Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Semin Cancer Biol ; 72: 36-45, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32619506

RESUMO

Breast cancer is the most common cancer in women with the highest mortality among this gender. Despite treatment strategies including surgery, hormone therapy and targeted therapy have recently advanced, innovative biomarkers are needed for the early detection, treatment and prognosis. An increasing number of non-coding RNAs (ncRNAs) have shown great potential as crucial players in different stages of the breast cancer tumorigenesis, influencing cell death, metabolism, epithelial-mesenchymal transition (EMT), metastasis and drug resistance. Long non-coding RNAs (lncRNAs), specifically, are a class of RNA transcripts with a length greater than 200 nucleotides, which have also been shown to exerts oncogenic or tumour suppressive roles in the pathogenesis of breast cancer. LncRNAs are implicated in different molecular mechanisms by regulating gene expressions and functions at transcriptional, translational, and post-translational levels. Here, we aim to briefly discuss the latest existing body of knowledge regarding the key functions and the molecular mechanisms of some of the most relevant lncRNAs in the pathogenesis, treatment and prognosis of breast cancer.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/terapia , Terapia de Alvo Molecular/métodos , RNA Longo não Codificante/genética , Animais , Neoplasias da Mama/genética , Gerenciamento Clínico , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos
2.
Leukemia ; 32(4): 911-919, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29209041

RESUMO

The E3 ubiquitin ligase (E3) WWP1 is an oncogenic factor implicated in the maintenance of different types of epithelial cancers. The role of WW domain-containing E3 ubiquitin protein ligase 1 (WWP1) in haematological neoplasms remains unknown. Acute myeloid leukaemia (AML) is characterized by the expansion of malignant myeloid cells blocked at different stages of differentiation. Here we report that the expression of WWP1 is significantly augmented in a large cohort of primary AML patients and in AML cell lines, compared with haematopoietic cells from healthy donors. We show that WWP1 inactivation severely impairs the growth of primary AML blasts and cell lines in vitro. In vivo, we observed a reduced leukaemogenic potential of WWP1-depleted AML cells upon transplantation into immunocompromised mice. Mechanistically, WWP1 inactivation induces the accumulation of its protein substrate p27Kip1, which ultimately contributes to G0/G1 cell cycle arrest of AML blasts. In addition, WWP1 depletion triggers the autophagy signalling and reduces survival of leukaemic cells. Collectively, our findings provide molecular insights into the anti-cancer potential of WWP1 inhibition, suggesting that this E3 is a promising biomarker and druggable target in AML.


Assuntos
Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fase G1/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Camundongos , Fase de Repouso do Ciclo Celular/fisiologia , Transdução de Sinais/fisiologia , Células U937 , Ubiquitinação/fisiologia
4.
Cell Death Dis ; 5: e1203, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24787015

RESUMO

Inhibition of distinct ubiquitin E3 ligases might represent a powerful therapeutic tool. ITCH is a HECT domain-containing E3 ligase that promotes the ubiquitylation and degradation of several proteins, including p73, p63, c-Jun, JunB, Notch and c-FLIP, thus affecting cell fate. Accordingly, ITCH depletion potentiates the effect of chemotherapeutic drugs, revealing ITCH as a potential pharmacological target in cancer therapy. Using high throughput screening of ITCH auto-ubiquitylation, we identified several putative ITCH inhibitors, one of which is clomipramine--a clinically useful antidepressant drug. Previously, we have shown that clomipramine inhibits autophagy by blocking autophagolysosomal fluxes and thus could potentiate chemotherapy in vitro. Here, we found that clomipramine specifically blocks ITCH auto-ubiquitylation, as well as p73 ubiquitylation. By screening structural homologs of clomipramine, we identified several ITCH inhibitors and putative molecular moieties that are essential for ITCH inhibition. Treating a panel of breast, prostate and bladder cancer cell lines with clomipramine, or its homologs, we found that they reduce cancer cell growth, and synergize with gemcitabine or mitomycin in killing cancer cells by blocking autophagy. We also discuss a potential mechanism of inhibition. Together, our study (i) demonstrates the feasibility of using high throughput screening to identify E3 ligase inhibitors and (ii) provides insight into how clomipramine and its structural homologs might interfere with ITCH and other HECT E3 ligase catalytic activity in (iii) potentiating chemotherapy by regulating autophagic fluxes. These results may have direct clinical applications.


Assuntos
Antidepressivos/farmacologia , Autofagia/efeitos dos fármacos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Sítios de Ligação , Linhagem Celular Tumoral , Clomipramina/análogos & derivados , Clomipramina/química , Clomipramina/farmacologia , Sinergismo Farmacológico , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , Reprodutibilidade dos Testes , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
5.
Cell Death Dis ; 4: e645, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23703390

RESUMO

p63 is a p53 family transcription factor, which besides unique roles in epithelial development, shares tumor suppressive activity with its homolog p53. The p63 gene has different transcriptional start sites, which generate two N-terminal isoforms (transactivation domain (TA)p63 and amino terminal truncated protein(ΔN)p63); in addition alternative splicing at the 5'-end give rise to at least five C-terminal isoforms. This complexity of gene structure has probably fostered the debate and controversy on p63 function in cancer, with TP63-harboring two distinctive promoters, codifying for the TAp63 and ΔNp63 isoforms, and having discrete functions. However, ΔNp63 also drives expression of target genes that have a relevant role in cancer and metastasis. In this study, we identified a novel p63 transcriptional target, caspase-1. Caspase-1 is proinflammatory caspase, which functions in tumor suppression. We show that both p63 isoforms promote caspase-1 expression by physical binding to its promoter. Consistent with our in vitro findings, we also identified a direct correlation between p63 and caspase-1 expression in human cancer data sets. In addition, survival estimation analysis demonstrated that functional interaction between p63 and caspase-1 represents a predictor of positive survival outcome in human cancers. Overall, our data report a novel p63 target gene involved in tumor suppression, and the clinical analysis underlines the biological relevance of this finding and suggests a further clinically predictive biomarker.


Assuntos
Caspase 1/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Caspase 1/genética , Linhagem Celular , Células HEK293 , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas Supressoras de Tumor/genética
6.
Oncogene ; 32(39): 4721-6, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-23085759

RESUMO

The Cullin4A (cul4A)-dependent ligase (CDL4A) E3 has been implicated in a variety of biological processes, including cell cycle progression and DNA damage response. Remarkably, CDL4A exerts its function through both proteolytic and non-proteolytic events. Here, we show that the p53 family member p73 is able to interact with the CDL4A complex through its direct binding to the receptor subunit DNA-binding protein 1 (DDB1). As a result, the CDL4A complex is able to monoubiquitylate p73. Modification of p73 by CDL4A-mediated ubiquitylation does not affect p73 protein stability, but negatively regulates p73-dependent transcriptional activity. Indeed, genetic or RNA interference-mediated depletion of DDB1 induces the expression of several p73 target genes in a p53-independent manner. In addition, by exploiting a bioinformatic approach, we found that elevated expression of Cul4A in human breast carcinomas is associated with repression of p73 target genes. In conclusion, our findings add a novel insight into the regulation of p73 by the CDL4A complex, through the inhibition of its transcriptional function.


Assuntos
Proteínas Culina/fisiologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/antagonistas & inibidores , Transcrição Gênica , Proteínas Supressoras de Tumor/antagonistas & inibidores , Animais , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Proteínas de Transporte/fisiologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Camundongos , Complexos Multiproteicos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Transcrição Gênica/fisiologia , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação
7.
Cold Spring Harb Perspect Biol ; 2(9): a004887, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20484388

RESUMO

p73 and p63 are two homologs of the tumor suppressive transcription factor p53. Given the high degree of structural similarity shared by the p53 family members, p73 and p63 can bind and activate transcription from the majority of the p53-responsive promoters. Besides overlapping functions shared with p53 (i.e., induction of apoptosis in response to cellular stress), the existence of extensive structural variability within the family determines unique roles for p63 and p73. Their crucial and specific functions in controlling development and differentiation are well exemplified by the p63 and p73 knockout mouse phenotypes. Here, we describe the contribution of p63 and p73 to human pathology with emphasis on their roles in tumorigenesis and development.


Assuntos
Proteínas de Ligação a DNA/genética , Evolução Molecular , Proteínas Nucleares/genética , Transativadores/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Animais , Humanos , Isoformas de Proteínas , Fatores de Transcrição , Proteína Tumoral p73
9.
Oncogene ; 28(35): 3157-66, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19581926

RESUMO

The transcription factor p73, a member of the p53 family, mediates cell-cycle arrest and apoptosis in response to DNA damage-induced cellular stress, acting thus as a proapoptotic gene. Similar to p53, p73 activity is regulated by post-translational modification, including phosphorylation, acetylation and ubiquitylation. In C. elegans, the F-box protein FSN-1 controls germline apoptosis by regulating CEP-1, the single ancestral p53 family member. Here we report that FBXO45, the human ortholog of FSN-1, binds specifically to p73 triggering its proteasome-dependent degradation. Importantly, SCF(FBXO45) ubiquitylates p73 both in vivo and in vitro. Moreover, siRNA-mediated depletion of FBXO45 stabilizes p73 and concomitantly induces cell death in a p53-independent manner. All together, these results show that the orphan F-box protein FBXO45 regulates the stability of p73, highlighting a conserved pathway evolved from nematode to human by which the p53 members are regulated by an SCF-dependent mechanism.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas F-Box/metabolismo , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Neoplasias da Mama/patologia , Células CHO , Morte Celular/genética , Linhagem Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Proteínas F-Box/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Hemaglutininas/metabolismo , Humanos , Rim/citologia , Leupeptinas/farmacologia , Mutação , Neuroblastoma/patologia , Inibidores de Proteassoma , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Especificidade por Substrato , Temperatura , Transfecção , Proteína Tumoral p73 , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
10.
Cell Death Differ ; 15(7): 1103-12, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18552861

RESUMO

The HECT-type E3 ubiquitin ligase (E3) Itch is absent in the non-agouti-lethal 18H or Itchy mice, which develop a severe immunological disease, including lung and stomach inflammation and hyperplasia of lymphoid and hematopoietic cells. The involvement of Itch in multiple signaling pathways and pathological conditions is presently an area of extensive scientific interest. This review aims to bring together a growing body of work exploring Itch-regulated biological processes, and to highlight recent discoveries on the regulatory mechanisms modulating its catalytic activity and substrate recognition capability. Our contribution is also an endeavor to correlate Itch substrate specificity with the pathological defects manifested by the mutant Itchy mice.


Assuntos
Sistema Imunitário/metabolismo , Neoplasias/enzimologia , Proteínas Repressoras/metabolismo , Pele/enzimologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Morte Celular , Receptores ErbB/metabolismo , Sistema Imunitário/patologia , Queratinócitos/metabolismo , Camundongos , Camundongos Mutantes , Neoplasias/imunologia , Neoplasias/patologia , Fosforilação , Transporte Proteico , Receptores de Quimiocinas/metabolismo , Proteínas Repressoras/imunologia , Transdução de Sinais , Pele/imunologia , Pele/patologia , Especificidade por Substrato , Canais de Cátion TRPC/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
11.
Cell Death Differ ; 9(9): 873-80, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12181738

RESUMO

By crossing Huntington's disease (HD) R6/1 transgenic mice with 'tissue' transglutaminase (TG2) knock-out mice, we have demonstrated that this multifunctional enzyme plays an important role in the neuronal death characterising this disorder in vivo. In fact, a large reduction in cell death is observed in R6/1, TG2(-/-) compared with R6/1 transgenic mice. In addition, we have shown that the formation of neuronal intranuclear inclusions (NII) is potentiated in absence of the 'tissue' transglutaminase. These phenomena are paralleled by a significant improvement both in motor performances and survival of R6/1, TG2(-/-) versus R6/1 mice. Taken together these findings suggest an important role for tissue transglutaminase in the regulation of neuronal cell death occurring in Huntington's disease.


Assuntos
Encéfalo/enzimologia , Morte Celular/genética , Proteínas de Ligação ao GTP/deficiência , Doença de Huntington/enzimologia , Degeneração Neural/enzimologia , Neurônios/enzimologia , Transglutaminases/deficiência , Animais , Encéfalo/patologia , Encéfalo/ultraestrutura , Modelos Animais de Doenças , Regulação para Baixo/genética , Feminino , Proteínas de Ligação ao GTP/genética , Guanosina Trifosfato/metabolismo , Doença de Huntington/genética , Doença de Huntington/mortalidade , Imuno-Histoquímica , Corpos de Inclusão/genética , Corpos de Inclusão/patologia , Corpos de Inclusão/ultraestrutura , Longevidade/genética , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Atividade Motora/genética , Neocórtex/enzimologia , Neocórtex/patologia , Neocórtex/ultraestrutura , Neostriado/enzimologia , Neostriado/patologia , Neostriado/ultraestrutura , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Proteína 2 Glutamina gama-Glutamiltransferase , Taxa de Sobrevida , Transglutaminases/genética
12.
Oncogene ; 20(32): 4305-16, 2001 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-11466611

RESUMO

Nitric oxide (NO), peroxynitrite, and 4-hydroxynonenal (HNE) may be involved in the pathological demise of cells via apoptosis. Apoptosis induced by these agents is inhibited by Bcl-2, suggesting the involvement of mitochondria in the death pathway. In vitro, NO, peroxynitrite and HNE can cause direct permeabilization of mitochondrial membranes, and this effect is inhibited by cyclosporin A, indicating involvement of the permeability transition pore complex (PTPC) in the permeabilization event. NO, peroxynitrite and HNE also permeabilize proteoliposomes containing the adenine nucleotide translocator (ANT), one of the key components of the PTPC, yet have no or little effects on protein-free control liposomes. ANT-dependent, NO-, peroxynitrite- or HNE-induced permeabilization is at least partially inhibited by recombinant Bcl-2 protein, as well as the antioxidants trolox and butylated hydroxytoluene. In vitro, none of the tested agents (NO, peroxynitrite, HNE, and tert-butylhydroperoxide) causes preferential carbonylation HNE adduction, or nitrotyrosylation of ANT. However, all these agents induced ANT to undergo thiol oxidation/derivatization. Peroxynitrite and HNE also caused significant lipid peroxidation, which was antagonized by butylated hydroxytoluene but not by recombinant Bcl-2. Transfection-enforced expression of vMIA, a viral apoptosis inhibitor specifically targeted to ANT, largely reduces the mitochondrial and nuclear signs of apoptosis induced by NO, peroxynitrite and HNE in intact cells. Taken together these data suggest that NO, peroxynitrite, and HNE may directly act on ANT to induce mitochondrial membrane permeabilization and apoptosis.


Assuntos
Aldeídos/farmacologia , Apoptose , Canais Iônicos , Translocases Mitocondriais de ADP e ATP/metabolismo , Nitratos/farmacologia , Óxido Nítrico/metabolismo , Oxidantes/farmacologia , Animais , Núcleo Celular/ultraestrutura , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose , Membranas Intracelulares/metabolismo , Células Jurkat , Peroxidação de Lipídeos , Proteínas de Membrana/fisiologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Permeabilidade , Proteínas/fisiologia , Proteolipídeos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia
13.
Med Pediatr Oncol ; 36(1): 115-7, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11464861

RESUMO

BACKGROUND AND PROCEDURE: The CD95/CD95 ligand (CD95L) system is a key regulator of apoptosis. To evaluate a possible role of the CD95/CD95L system in human neuroblastoma (NB) cells, we investigated the constitutive and interferongamma (INFgamma)-induced expression of CD95 and CD95L, and CD95-mediated cell death in the SK-N-BE(2) cell line. RESULTS: Modulation of CD95/CD95L expression and triggering of an autocrine apoptotic mechanism by IFNgamma suggest a potential role for INFgamma as a therapeutic agent for NB. CONCLUSIONS: The evidence that retinoids induce apoptosis via tissue transglutaminase (tTG) and that N-methyl-D-aspartate (NMDA) and gp120 act through the nitric oxide synthase (NOS) activation pathway, indicates the existence of different molecular mechanisms of action, whose pharmacological exploitation might be used in an additive fashion.


Assuntos
Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interferon gama/farmacologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Neuroblastoma/patologia , Receptor fas/fisiologia , Cálcio/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinergismo Farmacológico , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteína Ligante Fas , Proteínas de Ligação ao GTP/fisiologia , Proteína gp120 do Envelope de HIV/fisiologia , Humanos , Transporte de Íons , Glicoproteínas de Membrana/genética , N-Metilaspartato/farmacologia , Proteínas de Neoplasias/genética , Neuroblastoma/metabolismo , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase/fisiologia , Proteína 2 Glutamina gama-Glutamiltransferase , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Transglutaminases/fisiologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia , Receptor fas/genética
14.
J Cell Biochem ; 82(1): 123-33, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11400169

RESUMO

Nitric oxide (NO) plays an important anti-apoptotic role by inactivating both upstream and downstream apoptotic molecules. We now report that exogenously supplied NO protected Jurkat T cells from anti-CD95-stimulated apoptosis. We have recently shown that nitrosation of the activator protein-1 (AP-1) transcriptional factor is crucial for NO-mediated inhibition of cell death triggered by etoposide or ceramide. Since the inhibition of apoptosis by NO has been reported to involve AP-1, we evaluated its involvement in in CD95-mediated cell death. Cross-linking of CD95 enhanced AP-1 DNA binding activity and AP-1-dependent CD95L transactivation, which were both significantly reduced by different NO-donors compounds. However, AP-1 induction does not seem to significantly contribute to anti-CD95-triggered apoptosis, as cell death could not be prevented by using the recombinant Fas-Fc fusion protein which inhibits the CD95/CD95L interaction. We observed that caspase 3-like activity was negatively modulated by several NO-donors in vitro and that titratable thiol groups of purified caspases 3, 7, and 9 decreased in the presence of NO-releasing compounds. In conclusion, we demonstrated that NO-mediated inhibition of other targets, possibly caspases, but not AP-1, is a crucial event responsible for protection against anti-CD95-stimulated apoptosis. Even though NO affects multiple molecular mechanisms, the relevant target for exerting the cellular effects, may vary among different models.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Óxido Nítrico/farmacologia , Fator de Transcrição AP-1/efeitos dos fármacos , Apoptose/fisiologia , Caspases/metabolismo , Proteína Ligante Fas , Humanos , Células Jurkat , Glicoproteínas de Membrana/química , Óxido Nítrico/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Fator de Transcrição AP-1/metabolismo , Receptor fas/química , Receptor fas/metabolismo
15.
Med Pediatr Oncol ; 35(6): 663-8, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11107142

RESUMO

The RARbeta/gamma-selective retinoids fenretinide and CD437 induce caspase-dependent apoptosis but generate free radicals independently of caspases. Apoptosis, but not free radical generation, induced by these retinoids is inhibited by RARbeta/gamma-specific antagonists. Both fenretinide and CD437 induce apoptosis synergistically with cisplatin, carboplatin, or etoposide. However, antioxidants inhibit this synergy to the level obtained with chemotherapeutic drugs alone, and this implies that free radical generation is important in the synergistic response. Since apoptosis induced by fenretinide or CD437 is mediated by apoptotic pathways involving RARs and/or mitochondria and differs from mechanisms of chemotherapy-induced apoptosis this may explain the strong synergistic response seen between these synthetic retinoids and chemotherapeutic drugs. These results suggest that fenretinide or CD437 may be useful adjuncts to neuroblastoma therapy.


Assuntos
Antineoplásicos/uso terapêutico , Fenretinida/uso terapêutico , Neuroblastoma/tratamento farmacológico , Retinoides/uso terapêutico , Apoptose/efeitos dos fármacos , Criança , Sinergismo Farmacológico , Radicais Livres/metabolismo , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Células Tumorais Cultivadas
16.
Cell Mol Life Sci ; 57(4): 612-22, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11130461

RESUMO

Nitric oxide (NO) and its related molecules are important messengers that play central roles in pathophysiology. Redox modulation of thiol groups on protein cysteine residues by S-nitrosylation can modulate protein function. NO has emerged as a potent regulator of apoptosis in many cell types, either preventing cell death or driving an apoptotic response into a necrotic one. NO protects neuroblastoma cells from retinoid- and cisplatin-induced apoptosis, without significantly increasing necrotic cell damage. Nitrosylation of thiol groups of several critical factors may be important for cell survival. Indeed, S-nitrosylation of the active-site cysteine residue of apoptotic molecules, such as caspases and tissue transglutaminase, results in the inhibition of their catalytic activities and has important implications for the regulation of apoptosis by NO. On the other hand, NO is able to shift the anti-CD95- and ceramide-triggered apoptotic response of Jurkat T cells into necrotic cell death. In these apoptotic models, NO is therefore unable to solely inhibit cell death, indicating that it may act below the point of no return elicited by CD95-ligation and ceramide stimulation.


Assuntos
Apoptose , Necrose , Óxido Nítrico/fisiologia , Animais , Humanos , Óxido Nítrico/metabolismo
17.
Int J Cancer ; 88(6): 977-85, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11093824

RESUMO

Retinoic acid therapy improves the survival of children with neuroblastoma and 13-cis retinoic acid now forms an important component of treatment for residual disease of stage IV neuroblastoma after chemotherapy. However, although 13-cis retinoic acid induces differentiation, other retinoids are effective at inducing apoptosis of neuroblastoma in vitro, including the novel compounds fenretinide and CD437 and these may be alternative retinoids for neuroblastoma therapy. The aim of our study was to evaluate the ability of fenretinide, CD437 (6-¿3-(1-adamantyl)-4-hydroxyphenyl¿ -2-naphthalene carboxylic acid) and different retinoic acid isomers to induce apoptosis of neuroblastoma in conjunction with the chemotherapeutic drugs, cisplatin, etoposide and carboplatin. Neuroblastoma cell lines were treated with retinoids prior to treatment with chemotherapeutic agents and flow cytometry used to measure apoptosis and free radical generation. Pre-treatment of neuroblastoma cell lines with fenretinide or CD437 prior to treatment with cisplatin, etoposide or carboplatin synergistically increased apoptosis, an effect not seen with 13-cis, all-trans or 9-cis retinoic acid. Contrary to retinoic acid isomers or chemotherapeutic drugs, apoptosis of neuroblastoma cells induced by fenretinide or CD437 was accompanied by the generation of intracellular free radicals. Quenching of fenretinide- or CD437-induced free radicals with antioxidants abolished the synergistic response seen with the subsequent addition of chemotherapeutic agents. Therefore, the generation of free radicals by fenretinide or CD437 may be the key property of these retinoids leading to synergistic responses with chemotherapeutic drugs. Clearly, these synthetic retinoids provide new opportunities for novel neuroblastoma therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Carboplatina/uso terapêutico , Sobrevivência Celular , Cisplatino/uso terapêutico , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Etoposídeo/uso terapêutico , Fenretinida/administração & dosagem , Citometria de Fluxo , Radicais Livres/análise , Humanos , Neuroblastoma/fisiopatologia , Retinoides/administração & dosagem , Fatores de Tempo , Tretinoína/uso terapêutico , Células Tumorais Cultivadas/efeitos dos fármacos
18.
J Invest Dermatol ; 115(4): 731-9, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10998152

RESUMO

Epidermal keratinocytes undergo terminal differentiation to form the stratum corneum, which consists of many layers of flat dead cells. These cells assemble an insoluble cornified envelope composed of specific proteins deposited on the intracellular surface of the cell membrane. The proteins are crosslinked by the action of transglutaminases, which catalyze the formation of isodipeptide bonds between the epsilon-NH2 side chain of a lysine residue and the gamma-amide side chain of a glutamine residue. Transglutaminases share a conserved, highly reactive cysteine in their active site. In this study, we found that nitric-oxide-releasing compounds inhibited cornified envelope formation in cultured keratinocytes and the in vitro crosslinking of loricrin, a natural substrate of transglutaminases. The NO donors inhibited transglutaminase catalytic activity in a dose-dependent manner, in both purified enzymes and keratinocyte extracts. Titration of thiol groups of transglutaminases indicated that NO regulates their enzymatic activity by chemically modifying a cysteine residue, possibly by S-nitrosylation. NO was also found to inhibit DNA-binding activity of activating protein 1 in keratinocyte nuclear extracts, and to interfere with the transactivation of activating protein 1 responsive genes such as transglutaminase 1, involucrin, and loricrin, whose expression is regulated during epidermal differentiation. In conclusion, we propose that NO may inhibit keratinocyte differentiation, acting both at transcriptional level (inactivation of activating protein 1) and at post-translational level (inhibition of transglutaminase activity).


Assuntos
Queratinócitos/citologia , Óxido Nítrico/farmacologia , Fator de Transcrição AP-1/antagonistas & inibidores , Transglutaminases/metabolismo , Reagentes de Ligações Cruzadas/farmacologia , Ativação Enzimática/fisiologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/ultraestrutura , Proteínas de Membrana/metabolismo
19.
Exp Cell Res ; 260(1): 50-60, 2000 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-11010810

RESUMO

Fenretinide is an effective inducer of apoptosis in many malignancies but its precise mechanism(s) of action in the induction of apoptosis in neuroblastoma is unclear. To characterize fenretinide-induced apoptosis, neuroblastoma cell lines were treated with fenretinide and flow cytometry was used to measure apoptosis, free radical generation, and mitochondrial permeability changes. Fenretinide induced high levels of caspase-dependent apoptosis accompanied by an increase in free radicals and the release of cytochrome c in the absence of mitochondrial permeability transition. Apoptosis was blocked by two retinoic acid receptor (RAR)-beta/gamma-specific antagonists, but not by an RARalpha-specific antagonist. Free radical induction in response to fenretinide was not blocked by the caspase inhibitor ZVAD or by RAR antagonists and was only marginally reduced in cells selected for resistance to fenretinide. Therefore, free radical generation may be only one of a number of intracellular mechanisms of apoptotic signaling in response to fenretinide. These results suggest that the effector pathway of fenretinide-induced apoptosis of neuroblastoma is caspase dependent, involving mitochondrial release of cytochrome c independently of permeability changes, and mediated by specific RARs. As the mechanism of action of fenretinide may be different from other retinoids, this compound may be a valuable adjunct to neuroblastoma therapy with retinoic acid and conventional chemotherapeutic drugs.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fenretinida/farmacologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/fisiologia , Inibidores de Caspase , Caspases/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Grupo dos Citocromos c/metabolismo , Radicais Livres/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neuroblastoma/metabolismo , Oligopeptídeos/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Receptores do Ácido Retinoico/antagonistas & inibidores , Células Tumorais Cultivadas
20.
Cancer Res ; 60(9): 2377-83, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10811113

RESUMO

Several inducers of cytotoxic stress promote apoptotic cell death, which, at least in some cases, involves the CD95/CD95 ligand (CD95L) pathway. The induction of the CD95/CD95L pathway can be activated by the activator protein-1 (AP-1)-mediated up-regulation of the CD95L promoter, which is responsible for the induction of apoptosis elicited by stimuli such as etoposide. We show that nitric oxide (NO) represents a regulatory element able to block apoptosis by interfering with this loop. Etoposide- and C6-ceramide-induced apoptosis in Jurkat T cells with different kinetics. Cell death was accompanied by an increase in DNA-binding activity of the transcription factor AP-1, transactivation of the AP-1 site-containing CD95L promoter, and caspase 3-like protease activation. Using different NO-releasing compounds, we found that apoptosis was prevented in a dose-dependent manner. Furthermore, in both models of apoptosis, NO-releasing compounds dose-dependently reduced: (a) the number of the titratable thiol groups (cysteine residues) of c-Jun; (b) induction of AP-1 DNA-binding activity; (c) AP-1-driven transactivation of the CD95L promoter; and (d) caspase activation. In conclusion, our data demonstrate that NO can modulate cell death at an upstream level, by interfering with the ability of AP-1 to induce CD95L expression.


Assuntos
Apoptose , Glicoproteínas de Membrana/metabolismo , Óxido Nítrico/farmacologia , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Western Blotting , Núcleo Celular/metabolismo , Ceramidas/farmacologia , Fragmentação do DNA , Relação Dose-Resposta a Droga , Etoposídeo/farmacologia , Proteína Ligante Fas , Humanos , Células Jurkat , Modelos Biológicos , Dados de Sequência Molecular , Doadores de Óxido Nítrico/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...