Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33372143

RESUMO

The electron-transferring flavoprotein-menaquinone oxidoreductase ABCX (EtfABCX), also known as FixABCX for its role in nitrogen-fixing organisms, is a member of a family of electron-transferring flavoproteins that catalyze electron bifurcation. EtfABCX enables endergonic reduction of ferredoxin (E°' ∼-450 mV) using NADH (E°' -320 mV) as the electron donor by coupling this reaction to the exergonic reduction of menaquinone (E°' -80 mV). Here we report the 2.9 Å structure of EtfABCX, a membrane-associated flavin-based electron bifurcation (FBEB) complex, from a thermophilic bacterium. EtfABCX forms a superdimer with two membrane-associated EtfCs at the dimer interface that contain two bound menaquinones. The structure reveals that, in contrast to previous predictions, the low-potential electrons bifurcated from EtfAB are most likely directly transferred to ferredoxin, while high-potential electrons reduce the quinone via two [4Fe-4S] clusters in EtfX. Surprisingly, EtfX shares remarkable structural similarity with mammalian [4Fe-4S] cluster-containing ETF ubiquinone oxidoreductase (ETF-QO), suggesting an unexpected evolutionary link between bifurcating and nonbifurcating systems. Based on this structure and spectroscopic studies of a closely related EtfABCX, we propose a detailed mechanism of the catalytic cycle and the accompanying structural changes in this membrane-associated FBEB system.


Assuntos
Flavoproteínas Transferidoras de Elétrons/metabolismo , Quinona Redutases/metabolismo , Quinona Redutases/ultraestrutura , Proteínas de Bactérias/metabolismo , Catálise , Microscopia Crioeletrônica/métodos , Transporte de Elétrons , Elétrons , Ferredoxinas/metabolismo , Flavinas/metabolismo , Flavoproteínas/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Modelos Moleculares , NAD/metabolismo , Fixação de Nitrogênio/fisiologia , Oxirredução , Pyrococcus furiosus/metabolismo , Quinona Redutases/fisiologia , Vitamina K 2/metabolismo
2.
J Neurosci ; 40(45): 8698-8714, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33046554

RESUMO

The formation of memory for a novel experience is a critical cognitive capacity. The ability to form novel memories is sensitive to age-related pathologies and disease, to which prolonged metabolic stress is a major contributing factor. Presently, we describe a dopamine-dependent redox modulation pathway within the hippocampus of male mice that promotes memory consolidation. Namely, following novel information acquisition, quinone reductase 2 (QR2) is suppressed by miRNA-182 (miR-182) in the CA1 region of the hippocampus via dopamine D1 receptor (D1R) activation, a process largely facilitated by locus coeruleus activity. This pathway activation reduces ROS generated by QR2 enzymatic activity, a process that alters the intrinsic properties of CA1 interneurons 3 h following learning, in a form of oxidative eustress. Interestingly, novel experience decreases QR2 expression predominately in inhibitory interneurons. Additionally, we find that in aged animals this newly described QR2 pathway is chronically under activated, resulting in miR-182 underexpression and QR2 overexpression. This leads to accumulative oxidative stress, which can be seen in CA1 via increased levels of oxidized, inactivated potassium channel Kv2.1, which undergoes disulfide bridge oligomerization. This newly described interneuron-specific molecular pathway lies alongside the known mRNA translation-dependent processes necessary for long-term memory formation, entrained by dopamine in CA1. It is a process crucial for the distinguishing features of novel memory, and points to a promising new target for memory enhancement in aging and age-dependent diseases.SIGNIFICANCE STATEMENT One way in which evolution dictates which sensory information will stabilize as an internal representation, relies on information novelty. Dopamine is a central neuromodulator involved in this process in the mammalian hippocampus. Here, we describe for the first time a dopamine D1 receptor-dependent quinone reductase 2 pathway in interneurons. This is a targeted redox event necessary to delineate a novel experience to a robust long-term internal representation. Activation of this pathway alone can explain the effect novelty has on "flashbulb" memories, and it can become dysfunctional with age and diseases, such as Alzheimer's disease.


Assuntos
Região CA1 Hipocampal/fisiologia , Dopamina/fisiologia , Interneurônios/fisiologia , Memória/fisiologia , Quinona Redutases/fisiologia , Transdução de Sinais/fisiologia , Envelhecimento/fisiologia , Envelhecimento/psicologia , Animais , Região CA1 Hipocampal/crescimento & desenvolvimento , Antagonistas de Dopamina/farmacologia , Medo/psicologia , Masculino , Consolidação da Memória/fisiologia , Memória de Longo Prazo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/biossíntese , MicroRNAs/genética , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Reconhecimento Psicológico , Canais de Potássio Shab/metabolismo
3.
J Inherit Metab Dis ; 43(5): 1024-1036, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32160317

RESUMO

Hydrogen sulfide, a signaling molecule formed mainly from cysteine, is catabolized by sulfide:quinone oxidoreductase (gene SQOR). Toxic hydrogen sulfide exposure inhibits complex IV. We describe children of two families with pathogenic variants in SQOR. Exome sequencing identified variants; SQOR enzyme activity was measured spectrophotometrically, protein levels evaluated by western blotting, and mitochondrial function was assayed. In family A, following a brief illness, a 4-year-old girl presented comatose with lactic acidosis and multiorgan failure. After stabilization, she remained comatose, hypotonic, had neurostorming episodes, elevated lactate, and Leigh-like lesions on brain imaging. She died shortly after. Her 8-year-old sister presented with a rapidly fatal episode of coma with lactic acidosis, and lesions in the basal ganglia and left cortex. Muscle and liver tissue had isolated decreased complex IV activity, but normal complex IV protein levels and complex formation. Both patients were homozygous for c.637G > A, which we identified as a founder mutation in the Lehrerleut Hutterite with a carrier frequency of 1 in 13. The resulting p.Glu213Lys change disrupts hydrogen bonding with neighboring residues, resulting in severely reduced SQOR protein and enzyme activity, whereas sulfide generating enzyme levels were unchanged. In family B, a boy had episodes of encephalopathy and basal ganglia lesions. He was homozygous for c.446delT and had severely reduced fibroblast SQOR enzyme activity and protein levels. SQOR dysfunction can result in hydrogen sulfide accumulation, which, consistent with its known toxicity, inhibits complex IV resulting in energy failure. In conclusion, SQOR deficiency represents a new, potentially treatable, cause of Leigh disease.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Doença de Leigh/enzimologia , Mitocôndrias/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Quinona Redutases/fisiologia , Acidose Láctica/patologia , Encefalopatias/patologia , Pré-Escolar , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Família , Feminino , Homozigoto , Humanos , Sulfeto de Hidrogênio/química , Cinética , Doença de Leigh/metabolismo , Imageamento por Ressonância Magnética , Masculino , Oxirredução , Quinona Redutases/química
4.
Biochim Biophys Acta ; 1857(4): 473-82, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26721205

RESUMO

For Vibrio cholerae, the coordinated import and export of Na(+) is crucial for adaptation to habitats with different osmolarities. We investigated the Na(+)-extruding branch of the sodium cycle in this human pathogen by in vivo (23)Na-NMR spectroscopy. The Na(+) extrusion activity of cells was monitored after adding glucose which stimulated respiration via the Na(+)-translocating NADH:quinone oxidoreductase (Na(+)-NQR). In a V. cholerae deletion mutant devoid of the Na(+)-NQR encoding genes (nqrA-F), rates of respiratory Na(+) extrusion were decreased by a factor of four, but the cytoplasmic Na(+) concentration was essentially unchanged. Furthermore, the mutant was impaired in formation of transmembrane voltage (ΔΨ, inside negative) and did not grow under hypoosmotic conditions at pH8.2 or above. This growth defect could be complemented by transformation with the plasmid encoded nqr operon. In an alkaline environment, Na(+)/H(+) antiporters acidify the cytoplasm at the expense of the transmembrane voltage. It is proposed that, at alkaline pH and limiting Na(+) concentrations, the Na(+)-NQR is crucial for generation of a transmembrane voltage to drive the import of H(+) by electrogenic Na(+)/H(+) antiporters. Our study provides the basis to understand the role of the Na(+)-NQR in pathogenicity of V. cholerae and other pathogens relying on this primary Na(+) pump for respiration.


Assuntos
Quinona Redutases/fisiologia , Sódio/metabolismo , Vibrio cholerae/metabolismo , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Potenciais da Membrana
5.
Methods Enzymol ; 554: 255-70, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25725526

RESUMO

The first step in the mammalian metabolism of H2S is catalyzed by sulfide:quinone oxidoreductase (SQOR). Human SQOR is an integral membrane protein, which presumably interacts with the inner mitochondrial membrane in a monotopic fashion. The enzyme is a member of a family of flavoprotein disulfide oxidoreductases (e.g., glutathione reductase) that utilize a Cys-S-S-Cys disulfide bridge as an additional redox center. SQOR catalyzes a two-electron oxidation of H2S to sulfane sulfur using coenzyme Q as electron acceptor. The enzyme also requires a third substrate to act as the acceptor of the sulfane sulfur from a cysteine persulfide intermediate. Here, we describe a method for the bacterial expression of human SQOR as a catalytically active membrane-bound protein, procedures for solubilization and purification of the recombinant protein to >95% homogeneity, and spectrophotometric assays to monitor SQOR-mediated H2S oxidation in reactions with different sulfane sulfur acceptors.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Quinona Redutases/fisiologia , Biocatálise , Ensaios Enzimáticos , Escherichia coli , Humanos , Sulfeto de Hidrogênio/química , Cinética , Oxirredução , Quinona Redutases/química , Quinona Redutases/isolamento & purificação
6.
Cell Biol Int ; 37(8): 835-42, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23554006

RESUMO

We have examined the role of melatonin receptor MT2 and quinone reductase II in the regulation of the redox status of preadipocytes (3T3-L1) in vitro. 3T3-L1 cells were treated with melatonin at a physiological concentration (10(-9) mol/L) and a supraphysiological (pharmacological) concentration (10(-3) mol/L) for 24 h. Luzindole (10(-4) mol/L), an antagonist of MT2 receptor, and prazosin (10(-5) mol/L), an inhibitor of quinone reductase II, were added 30 min before subsequent exposure of the cells to melatonin. The level of oxidative stress was determined by the analysis of activities of enzymes neutralising reactive oxygen species, and determination of the malondialdehyde (MDA) content. Melatonin increased activities of manganese and copper-zinc superoxide dismutase (MnSOD, Cu/ZnSOD) and catalase (CAT) at both a physiological concentration (10(-9) mol/L) and a pharmacological concentration (10(-3) mol/L). MDA content was unchanged, whereas activities of glutathione peroxidase (GSH-Px) and glutathione reductase (GSSG-Rd) were increased only by the physiological concentration. Both effects were partially inhibited by luzindole, but not prazosin. These observations suggest that melatonin, acting at least partially via MT2 receptors, can increase antioxidant enzymes activities in 3T3-L1 preadipocytes.


Assuntos
Adipócitos/metabolismo , Quinona Redutases/fisiologia , Receptor MT2 de Melatonina/fisiologia , Células 3T3-L1 , Animais , Catalase/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Malondialdeído/metabolismo , Melatonina/fisiologia , Camundongos , Oxirredução , Estresse Oxidativo , Prazosina/farmacologia , Quinona Redutases/antagonistas & inibidores , Receptor MT2 de Melatonina/antagonistas & inibidores , Superóxido Dismutase/metabolismo , Triptaminas/farmacologia
7.
Biofactors ; 36(5): 360-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20623546

RESUMO

There is ample evidence that shows an inverse relationship between consumption of fruit/vegetable-rich diets and the risk of cancer at various anatomical sites. In this review, we will assess and summarize recent advances on cancer prevention by resveratrol, a natural stilbenoid present in red grapes, peanuts, some common drinks, and dietary supplements. We will focus on data published within the past few years on in vivo model tumor animal studies that reinforce the chemopreventive efficacy of resveratrol against a multitude of cancers, as well as on its sensitization/enhancing activities against tumor cells when used in combination with established chemotherapeutic and pharmaceutical agents. In addition, we will review examples resveratrol-target proteins, denoted RTPs, including the 24-kDa cytosolic protein quinone reductase 2 (NQO2) discovered in our laboratory that may confer resveratrol responsiveness to cancer cells. We will discuss the possible role of NQO2 in mediating cancer prevention by resveratrol. Our analysis of published data strengthen support that resveratrol displays novel roles in various cellular processes, and help to establish an expanded molecular framework for cancer prevention by resveratrol in vivo.


Assuntos
Anticarcinógenos/farmacologia , Neoplasias/prevenção & controle , Quinona Redutases/fisiologia , Estilbenos/farmacologia , Animais , Antineoplásicos Fitogênicos , Disponibilidade Biológica , Neoplasias da Mama/prevenção & controle , Dieta , Feminino , Humanos , Masculino , Neoplasias da Próstata/prevenção & controle , Radiossensibilizantes/farmacologia , Resveratrol , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia
8.
Biochim Biophys Acta ; 1797(8): 1500-11, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20398623

RESUMO

Sulfide (H2S) is an inhibitor of mitochondrial cytochrome oxidase comparable to cyanide. In this study, poisoning of cells was observed with sulfide concentrations above 20 microM. Sulfide oxidation has been shown to take place in organisms/cells naturally exposed to sulfide. Sulfide is released as a result of metabolism of sulfur containing amino acids. Although in mammals sulfide exposure is not thought to be quantitatively important outside the colonic mucosa, our study shows that a majority of mammalian cells, by means of the mitochondrial sulfide quinone reductase (SQR), avidly consume sulfide as a fuel. The SQR activity was found in mitochondria isolated from mouse kidneys, liver, and heart. We demonstrate the precedence of the SQR over the mitochondrial complex I. This explains why the oxidation of the mineral substrate sulfide takes precedence over the oxidation of other (carbon-based) mitochondrial substrates. Consequently, if sulfide delivery rate remains lower than the SQR activity, cells maintain a non-toxic sulfide concentration (<1 microM) in their external environment. In the colonocyte cell line HT-29, sulfide oxidation provided the first example of reverse electron transfer in living cells, such a transfer increasing sulfide tolerance. However, SQR activity was not detected in brain mitochondria and neuroblastoma cells. Consequently, the neural tissue would be more sensitive to sulfide poisoning. Our data disclose new constraints concerning the emerging signaling role of sulfide.


Assuntos
Colo/metabolismo , Sulfeto de Hidrogênio/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Transporte de Elétrons , Células HT29 , Humanos , Camundongos , Mitocôndrias/metabolismo , NAD/metabolismo , Oxirredução , Quinona Redutases/genética , Quinona Redutases/fisiologia , Rotenona/farmacologia , Transdução de Sinais
9.
J Bioenerg Biomembr ; 41(6): 493-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19904590

RESUMO

Mitochondrial impairment has been collecting more and more attention as a contributing factor to the etiology of Parkinson's disease. Above all, the NADH-quinone oxidoreductase, complex I, of the respiratory chain seems to be most culpable. Complex I dysfunction is translated to an increased production of reactive oxygen species and a decreased energy supply. In the brain, the dopaminergic neurons are one of the most susceptible cells. Their death is directly linked to the disease apparition. Developing an effective gene therapy is challenged by harmful actions of reactive oxygen species. To overcome this problem a therapeutic candidate must be able to restore the NADH-quinone oxidoreductase activity regardless of how complex I is impaired. Here we discuss the potency of the yeast alternative NADH dehydrogenase, the Ndi1 protein, to reinstate the mitochondrial respiratory chain compensating for disabled complex I and the benefit Ndi1 brings toward retardation of Parkinson's disease.


Assuntos
Complexo I de Transporte de Elétrons/fisiologia , Doença de Parkinson/terapia , Proteínas de Saccharomyces cerevisiae/uso terapêutico , Animais , Complexo I de Transporte de Elétrons/uso terapêutico , Terapia Genética , Humanos , Proteínas Mitocondriais , Doença de Parkinson/etiologia , Quinona Redutases/fisiologia
10.
Asian J Androl ; 11(6): 653-61, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19767760

RESUMO

Resveratrol is a dietary polyphenol espoused to have chemopreventive activity against a variety of human cancer types. We first reported that resveratrol significantly decreases the proliferation of both androgen-dependent and hormone-refractory prostate cancer cells. However, the effects of resveratrol in normal prostate epithelial and stromal cells, particularly with regard to its uptake, subcellular distribution and intracellular targets, have not been investigated. To advance the knowledge on accessibility and cellular disposition of resveratrol in prostate cells, [(3)H] resveratrol, fractionation of cell extracts into subcellular compartments, Western blot analysis, resveratrol affinity column chromatography and flow cytometry were used to study the uptake and intracellular distribution of resveratrol in normally cultured prostate stromal (PrSCs) and epithelial cells (PrECs). Pretreatment of both PrSCs and PrECs for 2 days with resveratrol modulated its uptake and selectively increased its distribution to the membrane and organelle compartments. Resveratrol affinity column chromatography studies showed differential expression of a previously identified resveratrol-targeting protein, quinone reductase 2 (QR2), in PrSCs and PrECs. Flow cytometric analysis comparing resveratrol-treated and untreated PrSCs showed a large decrease in G(1)-phase and a concomitant increase in S and G(2)/M-phases of the cell cycle. These results suggest that resveratrol suppresses PrSC proliferation by affecting cell cycle phase distribution, which may involve the participation by QR2.


Assuntos
NAD(P)H Desidrogenase (Quinona)/fisiologia , Próstata/metabolismo , Quinona Redutases/fisiologia , Estilbenos/metabolismo , Ciclo Celular/fisiologia , Células Cultivadas , Humanos , Masculino , Próstata/citologia , Resveratrol
11.
Biochemistry ; 48(40): 9516-24, 2009 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-19694431

RESUMO

Vibrio cholerae and many other marine and pathogenic bacteria possess a unique respiratory complex, the Na(+)-pumping NADH:quinone oxidoreductase (Na(+)-NQR), which pumps Na(+) across the cell membrane using the energy released by the redox reaction between NADH and ubiquinone. To function as a selective sodium pump, Na(+)-NQR must contain structures that (1) allow the sodium ion to pass through the hydrophobic core of the membrane and (2) provide cation specificity to the translocation system. In other sodium-transporting proteins, the structures that carry out these roles frequently include aspartate and glutamate residues. The negative charge of these residues facilitates binding and translocation of sodium. In this study, we have analyzed mutants of acid residues located in the transmembrane helices of subunits B, D, and E of Na(+)-NQR. The results are consistent with the participation of seven of these residues in the translocation process of sodium. Mutations at NqrB-D397, NqrD-D133, and NqrE-E95 produced a decrease of approximately >or=10-fold in the apparent affinity of the enzyme for sodium (Km(app)(Na+)), which suggests that these residues may form part of a sodium-binding site. Mutation at other residues, including NqrB-E28, NqrB-E144, NqrB-E346, and NqrD-D88, had a strong effect on the quinone reductase activity of the enzyme and its sodium sensitivity, but a weaker effect on the apparent sodium affinity, consistent with a possible role in sodium conductance pathways.


Assuntos
Ácido Aspártico , Proteínas de Bactérias/química , Ácido Glutâmico , Proteínas de Membrana/química , Quinona Redutases/química , Sódio/química , Sódio/metabolismo , Vibrio cholerae/enzimologia , Ácido Aspártico/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Sítios de Ligação/genética , Transporte Biológico Ativo/genética , Ácido Glutâmico/genética , Concentração de Íons de Hidrogênio , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Mutagênese Sítio-Dirigida , Quinona Redutases/genética , Quinona Redutases/fisiologia , Vibrio cholerae/genética
12.
FEBS J ; 276(16): 4313-24, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19624732

RESUMO

Quinone reductases are ubiquitous soluble enzymes found in bacteria, fungi, plants and animals. These enzymes utilize a reduced nicotinamide such as NADH or NADPH to reduce the flavin cofactor (either FMN or FAD), which then affords two-electron reduction of cellular quinones. Although the chemical nature of the quinone substrate is still a matter of debate, the reaction appears to play a pivotal role in quinone detoxification by preventing the generation of potentially harmful semiquinones. In recent years, an additional role of quinone reductases as regulators of proteasomal degradation of transcription factors and possibly intrinsically unstructured protein has emerged. To fulfil this role, quinone reductase binds to the core particle of the proteasome and recruits certain transcription factors such as p53 and p73alpha to the complex. The latter process appears to be governed by the redox state of the flavin cofactor of the quinone reductase, thus linking the stability of transcription factors to cellular events such as oxidative stress. Here, we review the current evidence for protein complex formation between quinone reductase and the 20S proteasome in eukaryotic cells and describe the regulatory role of this complex in stabilizing transcription factors by acting as inhibitors of their proteasomal degradation.


Assuntos
Flavoproteínas/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Quinona Redutases/fisiologia , Flavoproteínas/metabolismo , Oxirredução , Estabilidade Proteica , Quinona Redutases/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
13.
Clin Cancer Res ; 15(5): 1534-42, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19223498

RESUMO

PURPOSE: NRH:quinone oxidoreductase 2 (NQO2) is known to protect against myelogenous hyperplasia. However, the role of NQO2 in prevention of hematologic malignancies remains unknown. Present studies investigated in vivo role of NQO2 in prevention of myeloproliferative disease and lymphomas. EXPERIMENTAL DESIGN: Wild-type and NQO2-null mice were exposed to 0, 1, and 3 Gy gamma-radiation. One year later, the mice were analyzed for the development of myeloproliferative disease and lymphomas. Immunohistochemistry analysis determined the B- and T-cell origin of lymphomas. The mice were also sacrificed at 6 and 48 h after radiation exposure and bone marrow was collected and analyzed for p53, Bax, and B-cell apoptosis. Bone marrow cells were cultured and the rate of degradation of p53 was analyzed. RESULTS: Seventy-two percent NQO2-null mice showed development of B-cell lymphomas in multiple tissues compared with 11% in wild-type mice exposed to 3 Gy gamma-radiation. In contrast, only 22% NQO2-null mice showed myeloproliferation compared with none in wild-type mice. Further analysis revealed that bone marrow from NQO2-null mice contained lower levels of p53 compared with wild-type mice due to rapid degradation of p53. In addition, the exposure to radiation resulted in lower induction of p53 and Bax and decreased B-cell apoptosis in NQO2-null mice. CONCLUSION: NQO2-null mice are highly susceptible to develop radiation-induced B-cell lymphomas. The lack of significant induction of p53 and Bax and decrease in B-cell apoptosis presumably contributed to the development of lymphomas. NQO2 functions as endogenous factor in prevention against radiation-induced B-cell lymphomas.


Assuntos
Linfócitos B/efeitos da radiação , Raios gama , Linfoma de Células B/etiologia , Neoplasias Induzidas por Radiação/etiologia , Quinona Redutases/fisiologia , Animais , Apoptose/efeitos da radiação , Linfócitos B/metabolismo , Linfócitos B/patologia , Western Blotting , Medula Óssea/metabolismo , Medula Óssea/patologia , Células Cultivadas , Feminino , Citometria de Fluxo , Linfoma de Células B/enzimologia , Linfoma de Células B/patologia , Masculino , Camundongos , Camundongos Knockout , Neoplasias Induzidas por Radiação/enzimologia , Neoplasias Induzidas por Radiação/patologia , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo
14.
Clin Exp Pharmacol Physiol ; 35(12): 1419-25, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18671725

RESUMO

1. Resveratrol, a polyphenol in red wine, has a cardioprotective effect. Resveratrol-targeting protein (RTP) has been purified using a resveratrol affinity column (RAC) and has been identified as quinone reductase type 2 (NQO2). We hypothesize that NQO2 is the target protein of resveratrol in vascular smooth muscle cells (VSMC) and that resveratrol inhibits proliferation of VSMC through its action on NQO2. In the present study, we investigated the correlation between NQO2 regulation and cell proliferation in VSMC in response to resveratrol treatment. 2. The RTP was purified using RAC and was detected with a NQO2 polyclonal antibody. The VSMC were incubated with resveratrol (1, 10 and 50 micromol/L) for 24, 48 and 72 h. Cell proliferation was detected by cell counting and bromodeoxyuridine (BrdU) assay. A lentiviral vector incorporating NQO2 short interference (si) RNA of short hairpin design was constructed and transduced into VSMC. Real-time quantitative polymerase chain reaction was used to measure NQO2 mRNA levels; NQO2 expression was determined by western blot analysis. 3. Using RAC, we extracted a 26 kDa protein from aortic smooth muscle, which was referred to as RTP-26. Proliferation of VSMC was inhibited by resveratrol in a concentration- and time-dependent manner. The mRNA and protein expression of NQO2 was also repressed by resveratrol in a concentration- and time-dependent manner. A similar pattern of inhibition was observed for cells treated with resveratrol (25 micromol/L) as for cells transduced with a lentiviral vector containing siRNA sequences against NQO2. 4. Collectively, these data indicate that the suppression of VSMC proliferation mediated by resveratrol correlates with NQO2 downregulation.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Quinona Redutases/antagonistas & inibidores , Estilbenos/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo/fisiologia , Masculino , Quinona Redutases/biossíntese , Quinona Redutases/fisiologia , Coelhos , Ratos , Ratos Sprague-Dawley , Resveratrol , Fatores de Tempo
15.
Cell Mol Life Sci ; 65(1): 141-60, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17938860

RESUMO

Quinones are abundant cyclic organic compounds present in the environment as well as in pro- and eukaryotic cells. Several species have been shown to possess enzymes that afford the two-electron reduction to the hydroquinone form in an attempt to avoid the generation of one-electron reduced semiquinone known to cause oxidative stress. These enzymes utilize a flavin cofactor, either FMN or FAD, to transfer a hydride from an electron donor, such as NAD(P)H, to a quinone substrate. This family of flavin-dependent quinone reductases shares a flavodoxin-like structure and reaction mechanism pointing towards a common evolutionary origin. Recent studies of their physiological functions in eukaryotes suggest a role beyond detoxication of quinones and involvement in the oxygen stress response. Accordingly, mammalian quinone reductases emerge as central molecular switches that control the lifespan of transcription factors, such as p53, and hence participate in the development of apoptosis and cell transformation.


Assuntos
Flavinas/fisiologia , Quinona Redutases/fisiologia , Animais , Apoptose , Transformação Celular Neoplásica , Humanos , Estresse Oxidativo , Fatores de Transcrição
16.
Appl Environ Microbiol ; 73(8): 2479-85, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17293530

RESUMO

An NAD(P)H-nicotine blue (quinone) oxidoreductase was discovered as a member of the nicotine catabolic pathway of Arthrobacter nicotinovorans. Transcriptional analysis and electromobility shift assays showed that the enzyme gene was expressed in a nicotine-dependent manner under the control of the transcriptional activator PmfR and thus was part of the nicotine regulon of A. nicotinovorans. The flavin mononucleotide-containing enzyme uses NADH and, with lower efficiency, NADPH to reduce, by a two-electron transfer, nicotine blue to the nicotine blue leuco form (hydroquinone). Besides nicotine blue, several other quinones were reduced by the enzyme. The NAD(P)H-nicotine blue oxidoreductase may prevent intracellular one-electron reductions of nicotine blue which may lead to semiquinone radicals and potentially toxic reactive oxygen species.


Assuntos
Arthrobacter/fisiologia , Nicotina/metabolismo , Estresse Oxidativo , Quinona Redutases/genética , Quinona Redutases/metabolismo , Regulon , Arthrobacter/enzimologia , Arthrobacter/genética , Clonagem Molecular , Coenzimas/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Mononucleotídeo de Flavina/farmacologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Hidroquinonas/metabolismo , Modelos Biológicos , NAD/metabolismo , NADP/metabolismo , Oxirredução , Quinona Redutases/isolamento & purificação , Quinona Redutases/fisiologia , RNA Bacteriano/biossíntese , RNA Bacteriano/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Especificidade por Substrato
17.
J Biol Chem ; 281(41): 30917-24, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16905546

RESUMO

NAD(P)H:quinone oxidoreductase 1 (NQO1) and NRH:quinone oxidoreductase 2 (NQO2) are cytosolic enzymes that catalyze metabolic reduction of quinones and derivatives. NQO1-null and NQO2-null mice were generated that showed decreased lymphocytes in peripheral blood, myeloid hyperplasia, and increased sensitivity to skin carcinogenesis. In this report, we investigated the in vivo role of NQO1 and NQO2 in immune response and autoimmunity. Both NQO1-null and NQO2-null mice showed decreased B-cells in blood, lower germinal center response, altered B cell homing, and impaired primary and secondary immune responses. NQO1-null and NQO2-null mice also showed susceptibility to autoimmune disease as revealed by decreased apoptosis in thymocytes and pre-disposition to collagen-induced arthritis. Further experiments showed accumulation of NADH and NRH, cofactors for NQO1 and NQO2, indicating altered intracellular redox status. The studies also demonstrated decreased expression and lack of activation of immune-related factor NF-kappaB. Microarray analysis showed altered chemokines and chemokine receptors. These results suggest that the loss of NQO1 and NQO2 leads to altered intracellular redox status, decreased expression and activation of NF-kappaB, and altered chemokines. The results led to the conclusion that NQO1 and NQO2 are endogenous factors in the regulation of immune response and autoimmunity.


Assuntos
Formação de Anticorpos , Autoimunidade , Regulação Neoplásica da Expressão Gênica , Regulação da Expressão Gênica , NADPH Desidrogenase/fisiologia , Quinona Redutases/fisiologia , Animais , Apoptose , Artrite/metabolismo , Células da Medula Óssea/metabolismo , Citosol/metabolismo , Camundongos , NAD(P)H Desidrogenase (Quinona) , NADPH Desidrogenase/metabolismo , NF-kappa B/metabolismo , Oxirredução , Quinona Redutases/metabolismo
18.
Biochemistry (Mosc) ; 70(2): 143-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15807651

RESUMO

Structural and catalytic properties of bacterial Na+-translocating NADH:quinone oxidoreductases are briefly described. Special attention is given to studies on kinetics of the enzyme interaction with NADH and the role of sodium ions in this process. Based on the existing data, possible model mechanisms of sodium transfer by Na+-translocating NADH:quinone oxidoreductase are proposed.


Assuntos
Proteínas de Bactérias/fisiologia , Quinona Redutases/fisiologia , Proteínas de Bactérias/química , Catálise , Modelos Biológicos , Oxirredução , Quinona Redutases/química , Vibrio/enzimologia
19.
Biochemistry ; 44(9): 3562-71, 2005 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-15736965

RESUMO

The ND6 subunit is one of seven mitochondrial DNA-encoded subunits of the proton-translocating NADH-quinone oxidoreductase (complex I). Physiological importance of the ND6 subunit is becoming increasingly apparent because a number of mutations leading to amino acid changes in this subunit have been found to be associated with known mitochondrial diseases. Using the Escherichia coli enzyme (NDH-1), we have investigated the NuoJ subunit (the E. coli counterpart of ND6) by employing a chromosomal DNA manipulation technique. A series of point mutations was constructed directly on the nuoJ gene in the chromosome targeting at highly conserved residues. Analyses with blue-native gel electrophoresis and immunological methods revealed that, in all point mutants, the assembly of NDH-1 was normal and that the deamino-NADH-K(3)Fe(CN)(6) reductase activity of the membrane was essentially the same as that of the wild-type. However, energy-coupled NDH-1 activities were affected to varied extents. Among them, mutants of the Val-65 residue that is located in the most conserved transmembrane segment significantly lost the coupled electron-transfer activities and exhibited diminished membrane potential and proton translocation. This may suggest that Val-65 or the area around it is important for energy transduction of the coupling site 1. Together with the results on mutations related to human diseases, possible functional roles of the NuoJ subunit have been discussed.


Assuntos
Cromossomos Bacterianos/química , DNA Bacteriano/química , Proteínas de Escherichia coli/química , Proteínas de Membrana/química , Subunidades Proteicas/química , Quinona Redutases/química , Sequência de Aminoácidos , Cromossomos Bacterianos/genética , DNA Bacteriano/genética , DNA Mitocondrial/química , DNA Mitocondrial/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Potenciais da Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína/genética , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Quinona Redutases/genética , Quinona Redutases/fisiologia , Análise de Sequência de DNA
20.
Microbiol Mol Biol Rev ; 68(4): 603-16, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15590775

RESUMO

Type II NAD(P)H:quinone oxidoreductases (NDH-2) catalyze the two-electron transfer from NAD(P)H to quinones, without any energy-transducing site. NDH-2 accomplish the turnover of NAD(P)H, regenerating the NAD(P)(+) pool, and may contribute to the generation of a membrane potential through complexes III and IV. These enzymes are usually constituted by a nontransmembrane polypeptide chain of approximately 50 kDa, containing a flavin moiety. There are a few compounds that can prevent their activity, but so far no general specific inhibitor has been assigned to these enzymes. However, they have the common feature of being resistant to the complex I classical inhibitors rotenone, capsaicin, and piericidin A. NDH-2 have particular relevance in yeasts like Saccharomyces cerevisiae and in several prokaryotes, whose respiratory chains are devoid of complex I, in which NDH-2 keep the balance and are the main entry point of electrons into the respiratory chains. Our knowledge of these proteins has expanded in the past decade, as a result of contributions at the biochemical level and the sequencing of the genomes from several organisms. The latter showed that most organisms contain genes that potentially encode NDH-2. An overview of this development is presented, with special emphasis on microbial enzymes and on the identification of three subfamilies of NDH-2.


Assuntos
Células Procarióticas/enzimologia , Quinona Redutases/genética , Quinona Redutases/fisiologia , Sequência de Aminoácidos , Animais , Bactérias/enzimologia , Bactérias/genética , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Dados de Sequência Molecular , Quinona Redutases/química , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA