Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Ann Oncol ; 23(10): 2552-2561, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22431701

RESUMO

BACKGROUND: Predictive markers of response to chemotherapy are lacking in breast cancer patients. Forkhead Box Protein 3 (FOXP3) is an anti-oncogene whose absence in cancer cells could confer resistance to DNA damaging agent. So we made the hypothesis that FOXP3 expression predicts the response to anthracyclines in breast cancer patients and that adjuvant chemotherapy adding taxanes to anthracyclines confers an overall survival (OS) benefit over anthracyclines alone, in patients with FOXP3-negative tumors. PATIENTS AND METHODS: Expression of FOXP3 in cancer cells was evaluated by immunohistochemistry in tumor samples from 1097 patients who participated in the PACS01 randomized trial that evaluated in adjuvant setting the adjunction of docetaxel (Taxotere) to anthracyclines in patients with localized breast cancer. Kaplan-Meier analysis and Cox regression model were used to assess OS according to the presence or absence of FOXP3 expression in tumor cells. RESULTS: Four hundred and five tumors were found to express FOXP3 (37%). FOXP3 expression in breast cancer cells was associated with better OS (P = 0.003). Uni- and multivariate survival analyses according to treatment arm revealed that FOXP3 expression in breast cancer cells is independently associated with improved OS in patients treated with anthracycline-based adjuvant chemotherapy, but not in patients treated with sequential anthracycline-taxane. Moreover, in vitro experiments showed that FOXP3 induction in breast cancer cell lines using histone deacetylase inhibitor enhances anthracyclines efficacy. CONCLUSION: FOXP3 expression in tumor cells may be an accurate predictive biomarker of anthracycline efficacy in breast cancer.


Assuntos
Antraciclinas/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Fatores de Transcrição Forkhead/metabolismo , Biomarcadores Tumorais , Neoplasias da Mama/metabolismo , Quimioterapia Adjuvante , Feminino , Humanos
2.
Br J Cancer ; 105(3): 366-71, 2011 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-21750556

RESUMO

BACKGROUND: In HER2-overexpressing breast cancer, accumulating preclinical evidences suggest that some chemotherapies, like trastuzumab, but also taxanes, are able to trigger a T helper 1 (Th1) anticancer immune response that contribute to treatment success. T helper 1 immune response is characterised by the expression of the transcription factor T-bet in CD4 T lymphocytes. We hypothesised that the presence of such T cells in the tumour immune infiltrates following neoadjuvant chemotherapy would predict patient survival. METHODS: In a series of 102 consecutive HER2-overexpressing breast cancer patients treated by neoadjuvant chemotherapy incorporating antracyclines or taxane and trastuzumab, we studied by immunohistochemistry the peritumoral lymphoid infiltration by T-bet+ lymphocytes before and after chemotherapy in both treatment groups. Kaplan-Meier analysis and Cox modelling were used to assess relapse-free survival (RFS). RESULTS: Fifty-eight patients have been treated with trastuzumab-taxane and 44 patients with anthracyclines-based neoadjuvant chemotherapy. The presence of T-bet+ lymphocytes in peritumoral lymphoid structures after chemotherapy was significantly more frequent in patients treated with trastuzumab-taxane (P=0.0008). After a median follow-up of 40 months, the presence of T-bet+ lymphocytes after neoadjuvant chemotherapy confers significantly better RFS (log-rank test P=0.011) only in patients treated with trastuzumab-taxane. In this population, multivariate Cox regression model showed that only the presence of T-bet+ lymphocytes in peritumoral lymphoid structures after neoadjuvant chemotherapy was independently associated with improved RFS (P=0.04). CONCLUSION: These findings indicate that the tumour infiltration by T-bet+ Th1 lymphocytes following neoadjuvant trastuzumab-taxane may represent a new independent prognostic factor of improved outcome in HER2-overexpressing breast carcinoma.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Linfócitos T CD4-Positivos/metabolismo , Genes erbB-2 , Tecido Linfoide/metabolismo , Proteínas com Domínio T/metabolismo , Taxoides/administração & dosagem , Antraciclinas , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/mortalidade , Docetaxel , Feminino , Humanos , Pessoa de Meia-Idade , Terapia Neoadjuvante , Prognóstico , Trastuzumab
3.
Oncogene ; 25(58): 7635-45, 2006 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-16767158

RESUMO

Anticancer drugs can induce tumor cell death by caspase-dependent apoptosis. The observation that procaspase-10 expression decreased in leukemic cells from acute myeloblastic leukemia patients at first relapse led us to explore the role of caspase-10 in cytotoxic drug-induced apoptosis. We show that caspase-10 is activated in etoposide-treated cells in a dose- and time-dependent manner. A caspase-10 peptide inhibitor, a caspase-10 dominant-negative mutant or a small interfering RNA (siRNA)-mediated downregulation of the enzyme negatively interfere with drug-induced cell death and caspase-2, -3, -8 and -9 activation. The extrinsic pathway to apoptosis is not involved in drug-induced caspase-10 activation that occurs downstream of Bax redistribution to mitochondria and cytochrome c release from this organelle. siRNA-mediated downregulation of Apaf-1 prevents etoposide-mediated activation of caspase-10. In a cell-free assay, cytochrome c and dATP treatment of cell extracts after immunodepletion of either caspase-3 or caspase-9 indicates that caspase-10 is activated downstream of caspase-9. Then, caspase-10 is involved in a feedback amplification loop that amplifies caspase-9 and -3 activities. Altogether, these data indicate an active role for caspase-10 in cytotoxic drug-induced tumor cell death, downstream of the mitochondria.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose , Caspase 10/fisiologia , Etoposídeo/farmacologia , Transdução de Sinais , Apoptossomas/efeitos dos fármacos , Apoptossomas/metabolismo , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Leucemia Mieloide Aguda/diagnóstico , Mitocôndrias/metabolismo , Recidiva
4.
Oncogene ; 35(35): 4611-22, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-26853468

RESUMO

The omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) has anti-inflammatory and anti-cancer properties. Among pro-inflammatory mediators, tumor necrosis factor α (TNFα) plays a paradoxical role in cancer biology with induction of cancer cell death or survival depending on the cellular context. The objective of the study was to evaluate the role of TNFα in DHA-mediated tumor growth inhibition and colon cancer cell death. The treatment of human colorectal cancer cells, HCT-116 and HCT-8 cells, with DHA triggered apoptosis in autocrine TNFα-dependent manner. We demonstrated that DHA-induced increased content of TNFα mRNA occurred through a post-transcriptional regulation via the down-regulation of microRNA-21 (miR-21) expression. Treatment with DHA led to nuclear accumulation of Foxo3a that bounds to the miR-21 promoter triggering its transcriptional repression. Moreover, inhibition of RIP1 kinase and AMP-activated protein kinase α reduced Foxo3a nuclear-cytoplasmic shuttling and subsequent increase of TNFα expression through a decrease of miR-21 expression in DHA-treated colon cancer cells. Finally, we were able to show in HCT-116 xenograft tumor-bearing nude mice that a DHA-enriched diet induced a decrease of human miR-21 expression and an increase of human TNFα mRNA expression limiting tumor growth in a cancer cell-derived TNFα dependent manner. Altogether, the present work highlights a novel mechanism for anti-cancer action of DHA involving colon cancer cell death mediated through autocrine action of TNFα.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/administração & dosagem , MicroRNAs/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Animais , Apoptose/efeitos dos fármacos , Comunicação Autócrina , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Proteína Forkhead Box O3/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncogene ; 20(2): 260-9, 2001 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-11313953

RESUMO

Procaspase-2 is one of the cysteine aspartate proteases involved in apoptotic cell death. Alternative splicing of CASP-2 messenger RNA generates a long isoform, procaspase-2L, whose overexpression induces cell death and a truncated isoform, procaspase-2S, whose function remains poorly defined. The present study explored the consequences of procaspase-2S overexpression in U937 human leukemic cells exposed to the topoisomerase II inhibitor etoposide as an apoptotic stimulus. Overexpression of procaspase-2S in U937 cells partially prevented nuclear changes associated with etoposide-induced cell death, as determined by Hoechst 33342 staining of nuclear chromatin and electron microscopy studies. Procaspase-2S also prevented the maturation of apoptotic bodies, delayed phosphatidylserine externalization on the plasma membrane and prevented the cleavage and activation of procaspase-2L. These effects were not observed when the cysteine 289 in the consensus QACRG motif was mutated into a serine. Wild-type procaspase-2S overexpression did not influence the cleavage of procaspase-3, procaspase-7 and poly(ADP-ribose)polymerase nor the fragmentation of nuclear DNA into nucleosome-sized fragments. Altogether, these results indicate that the short isoform of procaspase-2 negatively interferes with selective features of apoptosis, an activity that is suppressed by mutation of the cysteine 289.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Cromatina/ultraestrutura , Precursores Enzimáticos/metabolismo , Fosfatidilserinas/metabolismo , Motivos de Aminoácidos , Apoptose/efeitos dos fármacos , Sequência de Bases , Caspase 2 , Caspases/genética , Cisteína , Fragmentação do DNA , Inibidores Enzimáticos/farmacologia , Precursores Enzimáticos/genética , Etoposídeo/farmacologia , Humanos , Isoenzimas , Leucemia , Dados de Sequência Molecular , Mutação , Inibidores da Topoisomerase II , Transfecção , Células Tumorais Cultivadas
6.
Therapie ; 56(5): 511-8, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11806287

RESUMO

Anticancer drugs can induce tumour cell death by apoptosis. The main pathway from specific damage induced by the drug to apoptosis involves activation of caspases in the cytosol by pro-apoptotic molecules such as cytochrome c released from the mitochondria. At least in some cell types, anticancer drugs also upregulate the expression of death receptors and sensitize tumour cells to their cognate ligands, which could be used to amplify the response to cytotoxic drugs. The Bcl-2 family of proteins, which includes anti- and pro-apoptotic molecules, regulates cell sensitivity at the mitochondrial level. Chemotherapeutic drugs modulate their expression (e.g. through p53-dependent gene transcription), their activity (e.g. by phosphorylation) and their subcellular localization (e.g. by translocation of pro-apoptotic proteins from the cytosol to the mitochondria). When interacting with tumour cells, anticancer drugs also activate lipid- and kinase-dependent signalling pathways that modulate the death response to specific damage. Protective pathways include activation of NF kappa B transcription factor, accumulation of heat shock proteins and activation of proteins involved in cell cycle regulation. The recent identification on these pathways to cell death has suggested several new strategies to improve the therapeutic efficacy of currently used anticancer drug regimens.


Assuntos
Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Citotoxinas/toxicidade , Neoplasias/tratamento farmacológico , Humanos , Neoplasias/patologia , Neoplasias/fisiopatologia , Células Tumorais Cultivadas
7.
Cell Death Differ ; 21(12): 1914-24, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25124554

RESUMO

Liver X receptors (LXRs) have been proposed to have some anticancer properties, through molecular mechanisms that remain elusive. Here we report for the first time that LXR ligands induce caspase-1-dependent cell death of colon cancer cells. Caspase-1 activation requires Nod-like-receptor pyrin domain containing 3 (NLRP3) inflammasome and ATP-mediated P2 × 7 receptor activation. Surprisingly, LXRß is mainly located in the cytoplasm and has a non-genomic role by interacting with pannexin 1 leading to ATP secretion. Finally, LXR ligands have an antitumoral effect in a mouse colon cancer model, dependent on the presence of LXRß, pannexin 1, NLRP3 and caspase-1 within the tumor cells. Our results demonstrate that LXRß, through pannexin 1 interaction, can specifically induce caspase-1-dependent colon cancer cell death by pyroptosis.


Assuntos
Apoptose , Receptores Nucleares Órfãos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antineoplásicos/farmacologia , Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Conexinas/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Hidrocarbonetos Fluorados/farmacologia , Receptores X do Fígado , Camundongos Endogâmicos BALB C , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transplante de Neoplasias , Proteínas do Tecido Nervoso/metabolismo , Receptores Nucleares Órfãos/agonistas , Sulfonamidas/farmacologia , Carga Tumoral/efeitos dos fármacos
9.
Blood ; 97(6): 1835-44, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11238127

RESUMO

Engagement of the plasma membrane receptor Fas can induce apoptosis of leukemic cells. Signaling through Fas requires the formation of a death-inducing signaling complex (DISC) that involves the cytoplasmic domain of Fas, the adaptor molecule FADD/MORT-1, and procaspase-8. The present study investigated whether another caspase, known as procaspase-2L, played a role in Fas-mediated cell death. A series of human leukemic variant cells was derived by stable transfection with a CASP2L antisense construct (CASP2L/AS). Specific down-regulation of procaspase-2L decreased the sensitivity of these cells to apoptosis induced by an agonistic anti-Fas antibody (Ab, clone CH11), as determined by studying DNA fragmentation, chromatin condensation, and externalization of phosphatidylserine on the plasma membrane. In leukemic cells transfected with an empty vector, anti-Fas Ab treatment activated caspase-8, decreased the expression of the BH3 domain-only protein Bid, triggered the release of cytochrome c from the mitochondria to the cytosol, and activated caspase-3. All these events could not be observed when CASP2L/AS cells were similarly treated with anti-Fas Abs. CASP2L/AS transfection did not inhibit the formation of the DISC and no direct interaction between procaspase-2L and either Fas or FADD or procaspase-8 was identified. Down-regulation of procaspase-2L inhibited anti-Fas Ab-mediated cleavage of c-FLIP (FLICE-inhibitory protein), a protein that interferes with the formation of a functional DISC. These results suggest that the long isoform of caspase-2 plays a role in the Fas-mediated pathway to cell death by contributing to caspase-8 activation at the DISC level.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Leucemia/tratamento farmacológico , Receptor fas/farmacologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Caspase 2 , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/efeitos dos fármacos , Caspases/genética , Caspases/metabolismo , Caspases/farmacologia , Grupo dos Citocromos c/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , DNA Antissenso/farmacologia , Humanos , Leucemia/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/farmacologia , Isoformas de Proteínas/fisiologia , Transfecção , Células Tumorais Cultivadas
10.
Blood ; 97(12): 3931-40, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11389037

RESUMO

Exposure of U937 human leukemic cells to the phorbol ester 12-O-tetradecanoylphorbol 13-acetate (TPA) induces their differentiation into monocyte/macrophage-like cells. This terminal differentiation is associated with a resistant phenotype to apoptosis induced by the topoisomerase II inhibitor etoposide. The inhibition occurs upstream of the mitochondrial release of cytochrome c and the activation of procaspase-2, -3, -6, -7, -8, and -9. By using cell-free systems, it was demonstrated that the mitochondrial pathway to cell death that involves mitochondrial membrane depolarization, cytochrome c release and cytosolic activation of procaspases by cytochrome c/dATP remains functional in TPA-differentiated U937 cells. Accordingly, 2 drugs recently shown to target the mitochondria, namely lonidamine and arsenic trioxide, bypass the resistance of TPA-differentiated U937 cells to classical anticancer drugs. Cell death induced by the 2 compounds is associated with mitochondrial membrane depolarization, release of cytochrome c and Smac/Diablo from the mitochondria, activation of caspases, poly(ADP-ribose) polymerase cleavage and internucleosomal DNA fragmentation. Moreover, the decreased glutathione content associated with the differentiation process amplifies the ability of arsenic trioxide to activate the mitochondrial pathway to cell death. Similar results were obtained by comparing undifferentiated and TPA-differentiated human HL60 leukemic cells. These data demonstrate that mitochondria-targeting agents bypass the resistance to classical anticancer drugs induced by TPA-mediated leukemic cell differentiation. (Blood. 2001;97:3931-3940)


Assuntos
Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Indazóis/farmacologia , Leucemia/patologia , Mitocôndrias/efeitos dos fármacos , Óxidos/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Antineoplásicos/farmacologia , Trióxido de Arsênio , Caspases/efeitos dos fármacos , Caspases/fisiologia , Diferenciação Celular/efeitos dos fármacos , Sistema Livre de Células/efeitos dos fármacos , Sistema Livre de Células/enzimologia , Resistência a Medicamentos , Etoposídeo/farmacologia , Humanos , Mitocôndrias/enzimologia , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células U937/efeitos dos fármacos , Células U937/enzimologia , Células U937/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA