Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Neuropharmacology ; 170: 108039, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32165217

RESUMO

The NAD(P)H:quinone oxidoreductase 1 (NQO1) gene encodes a cytosolic flavoenzyme that catalyzes the two-electron reduction of quinones to hydroquinones. A polymorphic form of NQO1 is associated with mood disorders such as schizophrenia. However, the role of NQO1 in dopaminergic system has not yet been elucidated. To determine the role of NQO1 in the dopaminergic system, we investigated pharmaco-behavioral effects of d-amphetamine using NQO1-deficienct mice. According to our comparative study involving NQO1+/+ and NQO1-/- mice, NQO1 deficiency increased d-amphetamine-induced psychomotor activity and psychological dependency compared to wild-type mice. Basal and d-amphetamine-induced dopamine levels were also enhanced by NQO1 deficiency. In NQO1-/- mice, neural activation induced by d-amphetamine was higher in dorsolateral striatum, but not in dorsomedial and ventral striata. Although protein level of CaMKIIα, which is a key player in amphetamine-induced dopamine efflux, was decreased in striata of NQO1-/- mice, phosphorylation of CaMKIIα was markedly enhanced in NQO1-/- mice compared to wild-type mice. Interestingly, experiments with pharmacological antagonist showed that D2 antagonist-induced suppression of locomotion required activation of NQO1. Moreover, the rewarding effect in response to D1 agonist was increased by NQO1 deficiency. These results suggest that striatal NQO1 is of considerable interest to understand the mechanism of dopaminergic regulation of psychiatric disorders.


Assuntos
Corpo Estriado/metabolismo , Dextroanfetamina/farmacologia , Dopamina/metabolismo , NAD(P)H Desidrogenase (Quinona)/deficiência , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Animais , Corpo Estriado/efeitos dos fármacos , Antagonistas dos Receptores de Dopamina D2/farmacologia , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D2/agonistas
2.
Cell Commun Signal ; 17(1): 168, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31842909

RESUMO

BACKGROUND: Our previous study has demonstrated that NAD(P)H: quinone oxidoreductase 1 (NQO1) is significantly upregulated in human liver cancer where it potentiates the apoptosis evasion of liver cancer cell. However, the underlying mechanisms of the oncogenic function of NQO1 in HCC have not been fully elucidated. METHODS: Expression of NQO1, SIRT6, AKT and X-linked inhibitor of apoptosis protein (XIAP) protein were measured by western blotting and immunohistochemistry. Additionally, the interaction between NQO1 and potential proteins were determined by immunoprecipitation assays. Furthermore, the effect of NQO1 and SIRT6 on tumor growth was determined in cell model and orthotopic tumor implantation model. RESULTS: We found that NQO1 overexpression in HCC enhanced SIRT6 protein stability via inhibiting ubiquitin-mediated 26S proteasome degradation. High level of SIRT6 reduced acetylation of AKT which resulted in increased phosphorylation and activity of AKT. Activated AKT subsequently phosphorylated anti-apoptotic protein XIAP at Ser87 which determined its protein stability. Reintroduction of SIRT6 or AKT efficiently rescued NQO1 knock-out-mediated inhibition of growth and induction of apoptosis. In orthotopic mouse model, NQO1 knock-out inhibited tumor growth and induced apoptosis while this effect was effectively rescued by SIRT6 overexpression or MG132 treatment partially. CONCLUSIONS: Collectively, these results reveal an oncogenic function of NQO1 in sustaining HCC cell proliferation through SIRT6/AKT/XIAP signaling pathway.


Assuntos
Apoptose , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Sirtuínas/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Neoplasias Hepáticas/patologia , NAD(P)H Desidrogenase (Quinona)/deficiência , Fosforilação , Estabilidade Proteica , Transdução de Sinais , Regulação para Cima
3.
J Exp Med ; 215(8): 2197-2209, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29934320

RESUMO

NAD(P)H:quinone oxidoreductase 1 (NQO1) protects cells against oxidative stress and toxic quinones. In this study, we found a novel role of NQO1 in suppressing Toll-like receptor (TLR)-mediated innate immune responses. NQO1-deficient macrophages selectively produced excessive amounts of IL-6, IL-12, and GM-CSF on LPS stimulation, and the deletion of NQO1 in macrophages exacerbated LPS-induced septic shock. NQO1 interacted with the nuclear IκB protein IκB-ζ, which is essential for the TLR-mediated induction of a subset of secondary response genes, including IL-6, and promoted IκB-ζ degradation in a ubiquitin-dependent manner. We demonstrated that PDLIM2, known as the ubiquitin E3 ligase, participates in NQO1-dependent IκB-ζ degradation. NQO1 augmented the association between PDLIM2 and IκB-ζ, resulting in increased IκB-ζ degradation. Collectively, this study describes a mechanism of the NQO1-PDLIM2 complex as a novel and important regulator in the innate immune signaling and suggests the therapeutic potential of NQO1 in TLR-mediated inflammation and disorders.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citocinas/biossíntese , NAD(P)H Desidrogenase (Quinona)/metabolismo , Proteínas Nucleares/metabolismo , Proteólise , Receptores Toll-Like/metabolismo , Animais , Núcleo Celular/metabolismo , Feminino , Proteínas com Domínio LIM/metabolismo , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , NAD(P)H Desidrogenase (Quinona)/deficiência , Poliubiquitina/metabolismo , Ligação Proteica , Estabilidade Proteica , Transporte Proteico , Células RAW 264.7 , Receptores de Hidrocarboneto Arílico/metabolismo , Sepse/metabolismo , Sepse/patologia , Ubiquitinação
4.
J Microbiol Biotechnol ; 26(8): 1446-51, 2016 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-27116994

RESUMO

Clostridium difficile toxin A causes acute gut inflammation in animals and humans. It is known to downregulate the tight junctions between colonic epithelial cells, allowing luminal contents to access body tissues and trigger acute immune responses. However, it is not yet known whether this loss of the barrier function is a critical factor in the progression of toxin A-induced pseudomembranous colitis. We previously showed that NADH:quinone oxidoreductase 1 (NQO1) KO (knockout) mice spontaneously display weak gut inflammation and a marked loss of colonic epithelial tight junctions. Moreover, NQO1 KO mice exhibited highly increased inflammatory responses compared with NQO1 WT (wild-type) control mice when subjected to DSS-induced experimental colitis. Here, we tested whether toxin A could also trigger more severe inflammatory responses in NQO1 KO mice compared with NQO1 WT mice. Indeed, our results show that C. difficile toxin A-mediated enteritis is significantly enhanced in NQO1 KO mice compared with NQO1 WT mice. The levels of fluid secretion, villus disruption, and epithelial cell apoptosis were also higher in toxin A-treated NQO1 KO mice compared with WT mice. The previous and present results collectively show that NQO1 is involved in the formation of tight junctions in the small intestine, and that defects in NQO1 enhance C. difficile toxin A-induced acute inflammatory responses, presumably via the loss of epithelial cell tight junctions.


Assuntos
Toxinas Bacterianas/toxicidade , Enterite/microbiologia , Enterite/fisiopatologia , Enterotoxinas/toxicidade , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/fisiologia , Animais , Apoptose , Toxinas Bacterianas/administração & dosagem , Clostridioides difficile/fisiologia , Enterite/patologia , Enterotoxinas/administração & dosagem , Células Epiteliais/patologia , Humanos , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , NAD(P)H Desidrogenase (Quinona)/deficiência , Junções Íntimas/patologia
5.
Hear Res ; 333: 235-246, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26341473

RESUMO

Ototoxicity is an important issue in patients receiving cisplatin chemotherapy. Numerous studies have demonstrated that cisplatin-induced ototoxicity is related to oxidative stress and DNA damage. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. The cofactor nicotinamide adenine dinucleotide (NAD(+)) has emerged as an important regulator of energy metabolism and cellular homeostasis. Here, we demonstrate that the levels and activities of sirtuin-1 (SIRT1) are suppressed by the reduction of intracellular NAD(+) levels in cisplatin-mediated ototoxicity. We provide evidence that the decreases in SIRT1 activity and expression facilitated by increasing poly(ADP-ribose) polymerase-1 (PARP-1) activation and microRNA-34a levels through cisplatin-mediated p53 activation aggravate the associated ototoxicity. Furthermore, we show that the induction of cellular NAD(+) levels using dunnione, which targets intracellular NQO1, prevents the toxic effects of cisplatin through the regulation of PARP-1 and SIRT1 activity. These results suggest that direct modulation of cellular NAD(+) levels by pharmacological agents could be a promising therapeutic approach for protection from cisplatin-induced ototoxicity.


Assuntos
Cisplatino , Cóclea/efeitos dos fármacos , Perda Auditiva/prevenção & controle , Audição/efeitos dos fármacos , NAD/metabolismo , Naftoquinonas/farmacologia , Substâncias Protetoras/farmacologia , Acetilação , Animais , Cóclea/metabolismo , Cóclea/fisiopatologia , Citoproteção , Modelos Animais de Doenças , Perda Auditiva/induzido quimicamente , Perda Auditiva/metabolismo , Perda Auditiva/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo
6.
Antioxid Redox Signal ; 21(2): 237-50, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-24512128

RESUMO

AIMS: ß-Lapachone (ß-lap), a novel radiosensitizer with potent antitumor efficacy alone, selectively kills solid cancers that over-express NAD(P)H: quinone oxidoreductase 1 (NQO1). Since breast or other solid cancers have heterogeneous NQO1 expression, therapies that reduce the resistance (e.g., NQO1(low)) of tumor cells will have significant clinical advantages. We tested whether NQO1-proficient (NQO1(+)) cells generated sufficient hydrogen peroxide (H2O2) after ß-lap treatment to elicit bystander effects, DNA damage, and cell death in neighboring NQO1(low) cells. RESULTS: ß-Lap showed NQO1-dependent efficacy against two triple-negative breast cancer (TNBC) xenografts. NQO1 expression variations in human breast cancer patient samples were noted, where ~60% cancers over-expressed NQO1, with little or no expression in associated normal tissue. Differential DNA damage and lethality were noted in NQO1(+) versus NQO1-deficient (NQO1(-)) TNBC cells and xenografts after ß-lap treatment. ß-Lap-treated NQO1(+) cells died by programmed necrosis, whereas co-cultured NQO1(-) TNBC cells exhibited DNA damage and caspase-dependent apoptosis. NQO1 inhibition (dicoumarol) or H2O2 scavenging (catalase [CAT]) blocked all responses. Only NQO1(-) cells neighboring NQO1(+) TNBC cells responded to ß-lap in vitro, and bystander effects correlated well with H2O2 diffusion. Bystander effects in NQO1(-) cells in vivo within mixed 50:50 co-cultured xenografts were dramatic and depended on NQO1(+) cells. However, normal human cells in vitro or in vivo did not show bystander effects, due to elevated endogenous CAT levels. Innovation and Conclusions: NQO1-dependent bystander effects elicited by NQO1 bioactivatable drugs (ß-lap or deoxynyboquinone [DNQ]) likely contribute to their efficacies, killing NQO1(+) solid cancer cells and eliminating surrounding heterogeneous NQO1(low) cancer cells. Normal cells/tissue are protected by low NQO1:CAT ratios.


Assuntos
Efeito Espectador/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Naftoquinonas/farmacologia , Quinonas/farmacologia , Radiossensibilizantes/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Feminino , Humanos , Camundongos , Camundongos Nus , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/genética , Oxirredução/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
BMB Rep ; 47(9): 494-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24393524

RESUMO

NADH:quinone oxidoreductase 1 (NQO1) is known to be involved in the regulation of energy synthesis and metabolism, and the functional studies of NQO1 have largely focused on metabolic disorders. Here, we show for the first time that compared to NQO1-WT mice, NQO1-KO mice exhibited a marked increase of permeability and spontaneous inflammation in the gut. In the DSS-induced colitis model, NQO1-KO mice showed more severe inflammatory responses than NQO1-WT mice. Interestingly, the transcript levels of claudin and occludin, the major tight junction molecules of gut epithelial cells, were significantly decreased in NQO1-KO mice. The colons of NQO1-KO mice also showed high levels of reactive oxygen species (ROS) and histone deacetylase (HDAC) activity, which are known to affect transcriptional regulation. Taken together, these novel findings indicate that NQO1 contributes to the barrier function of gut epithelial cells by regulating the transcription of tight junction molecules.


Assuntos
Células Epiteliais/enzimologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , Junções Íntimas/enzimologia , Animais , Permeabilidade da Membrana Celular , Células Cultivadas , Claudina-1/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Regulação para Baixo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Histona Desacetilases/metabolismo , Camundongos , Camundongos Knockout , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/genética , Ocludina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Junções Íntimas/metabolismo
8.
Free Radic Biol Med ; 67: 139-49, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24189322

RESUMO

Ischemia/reperfusion (I/R) is the most common cause of acute renal injury. I/R-induced reactive oxygen species (ROS) are thought to be a major factor in the development of acute renal injury by promoting the initial tubular damage. NAD(P)H: quinone oxidoreductase 1 (NQO1) is a well-known antioxidant protein that regulates ROS generation. The purpose of this study was to investigate whether NQO1 modulates the renal I/R injury (IRI) associated with NADPH oxidase (NOX)-derived ROS production in an animal model. We analyzed renal function, oxidative stress, and tubular apoptosis after IRI. NQO1(-/-) mice showed increased blood urea nitrogen and creatinine levels, tubular damage, oxidative stress, and apoptosis. In the kidneys of NQO1(-/-) mice, the cellular NADPH/NADP(+) ratio was significantly higher and NOX activity was markedly higher than in those of NQO1(+/+) mice. The activation of NQO1 by ß-lapachone (ßL) significantly improved renal dysfunction and reduced tubular cell damage, oxidative stress, and apoptosis by renal I/R. Moreover, the ßL treatment significantly lowered the cellular NADPH/NADP(+) ratio and dramatically reduced NOX activity in the kidneys after IRI. From these results, it was concluded that NQO1 has a protective role against renal injury induced by I/R and that this effect appears to be mediated by decreased NOX activity via cellular NADPH/NADP(+) modulation. These results provide convincing evidence that NQO1 activation might be beneficial for ameliorating renal injury induced by I/R.


Assuntos
Injúria Renal Aguda/enzimologia , Túbulos Renais/enzimologia , NAD(P)H Desidrogenase (Quinona)/genética , NADPH Oxidases/genética , Traumatismo por Reperfusão/enzimologia , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Ativadores de Enzimas/farmacologia , Expressão Gênica , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NAD(P)H Desidrogenase (Quinona)/deficiência , NADP/metabolismo , NADPH Oxidases/metabolismo , Naftoquinonas/farmacologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
9.
Toxicol Lett ; 219(1): 35-41, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23458895

RESUMO

NAD(P)H:quinone oxidoreductase 1 (NQO1) has been known to protect cells against stressors, including the diabetogenic reagent streptozotocin (STZ). The present study demonstrated that NQO1 deficiency resulted in increased pancreatic ß-cell death induced by multiple low dose of STZ (MLDS) injections. NQO1 knockout (KO) mice showed hyperglycemia, body weight loss, impaired glucose clearance rate and a lower plasma insulin level after MLDS treatment. Moreover, ß-cell mass and pancreatic insulin content were significantly lower in KO mice than in wild-type (WT) mice after MLDS treatment. Five days after the first STZ treatment, the islets of KO mice had substantially more TUNEL-positive ß-cells than those of WT mice, but there was no difference in the regeneration of ß-cells between KO mice and WT mice. At the same time, MLDS-treated KO mice showed significantly increased apoptotic markers in ß-cells, including cleaved caspase 3, Smac/DIABLO and AIF (apoptosis inducing factor) in the cytoplasm. These results suggest that mice deficient in NQO1 are vulnerable to MLDS-induced ß-cell destruction and diabetes, caused by increase of ß-cell apoptosis in pancreas.


Assuntos
Apoptose/efeitos dos fármacos , Diabetes Mellitus Experimental/enzimologia , Células Secretoras de Insulina/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/deficiência , Estreptozocina/farmacologia , Animais , Diabetes Mellitus Experimental/patologia , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Insulina/sangue , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NAD(P)H Desidrogenase (Quinona)/genética , Espécies Reativas de Oxigênio/metabolismo
10.
Free Radic Biol Med ; 52(3): 705-715, 2012 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-22198263

RESUMO

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Authors. Since learning of potential discrepancies between the raw data from the animal pulmonary physiology laboratory at Duke that were used to calculate the in vivo pulmonary mechanics and the re-exported machine-generated raw data, some studies published elsewhere have been replicated successfully. However it is not possible to replicate this study as the NQO1-deficient mice on the C57BL/6 background are no longer available from the NCI. The authors recognize that previous work to identify differences in alveolar size can vary dependent on background strain when comparing inbred mouse strains (Soutiere SE et al Resp Physiol Neurobiol 2004;140(3)183­91 doi: 10.1016/j.resp.2004.02.003). Because of the prolonged period of time required to successfully backcross NQO1-deficient animals onto C57BL/6J background and the time required to repeat studies presented in this manuscript the authors think it does not seem feasible to conduct replicate studies in a reasonable timeline. Therefore, the most appropriate course of action is to retract the report as it is the authors' goal to maintain accuracy of the scientific record to the best of their ability. The authors offer sincere apologies to the scientific community.


Assuntos
Pulmão/enzimologia , NAD(P)H Desidrogenase (Quinona)/deficiência , Enfisema Pulmonar/enzimologia , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Animais , Líquido da Lavagem Broncoalveolar , Células Cultivadas , Dinoprosta/análogos & derivados , Dinoprosta/metabolismo , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Pulmão/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Estresse Oxidativo , Carbonilação Proteica , Enfisema Pulmonar/tratamento farmacológico , Enfisema Pulmonar/patologia , Volume Residual
11.
PLoS One ; 6(5): e20590, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21655231

RESUMO

BACKGROUND: The etiology of myelodysplastic syndromes (MDS) is largely unknown. Exposure to cigarette smoke (CS) is reported to be associated with MDS risk. There is inconsistent evidence that deficiency of NAD(P)H-quinone: oxidoreductase 1 (NQO1) increases the risk of MDS. Earlier we had shown that CS induces toxicity only in marginal vitamin C-deficient guinea pigs but not in vitamin C-sufficient ones. We therefore considered that NQO1 deficiency along with marginal vitamin C deficiency might produce MDS in CS-exposed guinea pigs. METHODOLOGY AND PRINCIPAL FINDINGS: Here we show that CS exposure for 21 days produces MDS in guinea pigs having deficiency of NQO1 (fed 3 mg dicoumarol/day) conjoint with marginal vitamin C deficiency (fed 0.5 mg vitamin C/day). As evidenced by morphology, histology and cytogenetics, MDS produced in the guinea pigs falls in the category of refractory cytopenia with unilineage dysplasia (RCUD): refractory anemia; refractory thrombocytopenia that is associated with ring sideroblasts, micromegakaryocytes, myeloid hyperplasia and aneuploidy. MDS is accompanied by increased CD34(+) cells and oxidative stress as shown by the formation of protein carbonyls and 8-oxodeoxyguanosine. Apoptosis precedes MDS but disappears later with marked decrease in the p53 protein. MDS produced in the guinea pigs are irreversible. MDS and all the aforesaid pathophysiological events do not occur in vitamin C-sufficient guinea pigs. However, after the onset of MDS vitamin C becomes ineffective. CONCLUSIONS AND SIGNIFICANCE: CS exposure causes MDS in guinea pigs having deficiency of NQO1 conjoint with marginal vitamin C deficiency. The syndromes are not produced in singular deficiency of NQO1 or marginal vitamin C deficiency. Our results suggest that human smokers having NQO1 deficiency combined with marginal vitamin C deficiency are likely to be at high risk for developing MDS and that intake of a moderately large dose of vitamin C would prevent MDS.


Assuntos
Deficiência de Ácido Ascórbico/fisiopatologia , Síndromes Mielodisplásicas/induzido quimicamente , Síndromes Mielodisplásicas/etiologia , NAD(P)H Desidrogenase (Quinona)/deficiência , Poluição por Fumaça de Tabaco/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Ácido Ascórbico/sangue , Ácido Ascórbico/metabolismo , Medula Óssea/metabolismo , Citometria de Fluxo , Cobaias , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , NAD(P)H Desidrogenase (Quinona)/metabolismo , Espécies Reativas de Oxigênio/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L773-80, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21296895

RESUMO

The quinones duroquinone (DQ) and coenzyme Q(1) (CoQ(1)) and quinone reductase inhibitors have been used to identify reductases involved in quinone reduction on passage through the pulmonary circulation. In perfused rat lung, NAD(P)H:quinone oxidoreductase 1 (NQO1) was identified as the predominant DQ reductase and NQO1 and mitochondrial complex I as the CoQ(1) reductases. Since inhibitors have nonspecific effects, the goal was to use Nqo1-null (NQO1(-)/(-)) mice to evaluate DQ as an NQO1 probe in the lung. Lung homogenate cytosol NQO1 activities were 97 ± 11, 54 ± 6, and 5 ± 1 (SE) nmol dichlorophenolindophenol reduced·min(-1)·mg protein(-1) for NQO1(+/+), NQO1(+/-), and NQO1(-/-) lungs, respectively. Intact lung quinone reduction was evaluated by infusion of DQ (50 µM) or CoQ(1) (60 µM) into the pulmonary arterial inflow of the isolated perfused lung and measurement of pulmonary venous effluent hydroquinone (DQH(2) or CoQ(1)H(2)). DQH(2) efflux rates for NQO1(+/+), NQO1(+/-), and NQO1(-/-) lungs were 0.65 ± 0.08, 0.45 ± 0.04, and 0.13 ± 0.05 (SE) µmol·min(-1)·g dry lung(-1), respectively. DQ reduction in NQO1(+/+) lungs was inhibited by 90 ± 4% with dicumarol; there was no inhibition in NQO1(-/-) lungs. There was no significant difference in CoQ(1)H(2) efflux rates for NQO1(+/+) and NQO1(-/-) lungs. Differences in DQ reduction were not due to differences in lung dry weights, wet-to-dry weight ratios, perfusion pressures, perfused surface areas, or total DQ recoveries. The data provide genetic evidence implicating DQ as a specific NQO1 probe in the perfused rodent lung.


Assuntos
NAD(P)H Desidrogenase (Quinona)/metabolismo , Animais , Benzoquinonas/metabolismo , Benzoquinonas/farmacologia , Dicumarol/farmacologia , Pulmão/metabolismo , Camundongos , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/genética , Oxirredução , Circulação Pulmonar/efeitos dos fármacos , Ubiquinona/metabolismo , Ubiquinona/farmacologia
13.
J Pharmacol Exp Ther ; 334(1): 260-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20378716

RESUMO

NAD(P)H:quinone oxidoreductase 1 (NQO1) deficiency resulting from a homozygous NQO1*2 polymorphism has been associated with an increased risk of benzene-induced myeloid toxicity and a variety of de novo and therapy-induced leukemias. Endothelial cells in human bone marrow form one of the two known hematopoietic stem cell microenvironments and are one of the major cell types that express NQO1 in bone marrow. We have used a transformed human bone marrow endothelial cell (TrHBMEC) line to study the potential impact of a lack of NQO1 activity on adhesion molecule [endothelial leukocyte adhesion molecule 1 (E-selectin), vascular cell adhesion molecule (VCAM)-1, and intercellular adhesion molecule (ICAM)-1] expression and functional adhesion to bone marrow progenitor cells. We used both 5-methoxy-1,2-dimethyl-3-[(4-nitrophenoxy)methyl]indole-4,7-dione (ES936), a mechanism-based inhibitor of NQO1, and anti-NQO1 small interfering RNA to abrogate NQO1 activity. Real-time reverse transcription-polymerase chain reaction data demonstrated a significant inhibition of tumor necrosis factor (TNF)alpha-induced E-selectin mRNA levels after ES936 pretreatment. Immunoblot assays demonstrated a significant reduction in TNFalpha-stimulated E-selectin, VCAM-1, and ICAM-1 proteins after inhibition or knockdown of NQO1. The mechanisms underlying this effect remain undefined, but modulation of nuclear factor-kappaB (p65), c-Jun, and activating transcription factor 2, transcriptional regulators of adhesion molecules, were observed after inhibition or knockdown of NQO1. Decreased level of E-selectin, VCAM-1, and ICAM-1 also resulted in a functional deficit in adhesion. A parallel plate flow chamber study demonstrated a marked reduction in CD34(+) cell (KG1a) adhesion to NQO1-deficient TrHBMECs relative to controls. The reduced adhesive ability of TrHBMECs may affect the function of the vascular stem cell niche and also may contribute to the increased susceptibility of polymorphic individuals lacking NQO1 to leukemias and hematotoxicants such as benzene.


Assuntos
Antígenos CD34/metabolismo , Células da Medula Óssea/fisiologia , Moléculas de Adesão Celular/biossíntese , Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , NAD(P)H Desidrogenase (Quinona)/deficiência , Células da Medula Óssea/citologia , Células da Medula Óssea/enzimologia , Adesão Celular/fisiologia , Linhagem Celular Transformada , Selectina E/biossíntese , Selectina E/genética , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Inibidores Enzimáticos/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Immunoblotting , Indolquinonas/farmacologia , Molécula 1 de Adesão Intercelular/biossíntese , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese
14.
Cancer Res ; 70(3): 1006-14, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20103645

RESUMO

The cytosolic quinone oxidoreductases NQO1 and NQO2 protect cells against oxidative stress by detoxifying quinones and preventing redox cycling. In this study, we used double knockout (DKO) mice deficient for NQO1 and NQO2 to investigate the role of these antioxidative enzymes in a two-stage model of inflammatory skin carcinogenesis. In this model, tumors are caused by exposure to topical carcinogen dimethylbenz(a)anthracene or benzo(a)pyrene (BP) followed by twice weekly application of proinflammatory phorbol 12-myristate 13-acetate. On this classic chemical carcinogenesis protocol, DKO mice showed a significantly higher skin tumor frequency and multiplicity compared with control wild-type or single knockout mice. Analysis of skin from wild-type and DKO mice exposed to BP for 6, 12, or 24 hours revealed a relative delay in the activation of p53, p63, p19ARF, and apoptosis in DKO mice, consistent with a negative modifier role for NQO1/NQO2 in carcinogenesis. Our findings offer genetic evidence of the significance of quinone oxidoreductases NQO1 and NQO2 in limiting chemical skin carcinogenesis.


Assuntos
NAD(P)H Desidrogenase (Quinona)/genética , Quinona Redutases/genética , Neoplasias Cutâneas/genética , Animais , Apoptose/efeitos dos fármacos , Benzo(a)Antracenos , Benzo(a)pireno , Western Blotting , Caspase 3/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NAD(P)H Desidrogenase (Quinona)/deficiência , Ornitina Descarboxilase/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Quinona Redutases/deficiência , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Transativadores/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo
15.
Oncol Rep ; 21(5): 1169-74, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19360290

RESUMO

The NAD(P)H:quinone oxidoreductase 1 (NQO1) enzyme is implicated in protection against oxidative stress and carcinogenesis. NQO1 C609T genetic polymorphism was reported to be associated with an increased risk for cancers, including breast cancer. However, there is still lack of evidence whether higher oxidative stress occurs in breast tissues of patients with NQO1 609 C/T and/or T/T genotypes. Mitochondrial DNA (MtDNA) 4,977-bp deletion, the most common mutation of mtDNA, was frequently detected in post-mitotic tissues of aged subjects and associated with oxidative damage. In this study, we detected the mtDNA 4,977-bp deletion in 60 breast cancers and corresponding non-cancerous breast tissues. The incidence of the common 4,977-bp deletion in non-cancerous breast tissues (48.3%) was higher than that in breast cancer (5.0%). Moreover, 63.4% of the breast cancer patients with NQO1 C/T or T/T genotypes had the deletion in their non-cancerous breast tissues. The mtDNA deletion was more frequently detected in breast tissue of NQO1 C/T carriers (65.2%) and T/T carriers (61.1%) as compared with the NQO1 C/C carriers (15.8%, P=0.003). Similar results were observed in the <50 years old (y/o) group (P=0.06) and the > or =50 y/o group (P=0.005). Our findings suggest that mtDNA 4,977-bp deletion associated with NQO1 deficiency is involved in carcinogenesis and progression of breast cancer.


Assuntos
DNA Mitocondrial/genética , NAD(P)H Desidrogenase (Quinona)/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Progressão da Doença , Feminino , Deleção de Genes , Predisposição Genética para Doença , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/metabolismo , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo de Fragmento de Restrição
16.
J Mol Med (Berl) ; 83(10): 786-94, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16142472

RESUMO

Complex I has a vital role in the energy production of the cell, and the clinical spectrum of complex I deficiency varies from severe lactic acidosis in infants to muscle weakness in adults. It has been estimated that the cause of complex I deficiency, especially in children, is often a mutation in the nuclear-encoded genes and, more rarely, in the genes encoded by mitochondrial DNA. We sequenced nine complex I subunit coding genes, NDUFAB1, NDUFS1, NDUFS2, NDUFS3, NDUFS4, NDUFS7, NDUFS8, NDUFV1 and NDUFV2, in 13 children with defined complex I deficiency. Two novel substitutions were found: a synonymous replacement 201A>T in NDUFV2 and a non-synonymous base exchange 52C>T in NDUFS8. The 52C>T substitution produced the replacement Arg18Cys in the leading peptide of the TYKY subunit. This novel missense mutation was found as a heterozygote in one patient and her mother, but not among 202 healthy controls nor among 107 children with undefined encephalomyopathy. Bioinformatic analyses suggested that Arg18Cys could lead to marked changes in the physicochemical properties of the mitochondrial-targeting peptide of TYKY, but we could not see changes in the assembly or activity of complex I or in the transcription of NDUFS8 in the fibroblasts of our patient. We suggest that Arg18Cys in the leading peptide of the TYKY subunit is not solely pathogenic, and that other genetic factors contribute to the disease-causing potential of this mutation.


Assuntos
Complexo I de Transporte de Elétrons/genética , Variação Genética , Encefalomiopatias Mitocondriais/genética , NAD(P)H Desidrogenase (Quinona)/genética , Alelos , Substituição de Aminoácidos , Arginina/química , Arginina/genética , Criança , Biologia Computacional , Sequência Conservada , Cisteína/química , Cisteína/genética , Complexo I de Transporte de Elétrons/deficiência , Humanos , Encefalomiopatias Mitocondriais/enzimologia , Mutação , NAD(P)H Desidrogenase (Quinona)/deficiência , NADH Desidrogenase , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Análise de Sequência de DNA , Transcrição Gênica
17.
Chem Biol Interact ; 153-154: 147-57, 2005 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-15935811

RESUMO

Quinone oxidoreductases (NQO1 and NQO2) are cytosolic proteins that catalyze metabolic reduction of quinones and its derivatives to protect cells against redox cycling and oxidative stress. In humans, a high percentage of individuals with myeloid and other types of leukemia are homo- and heterozygous for a null mutant allele of NQO1. The NQO2 locus is also highly polymorphic in humans. Recently, we generated NQO1-/- and NQO2-/- mice deficient in NQO1 and NQO2 protein and activity, respectively. These mice showed no detectable developmental abnormalities and were indistinguishable from wild type mice. Interestingly, all the mice lacking expression of NQO1 and NQO2 protein demonstrated myelogenous hyperplasia of the bone marrow and increased granulocytes in the peripheral blood. Decreased apoptosis contributed to myelogenous hyperplasia. The studies on short-term exposure of NQO1-/- mice to benzene demonstrated substantially greater benzene-induced toxicity, as compared to wild type mice.


Assuntos
Benzeno/toxicidade , Medula Óssea/patologia , Transtornos Mieloproliferativos/enzimologia , NAD(P)H Desidrogenase (Quinona)/genética , Quinona Redutases/genética , Animais , Medula Óssea/enzimologia , Predisposição Genética para Doença , Humanos , Hiperplasia/induzido quimicamente , Hiperplasia/enzimologia , Hiperplasia/genética , Leucemia/induzido quimicamente , Leucemia/enzimologia , Leucemia/genética , Camundongos , Camundongos Knockout , Transtornos Mieloproliferativos/induzido quimicamente , Transtornos Mieloproliferativos/genética , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/metabolismo , Polimorfismo Genético , Quinona Redutases/deficiência , Quinona Redutases/metabolismo
18.
Chem Biol Interact ; 153-154: 159-64, 2005 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-15935812

RESUMO

Benzene induces bone marrow cytotoxicity and chromosomal breaks as a primary mode of action for the induction of bone marrow toxicity. Our research group has used genetically modified mouse models to examine metabolic and genomic response pathways involved in benzene induced cytotoxicity and genotoxicity in bone marrow and in hematopoietic stem cells (HSC). We review our studies using NQO1-/- mice and mEH-/- mice to examine the roles of these enzymes, NAD(P)H:quinone oxidoreductase-1 (NQO1) and microsomal epoxide hydrolase (mEH) in mediating benzene-induced toxicity. NQO1 catalyzes the detoxication of benzene quinone metabolites and mEH catalyzes the hydrolysis of benzene oxide. Our studies using gene expression profiling of bone marrow and enriched HSC populations isolated from the bone marrow of benzene-exposed mice demonstrate differential gene expression responses of key genes induced by inhaled benzene. These studies show that benzene toxicity is regulated by a number of genetic pathways that affect the production of reactive metabolites and DNA damage response pathways in a target tissue.


Assuntos
Benzeno/toxicidade , Medula Óssea/efeitos dos fármacos , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/efeitos dos fármacos , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Epóxido Hidrolases/deficiência , Epóxido Hidrolases/genética , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Knockout , Testes para Micronúcleos , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Reticulócitos/efeitos dos fármacos , Reticulócitos/patologia
19.
Cancer Res ; 63(18): 6016-25, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-14522930

RESUMO

Mitomycin C (MMC) is an anticancer drug that requires reductive activation to exert its toxicity. MMC is known to cross-link DNA that contributes significantly to the cytotoxicity and consequent cell death. Cytosolic NADPH:quinone oxidoreductase 1 (NQO1) and microsomal enzymes have been shown to mediate MMC-induced DNA cross-linking. However, NQO1 plays only a minor role, indicating presence of other cytosolic enzymes/proteins that contribute to this process. In this study, we have characterized a unique cytosolic activity in NQO1-null mice that catalyzed MMC-induced DNA cross-linking. This activity was cofactor independent and dicoumarol insensitive. The unique cytosolic activity was purified to homogeneity. The peptide sequencing of the purified protein identified the unique cytosolic activity as GRP58 (M(r) 58,000 glucose-regulatory protein), also known as GRp57/ER60/ERp61/HIP-70/Q2 and CPT. Immunodepletion of NQO1-null mice liver cytosol and partially purified fractions with anti-GRP58 antibody led to a complete loss of GRP58 protein and consequent significant reduction of MMC-induced DNA cross-linking. Mouse cDNA encoding GRP58 was isolated and sequenced. Chinese hamster ovary cells permanently overexpressing GRP58 showed increased MMC-induced DNA cross-linking and increased cytotoxicity on exposure to MMC. Bacterially expressed and purified GRP58 increased the MMC-induced DNA cross-linking when added to mouse cytosolic samples. A tissue array analysis indicated that GRP58 is ubiquitously expressed among mouse tissues, although at different levels. Expression analysis using matched human tumor/normal array revealed an up-regulation of GRP58 in breast, uterus, lung, and stomach tumors compared with normal tissues of similar origin.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Dano ao DNA/fisiologia , Proteínas de Choque Térmico/fisiologia , Isomerases/fisiologia , Mitomicina/farmacologia , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Reagentes de Ligações Cruzadas/farmacologia , Citosol/metabolismo , DNA/efeitos dos fármacos , DNA/metabolismo , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/metabolismo , Humanos , Isomerases/biossíntese , Isomerases/metabolismo , Fígado/enzimologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/genética , Isomerases de Dissulfetos de Proteínas , Homologia de Sequência de Aminoácidos
20.
Cancer Res ; 63(5): 929-35, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12615705

RESUMO

Enzymes that activate and detoxify benzene are likely genetic determinants of benzene-induced toxicity.NAD(P)H: quinone oxidoreductase-1 (NQO1) detoxifies benzoquinones, proposed toxic metabolites of benzene. NQO1 deficiency in humans is associated with an increased risk of leukemia, specifically acute myelogenous leukemia, and benzene poisoning. We examined the importance of NQO1 in benzene-induced toxicity by hypothesizing that NQO1-deficient (NQO1-/-) mice are more sensitive to benzene than mice with wild-type NQO1 (NQO1+/+; 129/Sv background strain). Male and female NQO1-/- and NQO1+/+ mice were exposed to inhaled benzene (0, 10, 50, or 100 ppm) for 2 weeks, 6 h/day, 5 days/week. Micronucleated peripheral blood cells were counted to assess genotoxicity. Peripheral blood counts and bone marrow histology were used to assess hematotoxicity and myelotoxicity. p21 mRNA levels in bone marrow cells were used as determinants of DNA damage response. Female NQO1-/- mice were more sensitive (6-fold) to benzene-induced genotoxicity than the female NQO1+/+ mice. Female NQO1-/- mice had a 9-fold increase (100 versus 0 ppm) in micronucleated reticulocytes compared with a 3-fold increase in the female NQO1+/+ mice. However, the induced genotoxic response in male mice was similar between the two genotypes (> or = 10-fold increase at 100 ppm versus 0 ppm). Male and female NQO1-/- mice exhibited greater hematotoxicity than NQO1+/+ mice. p21 mRNA levels were induced significantly in male mice (>10-fold) from both strains and female NQO1-/- mice (> 8-fold), which indicates an activated DNA damage response. These results indicate that NQO1 deficiency results in substantially greater benzene-induced toxicity. However, the specific patterns of toxicity differed between the male and female mice.


Assuntos
Benzeno/toxicidade , NAD(P)H Desidrogenase (Quinona)/metabolismo , Animais , Benzeno/farmacocinética , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Citocromo P-450 CYP2E1/metabolismo , DNA/efeitos dos fármacos , Dano ao DNA , Feminino , Predisposição Genética para Doença , Doenças Hematológicas/induzido quimicamente , Doenças Hematológicas/patologia , Inativação Metabólica/genética , Masculino , Camundongos , Microssomos Hepáticos/enzimologia , NAD(P)H Desidrogenase (Quinona)/deficiência , NAD(P)H Desidrogenase (Quinona)/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA