1
|
Nandha SR, Checker R, Patwardhan RS, Sharma D, Sandur SK. Anti-oxidants as therapeutic agents for oxidative stress associated pathologies: future challenges and opportunities. Free Radic Res 2025; 59:61-85. [PMID: 39764687 DOI: 10.1080/10715762.2025.2450504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/13/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Free radicals have been implicated in the pathogenesis of cancer along with cardiovascular, neurodegenerative, pulmonary and inflammatory disorders. Further, the relationship between oxidative stress and disease is distinctively established. Clinical trials using anti-oxidants for the prevention of disease progression have indicated some beneficial effects. However, these trials failed to establish anti-oxidants as therapeutic agents due to lack of efficacy. This is attributed to the fact that living systems are under dynamic redox control wherein their redox behavior is compartmentalized and simple aggregation of redox couples, distributed throughout the system, is of miniscule importance while determining their overall redox state. Further, free radical metabolism is intriguingly complex as they play plural roles segregated in a spatio-temporal manner. Depending on quality, quantity and site of generation, free radicals exhibit beneficial or harmful effects. Use of nonspecific, non-targeted, general ROS scavengers lead to systemic elimination of all types of ROS and interferes in cellular signaling. Failure of anti-oxidants to act as therapeutic agents lies in this oversimplification of extremely dynamic cellular redox environment as a static and non-compartmentalized redox state. Rather than generalizing the term "oxidative stress" if we can identify the "type of oxidative stress" in different types of diseases, a targeted and more specific anti-oxidant therapy may be developed. In this review, we discuss the concept of redox dynamics, role and type of oxidative stress in disease conditions, and current status of anti-oxidants as therapeutic agents. Further, we probe the possibility of developing novel, targeted and efficacious anti-oxidants with drug-like properties.
Collapse
Affiliation(s)
- Shivani R Nandha
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
2
|
Van Hall-Beauvais A, Poganik JR, Huang KT, Parvez S, Zhao Y, Lin HY, Liu X, Long MJC, Aye Y. Z-REX uncovers a bifurcation in function of Keap1 paralogs. eLife 2022; 11:e83373. [PMID: 36300632 PMCID: PMC9754640 DOI: 10.7554/elife.83373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022] Open
Abstract
Studying electrophile signaling is marred by difficulties in parsing changes in pathway flux attributable to on-target, vis-à-vis off-target, modifications. By combining bolus dosing, knockdown, and Z-REX-a tool investigating on-target/on-pathway electrophile signaling, we document that electrophile labeling of one zebrafish-Keap1-paralog (zKeap1b) stimulates Nrf2- driven antioxidant response (AR) signaling (like the human-ortholog). Conversely, zKeap1a is a dominant-negative regulator of electrophile-promoted Nrf2-signaling, and itself is nonpermissive for electrophile-induced Nrf2-upregulation. This behavior is recapitulated in human cells: (1) zKeap1b-expressing cells are permissive for augmented AR-signaling through reduced zKeap1b-Nrf2 binding following whole-cell electrophile treatment; (2) zKeap1a-expressing cells are non-permissive for AR-upregulation, as zKeap1a-Nrf2 binding capacity remains unaltered upon whole-cell electrophile exposure; (3) 1:1 ZKeap1a:zKeap1b-co-expressing cells show no Nrf2-release from the Keap1-complex following whole-cell electrophile administration, rendering these cells unable to upregulate AR. We identified a zKeap1a-specific point-mutation (C273I) responsible for zKeap1a's behavior during electrophilic stress. Human-Keap1(C273I), of known diminished Nrf2-regulatory capacity, dominantly muted electrophile-induced Nrf2-signaling. These studies highlight divergent and interdependent electrophile signaling behaviors, despite conserved electrophile sensing.
Collapse
Affiliation(s)
| | - Jesse R Poganik
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Kuan-Ting Huang
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
| | - Saba Parvez
- Department of Pharmacology and Toxicology, College of Pharmacy, University of UtahSalt Lake CityUnited States
| | - Yi Zhao
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
- BayRay Innovation Center, Shenzhen Bay LaboratoryShenzhenChina
| | - Hong-Yu Lin
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen UniversityXiamenChina
| | - Xuyu Liu
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
- School of Chemistry, The University of SydneySydneyAustralia
- The Heart Research Institute, NewtownNewtownAustralia
| | - Marcus John Curtis Long
- Department of Biochemistry, Faculty of Biology and Medicine, University of LausanneLausanneSwitzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
| |
Collapse
|
3
|
Abdelmageed N, Twafik WAA, Seddek AL, Morad SARF. Vinpocetine-based therapy is an attractive strategy against oxidative stress-induced hepatotoxicity in vitro by targeting Nrf2/HO-1 pathway. EXCLI JOURNAL 2021; 20:550-561. [PMID: 34121971 PMCID: PMC8192879 DOI: 10.17179/excli2021-3463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/11/2021] [Indexed: 12/27/2022]
Abstract
Vinpocetine (Vin), a synthetic-derivative of Vincamine, monoterpenoid indole alkaloid, has been reported to have various medicinal benefits. The purpose of our study was to investigate the pivotal role of “nuclear factor erythroid 2-related factor-2” (Nrf2)-mediated antioxidant protection of Vin against H2O2 and paracetamol (APAP)-induced liver toxicity. For this purpose, a normal human hepatic cell line (L02 cells) was incubated with cytotoxic concentrations of H2O2 or APAP in the presence or absence of Vin. To evaluate the responses, MTS Cell Viability assay, immunoblotting, biochemical assays, and molecular docking approach were used. Viability analysis showed that treatment of L02 cells with Vin prevented the cytotoxicity induced by H2O2 and APAP. It was evidenced by the fact that Vin dumped H2O2- and APAP-cytotoxicity and reactive oxygen species (ROS) generation. The immunoblotting analysis shows that Vin increased Nrf2 expression along with the expression of target protein, heme oxygenase-1 (HO-1), and increased intracellular glutathione (GSH) level. Interestingly, we found that Vin could protect the protein expression-level of Nrf2, which indicated the prospective interaction between Vin and Keap1 protein. Additionally, molecular docking-study revealed that Vin competed with Nrf2 for Keap1-binding site, with hydrogen and stearic interactions. Collectively, Vin effectively protects against H2O2 and APAP-induced cytotoxicity via executing Nrf2-mediated restoration of antioxidative/oxidative balance. Meanwhile, Vin interrupts protein-protein interaction between Nrf2 and Keap1, which might also contribute to decrease Nrf2 degradation and stabilize protein expression. Thus, Vin-based adjuvant therapy may represent a smart drug regimen to mitigate drug-induced oxidative stress and liver injuries.
Collapse
Affiliation(s)
- Noha Abdelmageed
- Department of Pharmacology, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Wael Ahmed-Anwar Twafik
- Department of Biochemistry, Animal Health Research Institute (AHRI), Qena branch, Qena, Egypt
| | - Abdel-Latif Seddek
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | | |
Collapse
|
4
|
Kao YT, Chen YS, Tang KW, Lee JC, Tseng CH, Tzeng CC, Yen CH, Chen YL. Discovery of 4-Anilinoquinolinylchalcone Derivatives as Potential NRF2 Activators. Molecules 2020; 25:molecules25143133. [PMID: 32650607 PMCID: PMC7396997 DOI: 10.3390/molecules25143133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
Activation of nuclear factor erythroid-2-related factor 2 (NRF2) has been proven to be an effective means to prevent the development of cancer, and natural curcumin stands out as a potent NRF2 activator and cancer chemopreventive agent. In this study, we have synthesized a series of 4-anilinoquinolinylchalcone derivatives, and used a NRF2 promoter-driven firefly luciferase reporter stable cell line, the HaCaT/ARE cells, to screen a panel of these compounds. Among them, (E)-3-{4-[(4-acetylphenyl)amino]quinolin-2-yl}-1-(4-fluorophenyl)prop-2-en-1-one (13b) significantly increased NRF2 activity in the HaCaT cell with a half maximal effective concentration (EC50) value of 1.95 μM. Treatment of compound 13b upregulated HaCaT cell NRF2 expression at the protein level. Moreover, the mRNA level of NRF2 target genes, heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and glucose-6-phosphate dehydrogenase (G6PD) were significantly increased in HaCaT cells upon the compound 13b treatment. The molecular docking results exhibited that the small molecule 13b is well accommodated by the bound region of Kelch-like ECH-associated protein 1 (Keap1)-Kelch and NRF2 through stable hydrogen bonds and hydrophobic interaction, which contributed to the enhancement of affinity and stability between the ligand and receptor. Compound 13b has been identified as the lead compound for further structural optimization.
Collapse
Affiliation(s)
- Yu-Tse Kao
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-T.K.); (C.-C.T.)
| | - Yi-Siao Chen
- Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Kai-Wei Tang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (K.-W.T.); (C.-H.T.)
| | - Jin-Ching Lee
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (K.-W.T.); (C.-H.T.)
| | - Cherng-Chyi Tzeng
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-T.K.); (C.-C.T.)
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (C.-H.Y.); (Y.-L.C.); Tel.: +886-7-3121101 (ext. 2684) (C.-H.Y.); Fax: +886-7-3125339 (C.-H.Y.)
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-T.K.); (C.-C.T.)
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (C.-H.Y.); (Y.-L.C.); Tel.: +886-7-3121101 (ext. 2684) (C.-H.Y.); Fax: +886-7-3125339 (C.-H.Y.)
| |
Collapse
|
5
|
The Keap1/Nrf2-ARE Pathway as a Pharmacological Target for Chalcones. Molecules 2018; 23:molecules23071803. [PMID: 30037040 PMCID: PMC6100069 DOI: 10.3390/molecules23071803] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 01/14/2023] Open
Abstract
Chalcones have shown a broad spectrum of biological activities with clinical potential against various diseases. The biological activities are mainly attributed to the presence in the chalcones of the α,β-unsaturated carbonyl system, perceived as a potential Michael acceptor. Chalcones could activate the Kelch-like ECH-associated protein 1 (Keap1)/Nuclear factor erythroid 2-related factor 2 (Nrf2) pathway through a Michael addition reaction with the cysteines of Keap1, which acts as a redox sensor and negative regulator of Nrf2. This modification allows the dissociation of Nrf2 from the cytoplasmic complex with Keap1 and its nuclear translocation. At this level, Nrf2 binds to the antioxidant response element (ARE) and activates the expression of several detoxification, antioxidant and anti-inflammatory genes as well as genes involved in the clearance of damaged proteins. In this regard, the Keap1/Nrf2–ARE pathway is a new potential pharmacological target for the treatment of many chronic diseases. In this review we summarize the current progress in the study of Keap1/Nrf2–ARE pathway activation by natural and synthetic chalcones and their potential pharmacological applications. Among the pharmacological activities highlighted, anti-inflammatory activity was more evident than others, suggesting a multi-target Michael acceptor mechanism for the chalcones involving key regulators of the Nrf2 and nuclear factor- κB (NF-κB) pathways.
Collapse
|
6
|
Gallic acid, a natural polyphenol, protects against tert-butyl hydroperoxide- induced hepatotoxicity by activating ERK-Nrf2-Keap1-mediated antioxidative response. Food Chem Toxicol 2017; 119:479-488. [PMID: 29066411 DOI: 10.1016/j.fct.2017.10.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 02/07/2023]
Abstract
Gallic acid (GA), a natural polyphenol, has been shown to exert a variety of heath promoting effects. We herein investigated the critical role of nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response in the protection of GA against tert-butyl hydroperoxide (t-BHP)-induced hepatotoxicity in L02 cells. Pretreatment of GA prevented the hepatocytotoxicity induced by t-BHP, as evidenced by the facts that GA suppressed t-BHP-induced cytotoxicity and reactive oxygen species (ROS) generation. GA induced nuclear translocation of Nrf2 along with expression of target proteins, including heme oxygenase-1 (HO-1) and glutamate cysteine ligase catalytic modify subunit (GCLC), and increased intracellular glutathione (GSH) content. Additionally, GA induced phosphorylated activation of extracellular regulated kinase (ERK), and ERK inhibitor PD98059 partially decreased GA-induced hepatoprotection, and downregulated the increased protein expressions of Nrf2, GCLC and HO-1 induced by GA. Interestingly, we found that GA could enhance the thermal stability of Keap1, which indicated the potential interaction between GA and Keap1. Furthermore, molecular docking indicated that GA possibly competed with Nrf2 for binding to Keap1. Collectively, GA effectively protects against t-BHP-induced hepatotoxicity via inducing ERK/Nrf2-mediated antioxidative signaling pathway. Meanwhile, GA disturbs protein-protein interaction between Keap1 and Nrf2 which might also contribute to nuclear translocation of Nrf2.
Collapse
|
7
|
Long MJ, Lin HY, Parvez S, Zhao Y, Poganik JR, Huang P, Aye Y. β-TrCP1 Is a Vacillatory Regulator of Wnt Signaling. Cell Chem Biol 2017; 24:944-957.e7. [PMID: 28736239 DOI: 10.1016/j.chembiol.2017.06.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/16/2017] [Accepted: 06/17/2017] [Indexed: 12/22/2022]
Abstract
Simultaneous hyperactivation of Wnt and antioxidant response (AR) are often observed during oncogenesis. However, it remains unclear how the β-catenin-driven Wnt and the Nrf2-driven AR mutually regulate each other. The situation is compounded because many players in these two pathways are redox sensors, rendering bolus redox signal-dosing methods uninformative. Herein we examine the ramifications of single-protein target-specific AR upregulation in various knockdown lines. Our data document that Nrf2/AR strongly inhibits β-catenin/Wnt. The magnitude and mechanism of this negative regulation are dependent on the direct interaction between β-catenin N terminus and β-TrCP1 (an antagonist of both Nrf2 and β-catenin), and independent of binding between Nrf2 and β-TrCP1. Intriguingly, β-catenin positively regulates AR. Because AR is a negative regulator of Wnt regardless of β-catenin N terminus, this switch of function is likely sufficient to establish a new Wnt/AR equilibrium during tumorigenesis.
Collapse
Affiliation(s)
- Marcus John Long
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Hong-Yu Lin
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Saba Parvez
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Yi Zhao
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jesse Richard Poganik
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Paul Huang
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Yimon Aye
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
8
|
Yun KL, Wang ZY. Target/signalling pathways of natural plant-derived radioprotective agents from treatment to potential candidates: A reverse thought on anti-tumour drugs. Biomed Pharmacother 2017; 91:1122-1151. [DOI: 10.1016/j.biopha.2017.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/15/2017] [Accepted: 05/01/2017] [Indexed: 02/07/2023] Open
|
9
|
McElroy PB, Sri Hari A, Day BJ, Patel M. Post-translational Activation of Glutamate Cysteine Ligase with Dimercaprol: A NOVEL MECHANISM OF INHIBITING NEUROINFLAMMATION IN VITRO. J Biol Chem 2017; 292:5532-5545. [PMID: 28202547 DOI: 10.1074/jbc.m116.723700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 01/25/2017] [Indexed: 11/06/2022] Open
Abstract
Neuroinflammation and oxidative stress are hallmarks of various neurological diseases. However, whether and how the redox processes control neuroinflammation is incompletely understood. We hypothesized that increasing cellular glutathione (GSH) levels would inhibit neuroinflammation. A series of thiol compounds were identified to elevate cellular GSH levels by a novel approach (i.e. post-translational activation of glutamate cysteine ligase (GCL), the rate-limiting enzyme in GSH biosynthesis). These small thiol-containing compounds were examined for their ability to increase intracellular GSH levels in a murine microglial cell line (BV2), of which dimercaprol (2,3-dimercapto-1-propanol (DMP)) was found to be the most effective compound. DMP increased GCL activity and decreased LPS-induced production of pro-inflammatory cytokines and inducible nitric-oxide synthase induction in BV2 cells in a concentration-dependent manner. The ability of DMP to elevate GSH levels and attenuate LPS-induced pro-inflammatory cytokine production was inhibited by buthionine sulfoximine, an inhibitor of GCL. DMP increased the expression of GCL holoenzyme without altering the expression of its subunits or Nrf2 target proteins (NQO1 and HO-1), suggesting a post-translational mechanism. DMP attenuated LPS-induced MAPK activation in BV2 cells, suggesting the MAPK pathway as the signaling mechanism underlying the effect of DMP. Finally, the ability of DMP to increase GSH via GCL activation was observed in mixed cerebrocortical cultures and N27 dopaminergic cells. Together, the data demonstrate a novel mechanism of GSH elevation by post-translational activation of GCL. Post-translational activation of GCL offers a novel targeted approach to control inflammation in chronic neuronal disorders associated with impaired adaptive responses.
Collapse
Affiliation(s)
- Pallavi B McElroy
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| | - Ashwini Sri Hari
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| | - Brian J Day
- the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Manisha Patel
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| |
Collapse
|
10
|
Zhang H, Davies KJA, Forman HJ. Oxidative stress response and Nrf2 signaling in aging. Free Radic Biol Med 2015; 88:314-336. [PMID: 26066302 PMCID: PMC4628850 DOI: 10.1016/j.freeradbiomed.2015.05.036] [Citation(s) in RCA: 615] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/29/2015] [Accepted: 05/31/2015] [Indexed: 12/20/2022]
Abstract
Increasing oxidative stress, a major characteristic of aging, has been implicated in a variety of age-related pathologies. In aging, oxidant production from several sources is increased, whereas antioxidant enzymes, the primary lines of defense, are decreased. Repair systems, including the proteasomal degradation of damaged proteins, also decline. Importantly, the adaptive response to oxidative stress declines with aging. Nrf2/EpRE signaling regulates the basal and inducible expression of many antioxidant enzymes and the proteasome. Nrf2/EpRE activity is regulated at several levels, including transcription, posttranslation, and interactions with other proteins. This review summarizes current studies on age-related impairment of Nrf2/EpRE function and discusses the changes in Nrf2 regulatory mechanisms with aging.
Collapse
Affiliation(s)
- Hongqiao Zhang
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology
| | - Kelvin J A Davies
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology; Division of Molecular & Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Henry Jay Forman
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology; School of Natural Science, University of California at Merced, Merced, CA 95344, USA.
| |
Collapse
|
11
|
Cuadrado A. Structural and functional characterization of Nrf2 degradation by glycogen synthase kinase 3/β-TrCP. Free Radic Biol Med 2015; 88:147-157. [PMID: 25937177 DOI: 10.1016/j.freeradbiomed.2015.04.029] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 12/30/2022]
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a master regulator of cellular homeostasis that controls the expression of more than 1% of human genes related to biotransformation reactions, redox homeostasis, energetic metabolism, DNA repair, and proteostasis. Its activity has a tremendous impact on physiology and pathology and therefore it is very tightly regulated, mainly at the level of protein stability. In addition to the very well established regulation by the ubiquitin E3 ligase adapter Keap1, recent advances have identified a novel mechanism based on signaling pathways that regulate glycogen synthase kinse-3 (GSK-3). This kinase phosphorylates specific serine residues in the Neh6 domain of Nrf2 to create a degradation domain that is then recognized by the ubiquitin ligase adapter β-TrCP and tagged for proteasome degradation by a Cullin1/Rbx1 complex. Here we review the mechanistic elements and the signaling pathways that participate in this regulation by GSK-3/β-TrCP. These pathways include those activated by ligands of tyrosine kinase, G protein-coupled, metabotropic, and ionotropic receptors that activate phosphatidyl inositol 3-kinase (PI3K)/ATK and by the canonical WNT signaling pathway, where a fraction of Nrf2 interacts with Axin1/GSK-3. Considering that free Nrf2 protein is localized in the nucleus, we propose a model termed "double flux controller" to explain how Keap1 and β-TrCP coordinate the stability of Nrf2 in several scenarios. The GSK-3/β-TrCP axis provides a novel therapeutic strategy to modulate Nrf2 activity.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigaciones Biomedicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain.
| |
Collapse
|
12
|
MDA-7/IL-24 inhibits Nrf2-mediated antioxidant response through activation of p38 pathway and inhibition of ERK pathway involved in cancer cell apoptosis. Cancer Gene Ther 2014; 21:416-26. [DOI: 10.1038/cgt.2014.45] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 11/08/2022]
|
13
|
Hou Y, Wang Y, Wang H, Xu Y. Induction of glutathione synthesis in human hepatocytes by acute and chronic arsenic exposure: differential roles of mitogen-activated protein kinases. Toxicology 2014; 325:96-106. [PMID: 25201354 DOI: 10.1016/j.tox.2014.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/01/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
Glutathione (GSH) is a vital component of antioxidant defense which protects cells from toxic insults. Previously we found intracellular GSH was involved in cell resistance against arsenic-induced cytotoxicity. However, molecular mechanisms of GSH homeostasis during arsenic exposure are largely undefined. Here, we investigated roles of mitogen-activated protein kinases (MAPKs) in GSH synthesis pathway with two arsenic exposure strategies by using Chang human hepatocytes. In one strategy, acute arsenic exposure (20 μM, 24 h) was applied, as MAPK signaling is generally considered to be transient. In the other one, chronic arsenic exposure (500 nM, 20 weeks) was applied, which mimicked the general human exposure to arsenic. We found that acute arsenic exposure activated extracellular signal-regulated 1/2 kinases (ERK1/2) and c-Jun N-terminal kinase (JNK) in parallel with increased transcription and nuclear translocation of factor-erythroid 2-related factor 2 (NRF2) and enhanced expression of γ-glutamyl cysteine ligase catalytic subunit (GCLC), resulting in elevated intracellular GSH levels. Specific ERK inhibitor abolished arsenic-induced NRF2 nuclear translocation and GSH synthesis. During chronic arsenic exposure which induced a malignant cellular phenotype, continuous p38 activation and NRF2 nuclear translocation were observed with enhanced GSH synthesis. Specific p38 inhibitor attenuated arsenic-enhanced GSH synthesis without changing NRF2 nuclear translocation. Taken together, our results indicate MAPK pathways play an important role in cellular GSH homeostasis in response to arsenic. However, the specific activation of certain MAPK is different between acute and chronic arsenic exposure. Furthermore, it appears that during chronic arsenic exposure, GSH synthesis is regulated by p38 at least in part independent of NRF2.
Collapse
Affiliation(s)
- Yongyong Hou
- Environmental Toxicology Program, School of Public Health, China Medical University, No. 92 North 2nd Road, Shenyang, Liaoning 110001, PR China.
| | - Yi Wang
- Environmental Toxicology Program, School of Public Health, China Medical University, No. 92 North 2nd Road, Shenyang, Liaoning 110001, PR China.
| | - Huihui Wang
- Environmental Toxicology Program, School of Public Health, China Medical University, No. 92 North 2nd Road, Shenyang, Liaoning 110001, PR China
| | - Yuanyuan Xu
- Environmental Toxicology Program, School of Public Health, China Medical University, No. 92 North 2nd Road, Shenyang, Liaoning 110001, PR China.
| |
Collapse
|
14
|
Szakács G, Hall MD, Gottesman MM, Boumendjel A, Kachadourian R, Day BJ, Baubichon-Cortay H, Di Pietro A. Targeting the Achilles heel of multidrug-resistant cancer by exploiting the fitness cost of resistance. Chem Rev 2014; 114:5753-74. [PMID: 24758331 PMCID: PMC4059772 DOI: 10.1021/cr4006236] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Magyar tudósok körútja 2, Budapest 1117, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Evolution has favored the utilization of dioxygen (O2) in the development of complex multicellular organisms. O2 is actually a toxic mutagenic gas that is highly oxidizing and combustible. It is thought that plants are largely to blame for polluting the earth's atmosphere with O2 owing to the development of photosynthesis by blue-green algae over 2 billion years ago. The rise of the plants and atmospheric O2 levels placed evolutionary stress on organisms to adapt or become extinct. This implies that all the surviving creatures on our planet are mutants that have adapted to the "abnormal biology" of O2. Much of the adaptation to the presence of O2 in biological systems comes from well-coordinated antioxidant and repair systems that focus on converting O2 to its most reduced form, water (H2O), and the repair and replacement of damaged cellular macromolecules. Biological systems have also harnessed O2's reactive properties for energy production, xenobiotic metabolism, and host defense and as a signaling messenger and redox modulator of a number of cell signaling pathways. Many of these systems involve electron transport systems and offer many different mechanisms by which antioxidant therapeutics can alternatively produce an antioxidant effect without directly scavenging oxygen-derived reactive species. It is likely that each agent will have a different set of mechanisms that may change depending on the model of oxidative stress, organ system, or disease state. An important point is that all biological processes of aerobes have coevolved with O2 and this creates a Pandora's box for trying to understand the mechanism(s) of action of antioxidants being developed as therapeutic agents.
Collapse
Affiliation(s)
- Brian J Day
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA.
| |
Collapse
|
16
|
Ishikado A, Sono Y, Matsumoto M, Robida-Stubbs S, Okuno A, Goto M, King GL, Keith Blackwell T, Makino T. Willow bark extract increases antioxidant enzymes and reduces oxidative stress through activation of Nrf2 in vascular endothelial cells and Caenorhabditis elegans. Free Radic Biol Med 2013; 65:1506-1515. [PMID: 23277146 PMCID: PMC3800243 DOI: 10.1016/j.freeradbiomed.2012.12.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/02/2012] [Accepted: 12/09/2012] [Indexed: 12/30/2022]
Abstract
Willow bark extract (WBE) is listed in the European Pharmacopoeia and has been traditionally used for treating fever, pain, and inflammation. Recent studies have demonstrated its clinical usefulness. This study investigated the antioxidative effects of WBE in human umbilical vein endothelial cells (HUVECs) and Caenorhabditis elegans. WBE prevented oxidative-stress-induced cytotoxicity of HUVECs and death of C. elegans. WBE dose-dependently increased mRNA and protein expression levels of the nuclear factor erythroid 2-related factor 2 (Nrf2) target genes heme oxygenase-1, γ-glutamylcysteine ligase modifier and catalytic subunits, and p62 and intracellular glutathione (GSH) in HUVECs. In the nematode C. elegans, WBE increased the expression of the gcs-1::green fluorescent protein reporter, a well-characterized target of the Nrf2 ortholog SKN-1, in a manner that was SKN-1-dependent. WBE increased intranuclear expression and DNA binding of Nrf2 and the activity of an antioxidant response element (ARE) reporter plasmid in HUVECs. WBE-induced expression of Nrf2-regulated genes and increased GSH levels in HUVECs were reduced by Nrf2 and p38 small interfering (si) RNAs and by the p38-specific inhibitor SB203580. Nrf2 siRNA reduced the cytoprotective effect of WBE against oxidative stress in HUVECs. Salicin, a major anti-inflammatory ingredient of WBE, failed to activate ARE-luciferase activity, whereas a salicin-free WBE fraction showed intensive activity. WBE induced antioxidant enzymes and prevented oxidative stress through activation of Nrf2 independent of salicin, providing a new potential explanation for the clinical usefulness of WBE.
Collapse
Affiliation(s)
| | - Yoko Sono
- R&D Department, Sunstar Inc., Osaka 569-1195, Japan
| | | | - Stacey Robida-Stubbs
- Section on Islet Cell & Regenerative Biology, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Aya Okuno
- R&D Department, Sunstar Inc., Osaka 569-1195, Japan
| | - Masashi Goto
- R&D Department, Sunstar Inc., Osaka 569-1195, Japan
| | - George L King
- Section on Vascular Cell Biology, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - T Keith Blackwell
- Section on Islet Cell & Regenerative Biology, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| | | |
Collapse
|
17
|
Champelovier P, Chauchet X, Hazane-Puch F, Vergnaud S, Garrel C, Laporte F, Boutonnat J, Boumendjel A. Cellular and molecular mechanisms activating the cell death processes by chalcones: Critical structural effects. Toxicol In Vitro 2013; 27:2305-15. [PMID: 24134853 DOI: 10.1016/j.tiv.2013.09.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/06/2013] [Accepted: 09/24/2013] [Indexed: 02/06/2023]
Abstract
Chalcones are naturally occurring compounds with diverse pharmacological activities. Chalcones derive from the common structure: 1,3-diphenylpropenone. The present study aims to better understand the mechanistic pathways triggering chalcones anticancer effects and providing evidences that minor structural difference could lead to important difference in mechanistic effect. We selected two recently investigated chalcones (A and B) and investigated them on glioblastoma cell lines. It was found that chalcone A induced an apoptotic process (type I PCD), via the activation of caspase-3, -8 and -9. Chalcone A also increased CDK1/cyclin B ratios and decreased the mitochondrial transmembrane potential (ΔΨm). Chalcone B induced an autophagic cell death process (type II PCD), ROS-related but independent of both caspases and protein synthesis. Both chalcones increased Bax/Bcl2 ratios and decreased Ki67 and CD71 antigen expressions. The present investigation reveals that despite the close structure of chalcones A and B, significant differences in mechanism of effect were found.
Collapse
Affiliation(s)
- Pierre Champelovier
- Laboratoire de Cytologie, Département d'Anatomie et de Cytologie Pathologiques, Institut de Biologie et de Pathologie, Centre Hospitalier Universitaire de Grenoble, Hôpital A. Michallon, CS10217, 38043 Grenoble cedex 09, France.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ma Y, Nie H, Sheng C, Chen H, Wang B, Liu T, Shao J, He X, Zhang T, Zheng C, Xia W, Ying W. Roles of oxidative stress in synchrotron radiation X-ray-induced testicular damage of rodents. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2012; 4:108-114. [PMID: 22837810 PMCID: PMC3403560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 05/17/2012] [Indexed: 06/01/2023]
Abstract
Synchrotron radiation (SR) X-ray has characteristic properties such as coherence and high photon flux, which has excellent potential for its applications in medical imaging and cancer treatment. However, there is little information regarding the mechanisms underlying the damaging effects of SR X-ray on biological tissues. Oxidative stress plays an important role in the tissue damage induced by conventional X-ray, while the role of oxidative stress in the tissue injury induced by SR X-ray remains unknown. In this study we used the male gonads of rats as a model to study the roles of oxidative stress in SR X-ray-induced tissue damage. Exposures of the testes to SR X-ray at various radiation doses did not significantly increase the lipid peroxidation of the tissues, assessed at one day after the irradiation. No significant decreases in the levels of GSH or total antioxidation capacity were found in the SR X-ray-irradiated testes. However, the SR X-ray at 40 Gy induced a marked increase in phosphorylated H2AX - a marker of double-strand DNA damage, which was significantly decreased by the antioxidant N-acetyl cysteine (NAC). NAC also attenuated the SR X-ray-induced decreases in the cell layer number of seminiferous tubules. Collectively, our observations have provided the first characterization of SR X-ray-induced oxidative damage of biological tissues: SR X-ray at high doses can induce DNA damage and certain tissue damage during the acute phase of the irradiation, at least partially by generating oxidative stress. However, SR X-ray of various radiation doses did not increase lipid peroxidation.
Collapse
Affiliation(s)
- Yingxin Ma
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University Shanghai 200030, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kachadourian R, Day BJ, Pugazhenti S, Franklin CC, Genoux-Bastide E, Mahaffey G, Gauthier C, Di Pietro A, Boumendjel A. A synthetic chalcone as a potent inducer of glutathione biosynthesis. J Med Chem 2012; 55:1382-8. [PMID: 22239485 DOI: 10.1021/jm2016073] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chalcones continue to attract considerable interest due to their anti-inflammatory and antiangiogenic properties. We recently reported the ability of 2',5'-dihydroxychalcone (2',5'-DHC) to induce both breast cancer resistance protein-mediated export of glutathione (GSH) and c-Jun N-terminal kinase-mediated increased intracellular GSH levels. Herein, we report a structure-activity relationship study of a series of 30 synthetic chalcone derivatives with hydroxyl, methoxyl, and halogen (F and Cl) substituents and their ability to increase intracellular GSH levels. This effect was drastically improved with one or two electrowithdrawing groups on phenyl ring B and up to three methoxyl and/or hydroxyl groups on phenyl ring A. The optimal structure, 2-chloro-4',6'-dimethoxy-2'-hydroxychalcone, induced both a potent NF-E2-related factor 2-mediated transcriptional response and an increased formation of glutamate cysteine ligase holoenzyme, as shown using a human breast cancer cell line stably expressing a luciferase reporter gene driven by antioxidant response elements.
Collapse
Affiliation(s)
- Remy Kachadourian
- Department of Medicine, National Jewish Health, Denver, Colorado 80206, United States.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
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: 12.7] [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
|