Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Oncotarget ; 8(50): 88244-88250, 2017 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-29152155

RESUMO

Chibby 1 (CBY1) is a small and evolutionarily conserved protein, which act as ß-catenin antagonist. CBY1 is encoded by C22orf2 (22q13.1) Its antagonistic function on ß-catenin involves the direct interaction with: The C-terminal activation domain of ß-catenin, which hinders ß-catenin binding with Tcf/Lef transcription factors hence repressing ß-catenin transcriptional activation. 14-3-3 scaffolding proteins (σ or ξ), which drive CBY1 nuclear export into a stable tripartite complex with ß-catenin. The relative proximity of C22orf2 gene encoding for CBY1 to the BCR breakpoint on chromosome 22q11, whose translocation and rearrangement with the c-ABL is the causative event of chronic myeloid leukemia (CML), suggested that gene haploinsufficiency may play a role in the disease pathogenesis and progression. We found CBY1 down-modulation associated with the BCR-ABL1, promoted by transcriptional mechanisms (promoter hyper-methylation) and post-transcriptional events, addressing the protein towards proteasome-dependent degradation through SUMOylation. CBY1 reduced expression in clonal progenitors and, more importantly, in leukemic stem cells (LSC), is contingent upon the tyrosine kinase (TK) activity of BCR-ABL1 fusion protein. Accordingly, its induction by Imatinib (IM) and second generation TK inhibitors contributes to ß-catenin inactivation through multiple events encompassing the activation of endoplasmic reticulum (ER) stress-associated unfolded protein response (UPR) and autophagy, eventually leading to apoptotic death. These findings support the advantage of combined regimens including drugs targeting DNA epigenetics and/or proteasome to eradicate the BCR-ABL1+ hematopoiesis.

3.
J Cell Biochem ; 118(11): 3968-3975, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28401599

RESUMO

FOXM1 transcription factor is a central component of tumor initiation, growth, and progression due to its multiple effects on cell cycle, DNA repair, angiogenesis and invasion, chromatin, protein anabolism, and cell adhesion. Moreover, FOXM1 interacts with ß-catenin promoting its nuclear import and transcriptional activation. Here, we show that FOXM1 is involved in the advantage of chronic myeloid leukemia hematopoiesis over the normal counterpart. FOXM1 hyper-activation associated with BCR-ABL1 results from phosphorylation by the fusion protein kinase-dependent activation of Polo-like kinase 1. FOXM1 phosphorylation lets its binding with ß-catenin and ß-catenin transcriptional activation, a key event for persistence of the leukemic stem cell compartment under tyrosine kinase inhibitor therapy. Polo-like kinase 1 inhibitor BI6727, already advanced for clinical use, breaks ß-catenin interaction with FOXM1, hence hampering FOXM1 phosphorylation, ß-catenin binding, nuclear import, and downstream signaling. In conclusion, our results support Polo-like kinase 1/FOXM1 axis as a complementary target to eradicate leukemic early progenitor/stem cell compartment in chronic myeloid leukemia. J. Cell. Biochem. 118: 3968-3975, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Proliferação de Células , Proteína Forkhead Box M1/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais , Feminino , Proteína Forkhead Box M1/genética , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/patologia
4.
PLoS One ; 10(7): e0131074, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26147002

RESUMO

The down-modulation of the ß-catenin antagonist Chibby 1 (CBY1) associated with the BCR-ABL1 fusion gene of chronic myeloid leukemia (CML) contributes to the aberrant activation of ß-catenin, particularly in leukemic stem cells (LSC) resistant to tyrosine kinase (TK) inhibitors. It is, at least partly, driven by transcriptional events and gene promoter hyper-methylation. Here we demonstrate that it also arises from reduced protein stability upon binding to 14-3-3σ adapter protein. CBY1/14-3-3σ interaction in BCR-ABL1+ cells is mediated by the fusion protein TK and AKT phosphorylation of CBY1 at critical serine 20, and encompasses the 14-3-3σ binding modes I and II involved in the binding with client proteins. Moreover, it is impaired by c-Jun N-terminal kinase (JNK) phosphorylation of 14-3-3σ at serine 186, which promotes dissociation of client proteins. The ubiquitin proteasome system UPS participates in reducing stability of CBY1 bound with 14-3-3σ through enhanced SUMOylation. Our results open new routes towards the research on molecular pathways promoting the proliferative advantage of leukemic hematopoiesis over the normal counterpart.


Assuntos
Proteínas 14-3-3/metabolismo , Biomarcadores Tumorais/metabolismo , Proteínas de Transporte/biossíntese , Exorribonucleases/metabolismo , Regulação Leucêmica da Expressão Gênica/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/biossíntese , Processamento de Proteína Pós-Traducional , Motivos de Aminoácidos , Benzamidas/farmacologia , Proteínas de Transporte/genética , Regulação para Baixo , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteína Oncogênica p65(gag-jun) , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirazóis/farmacologia , Frações Subcelulares/metabolismo , Sumoilação , beta Catenina/antagonistas & inibidores
5.
J Cell Biochem ; 116(4): 589-97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25389112

RESUMO

The decrease of Chibby1 (CBY1) contributes to ß catenin constitutive activation associated with the presence of the BCR-ABL1 fusion gene of chronic myeloid leukemia (CML). This is mediated by transcriptional events and driven by DNA hyper-methylation at promoter-associated CpG islands of the CBY1-encoding gene C22orf2. Moreover, CBY1 transcriptional induction proceeding from promoter de-methylation is a component of BCR-ABL1+ cell response to Imatinib (IM). Our study showed that DNA methyltransferase 1 (DNMT1) has a central role in the hyper-methylation at the C22orf2 promoter. Further investigation in leukemic hematopoietic progenitors from IM-responsive and IM-resistant CML patients at diagnosis failed to demonstrate any correlation between DNMT1-driven hyper-methylation of the C22orf2 promoter and response to IM. Notably, the response to IM was neither predicted by DNMT1-driven hyper-methylation of BCL2-like11 at diagnosis. In conclusion, the hypermethylation of C22orf2 and BCL2-like11 promoters proceeding from DNMT1 is a crucial component of their reduced expression, but it is not directly involved in CML resistance to IM. It might rather contribute to the disease evolution towards a drug-resistant phenotype in more advanced phases or blast crisis.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas de Transporte/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Resistencia a Medicamentos Antineoplásicos , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA/efeitos dos fármacos , Proteínas de Fusão bcr-abl/genética , Humanos , Mesilato de Imatinib/farmacologia , Mesilato de Imatinib/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , beta Catenina/antagonistas & inibidores
6.
Cell Signal ; 26(8): 1690-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24747551

RESUMO

Autophagy protects chronic myeloid leukemia stem cells from tyrosine kinase inhibitors hence supporting the disease persistence under therapy. However, the signals involved in autophagy regulation relative to BCR-ABL1 are still elusive. The autophagic flux proceeding from the inhibition of BCR-ABL1 tyrosine kinase represents a regulatory mechanism of ß-catenin stability through events encompassing the activation of calpain, which targets ß-catenin for proteasome-independent degradation. Accordingly, its inactivation may contribute to induce autophagy and autophagy induction may, in turn, promote ß-catenin autolysosomal degradation to originate a regulatory loop where ß-catenin plays a central role in cell decision between life and death. Here we proved that the cytoplasmic accumulation of ß-catenin driven by up-regulation of its antagonist Chibby1 is a component of autophagy induction in response to imatinib in BCR-ABL1+ cells opposing the apoptotic death. It is contingent upon ER stress and elevation of free Ca(2+) cytosolic concentration and results in the calpain cleavage into a 28kDa fragment implicated in ß-catenin proteasome-independent degradation. More important for BCR-ABL1+ cell survival and proliferation following IM treatment, might be the calpain-mediated cleavage of ß-catenin accumulated within the cytoplasmic compartment into a 75kDa fragment, still owning TCF-dependent transcriptional activity. Such a ß-catenin fragment might be crucial for BCR-ABL1+ cell survival following the fusion protein TK inhibition.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Benzamidas/farmacologia , Calpaína/metabolismo , Proteínas de Fusão bcr-abl/metabolismo , Piperazinas/farmacologia , Pirimidinas/farmacologia , beta Catenina/metabolismo , Cálcio/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático , Células Hep G2 , Humanos , Mesilato de Imatinib , Células K562 , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
7.
PLoS One ; 8(12): e81425, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24339928

RESUMO

Beta Catenin signaling is critical for the self-renewal of leukemic stem cells in chronic myeloid leukemia. It is driven by multiple events, enhancing beta catenin stability and promoting its transcriptional co-activating function. We investigated the impact of BCR-ABL1 on Chibby1, a beta catenin antagonist involved in cell differentiation and transformation. Relative proximity of the Chibby1 encoding gene (C22orf2) on chromosome 22q12 to the BCR breakpoint (22q11) lets assume its involvement in beta catenin activation in chronic myeloid leukemia as a consequence of deletions of distal BCR sequences encompassing one C22orf2 allele. Forty patients with chronic myeloid leukemia in chronic phase were analyzed for C22orf2 relocation and Chibby1 expression. Fluorescent in situ hybridization analyses established that the entire C22orf2 follows BCR regardless of chromosomes involved in the translocation. In differentiated hematopoietic progenitors (bone marrow mononuclear cell fractions) of 30/40 patients, the expression of Chibby1 protein was reduced below 50% of the reference value (peripheral blood mononuclear cell fractions of healthy persons). In such cell context, Chibby1 protein reduction is not dependent on C22orf2 transcriptional downmodulation; however, it is strictly dependent upon BCR-ABL1 expression because it was not observed at the moment of major molecular response under tyrosine kinase inhibitor therapy. Moreover, it was not correlated with the disease prognosis or response to therapy. Most importantly, a remarkable Chibby1 reduction was apparent in a putative BCR-ABL1+ leukemic stem cell compartment identified by a CD34+ phenotype compared to more differentiated hematopoietic progenitors. In CD34+ cells, Chibby1 reduction arises from transcriptional events and is driven by C22orf2 promoter hypermethylation. These results advance low Chibby1 expression associated with BCR-ABL1 as a component of beta catenin signaling in leukemic stem cells.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas Nucleares/metabolismo , beta Catenina/antagonistas & inibidores , Transporte Ativo do Núcleo Celular , Antígenos CD34/metabolismo , Proteínas de Transporte/genética , Núcleo Celular/metabolismo , Cromossomos Humanos Par 9/genética , Regulação para Baixo , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Modelos Moleculares , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/genética , Conformação de Ácido Nucleico , Transdução de Sinais , Ativação Transcricional , beta Catenina/genética , beta Catenina/metabolismo
8.
Cell Signal ; 25(9): 1820-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23707389

RESUMO

Chronic myeloid leukemia (CML) is a myeloproliferative disease caused by the constitutive tyrosine kinase (TK) activity of the BCR-ABL fusion protein. However, the phenotype of leukemic stem cells (LSC) is sustained by ß catenin rather than by the BCR-ABL TK. ß catenin activity in CML is contingent upon its stabilization proceeding from the BCR-ABL-induced phosphorylation at critical residues for interaction with the Adenomatous polyposis coli (APC)/Axin/glycogen synthase kinase 3 (GSK3) destruction complex or GSK3 inactivating mutations. Here we studied the impact of ß catenin antagonist Chibby (CBY) on ß catenin signaling in BCR-ABL1+ cells. CBY is a small conserved protein which interacts with ß catenin and impairs ß catenin-mediated transcriptional activation through two distinct molecular mechanisms: 1) competition with T cell factor (TCF) or lymphoid enhancer factor (LEF) for ß catenin binding; and 2) nuclear export of ß catenin via interaction with 14-3-3. We found that its enforced expression in K562 cell line promoted ß catenin cytoplasmic translocation resulting in inhibition of target gene transcription. Moreover, cytoplasmic accumulation of ß catenin activated the endoplasmic reticulum (ER) stress-associated pathway known as unfolded protein response (UPR). CBY-driven cytoplasmic accumulation of ß catenin is also a component of BCR-ABL1+ cell response to the TK inhibitor Imatinib (IM). It evoked the UPR activation leading to the induction of BCL2-interacting mediator of cell death (BIM) by UPR sensors. BIM, in turn, contributed to the execution phase of apoptosis in the activation of ER resident caspase 12 and mobilization of Ca(2+) stores.


Assuntos
Proteínas de Transporte/metabolismo , Citoplasma/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas Nucleares/metabolismo , Resposta a Proteínas não Dobradas , beta Catenina/metabolismo , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático , Humanos , Transporte Proteico , beta Catenina/análise
9.
J Cell Biochem ; 113(8): 2765-74, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22467095

RESUMO

The loss-of-function of ten-eleven-translocation (TET) 2, a Fe(2+) -oxoglutarate-dependent dioxygenase catalyzing 5 methyl cytosine (5mC) conversion into 5-hydroxymethylcytosine (5hmC), contributes to the hematopoietic transformation in vivo. The aim of our study was to elucidate its role in the phenotype of chronic myeloid leukemia (CML), a myeloproliferative disease caused by the Bcr-Abl rearranged gene. We first confirmed TET2 interaction with the Bcr-Abl protein predicted by a Fourier-based bioinformatic method. Such interaction led to TET2 cytoplasmatic compartmentalization in a complex tethered by the fusion protein tyrosine kinase (TK) and encompassing the Forkhead box O3a (FoxO3a) transcription factor. We then focused the impact of TET2 loss-of-function on epigenetic transcriptional regulation of Bcl2-interacting mediator (BIM), a pro-apoptotic protein transcriptionally regulated by FoxO3a. BIM downregulation is a critical component of CML progenitor extended survival and is also involved in the disease resistance to imatinib (IM). Here we reported that TET2 release from Bcr-Abl protein following TK inhibition in response to IM triggers a chain of events including TET2 nuclear translocation, re-activation of its enzymatic function at 5mC and recruitment at the BIM promoter followed by BIM transcriptional induction. 5hmC increment following TET2 re-activation was associated with the reduction of histone H3 tri-methylation at lysine 9 (H3K9me3), which may contribute with DNA de-methylation reported elsewhere to recast a permissive epigenetic "landscape" for FoxO3a transcriptional activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Citoplasma/metabolismo , Proteínas de Ligação a DNA/genética , Dioxigenases , Epigênese Genética/genética , Proteínas de Fusão bcr-abl/genética , Imunoprecipitação , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Camundongos , Proteínas Proto-Oncogênicas/genética
10.
Leuk Res ; 36(8): 1028-34, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22521726

RESUMO

The advantage of Aurora kinase (AK) inhibitors in chronic myeloid leukemia (CML) therapy mostly arises from "off-target" effects on tyrosine kinase (TK) activity of wild type (wt) or mutated Bcr-Abl proteins which drive the disease resistance to imatinib (IM). We proved that the AK inhibitor MK-0457 induces the growth arrest DNA damage-inducible (Gadd) 45a through recruitment of octamer-binding (Oct)-1 transcription factor at a critical promoter region for gene transcription and covalent modifications of histone H3 (lysine 14 acetylation, lysine 9 de-methylation). Such epigenetic chromatin modifications may depict a general mechanism promoting the re-activation of tumor suppressor genes silenced by Bcr-Abl.


Assuntos
Proteínas de Ciclo Celular/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Proteínas Nucleares/genética , Fator 1 de Transcrição de Octâmero/fisiologia , Piperazinas/farmacologia , Animais , Antineoplásicos/farmacologia , Aurora Quinases , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética/efeitos dos fármacos , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Proteínas Nucleares/metabolismo , Fator 1 de Transcrição de Octâmero/metabolismo , Piperazinas/uso terapêutico , Regiões Promotoras Genéticas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ativação Transcricional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
12.
Bioorg Med Chem Lett ; 21(22): 6867-71, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21962576

RESUMO

Targeting the binding site of 14-3-3 proteins lets the release of partner proteins involved in cell cycle progression, apoptosis, cytoskeletal rearrangement and transcriptional regulation and may therefore be regarded as an alternative strategy to integrate conventional therapeutic approaches against cancer. In the present work, we report the identification of two new small molecule inhibitors of 14-3-3σ/c-Abl protein-protein interaction (BV01 and BV101) discovered by means of computational methods. The most interesting compound (BV01) showed a lethal dose (LD(50)) in the low micromolar range against Ba/F3 murine cell lines expressing the Imatinib (IM)-sensitive wild type Bcr-Abl construct and the IM-resistant Bcr-Abl mutation T315I. BV01 interaction with 14-3-3σ was demonstrated by NMR studies and elucidated by docking. It blocked the binding domain of 14-3-3σ, hence promoting the release of the partner protein c-Abl (the one not involved in Bcr rearrangement), and its translocation to both the nuclear compartment and mitochondrial membranes to induce a pro-apoptotic response. Our results advance BV01 as a confirmed hit compound capable of eliciting apoptotic death of Bcr-Abl-expressing cells by interfering with 14-3-3σ/c-Abl protein-protein interaction.


Assuntos
Proteínas 14-3-3/metabolismo , Biomarcadores Tumorais/metabolismo , Desenho de Fármacos , Exonucleases/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas 14-3-3/antagonistas & inibidores , Proteínas 14-3-3/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/química , Linhagem Celular , Exonucleases/antagonistas & inibidores , Exonucleases/química , Exorribonucleases , Humanos , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Mapeamento de Interação de Proteínas , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores
13.
J Pharmacol Exp Ther ; 336(3): 596-604, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21041536

RESUMO

Resistance of chronic myeloid leukemia (CML) to tyrosine kinase inhibitor imatinib mesylate (IM) is most often due to point mutations in the Bcr-Abl fusion gene. T315I mutation (resulting in substitution of Ile for a Thr residue at the "gatekeeper" position 315) raises particular concern, because it also provides resistance to second-generation kinase inhibitors already approved for clinical use (nilotinib and dasatinib). Much effort is therefore focused on alternative molecular-based strategies. Previous studies proved that binding to 14-3-3 scaffolding proteins leads to cytoplasmic compartmentalization and suppression of proapoptotic and antiproliferative signals associated with Bcr-Abl protein kinase, hence contributing to leukemic clone expansion. Here we investigated the effect of 14-3-3 inhibition disruption on hematopoietic cells expressing the IM-sensitive wild type Bcr-Abl and the IM-resistant T315I mutation. Using a virtual screening protocol and docking simulations, we identified a nonpeptidic inhibitor of 14-3-3, named BV02, that exhibits a remarkable cytotoxicity against both cell types. c-Abl release from 14-3-3σ, promoting its relocation to nuclear compartment (where it triggers transcription of p73-dependent proapoptotic genes) and to mitochondrial membranes (where it induces the loss of mitochondrial transmembrane potential) combined with c-Abl enhanced association with caspase 9 (a critical step of sequential caspase activation further contributing to c-Abl pro-apoptotic function) has a prominent role in the effect of BV02 on Bcr-Abl-expressing cells. In conclusion, BV02 may be considered as a treatment option for CML and, in particular, for more advanced phases of the disease that developed IM resistance as a consequence of Bcr-Abl point mutations.


Assuntos
Proteínas 14-3-3/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Piperazinas/uso terapêutico , Pirazóis/farmacologia , Pirimidinas/uso terapêutico , Proteínas 14-3-3/química , Sequência de Aminoácidos , Animais , Antineoplásicos/farmacologia , Apoptose/fisiologia , Benzamidas/química , Benzamidas/uso terapêutico , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Camundongos , Dados de Sequência Molecular , Piperazinas/farmacologia , Pirazóis/química , Pirazóis/uso terapêutico , Pirimidinas/farmacologia
14.
Bioorg Med Chem Lett ; 20(20): 6133-7, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20832303

RESUMO

An in silico structure-based ligand design approach resulted in the identification of the first non-peptidic small molecule able to inhibit protein-protein interactions between 14-3-3 and c-Abl. This compound shows an anti-proliferative effect on human leukemia cells either sensitive or resistant to Imatinib, in consequence of the T315I mutation. It also mediates c-Abl release from 14-3-3 in a way similar to that found in response to Imatinib treatment.


Assuntos
Proteínas 14-3-3/antagonistas & inibidores , Antineoplásicos/química , Antineoplásicos/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas 14-3-3/metabolismo , Apoptose/efeitos dos fármacos , Benzamidas , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Modelos Moleculares , Piperazinas/farmacologia , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/farmacologia
15.
Cell Signal ; 22(8): 1247-53, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20388540

RESUMO

Slug, a Snail-related zinc-finger transcription factor implicated in the increased motility of mesenchymal cells during embryonic development and progression of cancer cells towards an invasive phenotype, plays a specific and critical role in the pathogenesis of Bcr-Abl-associated leukemias. Here we report that Slug over-expression associated with Bcr-Abl is conditional upon the tyrosine kinase (TK) activity of 210 fusion protein. Slug over-expression is driven by transcriptional events eventually integrated by post-transcriptional mechanisms leading to protein stabilization and is at least partly regulated by the ERK1/2 mitogen-activated protein kinase (MAPK). It contributes to apoptosis resistance of leukemic progenitors through the repression of pro-apoptotic Puma. Moreover, Slug is a component of leukemic progenitor resistance to imatinib mesylate (IM) driven by Bcr-Abl point mutations and, in particular, by T315I. Slug over-expression associated with p210 Bcr-Abl TK either in the wild type (wt) or mutated conformation results in a significant reduction of E-cadherin, the substrate of Beta catenin at cell membranes. In conclusion, our results suggest that Slug has a central role in a complex network involved in prolonged survival and IM resistance of CML progenitors.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Fatores de Transcrição/metabolismo , Animais , Antineoplásicos/farmacologia , Benzamidas , Caderinas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Proteínas de Fusão bcr-abl/metabolismo , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Camundongos , Piperazinas/farmacologia , Pirimidinas/farmacologia , Fatores de Transcrição da Família Snail
16.
Leuk Res ; 34(5): 641-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19643477

RESUMO

Constitutive tyrosine kinase (TK) activity of p210 BCR-ABL fusion protein of chronic myeloid leukemia (CML) usurps physiological functions of normal p145 c-ABL protein. Accordingly, its inhibition by imatinib mesylate (IM) lets p145 c-ABL translocate into the nuclear compartment, which drives cell growth arrest and apoptotic death. Here we show that IM and the mammalian target of rapamycin (mTOR) inhibitor RAD001 (Everolimus) have additive effects on BCR-ABL-expressing cells. Those effects are at least partly conditional upon the enhanced nuclear accumulation of p145 c-ABL through events encompassing post-translational modifications of p145 c-ABL (Thr(735) phosphorylation) precluding its nuclear export and of 14-3-3 sigma (Ser(186) phosphorylation by c-Jun N-terminal kinase [JNK]) promoting p145 c-ABL nuclear re-import.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Expressão Gênica/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas Proto-Oncogênicas c-abl/efeitos dos fármacos , Benzamidas , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Separação Celular , Everolimo , Citometria de Fluxo , Humanos , Mesilato de Imatinib , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Microscopia Confocal , Piperazinas/administração & dosagem , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/biossíntese , Pirimidinas/administração & dosagem , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Serina-Treonina Quinases TOR
18.
Traffic ; 10(6): 637-47, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19220809

RESUMO

Here we demonstrated that the 'loss of function' of not-rearranged c-ABL in chronic myeloid leukemia (CML) is promoted by its cytoplasmic compartmentalization bound to 14-3-3 sigma scaffolding protein. In particular, constitutive tyrosine kinase (TK) activity of p210 BCR-ABL blocks c-Jun N-terminal kinase (JNK) phosphorylation leading to 14-3-3 sigma phosphorylation at a critical residue (Ser(186)) for c-ABL binding in response to DNA damage. Moreover, it is associated with 14-3-3 sigma over-expression arising from epigenetic mechanisms (promoter hyper-acetylation). Accordingly, p210 BCR-ABL TK inhibition by the TK inhibitor Imatinib mesylate (IM) evokes multiple events, including JNK phosphorylation at Thr(183), p38 mitogen-activated protein kinase (MAPK) phosphorylation at Thr(180), c-ABL de-phosphorylation at Ser residues involved in 14-3-3 binding and reduction of 14-3-3 sigma expression, that let c-ABL release from 14-3-3 sigma and nuclear import, and address BCR-ABL-expressing cells towards apoptotic death. Informational spectrum method (ISM), a virtual spectroscopy method for analysis of protein interactions based on their structure, and mathematical filtering in cross spectrum (CS) analysis identified 14-3-3 sigma/c-ABL binding sites. Further investigation on CS profiles of c-ABL- and p210 BCR-ABL-containing complexes revealed the mechanism likely involved 14-3-3 precluded phosphorylation in CML cells.


Assuntos
Proteínas 14-3-3/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Humanos , Ligantes , MAP Quinase Quinase 4/metabolismo , Fosforilação , Transporte Proteico
19.
ChemMedChem ; 4(1): 118-26, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19039816

RESUMO

Docking simulations were used to predict the most favorable interaction between the T315I mutated form of Abl (invariably associated with resistance to the tyrosine kinase inhibitor imatinib mesylate, IM) and C6-unsubstituted and substituted pyrazolo[3,4-d]pyrimidines previously found to be dual Src/Abl inhibitors. Two C6-unsubstituted (1 and 2) and eight C6-substituted compounds (3-10) were selected and assayed for their effects on the Ba/F3 cell line transducing the wild-type p210Bcr-Abl construct, which is IM-sensitive, or three of the most common mutations associated with IM resistance in vivo (T315I, Y253F, and E255K), and driven to drug resistance by saturating doses of IL-3 or by the expression of the Bcr-Abl construct coding for the p185 protein of acute lymphoblastic leukemia. Compounds 1 and 2 were active against all cell lines assayed (LD(50) range: 0.7-4.3 microM), whereas C6-substituted compounds exhibited lower activity (LD(50) approximately 8 microM for compound 3 toward the T315I mutant). Notably, 1 and 2 were also effective toward the T315I mutation, which is insensitive to dual Src/Abl inhibitors. The cytotoxic effects of 1 and 2 on IM-sensitive and IM-resistant Ba/F3 cells were attributable, at least in part, to their pro-apoptotic activity. Taken together, such findings suggest that C6-unsubstituted pyrazolo[3,4-d]pyrimidines may represent useful inhibitors to target IM-resistant chronic myeloid leukemia.


Assuntos
Antineoplásicos/química , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Pirimidinas/química , Quinases da Família src/antagonistas & inibidores , Substituição de Aminoácidos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Benzamidas , Linhagem Celular Tumoral , Simulação por Computador , Resistencia a Medicamentos Antineoplásicos , Mesilato de Imatinib , Camundongos , Modelos Moleculares , Mutação , Fosforilação , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/toxicidade , Proteínas Proto-Oncogênicas c-abl/química , Pirimidinas/farmacologia , Pirimidinas/toxicidade , Quinases da Família src/química
20.
Int J Radiat Biol ; 84(7): 591-601, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18661375

RESUMO

PURPOSE: To investigate the impact of TP53 (tumor protein 53, p53) on genomic stability of osteosarcoma (OS). MATERIALS AND METHODS: In first instance, we expressed in OS cell line SAOS-2 (lacking p53) a wild type (wt) p53 construct, whose protein undergoes nuclear import and activation in response to ionizing radiations (IR). Thereafter, we investigated genomic imbalances (amplifications and deletions at genes or DNA regions most frequently altered in human cancers) associated with radio-resistance relative to p53 expression by mean of an array-based comparative genomic hybridization (aCGH) strategy. Finally we investigated a putative marker of radio-induced oxidative stress, a 4,977 bp deletion at mitochondrial (mt) DNA usually referred to as 'common' deletion, by mean of a polimerase chain reaction (PCR) strategy. RESULTS: In radio-resistant subclones generated from wt p53-transfected SAOS-2 cells DNA deletions were remarkably reduced and the accumulation of 'common' deletion at mtDNA (that may let the persistence of oxidative damage by precluding detoxification from reactive oxygen species [ROS]) completely abrogated. CONCLUSIONS: The results of our study confirm that wt p53 has a role in protection of OS cell DNA integrity. Multiple mechanisms involved in p53 safeguard of genomic integrity and prevention of deletion outcome are discussed.


Assuntos
DNA Mitocondrial/efeitos da radiação , Instabilidade Genômica/efeitos da radiação , Osteoblastos/efeitos da radiação , Osteossarcoma , Radiação Ionizante , Espécies Reativas de Oxigênio/efeitos da radiação , Proteína Supressora de Tumor p53/efeitos da radiação , Linhagem Celular Tumoral , DNA Mitocondrial/metabolismo , Instabilidade Genômica/genética , Instabilidade Genômica/fisiologia , Humanos , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Osteoblastos/citologia , Osteoblastos/fisiologia , Osteossarcoma/genética , Osteossarcoma/patologia , Espécies Reativas de Oxigênio/metabolismo , Seleção Genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...