Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Redox Rep ; 29(1): 2332038, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38563333

RESUMO

OBJECTIVES: Gentamicin is one of the most common ototoxic drugs that can lower patients' quality of life. Oxidative stress is a key factors inducing sensory hair cell death during gentamicin administration. So far, there are no effective drugs to prevent or treat gentamicin- induced hearing loss. A recent study found cystic fibrosis transmembrane conductance regulator (CFTR) as a new target to modulate cellular oxidative balance. The objective of this study was to estimate the effect of the CFTR activator ivacaftor on gentamicin-induced ototoxicity and determine its mechanism. METHODS: The hair cell count was analyzed by Myosin 7a staining. Apoptosis was analyzed by TUNEL Apoptosis Kit. Cellular reactive oxygen species (ROS) level was detected by DCFH-DA probes. The Nrf2 related proteins expression levels were analyzed by western blot. RESULTS: An in vitro cochlear explant model showed that gentamicin caused ROS accumulation in sensory hair cells and induced apoptosis, and this effect was alleviated by pretreatment with ivacaftor. Western blotting showed that ivacaftor administration markedly increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO1), and NAD(P)H:quinone oxidoreductase 1 (NQO1). The protective effect of ivacaftor was abolished by the Nrf2 inhibitor ML385. DISCUSSION: Our results indicate the protective role of the CFTR-Nrf2-HO1/NQO1 pathway in gentamicin-induced ototoxicity. Ivacaftor may be repositioned or repurposed towards aminoglycosides-induced hearing loss.


Assuntos
Aminofenóis , Perda Auditiva , Ototoxicidade , Quinolonas , Humanos , Gentamicinas/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Qualidade de Vida , Estresse Oxidativo , Apoptose , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia
2.
Ecotoxicol Environ Saf ; 270: 115853, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38128313

RESUMO

BACKGROUND: Manganese (Mn) and iron (Fe) are essential trace elements for humans, yet excessive exposure to Mn or Fe can accumulate in the central nervous system (CNS) and cause neurotoxicity. The purpose of this study was to investigate the effects of Mn and Fe exposure, alone or in combination, on inducing oxidative stress-induced neurological damage in rat cortical and SH-SY5Y cells, and to determine whether combined exposure to these metals increases their individual toxicity. METHODS: SH-SY5Y cells and male Sprague-Dawley rats were used to observe the effects of oxidative stress-induced neurological damage induced by exposure to manganese and iron alone or in combination. To detect the expression of anti-oxidative stress-related proteins, Nrf2, HO-1, and NQO1, and the apoptosis-related proteins, Bcl2 and Bax, and the neurological damage-related protein, α-syn. To detect reactive oxygen species generation and apoptosis. To detect the expression of the rat cortical protein Nrf2. To detect the production of proinflammatory cytokines. RESULTS: We demonstrate that juvenile developmental exposure to Mn and Fe and their combination impairs cognitive performance in rats by inducing oxidative stress causing neurodegeneration in the cortex. Mn, Fe, and their combined exposure increased the expression of ROS, Bcl2, Bax, and α-syn, activated the inflammatory factors IL-6 and IL-12, inhibited the activities of SOD and GSH, and induced oxidative stress-induced neurodegeneration both in rats and SH-SY5Y cells. Combined Mn-Fe exposure attenuated the oxidative stress induced by Mn and Fe exposure alone by increasing the expression of antioxidant factors Nrf2, HO-1, and NQO1. CONCLUSION: In both in vivo and in vitro studies, manganese and iron alone or in combination induced oxidative stress, leading to neuronal damage. In contrast, combined exposure to manganese and iron mitigated the oxidative stress induced by exposure to manganese and iron alone by increasing the expression of antioxidant factors. Therefore, studies to elucidate the main causes of toxicity and establish the molecular mechanisms of toxicity should help to develop more effective therapeutic modalities in the future.


Assuntos
Manganês , Neuroblastoma , Humanos , Masculino , Ratos , Animais , Manganês/toxicidade , Antioxidantes/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Ferro/metabolismo , Proteína X Associada a bcl-2/metabolismo , Ratos Sprague-Dawley , Estresse Oxidativo , Apoptose , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia
3.
J Nat Med ; 77(1): 12-27, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36074228

RESUMO

Hops, the dried female clusters from Humulus lupulus L., have traditionally been used as folk medicines for treating insomnia, neuralgia, and menopausal disorders. However, its pharmacological action on iron overload induced nerve damage has not been investigated. This study aims to evaluate the protective effects of hops extract (HLE) and its active constituent xanthohumol (XAN) on nerve injury induced by iron overload in vivo and in vitro, and to explore its underlying mechanism. The results showed that HLE and XAN significantly improved the memory impairment of iron overload mice, mainly manifested as shortened latency time, increased crossing platform times and spontaneous alternation ratio, and increased the expression of related proteins. Additionally, HLE and XAN significantly increased superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) activities, and remarkably decreased malondialdehyde (MDA) level in hippocampus. Also, HLE and XAN apparently reduced reactive oxygen species (ROS) content of PC12 cells induced by iron dextran (ID), and improved the oxidative stress level. Moreover, HLE and XAN significantly upregulated the expression of nuclear factor E2-related factor (Nrf2), NAD(P)H quinone oxidoreductase (NQO1), heme oxygenase-1 (HO-1), SOD, phosphorylated AKT (p-AKT), and phosphorylated GSK3ß (p-GSK3ß) both in hippocampus and PC12 cells. These findings demonstrated the protective effect of HLE and XAN against iron-induced memory impairment, which is attributed to its antioxidant profile by activation of AKT/GSK3ß and Nrf2/NQO1 pathways. Also, it was suggested that hops could be a potential candidate for iron overload-related neurological diseases treatment.


Assuntos
Humulus , Sobrecarga de Ferro , Ratos , Feminino , Camundongos , Animais , Humulus/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Estresse Oxidativo , Antioxidantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Sobrecarga de Ferro/induzido quimicamente , Sobrecarga de Ferro/tratamento farmacológico , Ferro/farmacologia , Heme Oxigenase-1/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-35805773

RESUMO

Arsenic is widely present in nature and is a common environmental poison that seriously damages human health. Chronic exposure to arsenic is a major environmental poisoning factor that promotes cell proliferation and leads to malignant transformation. However, its molecular mechanism remains unclear. In this study, we found that arsenite can promote the transformation of immortalized human keratinocyte cells (HaCaT) from the G0/G1 phase to S phase and demonstrated malignant phenotypes. This phenomenon is accompanied by obviously elevated levels of NRF2, NQO1, Cyclin E, and Cyclin-dependent kinase 2 (CDK2). Silencing the NRF2 expression with small interfering RNA (siRNA) in arsenite-transformed (T-HaCaT) cells was shown to reverse the malignant phenotype. Furthermore, the siRNA silencing of NQO1 significantly decreased the levels of the cyclin E-CDK2 complex, inhibiting the G0/G1 to S phase cell cycle progression and transformation to the T-HaCaT phenotypes. Thus, we hypothesized that the NRF2/NQO1 pathway played a key role in the arsenite-induced malignancy of HaCaT cells. By increasing the expression of Cyclin E-CDK2, the NRF2/NQO1 pathway can affect cell cycle progression and cell proliferation. A new common health effect mechanism of arsenic carcinogenesis has been identified; thus, it would contribute to the development of novel treatments to prevent and treat skin cancer caused by arsenic.


Assuntos
Arsênio , Arsenitos , Arsênio/metabolismo , Linhagem Celular , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Ciclina E/farmacologia , Humanos , Queratinócitos , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , RNA Interferente Pequeno/metabolismo
5.
Chemistry ; 26(12): 2713-2718, 2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-31814180

RESUMO

Human NAD(P)H: Quinone Oxidoreductase 1 (hNQO1) is an attractive enzyme for cancer therapeutics due to its significant overexpression in tumors compared to healthy tissues. Its unique catalytic mechanism involving the two-electron reduction of quinone-based compounds has made it a useful target to exploit in the design of hNQO1 fluorescent chemosensors and hNQO1-activatable-prodrugs. In this work, hNQO1 is exploited for an optical therapeutic. The probe uses the photosensitizer, phenalenone, which is initially quenched via photo-induced electron transfer by the attached quinone. Native phenalenone is liberated in the presence of hNQO1 resulting in the production of cytotoxic singlet oxygen upon irradiation. hNQO1-mediated activation in A549 lung cancer cells containing high levels of hNQO1 induces a dose-dependent photo-cytotoxic response after irradiation. In contrast, no photo-cytotoxicity was observed in the normal lung cell line, MRC9. By targeting hNQO1, this scaffold can be used to enhance the cancer selectivity of photodynamic therapy.


Assuntos
Antineoplásicos/química , NAD(P)H Desidrogenase (Quinona)/química , NAD/metabolismo , Fenalenos/química , Fármacos Fotossensibilizantes/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , NAD(P)H Desidrogenase (Quinona)/farmacologia , Fenalenos/farmacologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Quinonas/metabolismo , Oxigênio Singlete/química
6.
Prog Biophys Mol Biol ; 152: 35-44, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31733221

RESUMO

Cellular bioenergetics is an area showing promise for the development of new antimicrobials, antimalarials and cancer therapy. Enzymes involved in central carbon metabolism and energy generation are essential mediators of bacterial physiology, persistence and pathogenicity, lending themselves natural interest for drug discovery. In particular, succinate and malate are two major focal points in both the central carbon metabolism and the respiratory chain of Mycobacterium tuberculosis. Both serve as direct links between the citric acid cycle and the respiratory chain due to the quinone-linked reactions of succinate dehydrogenase, fumarate reductase and malate:quinone oxidoreductase. Inhibitors against these enzymes therefore hold the promise of disrupting two distinct, but essential, cellular processes at the same time. In this review, we discuss the roles and unique adaptations of these enzymes and critically evaluate the role that future inhibitors of these complexes could play in the bioenergetics target space.


Assuntos
Antituberculosos/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/farmacologia , Succinato Desidrogenase/farmacologia , Tuberculose/tratamento farmacológico , Benzoquinonas/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Descoberta de Drogas , Humanos , Malatos/metabolismo , Oxirredução , Ligação Proteica , Ácido Succínico/metabolismo
7.
Biomed Res Int ; 2019: 2076579, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31886179

RESUMO

Cutaneous squamous cell carcinoma (SCC) is a common cancer that significantly decreases the quality of life. It is known that external stimulus such as ultraviolet (UV) radiation induces cutaneous SCC via provoking oxidative stress. NAD(P)H dehydrogenase 1 (NQO1) is a ubiquitous flavoenzyme that functions as a guardian against oxidative stress. However, the effect of NQO1 on cutaneous SCC is not clearly elucidated. In this study, we investigated the effect of NQO1 on cutaneous SCC cells using the recombinant adenoviruses that can upregulate and/or downregulate NQO1 expression. Overexpression of NQO1 resulted in significant decrease of cell proliferation and colony forming activity of SCC lines (SCC12 and SCC13 cells). By contrast, knockdown of NQO1 increased the cell proliferation and colony forming activity. Accordingly, the levels of proliferation-related regulators, such as Cyclin D1, Cyclin E, PCNA, SOX2, and p63, were decreased by the overexpression of NQO1, while those were increased by knockdown of NQO1. In addition, NQO1 affected the invasion and migration of SCC cells in a very similar way, with the regulation of epithelial-mesenchymal transition- (EMT-) related molecules, including E-cadherin, N-cadherin, Vimentin, Snail, and Slug. Finally, the overexpression of NQO1 decreased the level of phosphorylated AKT, JNK, and p38 MAPK, while the knockdown of NQO1 increased the level of phosphorylated signaling molecules. Based on these data, NQO1 has tumor suppressive function in cutaneous SCC cells.


Assuntos
Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica/genética , NAD(P)H Desidrogenase (Quinona) , Neoplasias Cutâneas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Humanos , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Cutâneas/metabolismo
8.
Neurotoxicology ; 55: 10-12, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27168424

RESUMO

Astrocytes are exposed to aminochrome via the oxidation of dopamine that is taken up from the synaptic cleft after its release from dopaminergic neurons. Glutathione transferase M2-2 (GSTM2) has been shown to protect astrocytes from aminochrome-induced toxicity, but astrocytes also express DT-diaphorase, which has been shown to prevent aminochrome-induced neurotoxicity in dopaminergic neurons. Therefore, the question is whether DT-diaphorase also protects astrocytes from aminochrome-induced toxicity. DT-diaphorase is constitutively expressed in U373MG cells, and its inhibition by dicoumarol induced a significant increase of aminochrome-induced cell death. However, the inhibition of DT-diaphorase in U373MGsiGST6 cells, which have 74% of GSTM2 gene expression silenced, resulted in a more than 2-fold increase in cell death, suggesting that DT-diaphorase plays an important role in preventing aminochrome-induced toxicity in astrocytes.


Assuntos
Astrócitos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Indolquinonas/toxicidade , NAD(P)H Desidrogenase (Quinona)/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Transformada , Dicumarol/farmacologia , Inibidores Enzimáticos/farmacologia , Glutationa Transferase/metabolismo , Humanos , RNA Interferente Pequeno/metabolismo
9.
BMB Rep ; 48(11): 609-17, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26424559

RESUMO

NAD(P)H: quinone oxidoreductase (NQO1), an obligatory two-electron reductase, is a ubiquitous cytosolic enzyme that catalyzes the reduction of quinone substrates. The NQO1- mediated two-electron reduction of quinones can be either chemoprotection/detoxification or a chemotherapeutic response, depending on the target quinones. When toxic quinones are reduced by NQO1, they are conjugated with glutathione or glucuronic acid and excreted from the cells. Based on this protective effect of NQO1, the use of dietary compounds to induce the expression of NQO1 has emerged as a promising strategy for cancer prevention. On the other hand, NQO1-mediated two-electron reduction converts certain quinone compounds (such as mitomycin C, E09, RH1 and -lapachone) to cytotoxic agents, leading to cell death. It has been known that NQO1 is expressed at high levels in numerous human cancers, including breast, colon, cervix, lung, and pancreas, as compared with normal tissues. This implies that tumors can be preferentially damaged relative to normal tissue by cytotoxic quinone drugs. Importantly, NQO1 has been shown to stabilize many proteins, including p53 and p33ING1b, by inhibiting their proteasomal degradation. This review will summarize the biological roles of NQO1 in cancer, with emphasis on recent findings and the potential of NQO1 as a therapeutic target for the cancer therapy.


Assuntos
Antineoplásicos/farmacologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Animais , Humanos , NAD(P)H Desidrogenase (Quinona)/biossíntese , Quinonas/metabolismo
10.
Chin J Cancer ; 33(2): 80-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23845143

RESUMO

Hypoxia, a state of low oxygen, is a common feature of solid tumors and is associated with disease progression as well as resistance to radiotherapy and certain chemotherapeutic drugs. Hypoxic regions in tumors, therefore, represent attractive targets for cancer therapy. To date, five distinct classes of bioreactive prodrugs have been developed to target hypoxic cells in solid tumors. These hypoxia-activated prodrugs, including nitro compounds, N-oxides, quinones, and metal complexes, generally share a common mechanism of activation whereby they are reduced by intracellular oxidoreductases in an oxygen-sensitive manner to form cytotoxins. Several examples including PR-104, TH-302, and EO9 are currently undergoing phase II and phase III clinical evaluation. In this review, we discuss the nature of tumor hypoxia as a therapeutic target, focusing on the development of bioreductive prodrugs. We also describe the current knowledge of how each prodrug class is activated and detail the clinical progress of leading examples.


Assuntos
Antineoplásicos/farmacologia , Hipóxia Celular/efeitos dos fármacos , Neoplasias , Pró-Fármacos/farmacologia , Antraquinonas/química , Antraquinonas/farmacologia , Antineoplásicos/química , Aziridinas/química , Aziridinas/farmacologia , Humanos , Indolquinonas/química , Indolquinonas/farmacologia , Estrutura Molecular , NAD(P)H Desidrogenase (Quinona)/química , NAD(P)H Desidrogenase (Quinona)/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Compostos de Mostarda Nitrogenada/química , Compostos de Mostarda Nitrogenada/farmacologia , Nitroimidazóis/química , Nitroimidazóis/farmacologia , Mostardas de Fosforamida/química , Mostardas de Fosforamida/farmacologia , Pró-Fármacos/química , Tirapazamina , Triazinas/química , Triazinas/farmacologia
11.
Free Radic Biol Med ; 65: 1455-1463, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23912160

RESUMO

Mitochondrial dysfunction is associated with various forms of lung injury and disease that also involve alterations in pulmonary endothelial permeability, but the relationship, if any, between the two is not well understood. This question was addressed by perfusing isolated intact rat lung with a buffered physiological saline solution in the absence or presence of the mitochondrial complex I inhibitor rotenone (20 µM). Compared to control, rotenone depressed whole lung tissue ATP from 5.66 ± 0.46 (SEM) to 2.34 ± 0.15 µmol · g(-1) dry lung, with concomitant increases in the ADP:ATP and AMP:ATP ratios. Rotenone also increased lung perfusate lactate (from 12.36 ± 1.64 to 38.62 ± 3.14 µmol · 15 min(-1) perfusion · g(-1) dry lung) and the lactate:pyruvate ratio, but had no detectable impact on lung tissue GSH:GSSG redox status. The amphipathic quinone coenzyme Q1 (CoQ1; 50 µM) mitigated the impact of rotenone on the adenine nucleotide balance, wherein mitigation was blocked by NAD(P)H-quinone oxidoreductase 1 or mitochondrial complex III inhibitors. In separate studies, rotenone increased the pulmonary vascular endothelial filtration coefficient (Kf) from 0.043 ± 0.010 to 0.156 ± 0.037 ml · min(-1) · cm H2O(-1) · g(-1) dry lung, and CoQ1 protected against the effect of rotenone on Kf. A second complex I inhibitor, piericidin A, qualitatively reproduced the impact of rotenone on Kf and the lactate:pyruvate ratio. Taken together, the observations imply that pulmonary endothelial barrier integrity depends on mitochondrial bioenergetics as reflected in lung tissue ATP levels and that compensatory activation of whole lung glycolysis cannot protect against pulmonary endothelial hyperpermeability in response to mitochondrial blockade. The study further suggests that low-molecular-weight amphipathic quinones may have therapeutic utility in protecting lung barrier function in mitochondrial insufficiency.


Assuntos
Complexo I de Transporte de Elétrons/antagonistas & inibidores , Pulmão/irrigação sanguínea , Mitocôndrias/metabolismo , Rotenona/farmacologia , Ubiquinona/farmacologia , Trifosfato de Adenosina/análise , Animais , Antibacterianos/farmacologia , Barreira Alveolocapilar/efeitos dos fármacos , Permeabilidade Capilar , Complexo III da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Endotélio Vascular/metabolismo , Metabolismo Energético , Glicólise , Ácido Láctico/análise , Lesão Pulmonar , Masculino , NAD(P)H Desidrogenase (Quinona)/farmacologia , Oxirredução/efeitos dos fármacos , Piridinas/farmacologia , Ácido Pirúvico/análise , Ratos , Ratos Sprague-Dawley , Reperfusão , Desacopladores/farmacologia
12.
ACS Chem Biol ; 8(10): 2173-83, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23937670

RESUMO

A major goal of personalized medicine in oncology is the identification of drugs with predictable efficacy based on a specific trait of the cancer cell, as has been demonstrated with gleevec (presence of Bcr-Abl protein), herceptin (Her2 overexpression), and iressa (presence of a specific EGFR mutation). This is a challenging task, as it requires identifying a cellular component that is altered in cancer, but not normal cells, and discovering a compound that specifically interacts with it. The enzyme NQO1 is a potential target for personalized medicine, as it is overexpressed in many solid tumors. In normal cells NQO1 is inducibly expressed, and its major role is to detoxify quinones via bioreduction; however, certain quinones become more toxic after reduction by NQO1, and these compounds have potential as selective anticancer agents. Several quinones of this type have been reported, including mitomycin C, RH1, EO9, streptonigrin, ß-lapachone, and deoxynyboquinone (DNQ). However, no unified picture has emerged from these studies, and the key question regarding the relationship between NQO1 processing and anticancer activity remains unanswered. Here, we directly compare these quinones as substrates for NQO1 in vitro, and for their ability to kill cancer cells in culture in an NQO1-dependent manner. We show that DNQ is a superior NQO1 substrate, and we use computationally guided design to create DNQ analogues that have a spectrum of activities with NQO1. Assessment of these compounds definitively establishes a strong relationship between in vitro NQO1 processing and induction of cancer cell death and suggests these compounds are outstanding candidates for selective anticancer therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , NAD(P)H Desidrogenase (Quinona)/farmacologia , Quinonas/química , Western Blotting , Domínio Catalítico , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Moleculares , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Oxirredução , Especificidade por Substrato
13.
Org Lett ; 15(11): 2636-9, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23659457

RESUMO

The design, synthesis, and development of INDQ/NO, a novel nitric oxide (NO) prodrug targeted by a bioreductive trigger, are described. INDQ/NO, an indolequinone-diazeniumdiolate is found to be metabolized to produce NO by DT-diaphorase, a bioreductive enzyme that is overexpressed in certain cancers and hypoxic tumors. Cell-based assays revealed that INDQ/NO induces DNA damage and is a potent inhibitor of cancer cell proliferation.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Aziridinas/síntese química , Proliferação de Células/efeitos dos fármacos , Indolquinonas/síntese química , Indolquinonas/farmacologia , NAD(P)H Desidrogenase (Quinona)/química , NAD(P)H Desidrogenase (Quinona)/farmacologia , Óxido Nítrico/química , Óxido Nítrico/farmacologia , Pró-Fármacos/síntese química , Aziridinas/química , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Indolquinonas/química , NAD(P)H Desidrogenase (Quinona)/metabolismo , Óxido Nítrico/metabolismo , Pró-Fármacos/química
14.
Biochem Pharmacol ; 80(7): 977-81, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20599803

RESUMO

NAD(P)H quinone oxidoreductase (NQO1) has multiple functions in the cell including an ability to act as a detoxifying enzyme and as a protein chaperone. The latter property is particularly important in oncology as one of the client proteins of NQO1 is p53. The inhibitor, dicoumarol, is classically used to probe the biological properties of NQO1, but interpretation of enzyme function is compromised by the multiple "off-target" effects of this agent. Coumarin-based compounds that are more potent than dicoumarol as inhibitors of recombinant human NQO1 have been identified (Nolan et al., J Med Chem 2009;52:7142-56) The purpose of the work reported here is to demonstrate the functional activity of these agents for inhibiting NQO1 in cells. To do this, advantage was taken of the NQO1-mediated toxicity of the chemotherapeutic drug EO9 (Apaziquone). The toxicity of this drug is substantially reduced when the function of NQO1 is inhibited and many of the coumarin-based compounds are more efficient than dicoumarol for inhibiting EO9 toxicity. The ability to do this appears to be related to their capacity to inhibit NQO1 in cell free systems. In conclusion, agents have been identified that may be more pharmacologically useful than dicoumarol for probing the function of NQO1 in cells and tissues.


Assuntos
NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/metabolismo , Antineoplásicos/antagonistas & inibidores , Antineoplásicos/farmacologia , Aziridinas/antagonistas & inibidores , Aziridinas/farmacologia , Dicumarol/antagonistas & inibidores , Dicumarol/farmacologia , Humanos , Indolquinonas/farmacologia , NAD/antagonistas & inibidores , NAD/metabolismo , NAD/farmacologia , NAD(P)H Desidrogenase (Quinona)/farmacologia , Neoplasias , Proteínas/antagonistas & inibidores , Proteínas/farmacologia , Relação Estrutura-Atividade , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/farmacologia
15.
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
16.
Cancer Treat Rev ; 30(5): 437-49, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15245776

RESUMO

DT-diaphorase (DTD) is an obligate two-electron reductase which bioactivates chemotherapeutic quinones. DTD levels are elevated in a number of tumour types, including non-small cell lung carcinoma, colorectal carcinoma, liver cancers and breast carcinomas, when compared to the surrounding normal tissue. The differential in DTD between tumour and normal tissue should allow targeted activation of chemotherapeutic quinones in the tumour whilst minimising normal tissue toxicity. The prototypical bioreductive drug is Mitomycin C (MMC) which is widely used in clinical practice. However, MMC is actually a relatively poor substrate for DTD and its metabolism is pH-dependent. Other bioreductive drugs have failed because of poor solubility and inability to surpass other agents in use. RH1, a novel diaziridinylbenzoquinone, is a more efficient substrate for DTD. It has been demonstrated to have anti-tumour effects both in vitro and in vivo and demonstrates a relationship between DTD expression levels and drug response. RH1 has recently entered a phase I clinical trial in solid tumours under the auspices of Cancer Research UK. Recent work has demonstrated that DTD is present in the nucleus and is associated with both p53 and the heat shock protein, HSP-70. Furthermore, DTD is inducible by several non-toxic compounds and therefore much interest has focussed on increasing the differential in DTD levels between tumour and normal tissues.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacologia , Mitomicina/metabolismo , Mitomicina/farmacologia , NAD(P)H Desidrogenase (Quinona)/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Quinonas/metabolismo , Quinonas/farmacologia , Aziridinas/farmacologia , Benzoquinonas/farmacologia , Ensaios Clínicos como Assunto , Resistência a Medicamentos , Regulação Neoplásica da Expressão Gênica , Humanos , NAD(P)H Desidrogenase (Quinona)/biossíntese , Polimorfismo Genético , Proteína Supressora de Tumor p53
17.
Artigo em Chinês | MEDLINE | ID: mdl-15256146

RESUMO

OBJECTIVE: To explore the effects of interaction between environmental exposure factors and genetic polymorphism in toxicant metabolizing enzymes on risk of occupational chronic benzene poisoning. METHODS: One hundred and fifty-two cases of chronic benzene poisoning were analyzed for the risk by case-only study. RESULTS: The frequency of non-null GSTT1 gene in benzene poisoning workers with moderate benzene exposure level was higher than that in cases with lower benzene exposure (68.63% vs 38.00%, OR(adj) = 4.32, 95% CI 1.75 - 10.66, P = 0.002). The frequency of NQO1 C.609T/T gene in alcohol drinking group was higher than that in non-drinking group (61.11% vs 20.00%, OR(adj) = 8.03, 95% CI 2.28 - 28.25, P = 0.001), moreover, it was higher in workers with smoking and drinking than that in the rest group, and in drinking x exposure level workers than that in non-drinking x exposure level workers (85.71% vs 22.76%, OR(adj) = 18.62, 95% CI 2.01 - 172.72, P = 0.01 and 61.11% vs 20.00%, OR(adj) = 3.18, 95% CI 1.55 - 6.52, P = 0.002 respectively). The frequency of non-null GSTM1 gene was also higher in drinking x exposure level workers than that in non-drinking x exposure level workers (66.67% vs 47.06%, OR(adj) = 1.99, 95% CI 1.05 - 3.76, P = 0.036). CONCLUSION: There is interaction between the polymorphism of GSTT1 gene and moderate benzene exposure level; non-null GSTM1 gene and drinking x exposure level increase the risk of occupational chronic benzene poisoning; polymorphism of NQO1 gene C.609 also interacts with drinking, while polymorphism of NQO1 gene and drinking x smoking may further increase the risk of occupational chronic benzene poisoning.


Assuntos
Benzeno/envenenamento , Citocromo P-450 CYP2E1/genética , Glutationa Transferase/genética , Doenças Profissionais/genética , Polimorfismo de Fragmento de Restrição , Adulto , Benzeno/metabolismo , Citocromo P-450 CYP2E1/biossíntese , Citocromo P-450 CYP2E1/farmacologia , Feminino , Predisposição Genética para Doença , Genótipo , Glutationa Transferase/biossíntese , Glutationa Transferase/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , NAD(P)H Desidrogenase (Quinona)/biossíntese , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/farmacologia , Doenças Profissionais/enzimologia , Reação em Cadeia da Polimerase , Fatores de Risco
18.
Chem Res Toxicol ; 16(11): 1463-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14615973

RESUMO

Treatment for 48 h of murine Hepa 1c1c7 cells in culture with the cancer chemopreventive oltipraz (1) followed by addition of CD(3)I and immediate cell lysis yields, by LC/MS analysis, three isotopomers of the methylated pyrrolopyrazine (2), a known human metabolite of oltipraz. The major isotopomer (58%) is the one containing two CD(3)- groups attached to the pendant sulfur atoms of the pyrrolopyrazine ring, the others containing one CD(3)- and one CH(3)- group or two CH(3)- groups. It is concluded from this that the unmethylated pyrrolopyrazine (4) is the major metabolite of oltipraz. Prodrugs 5 and 6, which have been shown to rapidly generate 4 in the presence of GSH at physiological pH, induce the phase 2 enzyme NQO1 in Hepa 1c1c7 cells with potencies on par with oltipraz itself: CD(NQO1) = 14.4 +/- 1.3, 20.1 +/- 4.6, and 23.6 +/- 1.6 microM for oltipraz, 5, and 6, respectively. Pretreatment of oltipraz, 5, and 6 in cell culture media with 1 mM GSH, which is shown to immediately convert 5 and 6 to 4, followed by incubation with Hepa 1c1c7 cells shows similar potencies for oltipraz and the (decomposed) produrgs, with CD(NQO1) = 18.0 +/- 4.4 microM for 5, 17.8 +/- 0.2 microM for 6, and 13.5 +/- 1.4 microM for oltipraz. Treatment with compound 6 of murine hepatoma cells containing a luciferase gene under the control of the antioxidant response element (ARE) from the mouse heme oxygenase (ho-1) gene elicits induction of luciferase activity, CD = 35.8 +/- 2.8 microM, somewhat greater than the potency than oltipraz itself. Western blots of nuclear proteins isolated from Hepa 1c1c7 cells and probed with anti-Nrf2 indicate that as compared to vehicle DMSO, compound 6 stimulates nuclear translocation of Nrf2 from the cytosol. From this study, it is concluded that the major metabolite of the cancer chemopreventive oltipraz is a phase 2 enzyme inducer of comparable potency that activates the ARE and initiates nuclear translocation of transcription factor Nrf 2.


Assuntos
Indução Enzimática/efeitos dos fármacos , Pirazinas/metabolismo , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Indução Enzimática/fisiologia , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Luciferases/genética , Metilação , Camundongos , NAD(P)H Desidrogenase (Quinona)/farmacologia , Fator 2 Relacionado a NF-E2 , Pró-Fármacos/metabolismo , Pirazinas/química , Pirazinas/farmacologia , Pirróis/química , Pirróis/metabolismo , Tionas/química , Tionas/metabolismo , Tiofenos , Fatores de Tempo , Transativadores/genética , Transativadores/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Transfecção , Células Tumorais Cultivadas
19.
Environ Health Perspect ; 111(9): 1215-21, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12842776

RESUMO

Asphalt fumes are complex mixtures of various organic compounds, including polycyclic aromatic hydrocarbons (PAHs). PAHs require bioactivation by the cytochrome P-450 monooxygenase system to exert toxic/carcinogenic effects. The present study was carried out to characterize the acute pulmonary inflammatory responses and the alterations of pulmonary xenobiotic pathways in rats exposed to asphalt fumes by inhalation. Rats were exposed at various doses and time periods to air or to asphalt fumes generated at paving temperatures. To assess the acute damage and inflammatory responses, differential cell counts, acellular lactate dehydrogenase (LDH) activity, and protein content of bronchoalveolar lavage fluid were determined. Alveolar macrophage (AM) function was assessed by monitoring generation of chemiluminescence and production of tumor necrosis factor-alpha and interleukin-1. Alteration of pulmonary xenobiotic pathways was determined by monitoring the protein levels and activities of P-450 isozymes (CYP1A1 and CYP2B1), glutathioneS-transferase (GST), and NADPH:quinone oxidoreductase (QR). The results show that acute asphalt fume exposure did not cause neutrophil infiltration, alter LDH activity or protein content, or affect AM function, suggesting that short-term asphalt fume exposure did not induce acute lung damage or inflammation. However, acute asphalt fume exposure significantly increased the activity and protein level of CYP1A1 whereas it markedly reduced the activity and protein level of CYP2B1 in the lung. The induction of CYP1A1 was localized in nonciliated bronchiolar epithelial (Clara) cells, alveolar septa, and endothelial cells by immunofluorescence microscopy. Cytosolic QR activity was significantly elevated after asphalt fume exposure, whereas GST activity was not affected by the exposure. This induction of CYP1A1 and QR with the concomitant down-regulation of CYP2B1 after asphalt fume exposure could alter PAH metabolism and may lead to potential toxic effects in the lung.


Assuntos
Hidrocarbonetos/química , Exposição por Inalação , Pulmão/imunologia , Pulmão/patologia , Exposição Ocupacional , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Animais , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1A1/farmacologia , Citocromo P-450 CYP2B1/biossíntese , Citocromo P-450 CYP2B1/farmacologia , Indução Enzimática , Feminino , Glutationa Transferase/biossíntese , Glutationa Transferase/farmacologia , Inflamação , Interleucina-1/biossíntese , Pulmão/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/fisiologia , NAD(P)H Desidrogenase (Quinona)/biossíntese , NAD(P)H Desidrogenase (Quinona)/farmacologia , Hidrocarbonetos Policíclicos Aromáticos/farmacologia , Ratos , Ratos Sprague-Dawley , Temperatura , Fator de Necrose Tumoral alfa/biossíntese , Xenobióticos/metabolismo
20.
Cancer Biol Ther ; 2(2): 141-52, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12750552

RESUMO

Beta-lapachone (beta-Lap) triggers apoptosis in a number of human breast and prostate cancer cell lines through a unique apoptotic pathway that is dependent upon NQO1, a two-electron reductase. Recently, our laboratory showed that beta-lap-exposed MCF-7 cells exhibited an early increase in intracellular cytosolic Ca(2+) from endoplasmic reticulum stores, and that BAPTA-AM (an intracellular Ca(2+) chelator) blocked these early increases and partially inhibited all aspects of beta-lap-induced apoptosis. We now show that exposure of NQO1-expressing breast cancer cells to beta-lap stimulates a unique proteolytic apoptotic pathway involving mu-calpain activation. No apparent activation of m-calpain was noted. Upon activation, mu-calpain translocated to the nucleus concomitant with specific nuclear proteolytic events. Apoptotic responses in beta-lap-exposed NQO1-expressing cells were significantly delayed and survival enhanced by exogenous over-expression of calpastatin, a natural inhibitor of mu- and m-calpains. Furthermore, purified mu-calpain cleaved PARP to a unique fragment (approximately 60 kDa), not previously reported for calpains. We provide evidence that beta-lap-induced, mu-calpain-stimulated apoptosis does not involve any known apoptotic caspases; the activated fragments of caspases were not observed after beta-lap exposures, nor were there any changes in the pro-enzyme forms as measured by Western blot analyses. The ability of beta-lap to trigger an apparently novel, p53-independent, calpain-mediated apoptotic cell death further support the development of this drug for improved breast cancer therapy.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Calpaína/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia , Naftoquinonas/farmacologia , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/farmacologia , Calpaína/antagonistas & inibidores , Caspases/metabolismo , Núcleo Celular/metabolismo , Ensaio de Unidades Formadoras de Colônias , Inibidores de Cisteína Proteinase/farmacologia , Citosol/metabolismo , Ativação Enzimática , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Microscopia Confocal , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Transporte Proteico , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...