Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 288(51): 36484-91, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24194516

RESUMO

Keratinocyte differentiation is a key process in the formation and maintenance of the protective skin barrier. Dysregulation in the balance of reactive oxygen species homeostasis may play a role in keratinocyte differentiation. We have identified the mitochondrial deacetylase SIRT3 as a key regulator of mitochondrial reactive oxygen species in keratinocytes. Our studies demonstrate that SIRT3 expression is down-regulated during keratinocyte differentiation, consistent with an increase in mitochondrial superoxide levels. Importantly, loss of SIRT3 expression in keratinocytes increased superoxide levels and promoted the expression of differentiation markers, whereas overexpression decreased superoxide levels and reduced the expression of differentiation markers. These findings identify a new role for SIRT3 in the suppression of epidermal differentiation via lowering oxidative stress.


Assuntos
Diferenciação Celular , Queratinócitos/enzimologia , Estresse Oxidativo , Sirtuína 3/metabolismo , Linhagem Celular , Regulação para Baixo , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Sirtuína 3/genética , Superóxidos/metabolismo
2.
Mol Cancer Res ; 11(9): 1072-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23723075

RESUMO

UNLABELLED: Activating point mutations in K-RAS are extremely common in cancers of the lung, colon, and pancreas and are highly predictive of poor therapeutic response. One potential strategy for overcoming the deleterious effects of mutant K-RAS is to alter its posttranslational modification. Although therapies targeting farnesylation have been explored, and have ultimately failed, the therapeutic potential of targeting other modifications remains to be seen. Recently, it was shown that acetylation of lysine 104 attenuates K-RAS transforming activity by interfering with GEF-induced nucleotide exchange. Here, the deacetylases HDAC6 and SIRT2 were shown to regulate the acetylation state of K-RAS in cancer cells. By extension, inhibition of either of these enzymes has a dramatic impact on the growth properties of cancer cells expressing activation mutants of K-RAS. These results suggest that therapeutic targeting of HDAC6 and/or SIRT2 may represent a new way to treat cancers expressing mutant forms of K-RAS. IMPLICATIONS: This study suggests that altering K-RAS acetylation is a feasible approach to limiting tumorigenic potential.


Assuntos
Transformação Celular Neoplásica , Histona Desacetilases/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Sirtuína 2/metabolismo , Proteínas ras/metabolismo , Acetilação , Animais , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Desacetilase 6 de Histona , Histona Desacetilases/genética , Humanos , Camundongos , Terapia de Alvo Molecular , Células NIH 3T3 , Neoplasias/patologia , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Sirtuína 2/genética , Proteínas ras/genética
3.
Exp Gerontol ; 48(7): 634-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22964489

RESUMO

Mitochondria play a central role in the production of reactive oxygen species as byproducts of metabolism and energy production. In order to protect cellular structures from oxidative stress-induced damage, cells have evolved elegant mechanisms for mitochondrial ROS detoxification. The mitochondrial sirtuin, SIRT3, is emerging as a pivotal regulator of oxidative stress by deacetylation of substrates involved in both ROS production and detoxification. This review will summarize recent findings on the regulation of mitochondrial ROS homeostasis by SIRT3.


Assuntos
Envelhecimento/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 3/metabolismo , Fatores Etários , Animais , Homeostase , Humanos , Oxirredução , Transdução de Sinais
4.
Molecules ; 15(5): 3338-55, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20657484

RESUMO

Colorectal cancer (CRC) is a major cause of tumor-related morbidity and mortality worldwide. Recent research suggests that pharmacological intervention using dietary factors that activate the redox sensitive Nrf2/Keap1-ARE signaling pathway may represent a promising strategy for chemoprevention of human cancer including CRC. In our search for dietary Nrf2 activators with potential chemopreventive activity targeting CRC, we have focused our studies on trans-cinnamic aldehyde (cinnamaldeyde, CA), the key flavor compound in cinnamon essential oil. Here we demonstrate that CA and an ethanolic extract (CE) prepared from Cinnamomum cassia bark, standardized for CA content by GC-MS analysis, display equipotent activity as inducers of Nrf2 transcriptional activity. In human colon cancer cells (HCT116, HT29) and non-immortalized primary fetal colon cells (FHC), CA- and CE-treatment upregulated cellular protein levels of Nrf2 and established Nrf2 targets involved in the antioxidant response including heme oxygenase 1 (HO-1) and gamma-glutamyl-cysteine synthetase (gamma-GCS, catalytic subunit). CA- and CE-pretreatment strongly upregulated cellular glutathione levels and protected HCT116 cells against hydrogen peroxide-induced genotoxicity and arsenic-induced oxidative insult. Taken together our data demonstrate that the cinnamon-derived food factor CA is a potent activator of the Nrf2-orchestrated antioxidant response in cultured human epithelial colon cells. CA may therefore represent an underappreciated chemopreventive dietary factor targeting colorectal carcinogenesis.


Assuntos
Acroleína/análogos & derivados , Antioxidantes , Cinnamomum zeylanicum , Neoplasias do Colo/prevenção & controle , Fator 2 Relacionado a NF-E2/metabolismo , Acroleína/farmacologia , Linhagem Celular Tumoral , Quimioprevenção/métodos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Suplementos Nutricionais , Células Epiteliais , Humanos , Oxirredução , Transdução de Sinais
5.
Melanoma Res ; 20(2): 85-96, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20093988

RESUMO

Glyoxalase I [lactoylglutathione lyase (EC 4.4.1.5) encoded by GLO1] is a ubiquitous cellular defense enzyme involved in the detoxification of methylglyoxal, a cytotoxic byproduct of glycolysis. Accumulative evidence suggests an important role of GLO1 expression in protection against methylglyoxal-dependent protein adduction and cellular damage associated with diabetes, cancer, and chronological aging. On the basis of the hypothesis that GLO1 upregulation may play a functional role in glycolytic adaptations of cancer cells, we examined GLO1 expression status in human melanoma tissue. Quantitative reverse transcription polymerase chain reaction analysis of a cDNA tissue array containing 40 human melanoma tissues (stages III and IV) and 13 healthy controls revealed pronounced upregulation of GLO1 expression at the mRNA level. Immunohistochemical analysis of a melanoma tissue microarray confirmed upregulation of glyoxalase I protein levels in malignant melanoma tissue versus healthy human skin. Consistent with an essential role of GLO1 in melanoma cell defense against methylglyoxal cytotoxicity, siRNA interference targeting GLO1-expression (siGLO1) sensitized A375 and G361 human metastatic melanoma cells towards the antiproliferative, apoptogenic, and oxidative stress-inducing activity of exogenous methylglyoxal. Protein adduction by methylglyoxal was increased in siGLO1-transfected cells as revealed by immunodetection using a monoclonal antibody directed against the major methylglyoxal-derived epitope argpyrimidine that detected a single band of methylglyoxal-adducted protein in human LOX, G361, and A375 total cell lysates. Using two-dimensional proteomics followed by mass spectrometry the methylglyoxal-adducted protein was identified as heat shock protein 27 (Hsp27; HSPB1). Taken together, our data suggest a function of GLO1 in the regulation of detoxification and target adduction by the glycolytic byproduct methylglyoxal in malignant melanoma.


Assuntos
Lactoilglutationa Liase/biossíntese , Melanoma/metabolismo , Western Blotting , Separação Celular , Citometria de Fluxo , Expressão Gênica , Humanos , Imuno-Histoquímica , Lactoilglutationa Liase/genética , Melanoma/genética , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Transfecção
6.
Biochem Pharmacol ; 78(4): 344-54, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19394313

RESUMO

Altered redox homeostasis involved in the control of cancer cell survival and proliferative signaling represents a chemical vulnerability that can be targeted by prooxidant redox intervention. Here, we demonstrate that the redox dye 2,6-dichlorophenolindophenol (DCPIP) may serve as a prooxidant chemotherapeutic targeting human melanoma cells in vitro and in vivo. DCPIP-apoptogenicity observed in the human melanoma cell lines A375 and G361 was inversely correlated with NAD(P)H:quinone oxidoreductase (NQO1) expression levels. In A375 cells displaying low NQO1 activity, DCPIP induced apoptosis with procaspase-3 and PARP cleavage, whereas G361 cells expressing high levels of enzymatically active NQO1 were resistant to DCPIP-cytotoxicity. Genetic (siRNA) or pharmacological (dicoumarol) antagonism of NQO1 strongly sensitized G361 cells to DCPIP apoptogenic activity. DCPIP-cytotoxicity was associated with the induction of oxidative stress and rapid depletion of glutathione in A375 and NQO1-modulated G361 cells. Expression array analysis revealed a DCPIP-induced stress response in A375 cells with massive upregulation of genes encoding Hsp70B' (HSPA6), Hsp70 (HSPA1A), heme oxygenase-1 (HMOX1), and early growth response protein 1 (EGR1) further confirmed by immunodetection. Systemic administration of DCPIP displayed significant antimelanoma activity in the A375 murine xenograft model. These findings suggest feasibility of targeting tumors that display low NQO1 enzymatic activity using DCPIP.


Assuntos
2,6-Dicloroindofenol/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Melanoma/patologia , NAD(P)H Desidrogenase (Quinona)/farmacologia , Animais , Antineoplásicos/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Antagonismo de Drogas , Regulação Neoplásica da Expressão Gênica , Glutationa , Heme Oxigenase-1/metabolismo , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Camundongos SCID , NAD(P)H Desidrogenase (Quinona)/metabolismo , Oxirredução , Transdução de Sinais , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Free Radic Biol Med ; 46(2): 220-31, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19000754

RESUMO

Redox dysregulation in cancer cells represents a chemical vulnerability that can be targeted by pro-oxidant redox intervention. Dietary constituents that contain an electrophilic Michael acceptor pharmacophore may therefore display promising chemopreventive and chemotherapeutic anti-cancer activity. Here, we demonstrate that the cinnamon-derived dietary Michael acceptor trans-cinnamic aldehyde (CA) impairs melanoma cell proliferation and tumor growth. Feasibility of therapeutic intervention using high doses of CA (120 mg/kg, po, daily, 10 days) was demonstrated in a human A375 melanoma SCID mouse xenograft model. Low-micromolar concentrations (IC(50)< 10 microM) of CA, but not closely related CA derivatives devoid of Michael acceptor activity, suppressed proliferation of human metastatic melanoma cell lines (A375, G361, LOX) with G1 cell-cycle arrest, elevated intracellular ROS, and impaired invasiveness. Expression array analysis revealed that CA induced an oxidative stress response in A375 cells, up-regulating heme oxygenase 1, sulfiredoxin 1 homolog, thioredoxin reductase 1, and other genes, including the cell-cycle regulator and stress-responsive tumor suppressor gene cyclin-dependent kinase inhibitor 1A, a key mediator of G1-phase arrest. CA, but not Michael-inactive derivatives, inhibited NF-kappaB transcriptional activity and TNFalpha-induced IL-8 production in A375 cells. These findings support a previously unrecognized role of CA as a dietary Michael acceptor with potential anti-cancer activity.


Assuntos
Acroleína/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Melanoma/tratamento farmacológico , Reguladores de Crescimento de Plantas/farmacologia , Acroleína/farmacologia , Animais , Linhagem Celular Tumoral , Cinnamomum zeylanicum , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fase G1/efeitos dos fármacos , Perfilação da Expressão Gênica , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Interleucina-8/metabolismo , Melanoma/enzimologia , Melanoma/patologia , Camundongos , Camundongos SCID , Análise em Microsséries , NF-kappa B/antagonistas & inibidores , Invasividade Neoplásica/prevenção & controle , Transplante de Neoplasias , Estresse Oxidativo/efeitos dos fármacos , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tiorredoxina Redutase 1/genética , Tiorredoxina Redutase 1/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA