• Title/Summary/Keyword: NQO1 (NAD(P)H quinone oxidoreductase1)

Search Result 28, Processing Time 0.028 seconds

NAD(P)H Quinone Oxidoreductase 1 (NQO1) as a Cancer Therapeutic Target (암 치료 표적으로의 NAD(P)H Quinone Oxidoreductase 1 (NQO1))

  • Park, Eun Jung;Kwon, Taeg Kyu
    • Journal of Life Science
    • /
    • v.24 no.1
    • /
    • pp.98-103
    • /
    • 2014
  • NAD(P)H quinone oxidoreductase 1 (NQO1) is a flavoprotein that catalyzes the two electron reduction of diverse substrates, including quinones. It uses NADH or NADPH as a cofactor for enzymatic machinery. In the metabolism of quinones, NQO1 has two conflicting functions because of the different stability of converted hydroquinones. The stable form of hydroquinone is excreted from cells by conjugation with glutathione or glucuronic acid. The unstable form of hydroquinone induces cell death by induction of oxidative stress and DNA damage. Certain quinones known as bio-reductive agents have a cytotoxic function following reduction by NQO1. Bio-reductive agents, such as ${\beta}$-lapachone or mitomycin C, induce the depletion of NAD(P)H and the generation of oxidative stress in an NQO1-dependent manner. NQO1 is highly expressed in several cancer tissues. Therefore, NQO1 is a good therapeutic target for cancer treatment with bio-reductive agents.

Effects of 6-Arylamino-5,8-quinolinediones and 6-Chlore-7-ary-lamino-5,8-isoquinolinediones on NAD(P)H : Quinone Oxidoreductase (NQO1 ) Activity and Their Cytotoxic Potential

  • Ryu, Chung-Kyu;Jeong, Hyeh-Jean;Lee, Sang-Kook;You, Hee-Jung;Choi, Ko-Un;Shim, Ju-Yeon;Heo, Yeon-Hoi;Lee, Chong-Ock
    • Archives of Pharmacal Research
    • /
    • v.24 no.5
    • /
    • pp.390-396
    • /
    • 2001
  • Synthesized 6-arylamino-5,8-quinolinediones 4a-4j and 6-chloro-7-arylamino-5,8-isoquinolinediones 5a-5g were evaluated for effects on NAD(P)H quinone oxidoreductase (NQOl ) activity with the cytosolic fractions derived from cultured human lung cancer cells and their cytotoxicity in cultured several human solid cancer cell lines. The 5,8-quinolinediones 4 and 5,8-isoquinolinediones 5 affected the reduction potential by NQO1 activity and showed a potent cytotoxic activity against human cancer cell lines. The tested compounds 4a, 5c, 5f, and 5g were considered as more potent cytotoxic agents. The compounds 4d, 5b, 5c, 5e and 5g were comparable modulators of NQO1 activity.

  • PDF

NAD(P)H-quinone oxidoreductase-1 silencing modulates cytoprotection related protein expression in cisplatin cytotoxicity

  • Park, Se Ra;Jung, Ju Young;Kim, Young-Jung;Jung, Da Young;Lee, Mee Young;Ryu, Si Yun
    • Korean Journal of Veterinary Research
    • /
    • v.56 no.1
    • /
    • pp.15-21
    • /
    • 2016
  • NAD(P)H-quinone oxidoreductase-1 (NQO1) is a down-stream target gene of nuclear factor erythroid 2-related factor 2 (Nrf2), and performs diverse biological functions. Recently, NQO1 is recognized as an effective gene for the cytotoxic inserts with its diverse biological functions, which is focused on antioxidant properties. The aim of present study was to assess the impact of NQO1 knockdown on cytoprotection-related protein expression in cisplatin cytotoxicity by using small interfering (si) RNA targeted on NQO1 gene. Cytotoxicity of cisplatin on ACHN cells was assessed in a dose- and time-dependent manner after siScramble or siNQO1 treatment. After cisplatin treatment, cells were subjected to cell viability assay, western-blot analysis, and immunofluorescence study. The cell viability was decreased in the siNQO1 cells (50%) than the siScramble cells (70%) after 24 h of cisplatin ($20{\mu}M$) treatment. Moreover, cytoprotection-related protein expressions were markedly suppressed in the siNQO1 cells after cisplatin treatment. The expression of Nrf2 and Klotho were decreased by 20% and 40%, respectively, of that in siScramble cells. Nrf2 and Klotho activation were also decreased in cisplatin treated siNQO1 cells, confirmed by cytoplasm-tonuclear translocation. Our findings demonstrate that the increased cisplatin-induced cytotoxicity was accompanied by suppressed Nrf2 activation and Klotho expression in siNQO1 cells.

Inhibitory Effect of NAD(P)H:Quinone Oxidoreductase 1 on the Activation of Macrophages (NQO1 (NAD(P)H:quinone oxidoreductase 1)에 의한 대식세포 활성화 억제)

  • Hong, Ji;Zhang, Peng;Yoon, I Na;Kim, Ho
    • Journal of Life Science
    • /
    • v.27 no.8
    • /
    • pp.873-878
    • /
    • 2017
  • We previously reported that NAD(P)H:quinone oxidoreductase 1 (NQO1)-knockout (KO) mice exhibited spontaneous inflammation in the gut. We also found that NQO1-KO mice showed highly increased inflammatory responses compared with NQO1-WT control mice when subjected to DSS-induced experimental colitis. In a Clostridium difficile toxin-induced mouse enteritis model, NQO1-KO mice were also sensitive compared with NQO1-WT mice. Moreover, numerous studies have shown that NQO1 is functionally associated with immune regulation. Here, we assessed whether NQO1 defects can alter macrophage activation. We found that peritoneal macrophages isolated from NQO1-KO mice produced more IL-6 and $TNF-{\alpha}$ than those isolated from NQO1-WT mice. Moreover, the dicumarol-induced inhibition of NQO1 significantly increased IL-6 and $TNF-{\alpha}$ production in peritoneal macrophages isolated from NQO1-WT mice, as well as in the cultured mouse macrophage cell line, RAW264.7. These results indicate that NQO1 may negatively regulate the activation of macrophages. Knockout or chemical inhibition of NQO1 markedly reduced the expression of $I{\kappa}B$ (inhibitor of $NF{\kappa}B$) in both mouse peritoneal macrophages and RAW264.7 cells. Finally, RAW264.7 cells treated with dicumarol exhibited morphological changes reflecting macrophage activation. Our results suggest that NQO1 may suppress the $NF{\kappa}B$ pathways in macrophages, thereby suppressing the activation of these cells. Thus, immunosuppressive activity may be among the many possible functions of NQO1.

Implications of NQO1 in cancer therapy

  • Oh, Eun-Taex;Park, Heon Joo
    • BMB Reports
    • /
    • v.48 no.11
    • /
    • pp.609-617
    • /
    • 2015
  • 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 β-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.

Modulation of NAD(P)H:Quinone Oxidoreductase (NQO1) Activity Mediated by 5-Arylamino-2-methyl -4,7-dioxobenzothiazoles and their Cytotoxic Potential

  • Ryu, Chung-Kyu;Jeong, Hyeh-Jean;Lee, Sang-Kook;Kang, Hye-Young;Ko, Kyung-Min;Sun, Yang-Jung;Song, Eun-Ha;Hur, Yeon-Hoe;Lee, Chong-Ock
    • Archives of Pharmacal Research
    • /
    • v.23 no.6
    • /
    • pp.554-558
    • /
    • 2000
  • Synthesized 5-arylamino-2-methyl-4,7-dioxobenzothiazoles 3a-3o were evaluated for modulation of NAD(P)H: quinone oxidoreductase (NQOl) activity with the cytosolic fractions derived from cultured human lung cancer cells and their cytotoxicity in cultured several human solid cancer cell lines. The 4,7-dioxobenzothiazoles affected the reduction potential by NQOl activity and showed a potent cytotoxic activity against human cancer cell lines. The tested compounds 3a, 3b, 3g, 3h, 3n and 3o were considered as more potent cytotoxic agents, and comparable modulators of NQOl activity.

  • PDF

Dicumarol Inhibits PMA-Induced MMP-9 Expression through NQO1-independent manner in Human Renal Carcinoma Caki Cells (인간 신장암 Caki세포에서 dicumarol에 의한 PMA 매개 matrix metalloproteinase-9의 발현 억제 효과)

  • Park, Eun Jung;Kwon, Taeg Kyu
    • Journal of Life Science
    • /
    • v.26 no.2
    • /
    • pp.174-180
    • /
    • 2016
  • Dicumarol is a coumarin derivative isolated from sweet clover (Melilotus alba), and has anti-coagulant activity with the inhibitory activity of NAD(P)H quinone oxidoreductase1 (NQO1). NQO1 catalyzes the two-electron reduction of quinones to hydroquinones. Dicumarol competes with NAD(P)H for binding to NQO1, resulting in the inhibition of NQO1 enzymatic activity. The expression of matrix metalloproteinases (MMPs) has been implicated in the invasion and metastasis of cancer cells. The expression of MMPs is regulated by cytokines and signal transduction pathways, including those activated by phorbol myristate acetate (PMA). However, the effects of dicumarol on metalloproteinase (MMP)-9 expression and activity are not investigated here. This study investigated whether dicumarol inhibits MMP-9 expression and activity in PMA-treated human renal carcinoma Caki cells. Dicumarol markedly inhibited the PMA-induced MMP-9 mRNA expression and MMP-9 activity. NF-κB and AP1 promoter activity, which is important in MMP-9 expression, also decreased in dicumarol-treated cells. Furthermore, dicumarol markedly suppressed the ability of PMA-mediated migration in Caki cells. When the relevance of NQO1 in the dicumarol-mediated inhibitory effect on PMA-induced MMP9 activity was elucidated, knock-down of NQO1 with siRNA was found to have no effect on PMA-induced MMP9 activity, suggesting that the stimulating effect of dicumarol on PMA-induced MMP9 activity is independent of NQO1 activity. Taken together, the present studies suggested that dicumarol may inhibit PMA-induced migration via down-regulation of MMP-9 expression and activity.

Enhancement of radiation effect using beta-lapachone and underlying mechanism

  • Ahn, Ki Jung;Lee, Hyung Sik;Bai, Se Kyung;Song, Chang Won
    • Radiation Oncology Journal
    • /
    • v.31 no.2
    • /
    • pp.57-65
    • /
    • 2013
  • Beta-lapachone (${\beta}$-Lap; 3,4-dihydro-2, 2-dimethyl-2H-naphthol[1, 2-b]pyran-5,6-dione) is a novel anti-cancer drug under phase I/II clinical trials. ${\beta}$-Lap has been demonstrated to cause apoptotic and necrotic death in a variety of human cancer cells in vitro and in vivo. The mechanisms underlying the ${\beta}$-Lap toxicity against cancer cells has been controversial. The most recent view is that ${\beta}$-Lap, which is a quinone compound, undergoes two-electron reduction to hydroquinone form utilizing NAD(P)H or NADH as electron source. This two-electron reduction of ${\beta}$-Lap is mediated by NAD(P)H:quinone oxidoreductase (NQO1), which is known to mediate the reduction of many quinone compounds. The hydroquinone forms of ${\beta}$-Lap then spontaneously oxidizes back to the original oxidized ${\beta}$-Lap, creating futile cycling between the oxidized and reduced forms of ${\beta}$-Lap. It is proposed that the futile recycling between oxidized and reduced forms of ${\beta}$-Lap leads to two distinct cell death pathways. First one is that the two-electron reduced ${\beta}$-Lap is converted first to one-electron reduced ${\beta}$-Lap, i.e., semiquinone ${\beta}$-Lap $(SQ)^{{\cdot}-}$ causing production of reactive oxygen species (ROS), which then causes apoptotic cell death. The second mechanism is that severe depletion of NAD(P)H and NADH as a result of futile cycling between the quinone and hydroquinone forms of ${\beta}$-Lap causes severe disturbance in cellular metabolism leading to apoptosis and necrosis. The relative importance of the aforementioned two mechanisms, i.e., generation of ROS or depletion of NAD(P)H/NADH, may vary depending on cell type and environment. Importantly, the NQO1 level in cancer cells has been found to be higher than that in normal cells indicating that ${\beta}$-Lap may be preferentially toxic to cancer cells relative to non-cancer cells. The cellular level of NQO1 has been found to be significantly increased by divergent physical and chemical stresses including ionizing radiation. Recent reports clearly demonstrated that ${\beta}$-Lap and ionizing radiation kill cancer cells in a synergistic manner. Indications are that irradiation of cancer cells causes long-lasting elevation of NQO1, thereby sensitizing the cells to ${\beta}$-Lap. In addition, ${\beta}$-Lap has been shown to inhibit the repair of sublethal radiation damage. Treating experimental tumors growing in the legs of mice with irradiation and intraperitoneal injection of ${\beta}$-Lap suppressed the growth of the tumors in a manner more than additive. Collectively, ${\beta}$-Lap is a potentially useful anti-cancer drug, particularly in combination with radiotherapy.

Inhibition of NAD(P)H:Quinone Oxidoreductase 1 by Dicumarol Reduces Tight Junction in Human Colonic Epithelial Cells (인간 대장상피세포 밀착연접 형성과정에서 NQO1 저해 효과)

  • Hong, Ji;Zhang, Peng;Yoon, I Na;Kim, Ho
    • Journal of Life Science
    • /
    • v.26 no.5
    • /
    • pp.531-536
    • /
    • 2016
  • We previously showed that NAD(P)H:quinone oxidoreductase 1 (NQO1) knockout (KO) mice exhibited spontaneous inflammation with markedly increased mucosal permeability in the gut, and that NQO1 is functionally associated with regulating tight junctions in the mucosal epithelial cells that govern the mucosal barrier. Here, we confirm the role of NQO1 in the formation of tight junctions by human colonic epithelial cells (HT29). We treated HT29 cells with a chemical inhibitor of NQO1 (dicumarol; 10 μM), and examined the effect on the transepithelial resistance of epithelial cells and the protein expression levels of ZO1 and occludin (two known regulators of tight junctions between gut epithelial cells). The dicumarol-induced inhibition of NQO1 markedly reduced transepithelial resistance (a measure of tight junctions) and decreased the levels of the tested tight junction proteins. In vivo, luminal injection of dicumarol significantly increased mucosal permeability and decreased ZO1 and occludin protein expression levels in mouse guts. However, in contrast to the previous report that the epithelial cells of NQO1 KO mice showed marked down-regulations of the transcripts encoding ZO1 and occludin, these transcript levels were not affected in dicumarol-treated HT29 cells. This result suggests that the NQO1-depedent regulation of tight junction molecules may involve multiple processes, including both transcriptional regulation and protein degradation processes such as those governed by the ubiquitination/proteasomal, and/or lysosomal systems.

Induction of Anticarcinogenic Enzymes by Dichloromethane-soluble Fraction of Physalis alkekengi var. francheti Hort. in Mouse Hepatoma Cells

  • Seo, JiYeon;Kim, Hyo Jung;Kim, Jong-Sang
    • Current Research on Agriculture and Life Sciences
    • /
    • v.32 no.3
    • /
    • pp.119-124
    • /
    • 2014
  • Physalis alkekengi var. francheti Hort. is known as an insecticide and traditional remedy for liver related diseases. Therefore, this study investigated the chemopreventive effects of extracts and several solvent fractions (n-hexane, dichloromethane, n-butanol, water) of Physalis alkekengi var. francheti Hort. First, their cytotoxicity and NQO1 activity were measured using an MTT assay, plus a quinone reductase [NAD(P)H dehydrogenase (quinone); NAD(P)H: (quinone acceptor) oxidoreductase, EC 1.6.99.2]-inducing activity assay was performed using cultured murine hepatoma cells (Hepa1c1c7) and its mutant cells(BpRc1). The reduction of electrophilic quinones by NQO1 is an important detoxification pathway and major mechanism of chemoprevention. When compared with the other solvent soluble fractions with different polarities, the dichloromethane fraction of Physalis alkekengi var. francheti Hort. showed a higher NQO1-inducing activity that was also dose-dependent. Moreover, the dichloromethane fraction of Physalis alkekengi var. francheti Hort. induced ARE-luciferase activities in HepG2-C8 cells that were generated by transfecting the ARE-luciferase gene construct, suggesting the Nrf2-ARE-mediated induction of anti-oxidative enzymes. In conclusion, the dichloromethane-soluble fraction of Physalis alkekengi var. francheti Hort. showed a relatively strong induction of detoxifying enzymes, thereby meriting further study to identify the active components and evaluate their potential as cancer preventive agents.