151
|
Shay KP, Smith EJ, Hagen TM. Transcription factor Nrf2: examination of nuclear protein levels by immunoblotting and promoter response element binding by chromatin immunoprecipitation (ChIP). ACTA ACUST UNITED AC 2013; Chapter 17:Unit17.13. [PMID: 23045018 DOI: 10.1002/0471140856.tx1713s45] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nuclear factor erythroid 2 (NF-E2) related factor 2 (Nrf2) is a transcription factor that governs the expression of over a hundred so-called phase II detoxification and antioxidant genes that are regulated through the antioxidant response element (ARE). Loss of Nrf2 activity has been implicated in cardiovascular disease, inflammation, aging, and cancer. Nrf2 is induced to accumulate in the nucleus when the cell encounters an oxidative stress, a fact that has been exploited experimentally to test the conditions under which ARE-containing genes are expressed. The nuclear levels of Nrf2 give an indication of whether an experimental treatment results in Nrf2 localization and induction. mRNA levels of phase II genes may be measured as a follow-up, but in order to show a direct link between nuclear Nrf2 accumulation and increases in gene expression, it is useful to show that Nrf2 binds to AREs in the promoters of target genes. The simplest way to do this is to employ a chromatin immunoprecipitation (ChIP) assay along with an examination of cellular Nrf2 levels by immunoblotting.
Collapse
Affiliation(s)
- Kate Petersen Shay
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | | | | |
Collapse
|
152
|
Bryan HK, Olayanju A, Goldring CE, Park BK. The Nrf2 cell defence pathway: Keap1-dependent and -independent mechanisms of regulation. Biochem Pharmacol 2012; 85:705-17. [PMID: 23219527 DOI: 10.1016/j.bcp.2012.11.016] [Citation(s) in RCA: 779] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 11/24/2012] [Accepted: 11/27/2012] [Indexed: 12/13/2022]
Abstract
The transcription factor Nrf2 (NF-E2-related factor 2) plays a vital role in maintaining cellular homeostasis, especially upon the exposure of cells to chemical or oxidative stress, through its ability to regulate the basal and inducible expression of a multitude of antioxidant proteins, detoxification enzymes and xenobiotic transporters. In addition, Nrf2 contributes to diverse cellular functions including differentiation, proliferation, inflammation and lipid synthesis and there is an increasing association of aberrant expression and/or function of Nrf2 with pathologies including cancer, neurodegeneration and cardiovascular disease. The activity of Nrf2 is primarily regulated via its interaction with Keap1 (Kelch-like ECH-associated protein 1), which directs the transcription factor for proteasomal degradation. Although it is generally accepted that modification (e.g. chemical adduction, oxidation, nitrosylation or glutathionylation) of one or more critical cysteine residues in Keap1 represents a likely chemico-biological trigger for the activation of Nrf2, unequivocal evidence for such a phenomenon remains elusive. An increasing body of literature has revealed alternative mechanisms of Nrf2 regulation, including phosphorylation of Nrf2 by various protein kinases (PKC, PI3K/Akt, GSK-3β, JNK), interaction with other protein partners (p21, caveolin-1) and epigenetic factors (micro-RNAs -144, -28 and -200a, and promoter methylation). These and other processes are potentially important determinants of Nrf2 activity, and therefore may contribute to the maintenance of cellular homeostasis. Here, we dissect evidence supporting these Keap1-dependent and -independent mechanisms of Nrf2 regulation. Furthermore, we highlight key knowledge gaps in this important field of biology, and suggest how these may be addressed experimentally.
Collapse
Affiliation(s)
- Holly K Bryan
- MRC Centre for Drug Safety Science, Molecular & Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, L69 3GE, UK
| | | | | | | |
Collapse
|
153
|
Ampofo E, Sokolowsky T, Götz C, Montenarh M. Functional interaction of protein kinase CK2 and activating transcription factor 4 (ATF4), a key player in the cellular stress response. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:439-51. [PMID: 23123191 DOI: 10.1016/j.bbamcr.2012.10.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/05/2012] [Accepted: 10/23/2012] [Indexed: 01/16/2023]
Abstract
Protein kinase CK2 is a pleiotropic enzyme, which is implicated in the regulation of numerous biological processes. It seems to regulate the various functions by binding to other proteins and by phosphorylation of many different substrates. Here, we identified the activating transcription factor 4 (ATF4), an essential component of the ER stress signaling, as a new binding partner and a new substrate of CK2 in vitro and in vivo. Bifluorescence complementation analysis (BiFC) revealed that CK2α and ATF4 associate in the nucleus. By using mutants of ATF4 we identified serine 215 as the main CK2 phosphorylation site. The ATF4 S215A mutant turned out to be more stable than the wild-type form. We further noticed that an inhibition of CK2 caused an increased transcription of the ATF4 gene. Analyses of the transcription factor activity revealed an impaired activity of the CK2 phosphorylation mutant of ATF4. Thus, we show that (i) ATF4 is a binding partner of CK2α (ii) ATF4 is a substrate of CK2, (iii) the phosphorylation of ATF4 by CK2 influences the stability of ATF4, (iv) the transcription of ATF4 is regulated by CK2 and (v) the transcription factor activity of ATF4 is regulated by the CK2 phosphorylation of ATF4. Thus, CK2 plays an essential role in the regulation of the ER-stress induced signaling pathway.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Medizinische Biochemie und Molekularbiologie und Kompetenzzentrum, Molekulare Medizin, Universität des Saarlandes, Gebäude 44, 66424 Homburg, Germany
| | | | | | | |
Collapse
|
154
|
Saidu NEB, Touma R, Asali IA, Jacob C, Montenarh M. Diallyl tetrasulfane activates both the eIF2α and Nrf2/HO-1 pathways. Biochim Biophys Acta Gen Subj 2012; 1830:2214-25. [PMID: 23046979 DOI: 10.1016/j.bbagen.2012.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/09/2012] [Accepted: 10/03/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Diallyl polysulfanes have been shown to exert cell cycle arrest, anti-tumor and anti-inflammatory activities in a variety of in vitro and in vivo models. Although diallyl polysulfanes cause oxidative stress, little is known about the underlying signaling cascades leading to antioxidant defense or apoptosis. METHODS Cells were treated with DATTS at different concentrations and for different time periods. Reactive oxygen species and thiol concentrations were determined by commercially available kits. The expression levels of signal molecules were determined by Western Blot analysis. A direct influence of Nrf2 on the promoter of HO-1 gene was determined by a luciferase assay with the StRE promoter element from the HO-1 gene. RESULTS We found an immediate increase in the level of the superoxide anion radical O(2)(-) and hydrogen peroxide H(2)O(2) and an overall thiol depletion. DATTS treatment of HCT116 cells also caused an up-regulation of phospho-eIF2α, nuclear Nrf2 and HO-1 protein levels in a time and concentration-dependent manner. Pre-treatment of cells with antioxidants significantly reduced the elevated expression levels of these proteins. A direct contribution of Nrf2 was shown by its interaction with the stress-response element of the HO-1 promoter. CONCLUSIONS DATTS activates the ROS-eIF2α/Nrf2 HO-1 signaling cascades leading to the up-regulation of HO-1. However, this antioxidant defense is not sufficient to protect HCT116 cells from apoptosis. GENERAL SIGNIFICANCE This study shows for the first time a parallel but not equal activation of signaling pathways by DATTS with a competitive ultimate cellular outcome.
Collapse
|
155
|
Zhang M, An C, Gao Y, Leak RK, Chen J, Zhang F. Emerging roles of Nrf2 and phase II antioxidant enzymes in neuroprotection. Prog Neurobiol 2012; 100:30-47. [PMID: 23025925 DOI: 10.1016/j.pneurobio.2012.09.003] [Citation(s) in RCA: 452] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 09/04/2012] [Accepted: 09/20/2012] [Indexed: 12/13/2022]
Abstract
Phase II metabolic enzymes are a battery of critical proteins that detoxify xenobiotics by increasing their hydrophilicity and enhancing their disposal. These enzymes have long been studied for their preventative and protective effects against mutagens and carcinogens and for their regulation via the Keap1 (Kelch-like ECH associated protein 1)/Nrf2 (Nuclear factor erythroid 2 related factor 2)/ARE (antioxidant response elements) pathway. Recently, a series of studies have reported the altered expression of phase II genes in postmortem tissue of patients with various neurological diseases. These observations hint at a role for phase II enzymes in the evolution of such conditions. Furthermore, promising findings reveal that overexpression of phase II genes, either by genetic or chemical approaches, confers neuroprotection in vitro and in vivo. Therefore, there is a need to summarize the current literature on phase II genes in the central nervous system (CNS). This should help guide future studies on phase II genes as therapeutic targets in neurological diseases. In this review, we first briefly introduce the concept of phase I, II and III enzymes, with a special focus on phase II enzymes. We then discuss their expression regulation, their inducers and executors. Following this background, we expand our discussion to the neuroprotective effects of phase II enzymes and the potential application of Nrf2 inducers to the treatment of neurological diseases.
Collapse
Affiliation(s)
- Meijuan Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
156
|
Ma Q, He X. Molecular basis of electrophilic and oxidative defense: promises and perils of Nrf2. Pharmacol Rev 2012; 64:1055-81. [PMID: 22966037 DOI: 10.1124/pr.110.004333] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Induction of drug-metabolizing enzymes through the antioxidant response element (ARE)-dependent transcription was initially implicated in chemoprevention against cancer by antioxidants. Recent progress in understanding the biology and mechanism of induction revealed a critical role of induction in cellular defense against electrophilic and oxidative stress. Induction is mediated through a novel signaling pathway via two regulatory proteins, the nuclear factor erythroid 2-related factor 2 (Nrf2) and the Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1). Nrf2 binds to Keap1 at a two site-binding interface and is ubiquitinated by the Keap1/cullin 3/ring box protein-1-ubiquitin ligase, resulting in a rapid turnover of Nrf2 protein. Electrophiles and oxidants modify critical cysteine thiols of Keap1 and Nrf2 to inhibit Nrf2 ubiquitination, leading to Nrf2 activation and induction. Induction increases stress resistance critical for cell survival, because knockout of Nrf2 in mice increased susceptibility to a variety of toxicity and disease processes. Collateral to diverse functions of Nrf2, genome-wide search has led to the identification of a plethora of ARE-dependent genes regulated by Nrf2 in an inducer-, tissue-, and disease-dependent manner to control drug metabolism, antioxidant defense, stress response, proteasomal degradation, and cell proliferation. The protective nature of Nrf2 could also be hijacked in a number of pathological conditions by means of somatic mutation, epigenetic alteration, and accumulation of disruptor proteins, promoting drug resistance in cancer and pathologic liver features in autophagy deficiency. The repertoire of ARE inducers has expanded enormously; the therapeutic potential of the inducers has been examined beyond cancer prevention. Developing potent and specific ARE inducers and Nrf2 inhibitors holds certain new promise for the prevention and therapy against cancer, chronic disease, and toxicity.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute forOccupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia.
| | | |
Collapse
|
157
|
Kelsen SG. Respiratory epithelial cell responses to cigarette smoke: the unfolded protein response. Pulm Pharmacol Ther 2012; 25:447-52. [PMID: 22846757 DOI: 10.1016/j.pupt.2012.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 06/28/2012] [Accepted: 07/20/2012] [Indexed: 02/06/2023]
Abstract
Cigarette smoking exposes the respiratory epithelium to highly toxic, reactive oxygen nitrogen species which damage lung proteins in the endoplasmic reticulum (ER), the cell organelle in which all secreted and membrane proteins are processed. Accumulation of damaged or misfolded proteins in the ER, a condition termed ER stress, activates a complex cellular process termed the unfolded protein responses (UPR). The UPR acts to restore cellular protein homeostasis by regulating all aspects of protein metabolism including: protein translation and syntheses; protein folding; and protein degradation. However, activation of the UPR may also induce signaling pathways which induce inflammation and cell apoptosis. This review discusses the role of UPR in the respiratory epithelial cell response to cigarette smoke and the pathogenesis of lung diseases like COPD.
Collapse
Affiliation(s)
- Steven G Kelsen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Temple University School of Medicine, USA.
| |
Collapse
|
158
|
Cao S, Wang Y, Peng X. ROS-inducible DNA cross-linking agent as a new anticancer prodrug building block. Chemistry 2012; 18:3850-4. [PMID: 22378607 DOI: 10.1002/chem.201200075] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Sheng Cao
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 N. Cramer St., Milwaukee, WI 53211, USA
| | | | | |
Collapse
|
159
|
Rojo AI, Medina-Campos ON, Rada P, Zúñiga-Toalá A, López-Gazcón A, Espada S, Pedraza-Chaverri J, Cuadrado A. Signaling pathways activated by the phytochemical nordihydroguaiaretic acid contribute to a Keap1-independent regulation of Nrf2 stability: Role of glycogen synthase kinase-3. Free Radic Biol Med 2012; 52:473-87. [PMID: 22142471 DOI: 10.1016/j.freeradbiomed.2011.11.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 01/29/2023]
Abstract
Defense against oxidative stress is executed by an antioxidant program that is tightly controlled by the transcription factor Nrf2. The stability of Nrf2 involves the interaction of two degradation domains, designated Neh2 and Neh6, with the E3 ubiquitin ligase adaptors, Keap1 and β-TrCP, respectively. The regulation of Nrf2 through the Neh6 degron remains largely unexplored but requires GSK-3 to form a phosphodegron. In this study, the cancer-chemopreventive agent nordihydroguaiaretic acid (NDGA) increased the level of Nrf2 protein and expression of heme oxygenase-1 (HO-1) in kidney-derived LLC-PK1 and HEK293T cells and in wild-type mouse embryo fibroblasts (MEFs). However, NDGA did not induce HO-1 in Nrf2(-/-) MEFs, indicating that Nrf2 is required for induction. The relevance of the Nrf2/HO-1 axis to antioxidant protection was further demonstrated by the finding that the HO-1 inhibitor stannous-mesoporphyrin abolished protection against hydrogen peroxide conferred by NDGA. NDGA increased Nrf2 and HO-1 protein levels in Keap1(-/-) MEFs, implying that Keap1-independent mechanisms regulate Nrf2 stability. Mutants of the Neh2 or Nrh6 domain and chimeric proteins comprising cyan fluorescent protein fused to Neh2 and green fluorescent protein fused to Neh6 exhibited longer half-lives in the presence of NDGA, demonstrating that NDGA targets both the Neh2 and the Neh6 degrons. In common with other chemopreventive agents, NDGA activated the ERK1/2, p38, JNK, and PI3K pathways. By using selective kinase inhibitors we found that PI3K, JNK, and p38 were responsible for the stabilization of Nrf2 and induction of HO-1 by NDGA. To explain how NDGA might up-regulate Nrf2 in a Keap1-independent manner we explored the participation of GSK-3β because it controls the Neh6 phosphodegron. Importantly, NDGA caused inhibitory phosphorylation of GSK-3β at Ser9 and at Thr390, and this was associated with a substantial reduction in Neh6 phosphorylation. Our study demonstrates that NDGA activates Nrf2 through multiple signaling cascades and identifies GSK-3β as an integrator of these signaling pathways and a gatekeeper of Nrf2 stability at the level of the Neh6 phosphodegron.
Collapse
Affiliation(s)
- Ana I Rojo
- Departamento de Bioquímica e Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, Centro de Investigación en Red Sobre Enfermedades Neurodegenerativas, Instituto de Investigación Sanitaria la Paz, Madrid 28029, Spain
| | | | | | | | | | | | | | | |
Collapse
|
160
|
Pettazzoni P, Ciamporcero E, Medana C, Pizzimenti S, Dal Bello F, Minero VG, Toaldo C, Minelli R, Uchida K, Dianzani MU, Pili R, Barrera G. Nuclear factor erythroid 2-related factor-2 activity controls 4-hydroxynonenal metabolism and activity in prostate cancer cells. Free Radic Biol Med 2011; 51:1610-8. [PMID: 21816220 DOI: 10.1016/j.freeradbiomed.2011.07.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 06/30/2011] [Accepted: 07/07/2011] [Indexed: 01/17/2023]
Abstract
4-Hydroxynonenal (HNE) is an end product of lipoperoxidation with antiproliferative and proapoptotic properties in various tumors. Here we report a greater sensitivity to HNE in PC3 and LNCaP cells compared to DU145 cells. In contrast to PC3 and LNCaP cells, HNE-treated DU145 cells showed a smaller reduction in growth and did not undergo apoptosis. In DU145 cells, HNE did not induce ROS production and DNA damage and generated a lower amount of HNE-protein adducts. DU145 cells had a greater GSH and GST A4 content and GSH/GST-mediated HNE detoxification. Nuclear factor erythroid 2-related factor-2 (Nrf2) is a regulator of the antioxidant response. Nrf2 protein content and nuclear accumulation were higher in DU145 cells compared to PC3 and LNCaP cells, whereas the expression of KEAP1, the main negative regulator of Nrf2 activity, was lower. Inhibition of Nrf2 expression with specific siRNA resulted in a reduction in GST A4 expression and GS-HNE formation, indicating that Nrf2 controls HNE metabolism. In addition, Nrf2 knockdown sensitized DU145 cells to HNE-mediated antiproliferative and proapoptotic activity. In conclusion, we demonstrated that increased Nrf2 activity resulted in a reduction in HNE sensitivity in prostate cancer cells, suggesting a potential mechanism of resistance to pro-oxidant therapy.
Collapse
Affiliation(s)
- Piergiorgio Pettazzoni
- Section of General Pathology, Department of Medicine and Experimental Oncology, University of Turin, 10125 Turin, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Brigelius-Flohé R, Flohé L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid Redox Signal 2011; 15:2335-81. [PMID: 21194351 PMCID: PMC3166203 DOI: 10.1089/ars.2010.3534] [Citation(s) in RCA: 422] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Convincing concepts of redox control of gene transcription have been worked out for prokaryotes and lower eukaryotes, whereas the knowledge on complex mammalian systems still resembles a patchwork of poorly connected findings. The article, therefore, reviews principles of redox regulation with special emphasis on chemical feasibility, kinetic requirements, specificity, and physiological context, taking well investigated mammalian transcription factor systems, nuclear transcription factor of bone marrow-derived lymphocytes (NF-κB), and kelch-like ECH-associated protein-1 (Keap1)/Nrf2, as paradigms. Major conclusions are that (i) direct signaling by free radicals is restricted to O(2)•- and •NO and can be excluded for fast reacting radicals such as •OH, •OR, or Cl•; (ii) oxidant signals are H(2)O(2), enzymatically generated lipid hydroperoxides, and peroxynitrite; (iii) free radical damage is sensed via generation of Michael acceptors; (iv) protein thiol oxidation/alkylation is the prominent mechanism to modulate function; (v) redox sensors must be thiol peroxidases by themselves or proteins with similarly reactive cysteine or selenocysteine (Sec) residues to kinetically compete with glutathione peroxidase (GPx)- and peroxiredoxin (Prx)-type peroxidases or glutathione-S-transferases, respectively, a postulate that still has to be verified for putative mammalian sensors. S-transferases and Prxs are considered for system complementation. The impact of NF-κB and Nrf2 on hormesis, management of inflammatory diseases, and cancer prevention is critically discussed.
Collapse
Affiliation(s)
- Regina Brigelius-Flohé
- Department Biochemistry of Micronutrients, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, Nuthetal, Germany.
| | | |
Collapse
|
162
|
Ivanov AV, Smirnova OA, Ivanova ON, Masalova OV, Kochetkov SN, Isaguliants MG. Hepatitis C virus proteins activate NRF2/ARE pathway by distinct ROS-dependent and independent mechanisms in HUH7 cells. PLoS One 2011; 6:e24957. [PMID: 21931870 PMCID: PMC3172309 DOI: 10.1371/journal.pone.0024957] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 08/25/2011] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) is a highly pathogenic human virus associated with liver fibrosis, steatosis, and cancer. In infected cells HCV induces oxidative stress. Here, we show that HCV proteins core, E1, E2, NS4B, and NS5A activate antioxidant defense Nrf2/ARE pathway via several independent mechanisms. This was demonstrated by the analysis of transient co-expression in Huh7 cells of HCV proteins and luciferase reporters. Expression, controlled by the promoters of stress-response genes or their minimal Nrf2-responsive elements, was studied using luminescence assay, RT-qPCR and/or Western-blot analysis. All five proteins induced Nrf2 activation by protein kinase C in response to accumulation of reactive oxygen species (ROS). In addition, expression of core, E1, E2, NS4B, and NS5A proteins resulted in the activation of Nrf2 in a ROS-independent manner. The effect of core and NS5A was mediated through casein kinase 2 and phosphoinositide-3 kinase, whereas those of NS4B, E1, and E2, were not mediated by either PKC, CK2, PI3K, p38, or ERK. Altogether, on the earliest stage of expression HCV proteins induced a strong up-regulation of the antioxidant defense system. These events may underlie the harmful effects of HCV-induced oxidative stress during acute stage of hepatitis C.
Collapse
Affiliation(s)
- Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | | | | | | | | | | |
Collapse
|
163
|
Ren K, Xiang S, He F, Zhang W, Ding X, Wu Y, Yang L, Zhou J, Gao X, Zhang J. CK2 phosphorylates AP-2α and increases its transcriptional activity. BMB Rep 2011; 44:490-5. [DOI: 10.5483/bmbrep.2011.44.7.490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
164
|
Cheung KL, Yu S, Pan Z, Ma J, Wu TY, Kong ANT. tBHQ-induced HO-1 expression is mediated by calcium through regulation of Nrf2 binding to enhancer and polymerase II to promoter region of HO-1. Chem Res Toxicol 2011; 24:670-6. [PMID: 21443188 DOI: 10.1021/tx1004369] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Induction of Nrf2-mediated detoxifying/antioxidant enzymes is an effective strategy for cancer chemoprevention. The goal of this study was to examine the role of calcium [Ca(2+)] in regulating a well-known phenolic chemopreventive compound tertiary-butylhydroquinone (tBHQ) activation of Nrf2 and induction of Nrf2 downstream target gene heme-oxygenase (HO-1). tBHQ alone caused Nrf2 nuclear localization and induced HO-1 mRNA and protein expression in a dose-dependent manner. Using RT-PCR and Western blotting, we showed that tBHQ-induced transcription of HO-1 is Ca(2+)-dependent. Chelation of [Ca(2+)](ext) or [Ca(2+)](intra) by EGTA or BAPTA attenuated tBHQ-induced HO-1. Cotreatment of tBHQ with inhibitors of [Ca(2+)]-sensitive protein kinase C and camodulin kinase did not attenuate HO-1 induction. Nuclear translocation of Nrf2 induced by tBHQ was also not affected by treatment of EGTA or BAPTA. Additionally, EGTA and BAPTA treatments decreased basal nuclear phosphorylation of CREB and decreased tBHQ-induced Nrf2-CBP binding and Nrf2 binding to enhancer as well as polymerase II binding to the promoter of HO-1 gene. Furthermore, tBHQ in combination with higher [Ca(2+)](ext) augmented HO-1 induction both in vitro and in vivo, indicating that the modulation of [Ca(2+)](int) could be used as an adjuvant to increase the efficacy of chemopreventive agents. Taken together, our results indicated that in addition to tBHQ-induced oxidative stress-mediated Nrf2 translocation, HO-1 induction by tBHQ also appears to be dependent on a series of Ca(2+)-regulated mechanisms.
Collapse
Affiliation(s)
- Ka Lung Cheung
- Graduate Program in Pharmaceutical Science, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | | | | | | | | | | |
Collapse
|
165
|
Jian Z, Li K, Liu L, Zhang Y, Zhou Z, Li C, Gao T. Heme oxygenase-1 protects human melanocytes from H2O2-induced oxidative stress via the Nrf2-ARE pathway. J Invest Dermatol 2011; 131:1420-7. [PMID: 21412259 DOI: 10.1038/jid.2011.56] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Oxidative stress caused by hydrogen peroxide (H(2)O(2)) leads to cell death and has been implicated in the pathogenesis of vitiligo. The nuclear factor E2-related factor 2 (Nrf2)-antioxidant response element (ARE), a major antioxidant pathway, regulates oxidative stress-related cytoprotective genes. We hypothesized that the Nrf2-ARE pathway protects human melanocytes from H(2)O(2)-induced oxidative damage through the induction of downstream antioxidative genes. Thus, we used Nrf2 short interfering RNA (siRNA) and pCMV6-XL5-Nrf2 to downregulate or upregulate Nrf2 expression in immortalized human melanocyte cell line PIG1. The melanocytes were then analyzed under different oxidative stress conditions for cell viability and apoptosis. Our study demonstrated that heme oxygenase-1 (HO-1) was the most induced antioxidant gene in PIG1 cells after treatment with H(2)O(2). Knockdown of Nrf2 or zinc protoporphyrin IX (ZnPP) treatment increased cell death caused by H(2)O(2) in melanocytes, but upregulation of Nrf2 or hemin treatment reduced cell death caused by H(2)O(2) in melanocytes. In addition, the H(2)O(2)-induced Nrf2-ARE/HO-1 pathway was confirmed in primary cultured human melanocytes by examining the expression and translocation of Nrf2 and HO-1. These data suggested that regulation of the Nrf2/HO-1 pathway can reduce H(2)O(2)-induced oxidative damage in human melanocytes. Our data demonstrate that HO-1 protects human melanocytes from oxidative damage via the Nrf2-ARE pathway.
Collapse
Affiliation(s)
- Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | |
Collapse
|
166
|
Cheng X, Siow RCM, Mann GE. Impaired redox signaling and antioxidant gene expression in endothelial cells in diabetes: a role for mitochondria and the nuclear factor-E2-related factor 2-Kelch-like ECH-associated protein 1 defense pathway. Antioxid Redox Signal 2011; 14:469-87. [PMID: 20524845 DOI: 10.1089/ars.2010.3283] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Type 2 diabetes is an age-related disease associated with vascular pathologies, including severe blindness, renal failure, atherosclerosis, and stroke. Reactive oxygen species (ROS), especially mitochondrial ROS, play a key role in regulating the cellular redox status, and an overproduction of ROS may in part underlie the pathogenesis of diabetes and other age-related diseases. Cells have evolved endogenous defense mechanisms against sustained oxidative stress such as the redox-sensitive transcription factor nuclear factor E2-related factor 2 (Nrf2), which regulates antioxidant response element (ARE/electrophile response element)-mediated expression of detoxifying and antioxidant enzymes and the cystine/glutamate transporter involved in glutathione biosynthesis. We hypothesize that diminished Nrf2/ARE activity contributes to increased oxidative stress and mitochondrial dysfunction in the vasculature leading to endothelial dysfunction, insulin resistance, and abnormal angiogenesis observed in diabetes. Sustained hyperglycemia further exacerbates redox dysregulation, thereby providing a positive feedback loop for severe diabetic complications. This review focuses on the role that Nrf2/ARE-linked gene expression plays in regulating endothelial redox homeostasis in health and type 2 diabetes, highlighting recent evidence that Nrf2 may provide a therapeutic target for countering oxidative stress associated with vascular disease and aging.
Collapse
Affiliation(s)
- Xinghua Cheng
- Cardiovascular Division, School of Medicine, King's College London, London, United Kingdom
| | | | | |
Collapse
|
167
|
SCF/{beta}-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol Cell Biol 2011; 31:1121-33. [PMID: 21245377 DOI: 10.1128/mcb.01204-10] [Citation(s) in RCA: 612] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Regulation of transcription factor Nrf2 (NF-E2-related factor 2) involves redox-sensitive proteasomal degradation via the E3 ubiquitin ligase Keap1/Cul3. However, Nrf2 is controlled by other mechanisms that have not yet been elucidated. We now show that glycogen synthase kinase 3 (GSK-3) phosphorylates a group of Ser residues in the Neh6 domain of mouse Nrf2 that overlap with an SCF/β-TrCP destruction motif (DSGIS, residues 334 to 338) and promotes its degradation in a Keap1-independent manner. Nrf2 was stabilized by GSK-3 inhibitors in Keap1-null mouse embryo fibroblasts. Similarly, an Nrf2(ΔETGE) mutant, which cannot be degraded via Keap1, accumulated when GSK-3 activity was blocked. Phosphorylation of a Ser cluster in the Neh6 domain of Nrf2 stimulated its degradation because a mutant Nrf2(ΔETGE 6S/6A) protein, lacking these Ser residues, exhibited a longer half-life than Nrf2(ΔETGE). Moreover, Nrf2(ΔETGE 6S/6A) was insensitive to β-TrCP regulation and exhibited lower levels of ubiquitination than Nrf2(ΔETGE). GSK-3β enhanced ubiquitination of Nrf2(ΔETGE) but not that of Nrf2(ΔETGE 6S/6A). The Nrf2(ΔETGE) protein but not Nrf2(ΔETGE 6S/6A) coimmunoprecipitated with β-TrCP, and this association was enhanced by GSK-3β. Our results show for the first time that Nrf2 is targeted by GSK-3 for SCF/β-TrCP-dependent degradation. We propose a "dual degradation" model to describe the regulation of Nrf2 under different pathophysiological conditions.
Collapse
|
168
|
Kawai Y, Garduño L, Theodore M, Yang J, Arinze IJ. Acetylation-deacetylation of the transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) regulates its transcriptional activity and nucleocytoplasmic localization. J Biol Chem 2010; 286:7629-40. [PMID: 21196497 DOI: 10.1074/jbc.m110.208173] [Citation(s) in RCA: 241] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Activation of Nrf2 by covalent modifications that release it from its inhibitor protein Keap1 has been extensively documented. In contrast, covalent modifications that may regulate its action after its release from Keap1 have received little attention. Here we show that CREB-binding protein induced acetylation of Nrf2, increased binding of Nrf2 to its cognate response element in a target gene promoter, and increased Nrf2-dependent transcription from target gene promoters. Heterologous sirtuin 1 (SIRT1) decreased acetylation of Nrf2 as well as Nrf2-dependent gene transcription, and its effects were overridden by dominant negative SIRT1 (SIRT1-H355A). The SIRT1-selective inhibitors EX-527 and nicotinamide stimulated Nrf2-dependent gene transcription, whereas resveratrol, a putative activator of SIRT1, was inhibitory, mimicking the effect of SIRT1. Mutating lysine to alanine or to arginine at Lys(588) and Lys(591) of Nrf2 resulted in decreased Nrf2-dependent gene transcription and abrogated the transcription-activating effect of CREB-binding protein. Furthermore, SIRT1 had no effect on transcription induced by these mutants, indicating that these sites are acetylation sites. Microscope imaging of GFP-Nrf2 in HepG2 cells as well as immunoblotting for Nrf2 showed that acetylation conditions resulted in increased nuclear localization of Nrf2, whereas deacetylation conditions enhanced its cytoplasmic rather than its nuclear localization. We posit that Nrf2 in the nucleus undergoes acetylation, resulting in binding, with basic-region leucine zipper protein(s), to the antioxidant response element and consequently in gene transcription, whereas deacetylation disengages it from the antioxidant response element, thereby resulting in transcriptional termination and subsequently in its nuclear export.
Collapse
Affiliation(s)
- Yumiko Kawai
- Department of Physiology, Meharry Medical College, Nashville, Tennessee 37208-3599, USA
| | | | | | | | | |
Collapse
|
169
|
Giudice A, Arra C, Turco MC. Review of molecular mechanisms involved in the activation of the Nrf2-ARE signaling pathway by chemopreventive agents. Methods Mol Biol 2010; 647:37-74. [PMID: 20694660 DOI: 10.1007/978-1-60761-738-9_3] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human exposures to environmental toxicants have been associated with etiology of many diseases including inflammation, cancer, and cardiovascular and neurodegenerative disorders. To counteract the detrimental effect of environmental insults, mammalian cells have evolved a hierarchy of sophisticated sensing and signaling mechanisms to turn on or off endogenous antioxidant responses accordingly. One of the major cellular antioxidant responses is the induction of antioxidative and carcinogen-detoxification enzymes through the cytoplasmic oxidative stress system (Nrf2-Keap1) activated by a variety of natural and synthetic chemopreventive agents. Under normal conditions, Keap1 anchors the Nrf2 transcription factor within the cytoplasm targeting it for ubiquitination and proteasomal degradation to maintain low levels of Nrf2 that mediate the constitutive expression of Nrf2 downstream genes. When cells are exposed to chemopreventive agents and oxidative stress, a signal involving phosphorylation and/or redox modification of critical cysteine residues in Keap1 inhibits the enzymatic activity of the Keap1-Cul3-Rbx1 E3 ubiquitin ligase complex, resulting in decreased Nrf2 ubiquitination and degradation. As a consequence, free Nrf2 translocates into the nucleus and in combination with other transcription factors (e.g., sMaf, ATF4, JunD, PMF-1) transactivates the antioxidant response elements (AREs)/electrophile response elements (EpREs) of many cytoprotective genes, as well as Nrf2 itself. Upon recovery of cellular redox homeostasis, Keap1 travels into the nucleus to dissociate Nrf2 from the ARE. Subsequently, the Nrf2-Keap1 complex is exported out of the nucleus by the nuclear export sequence (NES) in Keap1. Once in the cytoplasm, the Nrf2-Keap1 complex associates with the Cul3-Rbx1 core ubiquitin machinery, resulting in degradation of Nrf2 and termination of the Nrf2/ARE signaling pathway. The discovery of multiple nuclear localization signals (NLSs) and nuclear export signals (NESs) in Nrf2 also suggests that the nucleocytoplasm translocation of transcription factors is the consequence of a dynamic equilibrium of multivalent NLSs and NESs. On the other hand, Keap1 may provide an additional regulation of the quantity of Nrf2 both in basal and inducible conditions. This chapter summarizes the current body of knowledge regarding the molecular mechanisms through which ARE inducers (chemopreventive agents) regulate the coordinated transcriptional induction of genes encoding phase II and antioxidant enzymes as well as other defensive proteins, via the nuclear factor-erythroid 2 (NF-E2-p45)-related factor 2(Nrf2)/(ARE) signaling pathway.
Collapse
Affiliation(s)
- Aldo Giudice
- G. Pascale Foundation National Cancer Institute, Naples, Italy.
| | | | | |
Collapse
|
170
|
Shlomai J. Redox control of protein-DNA interactions: from molecular mechanisms to significance in signal transduction, gene expression, and DNA replication. Antioxid Redox Signal 2010; 13:1429-76. [PMID: 20446770 DOI: 10.1089/ars.2009.3029] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Protein-DNA interactions play a key role in the regulation of major cellular metabolic pathways, including gene expression, genome replication, and genomic stability. They are mediated through the interactions of regulatory proteins with their specific DNA-binding sites at promoters, enhancers, and replication origins in the genome. Redox signaling regulates these protein-DNA interactions using reactive oxygen species and reactive nitrogen species that interact with cysteine residues at target proteins and their regulators. This review describes the redox-mediated regulation of several master regulators of gene expression that control the induction and suppression of hundreds of genes in the genome, regulating multiple metabolic pathways, which are involved in cell growth, development, differentiation, and survival, as well as in the function of the immune system and cellular response to intracellular and extracellular stimuli. It also discusses the role of redox signaling in protein-DNA interactions that regulate DNA replication. Specificity of redox regulation is discussed, as well as the mechanisms providing several levels of redox-mediated regulation, from direct control of DNA-binding domains through the indirect control, mediated by release of negative regulators, regulation of redox-sensitive protein kinases, intracellular trafficking, and chromatin remodeling.
Collapse
Affiliation(s)
- Joseph Shlomai
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Tropical and Infectious Diseases, Institute for Medical Research Canada-Israel, The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
171
|
Afonyushkin T, Oskolkova OV, Binder BR, Bochkov VN. Involvement of CK2 in activation of electrophilic genes in endothelial cells by oxidized phospholipids. J Lipid Res 2010; 52:98-103. [PMID: 20934988 DOI: 10.1194/jlr.m009480] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Oxidized phospholipids (OxPLs) are increasingly recognized as pleiotropic lipid mediators demonstrating a variety of biological activities. In particular, OxPLs induce electrophilic stress response and stimulate expression of NF-E2-related factor 2 (NRF2)-dependent genes. The mechanisms of NRF2 upregulation in response to OxPLs, however, are incompletely understood. Here we show that upregulation of NRF2 by OxPLs depends on the activity of the CK2 protein kinase. Inactivation of CK2 by chemical inhibitors or gene silencing resulted in diminished accumulation of NRF2 and its target genes, GCLM, HMOX1, and NQO1, downstream in response to OxPLs. Furthermore, inhibition of CK2 suppressed NRF2-dependent induction of ATF4 and its downstream gene VEGF. Thus, inactivation of CK2 in OxPL-treated endothelial cells results in inhibition of the NRF2-ATF4-VEGF axis and is likely to produce antiangiogenic effects. This work characterizes novel cross-talk between CK2 and cellular stress pathways, which may provide additional insights into the mechanisms of beneficial action and side-effects of CK2 inhibitors.
Collapse
Affiliation(s)
- Taras Afonyushkin
- Department of Vascular Biology and Thrombosis Research, Center for Biomolecular Medicine and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
172
|
Hur W, Sun Z, Jiang T, Mason DE, Peters EC, Zhang DD, Luesch H, Schultz PG, Gray NS. A small-molecule inducer of the antioxidant response element. ACTA ACUST UNITED AC 2010; 17:537-47. [PMID: 20534351 DOI: 10.1016/j.chembiol.2010.03.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 03/18/2010] [Accepted: 03/26/2010] [Indexed: 01/02/2023]
Abstract
Eukaryotic cells counteract oxidative and other environmental stress through the activation of Nrf2, the transcription factor that controls the expression of a host of protective enzymes by binding to the antioxidant response element (ARE). The electrophilic molecules that are able to activate Nrf2 and its downstream target genes have demonstrated therapeutic potential in carcinogen-induced tumor models. Using a high-throughput cellular screen, we discovered a class of ARE activator, which we named AI-1, that activates Nrf2 by covalently modifying Keap1, the negative regulator of Nrf2. Biochemical studies indicated that modification of Cys151 of Keap1 by AI-1 disrupted the ability of Keap1 to serve as an adaptor for Cul3-Keap1 ubiquitin ligase complex, thereby causing stabilization and transcriptional activation of Nrf2. AI-1 and its biotinylated derivative are useful pharmacological probes for investigating the molecular details of the cellular antioxidant response.
Collapse
Affiliation(s)
- Wooyoung Hur
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Sykiotis GP, Bohmann D. Stress-activated cap'n'collar transcription factors in aging and human disease. Sci Signal 2010; 3:re3. [PMID: 20215646 DOI: 10.1126/scisignal.3112re3] [Citation(s) in RCA: 615] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cap'n'collar (Cnc) transcription factors are conserved in metazoans and have important developmental and homeostatic functions. The vertebrate Nrf1, Nrf2, and Nrf3; the Caenorhabditis elegans SKN-1; and the Drosophila CncC comprise a subgroup of Cnc factors that mediate adaptive responses to cellular stress. The most studied stress-activated Cnc factor is Nrf2, which orchestrates the transcriptional response of cells to oxidative stressors and electrophilic xenobiotics. In rodent models, signaling by Nrf2 defends against oxidative stress and aging-associated disorders, such as neurodegeneration, respiratory diseases, and cancer. In humans, polymorphisms that decrease Nrf2 abundance have been associated with various pathologies of the skin, respiratory system, and digestive tract. In addition to preventing disease in rodents and humans, Cnc factors have life-span-extending and anti-aging functions in invertebrates. However, despite the pro-longevity and antioxidant roles of stress-activated Cnc factors, their activity paradoxically declines in aging model organisms and in humans suffering from progressive respiratory disease or neurodegeneration. We review the roles and regulation of stress-activated Cnc factors across species, present all reported instances in which their activity is paradoxically decreased in aging and disease, and discuss the possibility that the pharmacological restoration of Nrf2 signaling may be useful in the prevention and treatment of age-related diseases.
Collapse
Affiliation(s)
- Gerasimos P Sykiotis
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | |
Collapse
|
174
|
Ma Q. Transcriptional responses to oxidative stress: pathological and toxicological implications. Pharmacol Ther 2009; 125:376-93. [PMID: 19945483 DOI: 10.1016/j.pharmthera.2009.11.004] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 11/07/2009] [Indexed: 12/28/2022]
Abstract
The utilization of molecular oxygen as the terminal electron acceptor for energy production has in many ways shaped the evolution of complex life, physiology, and certain disease processes. The generation of reactive oxygen species (ROS), either as by-products of O(2) metabolism or by specialized enzymes, has the potential to damage cellular components and functions. Exposure to a variety of exogenous toxicants also promotes ROS production directly or through indirect means to cause toxicity. Oxidative stress activates the expression of a wide range of genes that mediate the pathogenic effect of ROS or are required for the detection and detoxification of the oxidants. In many cases, these are mediated by specific transcription factors whose expression, structure, stability, nuclear targeting, or DNA-binding affinity is regulated by the level of oxidative stress. This review examines major transcription factors that mediate transcriptional responses to oxidative stress, focusing on recent progress in the signaling pathways and mechanisms of activation of transcription factors by oxidative stress and the implications of this regulation in the development of disease and chemical toxicity.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States.
| |
Collapse
|
175
|
Cuadrado A, Moreno-Murciano P, Pedraza-Chaverri J. The transcription factor Nrf2 as a new therapeutic target in Parkinson's disease. Expert Opin Ther Targets 2009; 13:319-29. [DOI: 10.1517/13543780802716501] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
176
|
Chen-Roetling J, Li Z, Regan RF. Hemoglobin neurotoxicity is attenuated by inhibitors of the protein kinase CK2 independent of heme oxygenase activity. Curr Neurovasc Res 2009; 5:193-8. [PMID: 18691077 DOI: 10.2174/156720208785425684] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The heme oxygenase (HO) enzymes catalyze the rate-limiting step of heme breakdown, and may accelerate oxidative injury to neurons exposed to heme or hemoglobin. HO-1 and HO-2 are activated in vitro by the phos-phatidylinositol 3-kinase (PI3K)/Akt and protein kinase C (PKC)/CK2 pathways, respectively. The present study tested the hypotheses that CK2, PKC, and PI3K inhibitors would reduce both HO activity and neuronal vulnerability to hemoglobin in murine cortical cultures. Oxidative cell injury was quantified by LDH release and malondialdehyde assays. HO activity was assessed by carbon monoxide assay. Consistent with prior observations, treating primary cortical cultures with hemoglobin for 16h resulted in release of approximately half of neuronal LDH and a seven-fold increase in malondialdehyde. Both endpoints were significantly reduced by the CK2 inhibitors 4,5,6,7-tetrabromobenzotriazole (TBB) and 2-dimethyl-amino-4,5,6,7-tetrabromo-1H-benzimidazole (DMAT), and by the PKC inhibitor GF109203X; the PI3K inhibitors LY294002 and wortmannin had no effect. None of these inhibitors altered basal HO activity. The 1.9-fold activity increase observed after hemoglobin treatment was largely prevented by LY294002 and LY303511, a structural analog of LY294002 that does not inhibit PI3K activity. It was not reduced by wortmannin, TBB or GF109203X. These results suggest that the protective effect of CK2 and PKC inhibitors in this model is not dependent on reduction in HO activity. In this culture system that expresses both HO-1 and HO-2, HO activity does not appear to be primarily regulated by the PKC/CK2 or PI3K pathways.
Collapse
Affiliation(s)
- Jing Chen-Roetling
- Department of Emergency Medicine, Thomas Jefferson University, 1020 Sansom Street, Thompson Building Room 239, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|
177
|
Hayes JD, McMahon M. NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer. Trends Biochem Sci 2009; 34:176-88. [PMID: 19321346 DOI: 10.1016/j.tibs.2008.12.008] [Citation(s) in RCA: 673] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 12/08/2008] [Accepted: 12/10/2008] [Indexed: 12/26/2022]
Abstract
Transcription factor nuclear factor-erythroid 2-related factor 2 (NRF2) controls cellular adaptation to oxidants and electrophiles by inducing antioxidant and detoxification genes in response to redox stress. NRF2 is negatively regulated by Kelch-like ECH-associated protein 1 (KEAP1). Tumours from approximately 15% of patients with lung cancer harbour somatic mutations in KEAP1 that prevent effective NRF2 repression. Recently, two NRF2 mutation 'hot-spots' were identified in approximately 10% of patients with lung cancer, enabling the transcription factor to evade KEAP1-mediated repression. Somatic mutations in KEAP1 and NRF2 provide an insight into the molecular mechanisms by which NRF2 is regulated. Moreover, constitutive NRF2 activation might cause drug resistance in tumours, and an understanding of how the transcription factor is regulated indicates ways in which this could be overcome.
Collapse
Affiliation(s)
- John D Hayes
- Biomedical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK.
| | | |
Collapse
|
178
|
Clark J, Simon DK. Transcribe to survive: transcriptional control of antioxidant defense programs for neuroprotection in Parkinson's disease. Antioxid Redox Signal 2009; 11:509-28. [PMID: 18717631 DOI: 10.1089/ars.2008.2241] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a progressive, primarily motor disorder that is characterized by loss of dopaminergic (DA) neurons within the substantia nigra (SN). Cell death in PD has been associated with impaired mitochondrial function and increased oxidative stress. Strategies to reduce the oxidative load in DA cells may be beneficial in slowing the progression of PD. The transcription factor nuclear factor-erythroid 2 (NF-E2) related factor 2 (NRF2) is emerging as a master regulator of antioxidant defense systems, which makes it an attractive target for manipulations that aim to increase cellular resistance to oxidative stress. Peroxisome proliferator-activated receptor gamma (PPARgamma) coactivator-1 alpha (PGC1alpha) is a regulator of mitochondrial biogenesis genes that simultaneously upregulates many genes known to protect against oxidative stress. Pgc-1alpha knockout mice show enhanced susceptibility to SN neuronal loss following MPTP exposure, whilst overexpression of Pgc-1alpha appears to protect against oxidative stress in vitro. This makes PGC-1alpha a highly attractive target for neuroprotective therapies in PD. This review will explore the mechanisms behind the induction of NRF2 and PGC-1alpha in response to oxidative stress and identify common pathways that may provide targets for upregulating antioxidant defense programs.
Collapse
Affiliation(s)
- Joanne Clark
- Beth Israel Deaconess Medical Center, Department of Neurology, Boston, Massachusetts 02215, USA.
| | | |
Collapse
|
179
|
Ma Q. Xenobiotic-activated receptors: from transcription to drug metabolism to disease. Chem Res Toxicol 2008; 21:1651-71. [PMID: 18707139 DOI: 10.1021/tx800156s] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Xenobiotic-activated receptors (XARs) are a group of ligand-activated transcription factors that are evolutionally specialized to regulate genomic programs to protect the body against innumerable chemicals from the environment. XARs share unique properties, such as promiscuous ligand binding, conserved structural motifs, common protein partners, and overlapping target genes. These unique features of XARs clearly distinguish them from receptors that are activated by endogenous chemicals to regulate energy metabolism, reproduction, and growth and differentiation. XARs regulate xenobiotic metabolism and disposition by controlling the expression and induction of drug-metabolizing enzymes and transporters. Furthermore, XARs integrate a broad range of protective mechanisms, such as antioxidative response and immune/inflammatory functions, to antagonize foreign chemicals. As the primary means of xenobiotic sensing and defense, XARs are intimately involved in drug disposition, polymorphic drug clearance, drug-drug interaction, and pathogenesis of some chemically induced cancers and chronic diseases. As a consequence, some XAR characteristics have been exploited in drug development and safety evaluation of drugs and environmental carcinogens and toxicants. In this perspective, common features and recent advances in the structures, modes of action, and implications in disease and drug development of XARs are discussed.
Collapse
Affiliation(s)
- Qiang Ma
- Toxicology and Molecular Biology Branch, Health Effects Laboratory DiVision, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia 26505, USA.
| |
Collapse
|