Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 12(8): 785, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381018

RESUMO

Pediatric gliomas comprise a broad range of brain tumors derived from glial cells. While high-grade gliomas are often resistant to therapy and associated with a poor outcome, children with low-grade gliomas face a better prognosis. However, the treatment of low-grade gliomas is often associated with severe long-term adverse effects. This shows that there is a strong need for improved treatment approaches. Here, we highlight the potential for repurposing disulfiram to treat pediatric gliomas. Disulfiram is a drug used to support the treatment of chronic alcoholism and was found to be effective against diverse cancer types in preclinical studies. Our results show that disulfiram efficiently kills pediatric glioma cell lines as well as patient-derived glioma stem cells. We propose a novel mechanism of action to explain disulfiram's anti-oncogenic activities by providing evidence that disulfiram induces the degradation of the oncoprotein MLL. Our results further reveal that disulfiram treatment and MLL downregulation induce similar responses at the level of histone modifications and gene expression, further strengthening that MLL is a key target of the drug and explaining its anti-oncogenic properties.


Assuntos
Alcoolismo/tratamento farmacológico , Dissulfiram/uso terapêutico , Glioma/tratamento farmacológico , Glioma/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteólise , Auranofina/farmacologia , Auranofina/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Criança , Dissulfiram/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Reposicionamento de Medicamentos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/patologia , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação/efeitos dos fármacos , Gradação de Tumores , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
2.
Cell Death Discov ; 7(1): 87, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33931592

RESUMO

To sustain their malignancy, tumour cells acquire several metabolic adaptations such as increased oxygen, glucose, glutamine, and lipids uptake. Other metabolic processes are also enhanced as part of tumour metabolic reprogramming, for example, increased serine metabolism. Serine is a non-essential amino acid that supports several metabolic processes that are crucial for the growth and survival of proliferating cells, including protein, DNA, and glutathione synthesis. Indeed, increased activity of D-3-phosphoglycerate dehydrogenase (PHGDH), the enzyme rate-limiting de novo serine synthesis, has been extensively reported in several tumours. Therefore, selective inhibition of PHGDH may represent a new therapeutic strategy for over-expressing PHGDH tumours, owing to its downstream inhibition of essential biomass production such as one-carbon units and nucleotides. This perspective article will discuss the current status of research into small molecular inhibitors against PHGDH in colorectal cancer, breast cancer, and Ewing's sarcoma. We will summarise recent studies on the development of PHGDH-inhibitors, highlighting their clinical potential as new therapeutics. It also wants to shed a light on some of the key limitations of the use of PHGDH-inhibitors in cancer treatment which are worth taking into account.

3.
Cell Death Dis ; 10(11): 785, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31619667

RESUMO

Medulloblastoma (MB) is the most common malignant solid paediatric brain tumour. The standard treatment for MB is surgical resection of the tumour, radiation and chemotherapy. This therapy is associated with high morbidity and adverse side effects. Hence, more targeted and less toxic therapies are vitally needed to improve the quality of life of survivors. NPI-0052 is a novel proteasome inhibitor that irreversibly binds the 20S proteasome subunit. This compound has anti-tumour activity in metastatic solid tumours, glioblastoma and multiple myeloma with a good safety profile. Importantly, NPI-0052 has a lipophilic structure and can penetrate the blood-brain barrier, making it a suitable treatment for brain tumours. In the present study, we performed an in silico gene expression analysis to evaluate the proteasome subunit expression in MB. To evaluate the anticancer activity of NPI-0052, we used a range of MB patient-derived MB cells and cell lines. The synergistic cell death of NPI-0052 with γ-radiation was evaluated in tumour organoids derived from patient-derived MB cells. We show that high expression of proteasome subunits is a poor prognostic factor for MB patients. Also, our preclinical work demonstrated that NPI-0052 can inhibit proteasome activity and activate apoptosis in MB cells. Moreover, we observe that NPI-0052 has a synergistic apoptotic effect with γ-radiation, a component of the current MB therapy. Here, we present compelling preclinical evidence that NPI-0052 can be used as an adjuvant treatment for p53-family-expressing MB tumours.


Assuntos
Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/radioterapia , Raios gama/uso terapêutico , Lactonas/farmacologia , Meduloblastoma/tratamento farmacológico , Meduloblastoma/radioterapia , Pirróis/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Neoplasias Cerebelares/patologia , Quimiorradioterapia , Humanos , Meduloblastoma/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia
4.
Genes Dev ; 31(17): 1738-1753, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28971956

RESUMO

Medulloblastoma is the most common solid primary brain tumor in children. Remarkable advancements in the understanding of the genetic and epigenetic basis of these tumors have informed their recent molecular classification. However, the genotype/phenotype correlation of the subgroups remains largely uncharacterized. In particular, the metabolic phenotype is of great interest because of its druggability, which could lead to the development of novel and more tailored therapies for a subset of medulloblastoma. p73 plays a critical role in a range of cellular metabolic processes. We show overexpression of p73 in a proportion of non-WNT medulloblastoma. In these tumors, p73 sustains cell growth and proliferation via regulation of glutamine metabolism. We validated our results in a xenograft model in which we observed an increase in survival time in mice on a glutamine restriction diet. Notably, glutamine starvation has a synergistic effect with cisplatin, a component of the current medulloblastoma chemotherapy. These findings raise the possibility that glutamine depletion can be used as an adjuvant treatment for p73-expressing medulloblastoma.


Assuntos
Neoplasias Cerebelares/dietoterapia , Neoplasias Cerebelares/fisiopatologia , Glutamina/metabolismo , Meduloblastoma/dietoterapia , Meduloblastoma/fisiopatologia , Proteína Tumoral p73/genética , Proteína Tumoral p73/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/genética , Glutaminase/genética , Glutaminase/metabolismo , Xenoenxertos , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sobrevida , Serina-Treonina Quinases TOR/metabolismo , Resultado do Tratamento , Células Tumorais Cultivadas
5.
Cell Rep ; 20(2): 411-426, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28700942

RESUMO

Cell migration through the brain parenchyma underpins neurogenesis and glioblastoma (GBM) development. Since GBM cells and neuroblasts use the same migratory routes, mechanisms underlying migration during neurogenesis and brain cancer pathogenesis may be similar. Here, we identify a common pathway controlling cell migration in normal and neoplastic cells in the CNS. The nuclear scaffold protein promyelocytic leukemia (PML), a regulator of forebrain development, promotes neural progenitor/stem cell (NPC) and neuroblast migration in the adult mouse brain. The PML pro-migratory role is active also in transformed mouse NPCs and in human primary GBM cells. In both normal and neoplastic settings, PML controls cell migration via Polycomb repressive complex 2 (PRC2)-mediated repression of Slits, key regulators of axon guidance. Finally, a PML/SLIT1 axis regulates sensitivity to the PML-targeting drug arsenic trioxide in primary GBM cells. Taken together, these findings uncover a drug-targetable molecular axis controlling cell migration in both normal and neoplastic cells.


Assuntos
Sistema Nervoso Central/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/citologia , Glioblastoma/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Camundongos , Neurogênese/genética , Neurogênese/fisiologia , Lâmina Nuclear/metabolismo
6.
Neuron ; 74(1): 122-35, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22500635

RESUMO

Activity-dependent modifications of chromatin are believed to contribute to dramatic changes in neuronal circuitry. The mechanisms underlying these modifications are not fully understood. The histone variant H3.3 is incorporated in a replication-independent manner into different regions of the genome, including gene regulatory elements. It is presently unknown whether H3.3 deposition is involved in neuronal activity-dependent events. Here, we analyze the role of the histone chaperone DAXX in the regulation of H3.3 incorporation at activity-dependent gene loci. DAXX is found to be associated with regulatory regions of selected activity-regulated genes, where it promotes H3.3 loading upon membrane depolarization. DAXX loss not only affects H3.3 deposition but also impairs transcriptional induction of these genes. Calcineurin-mediated dephosphorylation of DAXX is a key molecular switch controlling its function upon neuronal activation. Overall, these findings implicate the H3.3 chaperone DAXX in the regulation of activity-dependent events, thus revealing a new mechanism underlying epigenetic modifications in neurons.


Assuntos
Proteínas de Transporte/metabolismo , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Potenciais da Membrana/fisiologia , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Proteínas Correpressoras , Epigênese Genética/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes Precoces , Camundongos , Camundongos Knockout , Chaperonas Moleculares , Rede Nervosa/crescimento & desenvolvimento , Rede Nervosa/metabolismo , Rede Nervosa/microbiologia , Condução Nervosa/fisiologia , Transcrição Gênica/fisiologia
7.
J Natl Cancer Inst ; 101(11): 828-32, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-19470951

RESUMO

Peptides that interfere with the natural resistance of cancer cells to genotoxin-induced apoptosis may improve the efficacy of anticancer regimens. We have previously reported that a cell-permeable RasGAP-derived peptide (TAT-RasGAP(317-326)) specifically sensitizes tumor cells to genotoxin-induced apoptosis in vitro. Here, we examined the in vivo stability of a protease-resistant D-form of the peptide, RI.TAT-RasGAP(317-326), and its effect on tumor growth in nude mice bearing subcutaneous human colon cancer HCT116 xenograft tumors. After intraperitoneal injection, RI.TAT-RasGAP(317-326) persisted in the blood of nude mice for more than 1 hour and was detectable in various tissues and subcutaneous tumors. Tumor-bearing mice treated daily for 7 days with RI.TAT-RasGAP(317-326) (1.65 mg/kg body weight) and cisplatin (0.5 mg/kg body weight) or doxorubicin (0.25 mg/kg body weight) displayed reduced tumor growth compared with those treated with either genotoxin alone (n = 5-7 mice per group; P = .004 and P = .005, respectively; repeated measures analysis of variance [ANOVA, two-sided]). This ability of the RI.TAT-RasGAP(317-326) peptide to enhance the tumor growth inhibitory effect of cisplatin was still observed at peptide doses that were at least 150-fold lower than the dose lethal to 50% of mice. These findings provide the proof of principle that RI.TAT-RasGAP(317-326) may be useful for improving the efficacy of chemotherapy in patients.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Proteínas Ativadoras de GTPase/farmacologia , Fragmentos de Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Antineoplásicos/administração & dosagem , Cisplatino/farmacologia , Neoplasias do Colo/patologia , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Sinergismo Farmacológico , Proteínas Ativadoras de GTPase/administração & dosagem , Células HCT116 , Humanos , Infusões Parenterais , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Mutagênicos/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Transplante Heterólogo
9.
Exp Cell Res ; 315(12): 2081-91, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19328779

RESUMO

The specific sensitization of tumor cells to the apoptotic response induced by genotoxins is a promising way of increasing the efficacy of chemotherapies. The RasGAP-derived fragment N2, while not regulating apoptosis in normal cells, potently sensitizes tumor cells to cisplatin- and other genotoxin-induced cell death. Here we show that fragment N2 in living cells is mainly located in the cytoplasm and only minimally associated with specific organelles. The cytoplasmic localization of fragment N2 was required for its cisplatin-sensitization property because targeting it to the mitochondria or the ER abrogated its ability to increase the death of tumor cells in response to cisplatin. These results indicate that fragment N2 requires a spatially constrained cellular location to exert its anti-cancer activity.


Assuntos
Apoptose , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Motivos de Aminoácidos , Animais , Antineoplásicos/farmacologia , Células Cultivadas , Cisplatino/farmacologia , Humanos , Camundongos , Proteínas Ativadoras de ras GTPase/genética
10.
J Photochem Photobiol B ; 88(1): 29-35, 2007 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-17560792

RESUMO

BACKGROUND: 5,10,15,20-Tetrakis(m-hydroxyphenyl)chlorin (mTHPC)-mediated photodynamic therapy (PDT) has shown insufficient tumor selectivity for the treatment of pleural mesothelioma. Tumor selectivity of mTHPC-PDT may be enhanced in the presence of the TAT-RasGAP(317-326) peptide which has the potential to specifically sensitize tumor cells to cytostatic agents. MATERIALS AND METHODS: H-meso-1 and human fibroblast cell cultures, respectively, were exposed to two different mTHPC doses followed by light delivery with and without TAT-RasGAP(317-326) administration. mTHPC was added to the cultures at a concentration of 0.04microg/ml and 0.10microg/ml, respectively, 24h before laser light illumination at 652nm (3J/cm(2), 40mW/cm(2)). TAT-RasGAP(317-326) was added to the cultures immediately after light delivery at a concentration of 20microM. The apoptosis rate was determined by scoring the cells displaying pycnotic nuclei. Cell viability was measured by using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. RESULTS: Light delivery associated with 0.04microg/ml mTHPC resulted in a significantly higher apoptosis rate in the presence of TAT-RasGAP(317-326) than without in H-meso-1 cells (p<0.05) but not in fibroblasts. In contrast, 1.0microg/ml mTHPC and light resulted in a significantly higher apoptosis rate in both H-meso-1 cells and fibroblasts as compared to controls (p<0.05) but the addition of TAT-RasGAP(317-326) did not lead to a further significant increase of the apoptosis rate of both H-meso-1 cells and fibroblasts as compared to mTHPC and light delivery alone. CONCLUSION: TAT-RasGAP(317-326) selectively enhanced the effect of mTHPC and light delivery on H-meso-1 cells but not on fibroblasts. However, this effect was mTHPC dose-dependent and occurred only at a low sensitizer dose.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas Ativadoras de GTPase/uso terapêutico , Mesoporfirinas/uso terapêutico , Mesotelioma/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Sobrevivência Celular , Fibroblastos/efeitos dos fármacos , Proteínas Ativadoras de GTPase/síntese química , Humanos , Mesotelioma/patologia , Fragmentos de Peptídeos/síntese química , Peptídeos/uso terapêutico , Células Tumorais Cultivadas
11.
Crit Rev Oncol Hematol ; 63(2): 160-71, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17544289

RESUMO

Agents that induce DNA damage in cells--the so-called genotoxins--have successfully been used for decades to treat patients with tumors. Genotoxins alter the DNA of cells, which is detected by DNA damage sensors and which leads to the activation of p53. Activation of p53 can lead to the death of cancer cells. The efficacy of genotoxins in humans is however limited by their toxicity to normal tissues. Specific sensitization of tumor cells to the action of genotoxins would reduce the efficacious doses of genotoxins to be used in patients, diminishing the detrimental side-effects of the drugs on normal tissues. A series of compounds able to sensitize cancer cells to DNA-damaging drugs have recently been identified that have the potential to increase the efficacy of currently used anti-cancer treatments.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Dano ao DNA/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias/metabolismo , Neoplasias/patologia
12.
Mol Cancer Res ; 5(5): 497-507, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17510315

RESUMO

Although chemotherapy has revolutionized cancer treatment, the associated side effects induced by lack of specificity to tumor cells remain a challenging problem. We have previously shown that TAT-RasGAP(317-326),a cell-permeable peptide derived from RasGAP, specifically sensitizes cancer cells to the action of genotoxins. The underlying mechanisms of this sensitization were not defined however. Here, we report that TAT-RasGAP(317-326) requires p53, but not the Ras effectors Akt and extracellular signal-regulated kinase, to mediate its tumor sensitization abilities. The TAT-RasGAP(317-326) peptide, although not modulating the transcriptional activity of p53 or its phosphorylation and acetylation status, nevertheless requires a functional p53 cellular status to increase the sensitivity of tumor cells to genotoxins. Genes regulated by p53 encode proapoptotic proteins, such as PUMA, and cell cycle control proteins, such as p21. The ability of TAT-RasGAP(317-326) to sensitize cancer cells was found to require PUMA but not p21. TAT-RasGAP(317-326) did not affect PUMA levels, however, but increased genotoxin-induced mitochondrial depolarization and caspase-3 activation. These results indicate that TAT-RasGAP(317-326) sensitizes tumor cells by activating signals that intersect with the p53 pathway downstream of, or at the level of, proapoptotic p53 target gene products to increase the activation of the mitochondrial death pathway.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Produtos do Gene tat/farmacologia , Mutagênicos/farmacologia , Neoplasias/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Ativadoras de ras GTPase/farmacologia , Acetilação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Produtos do Gene tat/química , Células HCT116 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias/metabolismo , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Termodinâmica , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteínas Ativadoras de ras GTPase/química
13.
Mol Biol Cell ; 16(8): 3511-20, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15901831

RESUMO

RasGAP bears two caspase-3 cleavage sites that are used sequentially as caspase activity increases in cells. When caspase-3 is mildly activated, RasGAP is first cleaved at position 455. This leads to the production of an N-terminal fragment, called fragment N, that activates the Ras-PI3K-Akt pathway and that promotes cell survival. At higher caspase activity, RasGAP is further cleaved at position 157 generating two small N-terminal fragments named N1 and N2. We have now determined the contribution of this second cleavage event in the regulation of apoptosis using cells in which the wild-type RasGAP gene has been replaced by a cDNA encoding a RasGAP mutant that cannot be cleaved at position 157. Our results show that cleavage of fragment N at position 157 leads to a marked reduction in Akt activity. This is accompanied by efficient processing of caspase-3 that favors cell death in response to various apoptotic stimuli. In nontumorigenic cells, fragments N1 and N2 do not modulate apoptosis. Therefore, the role of the second caspase-mediated cleavage of RasGAP is to allow the inactivation of the antiapoptotic function of fragment N so that caspases are no longer hampered in their ability to kill cells.


Assuntos
Apoptose , Caspases/metabolismo , Mutação/genética , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Caspase 3 , Linhagem Celular , Regulação para Baixo , Ativação Enzimática , Fibroblastos , Camundongos , Camundongos Knockout , Proteínas Ativadoras de ras GTPase/deficiência
14.
Mol Cell Biol ; 24(23): 10425-36, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15542850

RESUMO

Tight control of apoptosis is required for proper development and maintenance of homeostasis in multicellular organisms. Cells can protect themselves from potentially lethal stimuli by expressing antiapoptotic factors, such as inhibitors of apoptosis, FLICE (caspase 8)-inhibitory proteins, and members of the Bcl2 family. Here, we describe a mechanism that allows cells to survive once executioner caspases have been activated. This mechanism relies on the partial cleavage of RasGAP by caspase 3 into an amino-terminal fragment called fragment N. Generation of this fragment leads to the activation of the antiapoptotic Akt kinase, preventing further amplification of caspase activity. Partial cleavage of RasGAP is required for cell survival under stress conditions because cells expressing an uncleavable RasGAP mutant cannot activate Akt, cannot prevent amplification of caspase 3 activity, and eventually undergo apoptosis. Executioner caspases therefore control the extent of their own activation by a feedback regulatory mechanism initiated by the partial cleavage of RasGAP that is crucial for cell survival under adverse conditions.


Assuntos
Caspases/metabolismo , Proteínas Ativadoras de ras GTPase/química , Animais , Apoptose , Western Blotting , Caspase 3 , Caspases/química , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Eletroporação , Ativação Enzimática , Fibroblastos/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Células Jurkat , Lentivirus/genética , Camundongos , Modelos Genéticos , Mutação , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Estresse Fisiológico , Fatores de Tempo , Transfecção , Proteínas Ativadoras de ras GTPase/metabolismo
15.
Oncogene ; 23(55): 8971-8, 2004 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-15467750

RESUMO

Treatment of many cancers relies on the combined action of several genotoxins, but the detrimental effect of these drugs on normal cells can cause severe side effects. One major challenge in anticancer therapy is therefore to increase the selectivity of current treatments toward cancer cells in order to spare normal cells. We have recently demonstrated that a RasGAP caspase cleavage fragment is able to sensitize HeLa cells towards cisplatin-induced apoptosis. Here, we extend this observation by showing that this fragment also enhances cell death induced by adriamycin and mitoxantrone, two other widely used genotoxins. Furthermore, we have delineated a short sequence within this fragment that still bears the genotoxin-sensitization property. The peptide encoded by this sequence, when fused to the TAT cell permeation sequence, potently sensitized a number of tumors cells, but not normal cells, towards apoptosis induced by cisplatin, adriamycin and mitoxantrone. This sensitization effect was not mediated through modulation of NFkappaB activity or activation of the JNK and p38 MAPK pathways. Our results demonstrate the feasibility in enhancing the efficacy of currently used drugs to selectively kill cancer cells using peptides derived from pro-apoptotic caspase substrate fragments.


Assuntos
Proteínas Ativadoras de GTPase/química , Mutagênicos/metabolismo , Fragmentos de Peptídeos/química , Peptídeos/química , Proteínas Ativadoras de ras GTPase/metabolismo , Apoptose , Western Blotting , Linhagem Celular Tumoral , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Produtos do Gene tat/química , Genes Reporter , Células HeLa , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Luciferases/metabolismo , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mitoxantrona/farmacologia , NF-kappa B/metabolismo , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Domínios de Homologia de src
16.
Biochem Pharmacol ; 68(6): 1027-31, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15313397

RESUMO

Executioner caspases induce the biochemical and cellular changes characteristic of apoptosis. Activation of caspases is therefore regarded as "the kiss of death" resulting in the cell's demise. Recent reports indicate however that in some situations, caspase activation may induce other responses than apoptosis. These findings raise the question of how cells manage to counteract the killing activities of executioner caspases. Experiments performed in our laboratory have unraveled a mechanism that allows cells to survive in the presence of activated executioner caspases. This mechanism is based on the partial cleavage of RasGAP into an N-terminal fragment that activates the Ras-PI3K-Akt survival pathway. This protective pathway may be activated to allow cells to use executioner caspases for other purposes than inducing apoptosis.


Assuntos
Apoptose/fisiologia , Caspases/fisiologia , Proteínas Ativadoras de ras GTPase/fisiologia , Animais , Caspases/metabolismo , Sobrevivência Celular/fisiologia , Ativação Enzimática , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas Ativadoras de ras GTPase/uso terapêutico , Proteínas ras/metabolismo
17.
Carcinogenesis ; 25(6): 909-21, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14742324

RESUMO

p120 RasGTPase-activating protein (RasGAP), the main regulator of Ras GTPase family members, is cleaved at low caspase activity into an N-terminal fragment that triggers potent anti-apoptotic signals via activation of the Ras/PI-3 kinase/Akt pathway. When caspase activity is increased, RasGAP fragment N is further processed into two fragments that effectively potentiate apoptosis. Expression of RasGAP protein and its cleavage was assessed in human lung cancer cells with different histology and responsiveness to anticancer drug-induced apoptosis. Here we show that therapy-sensitive small lung carcinoma cell (SCLC) lines have lower RasGAP expression levels and higher spontaneous cleavage with formation of fragment N compared to therapy-resistant non-small cell lung carcinoma cell (NSCLC) lines. The first RasGAP cleavage event strongly correlated with the increased level of spontaneous apoptosis in SCLC. However, generation of protective RasGAP fragment N also related to the potency of SCLC to develop secondary therapy-resistance. In response to etoposide (ET), RasGAP fragment N was further cleaved in direct dependence on caspase-3 activity, which was more pronounced in NSCLC cells. Caspase inhibition, while effectively preventing the second cleavage of RasGAP, barely affected the first cleavage of RasGAP into fragment N that was always detectable in NSCLC and SCLC cells. These findings suggest that different levels of RasGAP and fragment N might play a significant role in the biology and different clinical course of both subtypes of lung neoplasms. Furthermore, constitutive formation of RasGAP fragment N can potentially contribute to primary resistance of NSCLC to anticancer therapy by ET but also to secondary therapy-resistance in SCLC.


Assuntos
Apoptose , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Pequenas/metabolismo , Caspases/metabolismo , Etoposídeo/farmacologia , Neoplasias Pulmonares/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Pequenas/enzimologia , Carcinoma de Células Pequenas/patologia , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA