wnt signaling pathway as well as by an up-regulation of HoxB4, a target gene of Wnt signaling. Both are known to directly stimulate the proliferation of HSC and to expand the HSC pool. In summary, we here show that valproic acid, known to induce differentiation or apoptosis in leukemic blasts, stimulates the proliferation of normal HSC, an effect with a potential effect on its future role in the treatment of acute myeloid leukemia.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Ácido Valproico/farmacologia , Animais , Antígenos CD34/biossíntese , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Fatores de Transcrição , Tretinoína/farmacologia , Regulação para Cima/efeitos dos fármacos
12.
Blood ; 103(9): 3535-43, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-14739224

RESUMO

Acute myeloid leukemia (AML) is characterized by the block of differentiation, deregulated apoptosis, and an increased self-renewal of hematopoietic precursors. It is unclear whether the self-renewal of leukemic blasts results from the cumulative effects of blocked differentiation and impaired apoptosis or whether there are mechanisms directly increasing self-renewal. The AML-associated translocation products (AATPs) promyelocytic leukemia/retinoic acid receptor alpha (PML/RAR alpha), promyelocytic leukemia zinc finger (PLZF)/RAR alpha (X-RAR alpha), and AML-1/ETO block hematopoietic differentiation. The AATPs activate the Wnt signaling by up-regulating gamma-catenin. Activation of the Wnt signaling augments self-renewal of hematopoietic stem cells (HSCs). Therefore, we investigated how AATPs influence self-renewal of HSCs and evaluated the role of gamma-catenin in the determination of the phenotype of HSCs expressing AATPs. Here we show that the AATPs directly activate the gamma-catenin promoter. The crucial role of gamma-catenin in increasing the self-renewal of HSCs upon expression of AATPs is demonstrated by (i) the abrogation of replating efficiency upon hindrance of gamma-catenin expression through RNA interference, and (ii) the augmentation of replating efficiency of HSCs upon overexpression of gamma-catenin itself. In addition, the inoculation of gamma-catenin-transduced HSCs into irradiated recipient mice establishes the clinical picture of AML. These data provide the first evidence that the aberrant activation of Wnt signaling by the AATP decisively contributes to the pathogenesis of AML.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Células-Tronco Hematopoéticas/patologia , Leucemia Mieloide/etiologia , Proteínas de Fusão Oncogênica/fisiologia , Proteínas de Peixe-Zebra , Doença Aguda , Animais , Divisão Celular , Transformação Celular Neoplásica , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/genética , Desmoplaquinas , Feminino , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Proteínas Proto-Oncogênicas/fisiologia , RNA Interferente Pequeno/farmacologia , Transdução Genética , Translocação Genética , Regulação para Cima , Proteínas Wnt , gama Catenina
13.
Oncogene ; 22(44): 6900-8, 2003 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-14534537

RESUMO

Since the 19th century, arsenic (As2O3) has been used in the treatment of chronic myelogenous leukemia (CML) characterized by the t(9;22) translocation. As2O3 induces complete remissions in patients with acute promyelocytic leukemia. The response to As2O3 is genetically determined by the t(15;17)-or the t(9;22)-specific fusion proteins PML/RARalpha or BCR/ABL. The PML portion of PML/RARalpha is crucial for the sensitivity to As2O3. PML is nearly entirely contained in PML/RARalpha. PML is upregulated by oncogenic RAS in primary fibroblasts. The aberrant kinase activity of BCR/ABL leads to constitutive activation of RAS. Therefore, we hypothesized that BCR/ABL could increase sensitivity to As2O2-induced apoptosis by modifying PML expression. To disclose the mechanism of As2O3-induced apoptosis in PML/RARalpha- and BCR/ABL-expressing cells, we focused on the role of PML for As2O3-induced cell death. Here we report that (i) sensitivity to As2O3-induced apoptosis of U937 cells can be increased either by overexpression of PML, or by conditional expression of activated RAS; (ii) also the expression of the t(8;21)-related AML-1/ETO increased sensitivity to As2O3-induced apoptosis; (iii) both BCR/ABL and AML-1/ETO activated RAS and modified the PML expression pattern; (iv) the expression of either BCR/ABL or AML-1/ETO rendered U937 cells sensitive to interferon alpha-induced apoptosis. In summary, these data suggest a crucial role of factors able to upregulate PML for As2O2-induced cell death.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Proteínas de Fusão bcr-abl/metabolismo , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Genes ras , Leucemia Promielocítica Aguda/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Apoptose/efeitos dos fármacos , Arsenicais/uso terapêutico , Benzamidas , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Proteínas de Fusão bcr-abl/genética , Humanos , Mesilato de Imatinib , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/genética , Monócitos/efeitos dos fármacos , Óxidos/farmacologia , Óxidos/uso terapêutico , Cromossomo Filadélfia , Piperazinas/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-abl/genética , Pirimidinas/farmacologia , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Transgenes , Translocação Genética , Células Tumorais Cultivadas , Células U937 , Regulação para Cima
14.
Blood ; 102(8): 2985-93, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12829585

RESUMO

Translocations involving the abl locus on chromosome 9 fuses the tyrosine kinase c-ABL to proteins harboring oligomerization interfaces such as BCR or TEL, enabling these ABL-fusion proteins (X-ABL) to transform cells and to induce leukemia. The ABL kinase activity is blocked by the ABL kinase inhibitor STI571 which abrogates transformation by X-ABL. To investigate the role of oligomerization for the transformation potential of X-ABL and for the sensitivity to STI571, we constructed ABL chimeras with oligomerization interfaces of proteins involved in leukemia-associated translocations such as BCR, TEL, PML, and PLZF. We assessed the capacity of these chimeras to form high molecular weight (HMW) complexes as compared with p185(BCR-ABL). There was a direct relationship between the size of HMW complexes formed by these chimeras and their capacity to induce factor independence in Ba/F3 cells, whereas there was an inverse relationship between the size of the HMW complexes and the sensitivity to STI571. The targeting of the oligomerization interface of p185(BCR-ABL) by a peptide representing the coiled coil region of BCR reduced its potential to transform fibroblasts and increased sensitivity to STI571. Our results indicate that targeting of the oligomerization interfaces of the X-ABL enhances the effects of STI571 in the treatment of leukemia caused by X-ABL.


Assuntos
Proteínas de Fusão bcr-abl/química , Proteínas Oncogênicas/metabolismo , Piperazinas/farmacologia , Proteínas Tirosina Quinases , Proteínas Proto-Oncogênicas , Pirimidinas/farmacologia , Animais , Antineoplásicos/farmacologia , Benzamidas , Western Blotting , Células COS , Linhagem Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica , DNA Complementar/metabolismo , Fibroblastos/metabolismo , Mesilato de Imatinib , Concentração Inibidora 50 , Leucemia/tratamento farmacológico , Camundongos , Microscopia de Fluorescência , Mutação , Fosforilação , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcr , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Fatores de Tempo
15.
Cancer Res ; 62(23): 7050-8, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12460926

RESUMO

Acute myeloid leukemia (AML)-associated chromosomal translocations result in formation of chimeric transcription factors, such as PML/RARalpha, PLZF/RARalpha, and AML-1/ETO, of which the components are involved in regulation of transcription by chromatin modeling through histone acetylation/deacetylation. The leukemic differentiation block is attributed to deregulated transcription caused by these chimeric fusion proteins, which aberrantly recruit histone-deacetylase (HDAC) activity. One essential differentiation pathway blocked by the leukemic fusion proteins is the vitamin (Vit) D(3) signaling. Here we investigated the mechanisms by which the leukemic fusion proteins interfere with VitD(3)-induced differentiation. The VitD(3)-receptor (VDR) is, like the retinoid receptors RAR, retinoid X receptor, and the thyroid hormone receptor (TR), a ligand-inducible transcription factor. In the absence of ligand, the transcriptional activity of TR and RAR is silenced by recruitment of HDAC activity through binding to corepressors. In the presence of ligand, TR and RAR activate transcription by releasing HDAC activity and by recruiting histone-acetyltransferase activity. Here we report that VDR binds corepressors in a ligand-dependent manner and that inhibition of HDAC activity increases VitD(3) sensitivity of HL-60 cells. Nevertheless, the inhibition of HDAC activity is unable to overcome the block of VitD(3)-induced differentiation caused by PLZF/RARalpha expression. Here we demonstrate that the expression of the translocation products PML/RARalpha and PLZF/RARalpha impairs the localization of VDR in the nucleus by binding to VDR. Furthermore, the overexpression of VDR in U937 cells expressing AML-related translocation products completely abolishes the block of VitD(3)-induced differentiation. Taken together these data indicate that the AML-associated translocation products block differentiation not only by interfering with chromatin-modeling but also by sequestering factors involved in the differentiation signaling pathways, such as VDR in the VitD(3)-induced differentiation.


Assuntos
Colecalciferol/antagonistas & inibidores , Leucemia Mieloide/patologia , Proteínas de Fusão Oncogênica/fisiologia , Receptores de Calcitriol/fisiologia , Diferenciação Celular/fisiologia , Colecalciferol/metabolismo , Colecalciferol/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core , Células HL-60 , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Estrutura Terciária de Proteína , Proteína 1 Parceira de Translocação de RUNX1 , Receptores de Calcitriol/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Transfecção , Translocação Genética , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA