1
|
Zhao M, Jiang Z, Yang Y, Wu S, Zhan Q, Zhou J, Liu Z, Zheng Y. Two Mycopolysaccharides from Hirsutella sinensis mycelia: Structural characterization and dual antioxidant-cardioprotective functions. Int J Biol Macromol 2025:145224. [PMID: 40516741 DOI: 10.1016/j.ijbiomac.2025.145224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 06/05/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
Cordyceps sinensis, a precious traditional Chinese herb, exhibits diverse polysaccharide structures and functions across strains. This study screened Hirsutella sinensis 02-3 and successfully extracted two mycopolysaccharides (fungal polysaccharides): ISP-AII (MW = 4.70 kDa) and ISP-BII (MW = 75.42 kDa). Chemical and monosaccharide composition analyses showed that ISP-AII comprised glucose, xylose, arabinose, galactose, and mannose (molar ratio 58.39:2.63:1.21:1.03:0.99), while ISP-BII contained additional fucose and exhibited a molar ratio of 57.82:1.07:1.06:0.39:0.37:0.32 for glucose, xylose, galactose, arabinose, fucose, and mannose, respectively. Techniques such as the Congo red test, Fourier transform infrared (FTIR), Scanning Electron Microscopy (SEM), and Nuclear Magnetic Resonance (NMR) were employed to characterize their structural features, suggesting both ISP-AII and ISP-BII were →4)-α-D-Glcp-(1 → main backbone. Functionally, ISP-BII had the highest 2,2-Diphenyl-1-Picrylhydrazyl (DPPH) scavenging rate of 61.7 %, and ISP-AII scavenged 56.12 % of hydroxyl radicals. Both also showed high reducing power and reactive oxygen species (ROS) - scavenging ability. Moreover, they protected cardiomyocytes from apoptosis by regulating key factors in the Phosphatidylinositol 3-Kinase/Protein Kinase B (PI3K/AKT) and mitochondrial BAX/Bcl-2/Caspase-3 apoptotic signaling pathways, and exerted antioxidant effects via the Keap1/Nrf2/ARE pathway. These results offer new insights into the cardioprotective functions of Cordyceps polysaccharides, and their potential applications in food, cosmetics, and pharmaceutical industries.
Collapse
Affiliation(s)
- Man Zhao
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Zhenhao Jiang
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Yingying Yang
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Shenghao Wu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Qiqi Zhan
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Junping Zhou
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Zhiqiang Liu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Yuguo Zheng
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| |
Collapse
|
2
|
Laddha AP, Seyednejad A, Donepudi AC, Goedken MJ, Manautou JE, Sartor GC. Inhibition of bromodomain and extra-terminal (BET) proteins with JQ1 exacerbates acetaminophen-induced hepatotoxicity by altering detoxification pathways and oxidative stress responses. Chem Biol Interact 2025; 413:111491. [PMID: 40157625 PMCID: PMC11998672 DOI: 10.1016/j.cbi.2025.111491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Acetaminophen (APAP) is a widely used over-the-counter drug for the treatment of fever and pain. At therapeutic doses, APAP has a relatively safe profile. However, at supratherapeutic doses it can produce liver injury and even fatal hepatotoxicity. Antioxidant genes regulated by the Kelch-like ECH-associated protein 1 (KEAP1)-nuclear factor, erythroid 2-like 2 (NRF2) pathway play a crucial role in hepatoprotection against APAP-induced hepatotoxicity and oxidative stress. Recent studies suggest that bromodomain and extra-terminal motif (BET) proteins, epigenetic readers, act as putative regulators of the KEAP1-NRF2 pathway, but their role in acute drug-induced liver injury (DILI) remains unclear. In this study, we employed complementary in vitro and in vivo approaches utilizing pharmacological inhibition and gene knockdown techniques to examine the role of BET proteins in APAP-induced toxicity. Our findings indicate that APAP treatment significantly alters the gene and protein expression of BET proteins in both mouse liver and the HC-04 cell line. Cytotoxicity analysis using lactate dehydrogenase (LDH) leakage assay revealed that treatment with the small molecule BET inhibitor JQ1 did not alter APAP-induced cytotoxicity. However, siRNA-mediated knockdown of the BET genes Brd3 and Brd4, but not Brd2, reduced APAP-induced cytotoxicity in HC-04 cells. In hepatic gene expression analysis experiments, JQ1 pretreatment in mice activated the Nrf2 pathway and altered antioxidant genes such as Gclc, Gclm, Ho-1, and Txnrd1, suggesting an enhancement of cellular defenses against APAP-induced oxidative stress at 12 h timepoint. However, by 24 h, histopathological findings revealed significant liver necrosis and inflammation in the JQ1-APAP treated group, indicating that while BET inhibition may confer early protection, it may not fully prevent long-term liver injury.
Collapse
Affiliation(s)
- A P Laddha
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, 06269, CT, USA
| | - A Seyednejad
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, 06269, CT, USA
| | - A C Donepudi
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, 06269, CT, USA
| | - M J Goedken
- Research Pathology Services, Rutgers University, Piscataway, NJ, 08854, USA
| | - J E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, 06269, CT, USA
| | - G C Sartor
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, 06269, CT, USA.
| |
Collapse
|
3
|
Dong Y, Jia R, Jiang Y, Li Q, Wang L, Ding W, Yan R, Qiu Y, Shi Z, Liu W, Wang J, Xu S, Li N. SMND-309 activates Nrf2 signaling to alleviate acetaminophen-induced hepatotoxicity and oxidative stress. PLoS One 2025; 20:e0310879. [PMID: 40163430 PMCID: PMC11957308 DOI: 10.1371/journal.pone.0310879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Acetaminophen (APAP) can be used for pain relief and fever alleviation, the overdose of which, however, may lead to the accumulation of N-acetyl-p-benzoquinone imine (NAPQI), inducing oxidative stress and liver damage. The natural compound SMND-309 has been shown to have hepatoprotective effects and potential antioxidant activity. However, its ability to alleviate acetaminophen-induced acute liver injury (AILI) has not been elucidated. OBJECTIVE To explore the protective effect of the natural compound SMND-309 against AILI and the potential mechanism. METHODS The AILI model was established using a mouse model and HepG2 cells for pathological evaluation and biochemical assays of mouse liver tissues to assess the level of liver injury. The effects of SMND-309 on cellular ROS levels and mitochondrial membrane potential were detected using DCFH-DA and JC-1 probes. Western blotting was performed to detect the expressions of Nrf2 signaling pathway and key proteins related to APAP metabolism in the combination of immunohistochemistry of liver tissues, with immunofluorescence assay used to detect whether Nrf2 undergoes nuclear translocation. Molecular docking, molecular dynamics simulation (MD) and biofilm layer interference (BLI) experiments were performed to detect the interaction of SMND-309 with Keap1. RESULTS SMND-309 improved histopathological changes in the liver, decreased alanine aminotransferase (ALT), aspartate aminotransferase (AST), and lactate dehydrogenase (LDH) levels, as well as attenuated oxidative stress injury and mitochondrial dysfunction in the HepG2 cell line. Further studies revealed that SMND-309 promoted nuclear translocation of Nrf2 and upregulated the expressions of glutamate-cysteine ligase catalytic subunit (GCLC), heme oxygenase 1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1). In addition, molecular docking and MD suggested that SMND-309 could bind Keap1 and identified possible binding modes, with BLI experiments confirming that SMND-309 directly interacted with Keap1. CONCLUSION SMND-309 exerts hepatoprotective effects against AILI in an Nrf2-ARE signaling pathway-dependent manner.
Collapse
Affiliation(s)
- Yao Dong
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Ru Jia
- Department of Obstetrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Yujie Jiang
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Qing Li
- Department of Obstetrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Lei Wang
- Department of Obstetrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Wensi Ding
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Rui Yan
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Yujie Qiu
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Zhengjie Shi
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Wenying Liu
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Jing Wang
- Department of Gastroenterology, Yantai Zhifu Hospital, Yantai, Shandong, P.R.China
| | - Sen Xu
- Binzhou Medical University, Yantai, Shandong, P.R.China
| | - Na Li
- Department of Orthopedics, Yantai Yantaishan Hospital, Yantai, Shandong, P.R.China
| |
Collapse
|
4
|
Duan M, Yu N, Liu J, Zhao Y, Zhang J, Song S, Wang S. Remimazolam Suppresses Oxidative Stress and Apoptosis in Cerebral Ischemia/Reperfusion Injury by Regulating AKT/GSK-3β/NRF2 Pathway. Drug Des Devel Ther 2025; 19:111-128. [PMID: 39807342 PMCID: PMC11725640 DOI: 10.2147/dddt.s478692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/25/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction The mechanism of remimazolam, a benzodiazepine that activates γ-aminobutyric acid a (GABAa) receptors, in cerebral ischemia/reperfusion (I/R) injury is not well understood. Therefore, we explored whether remimazolam activates protein kinase B (AKT)/glycogen synthase kinase-3β (GSK-3β)/nuclear factor erythroid 2-related factor 2 (NRF2) to attenuate brain I/R injury in transcerebral I/R-injured rats and transoxygenic glucose deprivation/reperfusion (OGD/R)-injured SY5Y cells. Material and Methods Remimazolam was added at the beginning of cell and rat reperfusion, and the PI3K/AKT inhibitor LY294002 was added to inhibit the AKT/GSK-3β/NRF2 pathway 24 h before cellular OGD/R treatment and 30 min before rat brain I/R treatment. The viability and apoptosis rate of SY5Y cells, neurological deficit score, cerebral infarction volume and morphological changes of rat brain cells as well as the protein expression of Bax, Bcl2, Caspase 3, Cleaved-Caspase 3 and the number of TdT-mediated dUTP Nick-End Labeling (TUNEL)-positive cells in the penumbral region were detected. Reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), NRF2, heme oxygenase 1 (HO-1), AKT, P-AKT, GSK-3β, P-GSK-3β protein expression, and nuclear translocation of NRF2 were measured in cell and animal assays. Results Reduced SY5Y cell viability and increased apoptosis caused by OGD/R injury, elevated neurological deficit scores and cerebral infarct volume induced by brain I/R injury in rats, cerebral cell injury, as well as elevated Bax, Cleaved-Caspase 3, decreased Bcl2, and increased number of TUNEL-positive cells in rat brain tissue were all moderated by remimazolam. Decreased GSH-Px, SOD and Elevated MDA, ROS induced by OGD/R-injured SY5Y cells and brain I/R-injured rats were moderated by remimazolam. Meanwhile, remimazolam increased NRF2, HO-1, P-AKT, P-GSK-3β, and the nuclear accumulation of NRF2. The PI3K/AKT inhibitor LY294002 reversed the role of remimazolam in brain I/R injury. Conclusion This study demonstrates that remimazolam activates the AKT/GSK-3β/NRF2 pathway, thereby attenuating oxidative stress and apoptosis to protect against brain I/R injury.
Collapse
Affiliation(s)
- Mei Duan
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Ning Yu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jia Liu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yang Zhao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jing Zhang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Siyi Song
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, People’s Republic of China
| | - Shilei Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
5
|
Wang T, Liu M, Li X, Zhang S, Gu H, Wei X, Wang X, Xu Z, Shen T. Naturally-derived modulators of the Nrf2 pathway and their roles in the intervention of diseases. Free Radic Biol Med 2024; 225:560-580. [PMID: 39368519 DOI: 10.1016/j.freeradbiomed.2024.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Cumulative evidence has verified that persistent oxidative stress is involved in the development of various chronic diseases, including pulmonary, neurodegenerative, kidney, cardiovascular, and liver diseases, as well as cancers. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a pivotal role in regulating cellular oxidative stress and inflammatory reactions, making it a focal point for disease prevention and treatment strategies. Natural products are essential resources for discovering leading molecules for new drug research and development. In this review, we comprehensively outlined the progression of the knowledge on the Nrf2 pathway, Nrf2 activators in clinical trials, the naturally-derived Nrf2 modulators (particularly from 2014-present), as well as their effects on the pathogenesis of chronic diseases.
Collapse
Affiliation(s)
- Tian Wang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Mingjie Liu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xinyu Li
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Sen Zhang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Haoran Gu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xuan Wei
- Shandong Center for Food and Drug Evaluation and Inspection, Jinan, Shandong, PR China
| | - Xiaoning Wang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Zhenpeng Xu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| | - Tao Shen
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| |
Collapse
|
6
|
Hayashi M, Okazaki K, Papgiannakopoulos T, Motohashi H. The Complex Roles of Redox and Antioxidant Biology in Cancer. Cold Spring Harb Perspect Med 2024; 14:a041546. [PMID: 38772703 PMCID: PMC11529857 DOI: 10.1101/cshperspect.a041546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Redox reactions control fundamental biochemical processes, including energy production, metabolism, respiration, detoxification, and signal transduction. Cancer cells, due to their generally active metabolism for sustained proliferation, produce high levels of reactive oxygen species (ROS) compared to normal cells and are equipped with antioxidant defense systems to counteract the detrimental effects of ROS to maintain redox homeostasis. The KEAP1-NRF2 system plays a major role in sensing and regulating endogenous antioxidant defenses in both normal and cancer cells, creating a bivalent contribution of NRF2 to cancer prevention and therapy. Cancer cells hijack the NRF2-dependent antioxidant program and exploit a very unique metabolism as a trade-off for enhanced antioxidant capacity. This work provides an overview of redox metabolism in cancer cells, highlighting the role of the KEAP1-NRF2 system, selenoproteins, sulfur metabolism, heme/iron metabolism, and antioxidants. Finally, we describe therapeutic approaches that can be leveraged to target redox metabolism in cancer.
Collapse
Affiliation(s)
- Makiko Hayashi
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA
| | - Keito Okazaki
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
7
|
Fitisemanu FM, Padilla-Benavides T. Emerging perspectives of copper-mediated transcriptional regulation in mammalian cell development. Metallomics 2024; 16:mfae046. [PMID: 39375833 PMCID: PMC11503025 DOI: 10.1093/mtomcs/mfae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Copper (Cu) is a vital micronutrient necessary for proper development and function of mammalian cells and tissues. Cu mediates the function of redox active enzymes that facilitate metabolic processes and signaling pathways. Cu levels are tightly regulated by a network of Cu-binding transporters, chaperones, and small molecule ligands. Extensive research has focused on the mammalian Cu homeostasis (cuprostasis) network and pathologies, which result from mutations and perturbations. There are roles for Cu-binding proteins as transcription factors (Cu-TFs) and regulators that mediate metal homeostasis through the activation or repression of genes associated with Cu handling. Emerging evidence suggests that Cu and some Cu-TFs may be involved in the regulation of targets related to development-expanding the biological roles of Cu-binding proteins. Cu and Cu-TFs are implicated in embryonic and tissue-specific development alongside the mediation of the cellular response to oxidative stress and hypoxia. Cu-TFs are also involved in the regulation of targets implicated in neurological disorders, providing new biomarkers and therapeutic targets for diseases such as Parkinson's disease, prion disease, and Friedreich's ataxia. This review provides a critical analysis of the current understanding of the role of Cu and cuproproteins in transcriptional regulation.
Collapse
|
8
|
Ramachandran A, Akakpo JY, Curry SC, Rumack BH, Jaeschke H. Clinically relevant therapeutic approaches against acetaminophen hepatotoxicity and acute liver failure. Biochem Pharmacol 2024; 228:116056. [PMID: 38346541 PMCID: PMC11315809 DOI: 10.1016/j.bcp.2024.116056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/15/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Liver injury and acute liver failure caused by an acetaminophen (APAP) overdose is a significant clinical problem in western countries. With the introduction of the mouse model of APAP hepatotoxicity in the 1970 s, fundamental mechanisms of cell death were discovered. This included the recognition that part of the APAP dose is metabolized by cytochrome P450 generating a reactive metabolite that is detoxified by glutathione. After the partial depletion of glutathione, the reactive metabolite will covalently bind to sulfhydryl groups of proteins, which is the initiating event of the toxicity. This insight led to the introduction of N-acetyl-L-cysteine, a glutathione precursor, as antidote against APAP overdose in the clinic. Despite substantial progress in our understanding of the pathomechanisms over the last decades viable new antidotes only emerged recently. This review will discuss the background, mechanisms of action, and the clinical prospects of the existing FDA-approved antidote N-acetylcysteine, of several new drug candidates under clinical development [4-methylpyrazole (fomepizole), calmangafodipir] and examples of additional therapeutic targets (Nrf2 activators) and regeneration promoting agents (thrombopoietin mimetics, adenosine A2B receptor agonists, Wharton's Jelly mesenchymal stem cells). Although there are clear limitations of certain therapeutic approaches, there is reason to be optimistic. The substantial progress in the understanding of the pathophysiology of APAP hepatotoxicity led to the consideration of several drugs for development as clinical antidotes against APAP overdose in recent years. Based on the currently available information, it is likely that this will result in additional drugs that could be used as adjunct treatment for N-acetylcysteine.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jephte Y Akakpo
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Steven C Curry
- Department of Medical Toxicology, Banner - University Medical Center Phoenix, Phoenix, AZ, USA; Department of Medicine, and Division of Clinical Data Analytics and Decision Support, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Barry H Rumack
- Department of Emergency Medicine and Pediatrics, University of Colorado School of Medicine, Denver, CO, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
9
|
Jaeschke H, Ramachandran A. Central Mechanisms of Acetaminophen Hepatotoxicity: Mitochondrial Dysfunction by Protein Adducts and Oxidant Stress. Drug Metab Dispos 2024; 52:712-721. [PMID: 37567742 PMCID: PMC11257690 DOI: 10.1124/dmd.123.001279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Acetaminophen (APAP) is an analgesic and antipyretic drug used worldwide, which is safe at therapeutic doses. However, an overdose can induce liver injury and even liver failure. Mechanistic studies in mice beginning with the seminal papers published by B.B. Brodie's group in the 1970s have resulted in important insight into the pathophysiology. Although the metabolic activation of APAP with generation of a reactive metabolite, glutathione depletion, and protein adduct formation are critical initiating events, more recently, mitochondria have come into focus as an important target and decision point of cell death. This review provides a comprehensive overview of the induction of mitochondrial superoxide and peroxynitrite formation and its propagation through a mitogen-activated protein kinase cascade, the mitochondrial permeability transition pore opening caused by iron-catalyzed protein nitration, and the mitochondria-dependent nuclear DNA fragmentation. In addition, the role of adaptive mechanisms that can modulate the pathophysiology, including autophagy, mitophagy, nuclear erythroid 2 p45-related factor 2 activation, and mitochondrial biogenesis, are discussed. Importantly, it is outlined how the mechanisms elucidated in mice translate to human hepatocytes and APAP overdose patients, and how this mechanistic insight explains the mechanism of action of the clinically approved antidote N-acetylcysteine and led to the recent discovery of a novel compound, fomepizole, which is currently under clinical development. SIGNIFICANCE STATEMENT: Acetaminophen (APAP)-induced liver injury is the most frequent cause of acute liver failure in western countries. Extensive mechanistic research over the last several decades has revealed a central role of mitochondria in the pathophysiology of APAP hepatotoxicity. This review article provides a comprehensive discussion of a) mitochondrial protein adducts and oxidative/nitrosative stress, b) mitochondria-regulated nuclear DNA fragmentation, c) adaptive mechanisms to APAP-induced cellular stress, d) translation of cell death mechanisms to overdose patients, and e) mechanism-based antidotes against APAP-induced liver injury.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
10
|
Zhang XN, Zhang YJ, Wang L, Hong SJ, Zhang CL, Zhao XL, Zeng T. NLRP3 inflammasome activation triggers severe inflammatory liver injury in N, N-dimethylformamide-exposed mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172653. [PMID: 38649053 DOI: 10.1016/j.scitotenv.2024.172653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
N,N-dimethylformamide (DMF) is a widely utilized chemical solvent with various industrial applications. Previous studies have indicated that the liver is the most susceptible target to DMF exposure, whereas the underlying mechanisms remain to be elucidated. This study aimed to investigate the role of NLRP3 inflammasome in DMF-induced liver injury in mice by using two NLRP3 inflammasome inhibitors, Nlrp3-/- mice, Nfe2l2-/- mice, and a macrophage-depleting agent. RNA sequencing revealed that endoplasmic reticulum (ER) stress and NLRP3 inflammasome-associated pathways were activated in the mouse liver after acute DMF exposure, which was validated by Western blotting. Interestingly, DMF-induced liver injury was effectively suppressed by two inflammasome inhibitors, MCC950 and Dapansutrile. In addition, knockout of Nlrp3 markedly attenuated DMF-induced liver injury without affecting the metabolism of DMF. Furthermore, silencing Nfe2l2 aggravated the liver injury and the NLRP3 inflammasome activation in mouse liver. Finally, the depletion of hepatic macrophages by clodronate liposomes significantly reduced the liver damage caused by DMF. These results suggest that NLRP3 inflammasome activation is the upstream molecular event in the development of acute liver injury induced by DMF.
Collapse
Affiliation(s)
- Xiu-Ning Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yan-Jing Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lin Wang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shu-Jun Hong
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Cui-Li Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiu-Lan Zhao
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
11
|
Mao Z, Mu J, Gao Z, Huang S, Chen L. Biological Functions and Potential Therapeutic Significance of O-GlcNAcylation in Hepatic Cellular Stress and Liver Diseases. Cells 2024; 13:805. [PMID: 38786029 PMCID: PMC11119800 DOI: 10.3390/cells13100805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
O-linked-β-D-N-acetylglucosamine (O-GlcNAc) glycosylation (O-GlcNAcylation), which is dynamically regulated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), is a post-translational modification involved in multiple cellular processes. O-GlcNAcylation of proteins can regulate their biological functions via crosstalk with other post-translational modifications, such as phosphorylation, ubiquitination, acetylation, and methylation. Liver diseases are a major cause of death worldwide; yet, key pathological features of the disease, such as inflammation, fibrosis, steatosis, and tumorigenesis, are not fully understood. The dysregulation of O-GlcNAcylation has been shown to be involved in some severe hepatic cellular stress, viral hepatitis, liver fibrosis, nonalcoholic fatty acid liver disease (NAFLD), malignant progression, and drug resistance of hepatocellular carcinoma (HCC) through multiple molecular signaling pathways. Here, we summarize the emerging link between O-GlcNAcylation and hepatic pathological processes and provide information about the development of therapeutic strategies for liver diseases.
Collapse
Affiliation(s)
- Zun Mao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China; (Z.M.); (Z.G.)
| | - Junpeng Mu
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou 221004, China;
| | - Zhixiang Gao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China; (Z.M.); (Z.G.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China; (Z.M.); (Z.G.)
| |
Collapse
|
12
|
Tan VWT, Salmi TM, Karamalakis AP, Gillespie A, Ong AJS, Balic JJ, Chan YC, Bladen CE, Brown KK, Dawson MA, Cox AG. SLAM-ITseq identifies that Nrf2 induces liver regeneration through the pentose phosphate pathway. Dev Cell 2024; 59:898-910.e6. [PMID: 38366599 DOI: 10.1016/j.devcel.2024.01.024] [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: 04/28/2023] [Revised: 12/07/2023] [Accepted: 01/24/2024] [Indexed: 02/18/2024]
Abstract
The liver exhibits a remarkable capacity to regenerate following injury. Despite this unique attribute, toxic injury is a leading cause of liver failure. The temporal processes by which the liver senses injury and initiates regeneration remain unclear. Here, we developed a transgenic zebrafish model wherein hepatocyte-specific expression of uracil phosphoribosyltransferase (UPRT) enabled the implementation of SLAM-ITseq to investigate the nascent transcriptome during initiation of liver injury and regeneration. Using this approach, we identified a rapid metabolic transition from the fed to the fasted state that was followed by induction of the nuclear erythroid 2-related factor (Nrf2) antioxidant program. We find that activation of Nrf2 in hepatocytes is required to induce the pentose phosphate pathway (PPP) and improve survival following liver injury. Mechanistically, we demonstrate that inhibition of the PPP disrupts nucleotide biosynthesis to prevent liver regeneration. Together, these studies provide fundamental insights into the mechanism by which early metabolic adaptation to injury facilitates tissue regeneration.
Collapse
Affiliation(s)
- Vicky W T Tan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Talhah M Salmi
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Anthony P Karamalakis
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Andrea Gillespie
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Athena Jessica S Ong
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jesse J Balic
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Yih-Chih Chan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Cerys E Bladen
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kristin K Brown
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mark A Dawson
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia; Department of Clinical Haematology, Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, VIC 3000, Australia.
| | - Andrew G Cox
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
13
|
Layman AJ, Alsbrook SM, Koturbash IK, McGill MR. Natural Products That Protect Against Acetaminophen Hepatotoxicity: A Call for Increased Rigor in Preclinical Studies of Dietary Supplements. J Diet Suppl 2024; 22:105-122. [PMID: 38562009 PMCID: PMC11442681 DOI: 10.1080/19390211.2024.2335573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Acetaminophen (APAP) overdose is one of the most common causes of acute liver injury. The current standard-of-care treatment for APAP hepatotoxicity, N-acetyl-l-cysteine, is highly effective when administered early after overdose, but loses efficacy in later-presenting patients. As a result, there is interest in the identification of new treatments for APAP overdose patients. Natural products are a promising source of new treatments because many are purported to have hepatoprotective effects. In fact, a great deal of research has been done to identify natural products that can protect against APAP-induced liver injury. However, serious concerns have been raised about the rigor and human relevance of these studies. Here, we systematically reviewed the APAP-natural product literature from 2013 to 2023 to determine the veracity of these concerns and the scope of the potential problem. The results substantiate the concerns that have been previously raised and point to concrete steps that can be taken to improve APAP-natural product research.
Collapse
Affiliation(s)
- Alexander J. Layman
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Scott M. Alsbrook
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Igor K. Koturbash
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Mitchell R. McGill
- Dept. of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Dept. of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR USA
- Dept. of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR USA
| |
Collapse
|
14
|
Masubuchi Y, Mikami K. Efficacy of oltipraz in preventing acetaminophen-induced liver injury in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:923-930. [PMID: 37535075 DOI: 10.1007/s00210-023-02649-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
Oltipraz (OPZ) is a synthetic dithiolethione with potential as a cancer chemopreventive agent, which can work by inducing detoxification enzymes. OPZ is an activator of nuclear factor erythroid 2-related factor 2 (Nrf2), suggesting its involvement in enzyme induction and possible protection against drug-induced liver injury. In this study, we present OPZ-mediated protection of mice against acetaminophen (APAP)-induced liver injury and discuss its possible contributing factors. Overnight-fasted male CD-1 mice were administered APAP intraperitoneally, and some mice were administered OPZ 16 h before APAP. Hepatotoxicity was assessed by measuring serum alanine aminotransferase leakage and histopathological evaluation. The hepatic mRNA expressions of CYP2E1, glutamate cysteine ligase (GCL), and NAD(P)H:quinone oxidoreductase (NQO1) were measured by real-time reverse-transcription polymerase chain reaction. OPZ protected mice from APAP-induced liver injury in a dose-dependent manner, but did not alter hepatic glutathione (GSH) content or GCL expression in control mice, indicating that its hepatoprotective effect is not due to changes in basal GSH levels. OPZ did not affect CYP2E1 expression or APAP-induced early GSH depletion, suggesting it does not inhibit the metabolic activation of APAP to produce N-acetyl-p-benzoquinone imine. In contrast, after GSH depletion, OPZ accelerated hepatic GSH recovery. APAP significantly increased GCL expression during liver injury, but OPZ treatment only led to additional NQO1 expression. This suggests that NQO1 is responsible for the enhanced GSH recovery and protection against APAP-induced liver injury seen in OPZ-treated mice. In summary, OPZ protects against APAP-induced liver injury by inducing NQO1 expression and resulting in improved GSH recovery.
Collapse
Affiliation(s)
- Yasuhiro Masubuchi
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Chiba Institute of Science, 15-8 Shiomi-Cho, Choshi, Chiba, 288-0025, Japan.
| | - Kenji Mikami
- Laboratory of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Chiba Institute of Science, 15-8 Shiomi-Cho, Choshi, Chiba, 288-0025, Japan
| |
Collapse
|
15
|
Jaeschke H, Ramachandran A. Acetaminophen Hepatotoxicity: Paradigm for Understanding Mechanisms of Drug-Induced Liver Injury. ANNUAL REVIEW OF PATHOLOGY 2024; 19:453-478. [PMID: 38265880 PMCID: PMC11131139 DOI: 10.1146/annurev-pathmechdis-051122-094016] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Acetaminophen (APAP) overdose is the clinically most relevant drug hepatotoxicity in western countries, and, because of translational relevance of animal models, APAP is mechanistically the most studied drug. This review covers intracellular signaling events starting with drug metabolism and the central role of mitochondrial dysfunction involving oxidant stress and peroxynitrite. Mitochondria-derived endonucleases trigger nuclear DNA fragmentation, the point of no return for cell death. In addition, adaptive mechanisms that limit cell death are discussed including autophagy, mitochondrial morphology changes, and biogenesis. Extensive evidence supports oncotic necrosis as the mode of cell death; however, a partial overlap with signaling events of apoptosis, ferroptosis, and pyroptosis is the basis for controversial discussions. Furthermore, an update on sterile inflammation in injury and repair with activation of Kupffer cells, monocyte-derived macrophages, and neutrophils is provided. Understanding these mechanisms of cell death led to discovery of N-acetylcysteine and recently fomepizole as effective antidotes against APAP toxicity.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA; ,
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA; ,
| |
Collapse
|
16
|
Liu M, Wu H, Li Q, Liu H, Chen C, Yin F, Wang H, Zha Z, Wang F. Mn 3O 4 nanozymes prevent acetaminophen-induced acute liver injury by attenuating oxidative stress and countering inflammation. J Colloid Interface Sci 2024; 654:83-95. [PMID: 37837854 DOI: 10.1016/j.jcis.2023.10.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/16/2023]
Abstract
Acetaminophen (APAP) overdose is steadily becoming the chief reason for drug-induced acute liver failure, yet limited treatment is currently clinically available. Considering that the mechanism of APAP-induced hepatotoxicity is inseparable from oxidative stress and inflammation, a biocompatible Mn3O4 nanozyme mimicking superoxide dismutase (SOD) and catalase (CAT) activities and possessing reactive oxygen species (ROS)-scavenging capacity and antiapoptotic properties, is reported herein as a promising nanodrug to treat APAP-induced liver injury (AILI). Possessing bioactive enzyme-like functions, Mn3O4 nanoparticles (NPs) can not only reduce the oxidative stress on the liver by decreasing ROS accumulation but also downregulate the infiltration of inflammatory macrophages that secrete proinflammatory cytokines (tumor necrosis factor-α, interleukin-1β, and interleukin-6). Notably, the bifunctional Mn3O4 NPs mediate nuclear factor-erythroid 2 p45-related factor 2 signaling pathway activation and nuclear factor kappa B signaling pathway inhibition to effectively prevent the already fragile APAP-overdosed murine hepatocytes from being attacked again, thus mitigating hepatocyte apoptosis and alleviating APAP-induced liver damage. Thus, the Mn3O4 nanozyme (Mn3O4 NPs) evaluated in this study has potential preventive and therapeutic effects on AILI.
Collapse
Affiliation(s)
- Menghua Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Haitao Wu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Qianhui Li
- China Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Hang Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Chongqing Chen
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Fan Yin
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| | - Fei Wang
- China Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
17
|
Ghosh P, Magee N, Akakpo JY, Ahamed F, Eppler N, Jones E, Yu Y, He L, Lebofsky M, Dai H, Jaeschke H, Ding WX, Zhang Y. Hepatocyte-specific deletion of small heterodimer partner protects mice against acetaminophen-induced hepatotoxicity via activation of Nrf2. Toxicol Sci 2023; 197:53-68. [PMID: 37792503 PMCID: PMC10734614 DOI: 10.1093/toxsci/kfad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023] Open
Abstract
Acetaminophen (APAP) overdose stands as the primary cause of acute liver failure in the United States. APAP hepatotoxicity involves hepatic glutathione (GSH) depletion and mitochondrial damage. To counteract the toxicity of APAP, the nuclear factor erythroid 2 like 2 (Nrf2) activates the expression of genes responsible for drug detoxification and GSH synthesis. In this study, we present evidence that the elimination of hepatocyte small heterodimer partner, a critical transcriptional repressor for liver metabolism, results in Nrf2 activation and protects mice from APAP-induced acute liver injury. Initial investigations conducted on wildtype (WT) mice revealed a swift downregulation of Shp mRNA within the first 24 h after APAP administration. Subsequent treatment of hepatocyte-specific Shp knockout (ShpHep-/-) mice with 300 mg/kg APAP for 2 h exhibited comparable bioactivation of APAP with that observed in the WT controls. However, a significant reduction in liver injury was observed in ShpHep-/- after APAP treatment for 6 and 24 h. The decreased liver injury correlated with a faster recovery of GSH, attributable to heightened expression of Nrf2 target genes involved in APAP detoxification and GSH synthesis. Moreover, in vitro studies revealed that SHP protein interacted with NRF2 protein, inhibiting the transcription of Nrf2 target genes. These findings hold relevance for humans, as overexpression of SHP hindered APAP-induced NRF2 activation in primary human hepatocytes. In conclusion, our studies have unveiled a novel regulatory axis involving SHP and NRF2 in APAP-induced acute liver injury, emphasizing SHP as a promising therapeutic target in APAP overdose-induced hepatotoxicity.
Collapse
Affiliation(s)
- Priyanka Ghosh
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Nancy Magee
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Jephte Y Akakpo
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Forkan Ahamed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Natalie Eppler
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Elizabeth Jones
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Yifan Yu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Lily He
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Margitta Lebofsky
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Hongyan Dai
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
18
|
Etemadi Y, Akakpo JY, Ramachandran A, Jaeschke H. Nrf2 as a therapeutic target in acetaminophen hepatotoxicity: A case study with sulforaphane. J Biochem Mol Toxicol 2023; 37:e23505. [PMID: 37598316 PMCID: PMC10842847 DOI: 10.1002/jbt.23505] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/24/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023]
Abstract
Acetaminophen (APAP) overdose can cause severe liver injury and acute liver failure. The only clinically approved antidote, N-acetylcysteine (NAC), is highly effective but has a narrow therapeutic window. In the last 2 decades, activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates acute phase proteins and antioxidant defense genes, has emerged as a putative new therapeutic target against APAP hepatotoxicity. However, virtually all studies that propose Nrf2 activation as mechanism of protection used prolonged pretreatment, which is not a clinically feasible approach to treat a drug overdose. Therefore, the objective of this study was to assess if therapeutic activation of Nrf2 is a viable approach to treat liver injury after APAP overdose. We used the water-soluble Nrf2 activator sulforaphane (SFN; 5 mg/kg) in a murine model of APAP hepatotoxicity (300 mg/kg). Our results indicate that short-term treatment (≤3 h) with SFN alone did not activate Nrf2 or its target genes. However, posttreatment with SFN after APAP partially protected at 6 h likely due to more rapid activation of the Nrf2-target gene heme oxygenase-1. A direct comparison of SFN with NAC given at 1 h after APAP showed a superior protection with NAC, which was maintained at 24 h unlike with SFN. Thus, Nrf2 activators have inherent problems like the need to create a cellular stress to activate Nrf2 and delayed adaptive responses which may hamper sustained protection against APAP hepatotoxicity. Thus, compared to the more direct acting antidote NAC, Nrf2 activators are less suitable for this indication.
Collapse
Affiliation(s)
- Yasaman Etemadi
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jephte Y Akakpo
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
19
|
Zhou W, He H, Wei Q, Che L, Zhao X, Liu W, Yan Y, Hu L, Du Y, Yin Z, Shuai Y, Yang L, Feng R. Puerarin protects against acetaminophen-induced oxidative damage in liver through activation of the Keap1/Nrf2 signaling pathway. Food Sci Nutr 2023; 11:6604-6615. [PMID: 37823166 PMCID: PMC10563760 DOI: 10.1002/fsn3.3609] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/08/2023] [Accepted: 07/22/2023] [Indexed: 10/13/2023] Open
Abstract
Puerarin (Pue) is a kind of isoflavone compound extracted from Pueraria lobata, which has significant antioxidant activity. Excessive use of acetaminophen (APAP) can cause oxidative stress in the liver and eventually lead to acute liver injury. The purpose of this study was to investigate the protective effect and the mechanism of puerarin on APAP-induced liver oxidative damage. In in vitro experiments, puerarin significantly increased the cell activity of HepG2 cells, reduced the ROS accumulation, alleviated the oxidative damage and mitochondrial dysfunction. In in vivo studies, our results showed that puerarin enhanced antioxidant activity and alleviated histopathological damage. Further studies showed that puerarin decreased the expression of Keap1, promoted the nuclear migration of Nrf2, and up-regulated the expression of GCLC, GCLM, HO-1 and NQO1. This study demonstrated that puerarin can protect APAP-induced liver injury via alleviating oxidative stress and mitochondrial dysfunction by affecting the nuclear migration of Nrf2 via inhibiting Keap1.
Collapse
Affiliation(s)
- Wanhai Zhou
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Heng He
- Natural Medicine Research Center, College of Veterinary MedicineSichuan Agricultural UniversityChengduChina
| | - Qin Wei
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| | - Litao Che
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Xin Zhao
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Wenwen Liu
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Yue Yan
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Lianqing Hu
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Yonghua Du
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary MedicineSichuan Agricultural UniversityChengduChina
| | - Yongkang Shuai
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| | - Li Yang
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| | - Ruizhang Feng
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| |
Collapse
|
20
|
Zhang S, Li Y, Liu X, Guo S, Jiang L, Huang Y, Wu Y. Carnosine alleviates kidney tubular epithelial injury by targeting NRF2 mediated ferroptosis in diabetic nephropathy. Amino Acids 2023; 55:1141-1155. [PMID: 37450047 DOI: 10.1007/s00726-023-03301-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Diabetic nephropathy (DN) can promote the occurrence of end-stage renal disease (ESRD). The injury of renal tubular epithelial cells is a significant reason for the occurrence of ESRD. A recent research demonstrated that ferroptosis was associated with renal tubular injury in DN. Ferroptosis is a kind of cell death brought on by the buildup of iron ions and lipid peroxidation brought on by ROS. Because carnosine (CAR) is a scavenger of iron ions and reactive oxygen species, we investigated whether CAR can improve DN by regulating ferroptosis. The results show that both CAR and Fer-1 significantly reduced kidney damage and inhibited ferroptosis in STZ mice. In addition, ferroptosis caused by HG or erastin (an inducer of ferroptosis) in human kidney tubular epithelial cell (HK2) was also rescued by CAR treatment. It was discovered that the protective effect of CAR against HG-induced ferroptosis was abolished when NRF2 was specifically knocked down in HK2 cells.
Collapse
Affiliation(s)
- Song Zhang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yuanyuan Li
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Xueqi Liu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Shanshan Guo
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Ling Jiang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yuebo Huang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Yonggui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
- Center for Scientific Research of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
21
|
Kim JS, Jegal KH, Park HR, Choi BR, Kim JK, Ku SK. A Mixture of Fermented Schizandrae Fructus Pomace and Hoveniae Semen cum Fructus Extracts Synergistically Protects against Oxidative Stress-Mediated Liver Injury. Antioxidants (Basel) 2023; 12:1556. [PMID: 37627551 PMCID: PMC10451536 DOI: 10.3390/antiox12081556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Schizandrae Fructus (SF) and Hoveniae Semen cum Fructus (HSCF) have long been used as medicinal herbs for treating various diseases in Asian traditional medicine. In the current study, we investigated the protective effect of fermented SF pomace and HSCF extract 1:1 (w:w) combination mixture (MSH) against carbon tetrachloride (CCl4)-induced acute liver injury mice. After MSH (50-200 mg/kg) oral administration for 7 consecutive days, animals were injected intraperitoneally with CCl4 (0.5 mL/kg). Histopathological observation revealed that administration of MSH synergistically decreased the degeneration of hepatocytes and the infiltration of inflammatory cells induced by CCl4. Moreover, MSH administration reduced the activities of alanine aminotransferase, aspartate aminotransferase, and γ-glutamyl transpeptidase in serum, and mitigated apoptotic cell death in hepatic parenchyma. In addition, MSH alleviated CCl4-mediated lipid peroxidation by restoring endogenous antioxidants capacities including glutathione contents, superoxide dismutase, and catalase activities. In vitro assessments using tert-butyl hydroperoxide-induced oxidative stress in HepG2 cells revealed that MSH protects hepatocytes by lowering ROS generation and lipid peroxidation via upregulating the transcriptional activity of nuclear factor erythroid-2-related factor 2 and the expression of antioxidant genes. Furthermore, MSH synergistically attenuated the expression of proinflammatory cytokines in CCl4-injured liver and lipopolysaccharide-stimulated RAW 264.7 cells. Taken together, these findings suggest that MSH has the potential to prevent acute liver damage by effectively suppressing oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jang-Soo Kim
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan-si 38610, Republic of Korea; (J.-S.K.); (H.-R.P.)
| | - Kyung-Hwan Jegal
- Department of Korean Medical Classics, College of Korean Medicine, Daegu Haany University, Gyeongsan-si 38610, Republic of Korea;
| | - Hye-Rim Park
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan-si 38610, Republic of Korea; (J.-S.K.); (H.-R.P.)
- Nutracore Co., Ltd., Suwon-si 16514, Republic of Korea;
| | - Beom-Rak Choi
- Nutracore Co., Ltd., Suwon-si 16514, Republic of Korea;
| | - Jae-Kwang Kim
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Gyeongsan-si 38610, Republic of Korea
| | - Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan-si 38610, Republic of Korea; (J.-S.K.); (H.-R.P.)
| |
Collapse
|
22
|
Gatbonton-Schwager T, Yagishita Y, Joshi T, Wakabayashi N, Srinivasan H, Suzuki T, Yamamoto M, Kensler TW. A Point Mutation at C151 of Keap1 of Mice Abrogates NRF2 Signaling, Cytoprotection in Vitro, and Hepatoprotection in Vivo by Bardoxolone Methyl (CDDO-Me). Mol Pharmacol 2023; 104:51-61. [PMID: 37188495 PMCID: PMC10353147 DOI: 10.1124/molpharm.123.000671] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023] Open
Abstract
Bardoxolone methyl (CDDO-Me) is an oleanane triterpenoid in late-stage clinical development for the treatment of patients with diabetic kidney disease. Preclinical studies in rodents demonstrate the efficacy of triterpenoids against carcinogenesis and other diseases, including renal ischemia-reperfusion injury, hyperoxia-induced acute lung injury, and immune hepatitis. Genetic disruption of Nrf2 abrogates protection by triterpenoids, suggesting that induction of the NRF2 pathway may drive this protection. Herein, we examined the effect of a point mutation (C151S) in KEAP1, a repressor of NRF2 signaling, at cysteine 151 in mouse embryo fibroblasts and mouse liver. Induction of target gene transcripts and enzyme activity by CDDO-Me was lost in C151S mutant fibroblasts compared with wild-type. Protection against menadione toxicity was also nullified in the mutant fibroblasts. In mouse liver, CDDO-Me evoked the nuclear translocation of NRF2, followed by increased transcript and activity levels of a prototypic target gene, Nqo1, in wild-type, but not C151S mutant, mice. To test the role of KEAP1 Cys151 in governing the broader pharmacodynamic action of CDDO-Me, wild-type and C151S mutant mice were challenged with concanavalin A to induce immune hepatitis. Strong protection was seen in wild-type but not C151S mutant mice. RNA-seq analysis of mouse liver from wild-type, C151S mutant, and Nrf2-knockout mice revealed a vigorous response of the NRF2 transcriptome in wild-type, but in neither C151S mutant nor Nrf2-knockout, mice. Activation of "off-target" pathways by CDDO were not observed. These data highlight the singular importance of the KEAP1 cysteine 151 sensor for activation of NRF2 signaling by CDDO-Me. SIGNIFICANCE STATEMENT: KEAP1 serves as a key sensor for induction of the cytoprotective signaling pathway driven by the transcription factor NRF2. Mutation of a single cysteine (C151) in KEAP1 abrogates the induction of NRF2 signaling and its downstream cytoprotective actions in vitro and in vivo by bardoxolone methyl (CDDO-Me), a drug in late-stage clinical development. Further, at these bioeffective concentrations/doses, activation of "off-target" pathways by CDDO-Me are not observed, highlighting the singular importance of NRF2 in its mode of action.
Collapse
Affiliation(s)
- Tonibelle Gatbonton-Schwager
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| | - Yoko Yagishita
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| | - Tanvi Joshi
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| | - Nobunao Wakabayashi
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| | - Harini Srinivasan
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| | - Takafumi Suzuki
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| | - Masayuki Yamamoto
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| | - Thomas W Kensler
- Translational Research Program (T.G.-S., Y.Y., T.J., N.W., T.W.K.) and Genomics & Bioinformatics (H.S.), Fred Hutchinson Cancer Center, Seattle, Washington and Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan (T.S., M.Y.)
| |
Collapse
|
23
|
Zhou Q, Zhou Q, Xia R, Zhang P, Xie Y, Yang Z, Khan A, Zhou Z, Tan W, Liu L. Swertiamarin or heat-transformed products alleviated APAP-induced hepatotoxicity via modulation of apoptotic and Nrf-2/NF- κB pathways. Heliyon 2023; 9:e18746. [PMID: 37554797 PMCID: PMC10404768 DOI: 10.1016/j.heliyon.2023.e18746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023] Open
Abstract
OBJECTIVE Swertiamarin (STM) belongs to iridoid class of compounds, and the heat-transformed products (HTPS) are produced by STM in the process of drug processing. The purpose of this study was to explore the protective effect and mechanism of STM or HTPS on acetaminophen (APAP)-induced hepatotoxicity. METHODS Mice and L-O2 cells were given APAP to establish the hepatotoxicity model in vivo and in vitro. The effects of STM or HTPS on oxidative stress, inflammation, and apoptosis induced by APAP were evaluated, with N-acetylcysteine (NAC) as a positive control. RESULTS STM or HTPS reduced the APAP-induced apoptosis of L-O2 cells and significantly alleviated the liver injury index induced by APAP (p < 0.01, 0.005) Interestingly, HTPS had better protective effect against APAP-induced hepatotoxicity than STM (p < 0.05). In addition STM or HTPS improved the histological abnormalities; inhibited lipid peroxidation and reduced the level of inflammatory mediators. They also activated the defense system of nuclear factor erythroid 2 related factor 2 (Nrf-2) and inhibited nuclear factor-κ B (NF-κB).
Collapse
Affiliation(s)
- Qian Zhou
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Qixiu Zhou
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Rui Xia
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Peng Zhang
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Yanqing Xie
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Zhuya Yang
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Afsar Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Zhihong Zhou
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Wenhong Tan
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Lu Liu
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| |
Collapse
|
24
|
Fuertes-Agudo M, Luque-Tévar M, Cucarella C, Martín-Sanz P, Casado M. Advances in Understanding the Role of NRF2 in Liver Pathophysiology and Its Relationship with Hepatic-Specific Cyclooxygenase-2 Expression. Antioxidants (Basel) 2023; 12:1491. [PMID: 37627486 PMCID: PMC10451723 DOI: 10.3390/antiox12081491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Oxidative stress and inflammation play an important role in the pathophysiological changes of liver diseases. Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor that positively regulates the basal and inducible expression of a large battery of cytoprotective genes, thus playing a key role in protecting against oxidative damage. Cyclooxygenase-2 (COX-2) is a key enzyme in prostaglandin biosynthesis. Its expression has always been associated with the induction of inflammation, but we have shown that, in addition to possessing other benefits, the constitutive expression of COX-2 in hepatocytes is beneficial in reducing inflammation and oxidative stress in multiple liver diseases. In this review, we summarized the role of NRF2 as a main agent in the resolution of oxidative stress, the crucial role of NRF2 signaling pathways during the development of chronic liver diseases, and, finally we related its action to that of COX-2, where it appears to operate as its partner in providing a hepatoprotective effect.
Collapse
Affiliation(s)
- Marina Fuertes-Agudo
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain; (M.F.-A.); (M.L.-T.); (C.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - María Luque-Tévar
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain; (M.F.-A.); (M.L.-T.); (C.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Carme Cucarella
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain; (M.F.-A.); (M.L.-T.); (C.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Paloma Martín-Sanz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas (IIB) “Alberto Sols”, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Marta Casado
- Instituto de Biomedicina de Valencia (IBV), CSIC, Jaume Roig 11, 46010 Valencia, Spain; (M.F.-A.); (M.L.-T.); (C.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| |
Collapse
|
25
|
Mukherjee AG, Gopalakrishnan AV. The mechanistic insights of the antioxidant Keap1-Nrf2 pathway in oncogenesis: a deadly scenario. Med Oncol 2023; 40:248. [PMID: 37480500 DOI: 10.1007/s12032-023-02124-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/06/2023] [Indexed: 07/24/2023]
Abstract
The Nuclear factor erythroid 2-related factor 2 (Nrf2) protein has garnered significant interest due to its crucial function in safeguarding cells and tissues. The Nrf2 protein is crucial in preserving tissue integrity by safeguarding cells against metabolic, xenobiotic and oxidative stress. Due to its various functions, Nrf2 is a potential pharmacological target for reducing the incidence of diseases such as cancer. However, mutations in Keap1-Nrf2 are not consistently favored in all types of cancer. Instead, they seem to interact with specific driver mutations of tumors and their respective tissue origins. The Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 pathway mutations are a powerful cancer adaptation that utilizes inherent cytoprotective pathways, encompassing nutrient metabolism and ROS regulation. The augmentation of Nrf2 activity elicits significant alterations in the characteristics of neoplastic cells, such as resistance to radiotherapy and chemotherapy, safeguarding against apoptosis, heightened invasiveness, hindered senescence, impaired autophagy and increased angiogenesis. The altered activity of Nrf2 can arise from diverse genetic and epigenetic modifications that instantly impact Nrf2 regulation. The present study aims to showcase the correlation between the Keap1-Nrf2 pathway and the progression of cancers, emphasizing genetic mutations, metabolic processes, immune regulation, and potential therapeutic strategies. This article delves into the intricacies of Nrf2 pathway anomalies in cancer, the potential ramifications of uncontrolled Nrf2 activity, and therapeutic interventions to modulate the Keap1-Nrf2 pathway.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
26
|
Cai C, Ma H, Peng J, Shen X, Zhen X, Yu C, Zhang P, Ji F, Wang J. USP25 regulates KEAP1-NRF2 anti-oxidation axis and its inactivation protects acetaminophen-induced liver injury in male mice. Nat Commun 2023; 14:3648. [PMID: 37339955 PMCID: PMC10282087 DOI: 10.1038/s41467-023-39412-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/13/2023] [Indexed: 06/22/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor responsible for mounting an anti-oxidation gene expression program to counter oxidative stress. Under unstressed conditions, Kelch-like ECH-associated protein 1 (KEAP1), an adaptor protein for CUL3 E3 ubiquitin ligase, mediates NRF2 ubiquitination and degradation. We show here that the deubiquitinase USP25 directly binds to KEAP1 and prevents KEAP1's own ubiquitination and degradation. In the absence of Usp25 or if the DUB is inhibited, KEAP1 is downregulated and NRF2 is stabilized, allowing the cells to respond to oxidative stress more readily. In acetaminophen (APAP) overdose-induced oxidative liver damage in male mice, the inactivation of Usp25, either genetically or pharmacologically, greatly attenuates liver injury and reduces the mortality rates resulted from lethal doses of APAP.
Collapse
Affiliation(s)
- Changzhou Cai
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Huailu Ma
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Jin Peng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Xiang Shen
- Chaser Therapeutics, Inc., Hangzhou, Zhejiang, 310018, China
| | - Xinghua Zhen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Pumin Zhang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Feng Ji
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Jiewei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
27
|
Gao Z, Zhan H, Zong W, Sun M, Linghu L, Wang G, Meng F, Chen M. Salidroside alleviates acetaminophen-induced hepatotoxicity via Sirt1-mediated activation of Akt/Nrf2 pathway and suppression of NF-κB/NLRP3 inflammasome axis. Life Sci 2023:121793. [PMID: 37224954 DOI: 10.1016/j.lfs.2023.121793] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023]
Abstract
Acetaminophen (APAP) overdose-induced hepatotoxicity is the most common cause of drug-induced liver injury worldwide, which is significantly linked to oxidative stress and sterile inflammation. Salidroside is the main active component extracted from Rhodiola rosea L., with anti-oxidative and anti-inflammatory activities. Herein, we investigated the protective effects of salidroside on APAP-induced liver injury and its underlying mechanisms. Pretreatment with salidroside reversed the impacts of APAP on cell viability, LDH release, and cell apoptosis in L02 cells. Moreover, the phenomena of ROS accumulation and MMP collapse caused by APAP were reverted by salidroside. Salidroside elevated the levels of nuclear Nrf2, HO-1, and NQO1. Using PI3k/Akt inhibitor LY294002 further confirmed that salidroside mediated the Nrf2 nuclear translocation through the Akt pathway. Pretreatment with Nrf2 siRNA or LY294002 markedly prevented the anti-apoptotic effect of salidroside. Additionally, salidroside reduced the levels of nuclear NF-κB, NLRP3, ASC, cleaved caspase-1, and mature IL-1β elevated by APAP. Moreover, salidroside pretreatment increased Sirt1 expression, whereas Sirt1 knock-down diminished the protective activities of salidroside, simultaneously reversing the up-regulation of the Akt/Nrf2 pathway and the down-regulation of NF-κB/NLRP3 inflammasome axis mediated by salidroside. We then used C57BL/6 mice to establish APAP-induced liver injury models and found that salidroside significantly alleviated liver injury. Furthermore, western blot analyses showed that salidroside promoted the Sirt1 expression, activated the Akt/Nrf2 pathway, and inhibited the NF-κB/NLRP3 inflammasome axis in APAP-treated mice. The findings of this study support a possible application of salidroside in the amelioration of APAP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Zhengshan Gao
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Honghong Zhan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Wei Zong
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Miaomiao Sun
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Lang Linghu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Guowei Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Fancheng Meng
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Min Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Southwest University, Ministry of Education, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
28
|
Yumimoto K, Sugiyama S, Motomura S, Takahashi D, Nakayama KI. Molecular evolution of Keap1 was essential for adaptation of vertebrates to terrestrial life. SCIENCE ADVANCES 2023; 9:eadg2379. [PMID: 37205751 DOI: 10.1126/sciadv.adg2379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Reactive oxygen species (ROS) posed a risk for the transition of vertebrates from aquatic to terrestrial life. How ancestral organisms adapted to such ROS exposure has remained a mystery. Here, we show that attenuation of the activity of the ubiquitin ligase CRL3Keap1 for the transcription factor Nrf2 during evolution was key to development of an efficient response to ROS exposure. The Keap1 gene was duplicated in fish to give rise to Keap1A and the only remaining mammalian paralog Keap1B, the latter of which shows a lower affinity for Cul3 and contributes to robust Nrf2 induction in response to ROS exposure. Mutation of mammalian Keap1 to resemble zebrafish Keap1A resulted in an attenuated Nrf2 response, and most knock-in mice expressing such a Keap1 mutant died on exposure as neonates to sunlight-level ultraviolet radiation. Our results suggest that molecular evolution of Keap1 was essential for adaptation to terrestrial life.
Collapse
Affiliation(s)
- Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Saori Motomura
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Daisuke Takahashi
- Department of Protein Structure, Function, and Design, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
29
|
Mišík M, Staudinger M, Kundi M, Worel N, Nersesyan A, Ferk F, Dusinska M, Azqueta A, Møller P, Knasmueller S. Use of the Single Cell Gel Electrophoresis Assay for the Detection of DNA-protective Dietary Factors: Results of Human Intervention Studies. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2023; 791:108458. [PMID: 37031732 DOI: 10.1016/j.mrrev.2023.108458] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/14/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
The single cell gel electrophoresis technique is based on the measurement of DNA migration in an electric field and enables to investigate via determination of DNA-damage the impact of foods and their constituents on the genetic stability. DNA-damage leads to adverse effects including cancer, neurodegenerative disorders and infertility. In the last 25 years approximately 90 human intervention trials have been published in which DNA-damage, formation of oxidized bases, alterations of the sensitivity towards reactive oxygen species and chemicals and of repair functions were investigated with this technique. In approximately 50% of the studies protective effects were observed. Pronounced protection was found with certain plant foods (spinach, kiwi fruits, onions), coffee, green tea, honey and olive oil. Also diets with increased contents of vegetables caused positive effects. Small amounts of certain phenolics (gallic acid, xanthohumol) prevented oxidative damage of DNA; with antioxidant vitamins and cholecalciferol protective effects were only detected after intake of doses that exceed the recommended daily uptake values. The evaluation of the quality of the studies showed that many have methodological shortcomings (lack of controls, no calibration of repair enzymes, inadequate control of the compliance and statistical analyses) which should be avoided in future investigations.
Collapse
Affiliation(s)
- Miroslav Mišík
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Marlen Staudinger
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Michael Kundi
- Center for Public Health, Department of Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Nadine Worel
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Armen Nersesyan
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Franziska Ferk
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry, NILU-Norwegian Institute for Air Research, Instituttveien 18, 2002 Kjeller, Norway
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona, Spain
| | - Peter Møller
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Denmark
| | - Siegfried Knasmueller
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A 1090 Vienna, Austria.
| |
Collapse
|
30
|
Roberts JA, Rainbow RD, Sharma P. Mitigation of Cardiovascular Disease and Toxicity through NRF2 Signalling. Int J Mol Sci 2023; 24:ijms24076723. [PMID: 37047696 PMCID: PMC10094784 DOI: 10.3390/ijms24076723] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Cardiovascular toxicity and diseases are phenomena that have a vastly detrimental impact on morbidity and mortality. The pathophysiology driving the development of these conditions is multifactorial but commonly includes the perturbance of reactive oxygen species (ROS) signalling, iron homeostasis and mitochondrial bioenergetics. The transcription factor nuclear factor erythroid 2 (NFE2)-related factor 2 (NRF2), a master regulator of cytoprotective responses, drives the expression of genes that provide resistance to oxidative, electrophilic and xenobiotic stresses. Recent research has suggested that stimulation of the NRF2 signalling pathway can alleviate cardiotoxicity and hallmarks of cardiovascular disease progression. However, dysregulation of NRF2 dynamic responses can be severely impacted by ageing processes and off-target toxicity from clinical medicines including anthracycline chemotherapeutics, rendering cells of the cardiovascular system susceptible to toxicity and subsequent tissue dysfunction. This review addresses the current understanding of NRF2 mechanisms under homeostatic and cardiovascular pathophysiological conditions within the context of wider implications for this diverse transcription factor.
Collapse
Affiliation(s)
- James A. Roberts
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Richard D. Rainbow
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| | - Parveen Sharma
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| |
Collapse
|
31
|
Xu Y, Peng T, Zhou Q, Zhu J, Liao G, Zou F, Meng X. Evaluation of the oxidative toxicity induced by lead, manganese, and cadmium using genetically modified nrf2a-mutant zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2023; 266:109550. [PMID: 36717045 DOI: 10.1016/j.cbpc.2023.109550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/03/2023] [Accepted: 01/15/2023] [Indexed: 01/30/2023]
Abstract
Heavy metal pollution has become a serious environmental concern and a threat to public health. Three of the most common heavy metals are cadmium (Cd), lead (Pb), and manganese (Mn). Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important transcription factor activated in the response to oxidative stress. In this study, mutant zebrafish with an nrf2a deletion of 7 bp were constructed by the CRISPR/Cas9 system to investigate the oxidative toxicity of these three heavy metals. The results of general toxicity tests showed that Pb exposure did not cause significant damage to mutant zebrafish compared with wild-type (WT) zebrafish. However, high Mn exposure increased mortality and malformation rates in mutant zebrafish. Of concern, Cd exposure caused significant toxic damage, including increased mortality and malformation rates, apoptosis of brain neurons, and severe locomotor behavior aberration in mutant zebrafish. The results of qRT-PCR indicated that Cd exposure could induce the activation of genes related to oxidative stress resistance in WT zebrafish, while the expression of these genes was inhibited in mutant zebrafish. This study showed that of the three heavy metals, Cd had the strongest oxidative toxicity, Mn had medium toxicity, and Pb had the weakest toxicity.
Collapse
Affiliation(s)
- Yongjie Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Peng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Qin Zhou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zhu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Gengze Liao
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
32
|
Abstract
Significance: Central nervous system (CNS) diseases are disorders of the brain and/or spinal cord and include neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor belonging to the cap-n-collar family that harbors a unique basic leucine zipper motif and plays as a master regulator of homeostatic responses. Recent Advances: Kelch-like ECH-associated protein 1 (KEAP1) is an adaptor of the Cullin3 (CUL3)-based ubiquitin E3 ligase that enhances the ubiquitylation of NRF2, which promotes the degradation of NRF2 to suppress its transcriptional activity in the absence of stress. Cysteine residues of KEAP1 are modified under stress conditions, and NRF2 degradation is attenuated, allowing it to accumulate and induce the expression of target genes. This regulatory system is referred to as the KEAP1-NRF2 system and plays a central role in protecting cells against various stresses. NRF2 also negatively regulates the expression of inflammatory cytokine and chemokine genes and suppresses pathological inflammation. As oxidative stress, inflammation, and proteostasis are known to contribute to neurodegenerative diseases, the KEAP1-NRF2 system is an attractive target for the treatment of these diseases. Critical Issues: In mouse models of neurodegenerative diseases, Nrf2 depletion exacerbates symptoms and enhances oxidative damage and inflammation in the CNS. In contrast, chemical or genetic NRF2 activation improves these symptoms. Indeed, the NRF2-activating chemical dimethyl fumarate is now widely used for the clinical treatment of MS. Future Directions: The KEAP1-NRF2 system is a promising therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Akira Uruno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
33
|
Sodium butyrate protects against rotavirus-induced intestinal epithelial barrier damage by activating AMPK-Nrf2 signaling pathway in IPEC-J2 cells. Int J Biol Macromol 2023; 228:186-196. [PMID: 36565836 DOI: 10.1016/j.ijbiomac.2022.12.219] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Rotavirus (RV) mainly infects intestinal epithelial cells, which leads to diarrhea in newborn piglets with dysfunction in the intestinal mucosal mechanical barrier. Sodium butyrate (SB) is one of the metabolites excreted by gut microbes. However, the protective effect of SB on RV infection induced intestinal mucosal mechanical barrier injury and its potential mechanism has not been well elucidated. In the present study, IPEC-J2 cells with RV infection was a model of intestinal mucosal mechanical barrier injury. Our results demonstrated that the appropriate concentration of SB can effectively alleviate TJ structural damage and up-regulating the expression of TJ-related genes. Furthermore, the appropriate concentration of SB can effectively reverse the increase of intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) level induced by RV infection. Meanwhile, the levels of antioxidant enzymes superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-px) and antioxidant proteins NAD(P)H dehydrogenase quinone 1 (NQO1) and heme oxygenase-1 (HO-1) were increased through SB treatment. In addition, we found that SB increased cellular antioxidant capacity by activating the adenosine monophosphate-activated protein kinase (AMPK)-nuclear factor erythroid 2-related factor (Nrf2) signaling pathway and the cytoprotective effect of SB is limited by GPR109A siRNA. Thus, our findings revealed that SB reduces oxidative stress caused by RV infection and restores the intestinal mucosal mechanical barrier function by activating the AMPK-Nrf2 signal pathway mediated by the receptor GPR109A.
Collapse
|
34
|
Uruno A, Yamamoto M. The KEAP1-NRF2 system and neurodegenerative diseases. Antioxid Redox Signal 2023. [PMID: 36734430 DOI: 10.1089/ars.2023.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Significance: Central nervous system (CNS) diseases are disorders of the brain and/or spinal cord and include neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). NF-E2-related factor 2 (NRF2) is a transcription factor belonging to the cap-n-collar (CNC) family that harbors a unique basic leucine zipper motif and plays as a master regulator of homeostatic responses. Recent Advances: Kelch-like ECH-associated protein 1 (KEAP1) is an adaptor of the Cullin3 (CUL3)-based ubiquitin E3 ligase that enhances the ubiquitylation of NRF2, which promotes the degradation of NRF2 to suppress its transcriptional activity in the absence of stress. Cysteine residues of KEAP1 are modified under stress conditions, and NRF2 degradation is attenuated, allowing it to accumulate and induce the expression of target genes. This regulatory system is referred to as the KEAP1-NRF2 system and plays a central role in protecting cells against various stresses. NRF2 also negatively regulates the expression of inflammatory cytokine and chemokine genes and suppresses pathological inflammation. As oxidative stress, inflammation, and proteostasis are known to contribute to neurodegenerative diseases, the KEAP1-NRF2 system is an attractive target for the treatment of these diseases. Critical Issues: In mouse models of neurodegenerative diseases, Nrf2 depletion exacerbates symptoms and enhances oxidative damage and inflammation in the CNS. In contrast, chemical or genetic NRF2 activation improves these symptoms. Indeed, the NRF2-activating chemical dimethyl fumarate (DMF) is now widely used for the clinical treatment of MS. Future Directions: The KEAP1-NRF2 system is a promising therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Akira Uruno
- Tohoku University, 13101, 2-1 Seiryo-cho, Aoba-ku, Sendai, Sendai, Miyagi, Japan, 980-8577;
| | - Masayuki Yamamoto
- Tohoku University Graduate School of Medicine, Department of Medical Biochemistry, 2-1 Seiryo-machi, Aoba-ku, Sendai, Sendai, Japan, 980-8575;
| |
Collapse
|
35
|
Xu W, Liu Y, Liu Q, Chen H, Lei L, Shen X, Liu L. Procyanidins Ameliorate Acetaminophen-induced Acute Liver Injury via Activating the Nrf-2/SOD-1 Signal Pathway. Pharmacogn Mag 2023. [DOI: 10.1177/09731296221144812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background and Objectives An overdose of acetaminophen (APAP) usually leads to acute liver injury, and oxidative stress is one of the fundamental mechanisms used to characterize it. Procyanidins (PCs) can reduce the oxidative stress in the liver of mice. This study aimed to investigate the potential protective role of PCs against APAP-induced acute liver injury. Materials and Methods Experiments were performed on male Kunming mice in six groups: phosphate-buffered saline, PCs, APAP, and PCs pretreated with 10, 50, and 100 mg/kg. The mice were peritoneally injected with PCs 30 min before the administration of APAP. First, survival rates of mice were scored every 12 hr for three days in succession. Furthermore, serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (T-Bil), total cholesterol (TC), triglyceride (TG), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-1 (IL-6) were determined. Additionally, histological analysis and hepatic oxidative stress including the levels of superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione (GSH) were assessed. Finally, the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and SOD-1 was detected by Western blotting. Results The data indicated that PCs improved survival rates of APAP-induced liver injury in mice models. Moreover, PCs could reduce the elevated serum levels of ALT, AST, T-Bil, TC, TG, TNF-α, IL-1β, and IL-6 due to APAP exposure with a dose-dependent manner. Besides, PCs pretreatment attenuated hepatic histopathological damage and oxidative stress which manifested the increases of SOD and GSH, whereas the decrease of MDA. Furthermore, PCs enhanced the protein expression of Nrf2 and SOD-1 in the PCs pretreatment groups compared with the APAP group. Conclusion PCs ameliorated APAP-induced acute liver injury, and Nrf2 signaling pathway modulating antioxidative stress might be involved in it.
Collapse
Affiliation(s)
- Wanting Xu
- Department of Pediatrics, Chengdu Second People’s Hospital, Chengdu, People’s Republic of China
| | - Yan Liu
- Department of Pediatrics, Chengdu Second People’s Hospital, Chengdu, People’s Republic of China
| | - Qun Liu
- Department of Pediatrics, Chengdu Second People’s Hospital, Chengdu, People’s Republic of China
| | - Huan Chen
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Langhuan Lei
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xiaojia Shen
- Department of Pediatrics, Chengdu Second People’s Hospital, Chengdu, People’s Republic of China
| | - Li Liu
- Department of Pediatrics, Chengdu Second People’s Hospital, Chengdu, People’s Republic of China
| |
Collapse
|
36
|
Jiang H, Shang Z, You L, Zhang J, Jiao J, Qian Y, Lin J, Wang F, Gao Y, Kong X, Sun X. Electroacupuncture Pretreatment at Zusanli (ST36) Ameliorates Hepatic Ischemia/Reperfusion Injury in Mice by Reducing Oxidative Stress via Activating Vagus Nerve-Dependent Nrf2 Pathway. J Inflamm Res 2023; 16:1595-1610. [PMID: 37092126 PMCID: PMC10120822 DOI: 10.2147/jir.s404087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Background and Purpose Current pharmacological approaches to prevent hepatic ischemia/reperfusion injury (IRI) are limited. To mitigate hepatic injury, more research is needed to improve the understanding of hepatic IRI. Depending on traditional Chinese medicine (TCM) theory, acupuncture therapy has been used for the treatment of ischemic diseases with good efficacy. However, the efficacy and mechanism of acupuncture for hepatic IRI are still unclear. Methods Blood provided to the left and middle lobe of mice livers was blocked with a non-invasive clamp and then the clamps were removed for reperfusion to establish a liver IRI model. Quantitative proteomics approach was used to evaluate the impact of EA pretreatment on liver tissue proteome in the IRI group. Serum biochemistry was used to detect liver injury, inflammation, and oxidative stress levels. H&E staining and TUNEL staining were used to detect hepatocyte injury and apoptosis. Immunohistochemistry and ELISA were used to detect the degree of inflammatory cell infiltration and the level of inflammation. The anti-inflammatory and antioxidant capacities were detected by Quantitative RT-PCR and Western blotting. Results We found that EA at Zusanli (ST36) has a protective effect on hepatic IRI in mice by alleviating oxidative stress, hepatocyte death, and inflammation response. Nuclear factor E2-related factor 2 (Nrf2) as a crucial target was regulated by EA and was then successfully validated. The Nrf2 inhibitor ML385 and cervical vagotomy eliminated the protective effect in the EA treatment group. Conclusion This study firstly demonstrated that EA pretreatment at ST36 significantly ameliorates hepatic IRI in mice by inhibiting oxidative stress via activating the Nrf2 signal pathway, which was vagus nerve-dependent.
Collapse
Affiliation(s)
- Haochen Jiang
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhi Shang
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Liping You
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jinghao Zhang
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Junzhe Jiao
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yihan Qian
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jiacheng Lin
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Fang Wang
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yueqiu Gao
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Xiaoni Kong, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China, Email
| | - Xuehua Sun
- Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Correspondence: Xuehua Sun, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China, Email
| |
Collapse
|
37
|
MafG-like contribute to copper and cadmium induced antioxidant response by regulating antioxidant enzyme in Procambarus clarkii. Gene 2022; 847:146848. [PMID: 36096331 DOI: 10.1016/j.gene.2022.146848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/11/2022] [Accepted: 08/25/2022] [Indexed: 11/23/2022]
Abstract
Avian musculoaponeurotic fibrosarcoma (Maf) proteins play an important role in Nrf2/Keap1 signaling pathway, which mainly resist the oxidant stress. The members of sMaf have a high homology basic leucine zipper (bZIP) and lack trans activation domain, and could interact with other transcriptional regulatory factors as a molecular chaperone. In this study, a full-length MafG-like gene was cloned from Procambarus Clarkii, designated as PcMafG-like, which consisted of an ORF length of 246 bp encoding 82 amino acids, a 5' untranslated region (UTR) of 483 bp, and a 3' UTR of 111 bp. The domain of PcMafG-like had a bZIP-Maf domain that binds to DNA. The cDNA sequence of PcMafG-like was 99 % similar to that of Penaeus vannamei. The mRNA of PcMafG-like was expressed in all tested tissues, and the highest expression was in muscle tissue. Under stimulation of Cu2+ and Cd2+, PcMafG-like was significantly up-regulated in hepatopancreas and gill, and the same result was testified by situ hybridization. The representative antioxidant genes, CAT, GPx and CZ-SOD, were significantly induced by Cu2+; CAT and GPx was induced by Cd2+. PcMafG-dsRNA significantly inhibited the expression of these up-regulated genes, but also inhibited the expression of other detected genes CZ-SOD, GST-θ and GST-1like. The antioxidant effect of PcMafG-like was further verified by oxidative stress markers (T-SOD, CuZnSOD, GPx, CAT, GSH and MDA) kits. Cu2+ and Cd2+ could induce the contents of these oxidative stress markers (MDA, GSH, CZ-SOD, CAT in Cu2+/Cd2+ treated group, and GSH-Px in Cd2+ group), while interference of PcMafG-like significantly inhibited the up-regulation. Furthermore, hematoxylin-eosin staining experiments showed that the degree of pathological damage was dose-dependent and time-dependent, and the pathological damage was more serious after dsRNA interfered with PcMafG-like. In addition, subcellular localization showed that PcMafG-like gene existed in nucleus. The recombinant protein PcMafG-like was expressed and purified in prokaryotic expression. The affinity analysis of promoter by agarose gel electrophoresis suggested that PcMafG-like could bind with CAT promoter in vitro. This indicated that PcMafG-like could activate antioxidant genes.
Collapse
|
38
|
Puhari SSM, Yuvaraj S, Vasudevan V, Ramprasath T, Rajkumar P, Arunkumar K, Amutha C, Selvam GS. Isolation and characterization of fucoidan from four brown algae and study of the cardioprotective effect of fucoidan from Sargassum wightii against high glucose-induced oxidative stress in H9c2 cardiomyoblast cells. J Food Biochem 2022; 46:e14412. [PMID: 36121745 DOI: 10.1111/jfbc.14412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/29/2022] [Accepted: 08/17/2022] [Indexed: 01/13/2023]
Abstract
Oxidative stress plays a vital role in the initiation and progression of diabetic cardiomyopathy (DCM). Increased cardiac dysfunction and apoptosis in DCM are independent factors associated with hypertension or coronary artery disease. Fucoidan, a class of sulfated polysaccharides, is widely used as food supplements and reported to have various pharmacological properties. However, the pharmacological property of Indian seaweeds remains unexplored. The present study is focused on isolating and characterizing the fucoidan from four brown seaweeds such as Sargassum wightii (SwF), Sargassum swartzii (SsF), Sargassum polycystum (SpF), Turbinaria ornata (ToF), and aimed to investigate cardioprotective effect of fucoidan against High Glucose (HG) induced oxidative stress in H9c2 cells. The mild acid hydrolysis method was used to isolate crude fucoidan from four brown seaweeds purified by the FPLC system. The biochemical composition analysis showed that SwF had a high content of fucoidan and sulfate, followed by SsF, SpF, and ToF. Further, FTIR, XRD, NMR, and SEM analysis confirmed the isolated fucoidan structures. SwF showed higher DPPH activity compared to another isolated fucoidan. In vitro studies with SwF revealed significantly decreased cytotoxicity, prevented the loss of MMP, reduced lipid peroxidation, and increased cellular enzymatic and non-enzymatic activity. qRT-PCR results showed SwF significantly upregulated the Nrf2, HO-1, NQO1, and Bcl2 and down-regulated the Bax and Caspase-3 mRNA expression compared to HG-treated cells. In conclusion, SwF could be used to develop functional foods for diabetic-mediated CVD complications compared to another isolated fucoidan. PRACTICAL APPLICATIONS: Bioactive carbohydrates have gained significant interest among researchers to improve human health. The biomedical field showed great interest in seaweed research in managing various diseases. In particular, seaweeds contain many bioactive compounds because of their chemical and biological diversity. Despite the various beneficial effects of fucoidan in CVD, the therapeutic potential of Indian seaweeds remains largely unexplored. Hence, this study isolated fucoidan from four brown seaweeds and studied their bioactive properties. Results revealed that SwF showed higher free radical scavenging activity compared to another isolated fucoidan. Therefore, SwF was selected for the in vitro study. SwF increased the cytoprotection through increasing antioxidant levels against oxidative stress in H9c2 cells. Staining analysis showed SwF increased cellular protection via inhibiting ROS protection and increasing MMP. Overall, fucoidan from SwF could be developed as a functional food for CVD.
Collapse
Affiliation(s)
- Shanavas Syed Mohamed Puhari
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Subramani Yuvaraj
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Varadaraj Vasudevan
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Tharmarajan Ramprasath
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Prabhakaran Rajkumar
- Department of Animal Sciences, Manonmanium Sundaranar University, Tirunelveli, India
| | - Kulanthaiyesu Arunkumar
- Department of Plant Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Chinnaiah Amutha
- Department of Animal Behaviour & Physiology, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Govindan Sadasivam Selvam
- Molecular Cardiology Unit, Department of Biochemistry, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| |
Collapse
|
39
|
Nrf2 and Oxidative Stress: A General Overview of Mechanisms and Implications in Human Disease. Antioxidants (Basel) 2022; 11:antiox11122345. [PMID: 36552553 PMCID: PMC9774434 DOI: 10.3390/antiox11122345] [Citation(s) in RCA: 317] [Impact Index Per Article: 105.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Organisms are continually exposed to exogenous and endogenous sources of reactive oxygen species (ROS) and other oxidants that have both beneficial and deleterious effects on the cell. ROS have important roles in a wide range of physiological processes; however, high ROS levels are associated with oxidative stress and disease progression. Oxidative stress has been implicated in nearly all major human diseases, from neurogenerative diseases and neuropsychiatric disorders to cardiovascular disease, diabetes, and cancer. Antioxidant defence systems have evolved as a means of protection against oxidative stress, with the transcription factor Nrf2 as the key regulator. Nrf2 is responsible for regulating an extensive panel of antioxidant enzymes involved in the detoxification and elimination of oxidative stress and has been extensively studied in the disease contexts. This review aims to provide the reader with a general overview of oxidative stress and Nrf2, including basic mechanisms of Nrf2 activation and regulation, and implications in various major human diseases.
Collapse
|
40
|
Habeos GI, Filippopoulou F, Habeos EE, Kalaitzopoulou E, Skipitari M, Papadea P, Lagoumintzis G, Niarchos A, Georgiou CD, Chartoumpekis DV. Maternal Calorie Restriction Induces a Transcriptional Cytoprotective Response in Embryonic Liver Partially Dependent on Nrf2. Antioxidants (Basel) 2022; 11:2274. [PMID: 36421460 PMCID: PMC9687455 DOI: 10.3390/antiox11112274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Calorie restriction is known to enhance Nrf2 signaling and longevity in adult mice, partially by reducing reactive oxygen species, but calorie restriction during pregnancy leads to intrauterine growth retardation. The latter is associated with fetal reprogramming leading to increased incidence of obesity, metabolic syndrome and diabetes in adult life. Transcription factor Nrf2 is a central regulator of the antioxidant response and its crosstalk with metabolic pathways is emerging. We hypothesized that the Nrf2 pathway is induced in embryos during calorie restriction in pregnant mothers. METHODS From gestational day 10 up to day 16, 50% of the necessary mouse diet was provided to Nrf2 heterozygous pregnant females with fathers being of the same genotype. Embryos were harvested at the end of gestational day 16 and fetal liver was used for qRT-PCR and assessment of oxidative stress (OS). RESULTS Intrauterine calorie restriction led to upregulation of mRNA expression of antioxidant genes (Nqo1, Gsta1, Gsta4) and of genes related to integrated stress response (Chac1, Ddit3) in WT embryos. The expression of a key gluconeogenic (G6pase) and two lipogenic genes (Acacb, Fasn) was repressed in calorie-restricted embryos. In Nrf2 knockout embryos, the induction of Nqo1 and Gsta1 genes was abrogated while that of Gsta4 was preserved, indicating an at least partially Nrf2-dependent induction of antioxidant genes after in utero calorie restriction. Measures of OS showed no difference (superoxide radical and malondialdehyde) or a small decrease (thiobarbituric reactive substances) in calorie-restricted WT embryos. CONCLUSIONS Calorie restriction during pregnancy elicits the transcriptional induction of cytoprotective/antioxidant genes in the fetal liver, which is at least partially Nrf2-dependent, with a physiological significance that warrants further investigation.
Collapse
Affiliation(s)
- George I. Habeos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Fotini Filippopoulou
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Evagelia E. Habeos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Electra Kalaitzopoulou
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Marianna Skipitari
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Polyxeni Papadea
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - George Lagoumintzis
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
- Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - Athanasios Niarchos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Christos D. Georgiou
- Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Dionysios V. Chartoumpekis
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
41
|
Cardamom Extract Alleviates the Oxidative Stress, Inflammation and Apoptosis Induced during Acetaminophen-Induced Hepatic Toxicity via Modulating Nrf2/HO-1/NQO-1 Pathway. Curr Issues Mol Biol 2022; 44:5390-5404. [PMID: 36354677 PMCID: PMC9688982 DOI: 10.3390/cimb44110365] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Acetaminophen (APAP) is the most extensively used and safest analgesic and antipyretic drug worldwide; however, its toxicity is associated with life-threatening acute liver failure. Cardamom (CARD), a sweet, aromatic, commonly used spice, has several pharmacological actions. In the current study, we tried to explore the chemical composition and the hepato-protective effect of ethanolic aqueous extract of CARD to mitigate APAP-induced hepatic toxicity and elucidate its underlying mechanism of action. Material and methods: Aqueous CARD extract was subjected to LC-TOF-MS analysis to separate and elucidate some of its components. In vivo animal experiments involved five groups of animals. In the normal and cardamom groups, mice were administered either saline or CARD (200 mg/kg), respectively, orally daily for 16 days. In the APAP group, the animals were administered saline orally daily for 15 days, and on the 16th day, animals were administered APAP (300 mg/kg) IP for the induction of acute hepatic failure. In the CARD 200 + APAP group, mice were administered CARD (200 mg/kg) for 15 days, followed by APAP on the 16th day. Results: The aqueous extract of CARD showed several compounds, belonging to polyphenol, flavonoids, cinnamic acid derivatives and essential oil components. In the in vivo investigations, APAP-induced impaired liver function, several histopathological alterations, oxidative stress and inflammatory and apoptotic status signified severe hepatic failure. Whereas, pretreatment with the CARD extract prior to APAP administration diminished serum levels of the hepatic function test and augmented Nrf2 nucleoprotein and HO-1 and NQO-1. CARD down-regulated MDA, inflammatory mediators (IL-1β, IL-6, TNF-α and NF-κB) and apoptotic markers (caspase 3 and 9 and Bax) and amplified the activities of SOD, catalase, GSH-Px and GSH-R in hepatic tissue samples. Conclusion: CARD extract mitigated the hepatic toxicity induced by APAP. The underlying mechanism of action of such hepato-protective action may be through upregulation of the Nrf2/HO-1/NQO-1 pathway with subsequent alleviation of the oxidative stress, inflammation and apoptosis induced by APAP. Many of the compounds identified in the CARD extract could be attributed to this pharmacological action of the extract.
Collapse
|
42
|
Yin N, Zhang W, Wei R, Yang Q, He F, Guo L, Feng M. Liposome cocktail activator modulates hepatocytes and remodels the microenvironment to mitigate acute liver failure. Asian J Pharm Sci 2022; 17:867-879. [PMID: 36600898 PMCID: PMC9800940 DOI: 10.1016/j.ajps.2022.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/09/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
Acute liver failure (ALF) is a mortal and critical hepatic disease, in which oxidative stress, inflammation storm and hepatocyte death are crucial in the pathogenesis. Hence, in contrast to the control of a single link, a combination therapy targeting multiple pathogenic links of the disease will be a favorable means to control the progression of the disease. In this study, we constructed dimethyl itaconate-loaded liposomes modified with dodecyl gallate as a cocktail activator to investigate its functional role in acetaminophen (APAP)-induced ALF. Our results demonstrated that the cocktail activator acted on hepatocytes and triggered cocktail efficacy, thereby simultaneously attenuating APAP-induced hepatocyte damage and remodeling the damage microenvironment. The cocktail activator could effectively scavenge reactive oxygen species, inhibit excessive inflammatory responses and reduce cell death in impaired hepatocytes for detoxification. More importantly, the cocktail activator could remodel the damage microenvironment, thus further promoting hepatocyte expansion and specifically switching macrophages from the M1 to M2 phenotype for a favorable liver regeneration of ALF. Furthermore, in APAP-induced ALF mouse model, the cocktail activator improved liver function, alleviated histopathological damage and increased survival rate. In summary, these findings indicate that the cocktail activator may provide a promising therapeutic approach for ALF treatment as a nanomedicine.
Collapse
Affiliation(s)
- Na Yin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Wenjun Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Runxiu Wei
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Qiang Yang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Fengming He
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ling Guo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Corresponding authors.
| | - Min Feng
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
- Corresponding authors.
| |
Collapse
|
43
|
Zhou Z, Wu Y, Hua W, Yan X, Li L, Zhu A, Qi J. Sappanone A ameliorates acetaminophen-induced acute liver injury in mice. Toxicology 2022; 480:153336. [PMID: 36126895 DOI: 10.1016/j.tox.2022.153336] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 10/14/2022]
Abstract
Sappanone A (SA), a homoisoflavonoid compound extracted from the heartwood of Caesalpinia sappan Linn., exerts anti-inflammatory and antioxidant activities. However, the effects of SA on acetaminophen (APAP) overdose-induced acute liver injury (ALI) have not been determined yet. This study aims to explore the protective effects of SA and the potential mechanisms of action. Mice were pretreated with SA (25, 50, and 100 mg/kg) by intraperitoneal (i.p.) injection for seven days prior to APAP (300 mg/kg, i.p.) administration. At 12 h after APAP injection, serum and liver samples were collected. Primary murine hepatocytes were used to investigate the underlying mechanisms. SA pretreatment dose-dependently attenuated APAP-induced ALI, as validated by reduced serum alanine/aspartate aminotransferase levels, histopathologic lesions, and oxidative stress. Consistently, pretreatment with SA reduced the formation of APAP protein adducts in damaged livers of mice. Mechanistically, SA could facilitate the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and thus promote cellular glutathione (GSH) synthesis. The hepatoprotective outcomes provided by SA were significantly abolished by treatment with ML385, a Nrf2 inhibitor. Besides, anti-inflammatory property of SA reduced inflammatory reaction in injured livers of mice. Of note, posttreatment with SA reveals significant therapeutic influences against APAP-induced ALI in mice. Collectively, our findings demonstrated that pretreated-SA ameliorated APAP-mediated ALI in mice, at least in part, by reducing the generation of APAP protein adducts via Nrf2-enhanced GSH synthesis, and by diminishing hepatic inflammation. Therefore, SA could be a potential hepatoprotective agent for treating ALI.
Collapse
Affiliation(s)
- Zixiong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China; Department of Pathology and Institute of Oncology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Yong Wu
- Department of Pathology and Institute of Oncology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Wenxi Hua
- Department of Pathology and Institute of Oncology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Xueqing Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Lanqian Li
- Department of Pathology and Institute of Oncology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - An Zhu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China.
| | - Jing Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, Fujian, China.
| |
Collapse
|
44
|
Amato CM, Fricke A, Marella S, Mogus JP, Bereman M, McCoy KA. An experimental evaluation of the efficacy of perinatal sulforaphane supplementation to decrease the incidence and severity of vinclozolin-induced hypospadias in the mouse model. Toxicol Appl Pharmacol 2022; 451:116177. [PMID: 35905821 PMCID: PMC9450412 DOI: 10.1016/j.taap.2022.116177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022]
Abstract
Determining the mechanisms of toxicity induced by pollutants has long been a research priority in lieu of considering the mechanisms of resilience that prevent deleterious impacts. Protective mechanisms in many taxa can be therapeutically targeted to enhance resilience to synthetic toxicants. For example, the environmental sensor, Nuclear factor (erythroid-derived 2)-like 2 (Nfe2l2 or Nrf2), a transcription factor, facilitates transcription of many protective genes. Hypospadias is a common malformation of the penis. The risk of being born with hypospadias increases with pollutant exposure. We use vinclozolin-induced hypospadias in the mouse as a model to test the hypothesis that pollutant-induced birth defects can be prevented and reduced in severity by augmenting natural mechanisms of resilience. Pregnant mice were exposed to the demasculinizing toxicant, vinclozolin, in combination with increasing doses of the NRF2 activator, sulforaphane. The sulforaphane dose that most effectively increased masculinization (anogenital distance) was identified and used to test the hypothesis that sulforaphane reduces the hypospadias-inducing potency of vinclozolin. Finally, a Nrf2 knockout study was conducted to test whether NRF2 was required for the sulforaphane-induced rescue effects. Sulforaphane supplementation to vinclozolin exposed embryos increased anogenital distance in a nonlinear fashion typical of Nrf2 activators. The most effective dose of sulforaphane (45 mg/kg) reduced the occurrence and severity of vinclozolin-induced hypospadias and corrected penis morphogenesis. The sulforaphane-induced rescue effect was dependent on the presence of Nrf2. Nrf2 plays a critical role in protecting the fetus from vinclozolin and reduces the incidence and severity of hypospadias, the most common birth defect in boys in many countries. This work lays a foundation for developing prenatal supplements that will protect the fetus from pollutant-induced hypospadias. Studying the protective mechanisms that drive resilience to toxicants will facilitate innovation of protective therapies.
Collapse
Affiliation(s)
- Ciro M Amato
- Department of Biology, East Carolina University, Greenville, North Carolina 27858, USA
| | - Ariel Fricke
- Department of Biology, East Carolina University, Greenville, North Carolina 27858, USA
| | - Sahiti Marella
- Department of Biology, East Carolina University, Greenville, North Carolina 27858, USA
| | - Joshua P Mogus
- Department of Biology, East Carolina University, Greenville, North Carolina 27858, USA
| | - Michael Bereman
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| | - Krista A McCoy
- Department of Biology, East Carolina University, Greenville, North Carolina 27858, USA; Harbor Branch Oceanographic Institute, Center for Coastal and Human Health, Florida Atlantic University, Fort Pierce, FL, USA.
| |
Collapse
|
45
|
Molecular mechanisms associated with the chemoprotective role of protocatechuic acid and its potential benefits in the amelioration of doxorubicin-induced cardiotoxicity: A review. Toxicol Rep 2022; 9:1713-1724. [PMID: 36561952 PMCID: PMC9764176 DOI: 10.1016/j.toxrep.2022.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/01/2022] [Accepted: 09/03/2022] [Indexed: 12/25/2022] Open
Abstract
Since its discovery in the 1960 s, doxorubicin (DOX) has constantly elicited the broadest spectrum of cancerocidal activity against human cancers. However, cardiotoxicity caused by DOX directly as well as its metabolites is a great source of concern over the continuous use of DOX in chemotherapy. While the exact mechanism of DOX-induced cardiotoxicity is yet to be completely understood, recent studies indicate oxidative stress, inflammation, and several forms of cell death as key pathogenic mechanisms that underpin the etiology of doxorubicin-induced cardiotoxicity (DIC). Notably, these key mechanistic events are believed to be negatively regulated by 3,4-dihydroxybenzoic acid or protocatechuic acid (PCA)-a plant-based phytochemical with proven anti-oxidant, anti-inflammatory, and anti-apoptotic properties. Here, we review the experimental findings detailing the potential ameliorative effects of PCA under exposure to DOX. We also discuss molecular insights into the pathophysiology of DIC, highlighting the potential intervention points where the use of PCA as a veritable chemoprotective agent may ameliorate DOX-induced cardiotoxicities as well as toxicities due to other anticancer drugs like cisplatin. While we acknowledge that controlled oral administration of PCA during chemotherapy may be insufficient to eliminate all toxicities due to DOX treatment, we propose that the ability of PCA to block oxidative stress, attenuate inflammation, and abrogate several forms of cardiomyocyte cell death underlines its great promise in the amelioration of DIC.
Collapse
|
46
|
Jyothidasan A, Sunny S, Murugesan S, Quiles JM, Challa AK, Dalley B, Cinghu SK, Nanda V, Rajasekaran NS. Transgenic Expression of Nrf2 Induces a Pro-Reductive Stress and Adaptive Cardiac Remodeling in the Mouse. Genes (Basel) 2022; 13:1514. [PMID: 36140682 PMCID: PMC9498410 DOI: 10.3390/genes13091514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Nuclear factor, erythroid 2 like 2 (Nfe2l2 or Nrf2), is a transcription factor that protects cells by maintaining a homeostatic redox state during stress. The constitutive expression of Nrf2 (CaNrf2-TG) was previously shown to be pathological to the heart over time. We tested a hypothesis that the cardiac-specific expression of full length Nrf2 (mNrf2-TG) would moderately increase the basal antioxidant defense, triggering a pro-reductive environment leading to adaptive cardiac remodeling. Transgenic and non-transgenic (NTG) mice at 7−8 months of age were used to analyze the myocardial transcriptome, structure, and function. Next generation sequencing (NGS) for RNA profiling and qPCR-based validation of the NGS data, myocardial redox levels, and imaging (echocardiography) were performed. Transcriptomic analysis revealed that out of 14,665 identified mRNAs, 680 were differently expressed (DEG) in TG hearts. Of 680 DEGs, 429 were upregulated and 251 were downregulated significantly (FC > 2.0, p < 0.05). Gene set enrichment analysis revealed that the top altered pathways were (a) Nrf2 signaling, (b) glutathione metabolism and (c) ROS scavenging. A comparative analysis of the glutathione redox state in the hearts demonstrated significant differences between pro-reductive vs. hyper-reductive conditions (233 ± 36.7 and 380 ± 68.7 vs. 139 ± 8.6 µM/mg protein in mNrf2-TG and CaNrf2-TG vs. NTG). Genes involved in fetal development, hypertrophy, cytoskeletal rearrangement, histone deacetylases (HDACs), and GATA transcription factors were moderately increased in mNrf2-TG compared to CaNrf2-TG. Non-invasive echocardiography analysis revealed an increase in systolic function (ejection fraction) in mNrf2-TG, suggesting an adaptation, as opposed to pathological remodeling in CaNrf2-TG mice experiencing a hyper-reductive stress, leading to reduced survival (40% at 60 weeks). The effects of excess Nrf2-driven antioxidant transcriptome revealed a pro-reductive condition in the myocardium leading to an adaptive cardiac remodeling. While pre-conditioning the myocardial redox with excess antioxidants (i.e., pro-reductive state) could be beneficial against oxidative stress, a chronic pro-reductive environment in the myocardium might transition the adaptation to pathological remodeling.
Collapse
Affiliation(s)
- Arun Jyothidasan
- Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sini Sunny
- Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Saravanakumar Murugesan
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Justin M. Quiles
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA 92093, USA
| | - Anil Kumar Challa
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Brian Dalley
- Huntsman Cancer Center-Genomic Core Facility, University of Utah, Salt Lake City, UT 84112, USA
| | - Senthil Kumar Cinghu
- Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Vivek Nanda
- Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Namakkal-Soorappan Rajasekaran
- Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Center for Free Radical Biology (CFRB), University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
47
|
Bathish B, Robertson H, Dillon JF, Dinkova-Kostova AT, Hayes JD. Nonalcoholic steatohepatitis and mechanisms by which it is ameliorated by activation of the CNC-bZIP transcription factor Nrf2. Free Radic Biol Med 2022; 188:221-261. [PMID: 35728768 DOI: 10.1016/j.freeradbiomed.2022.06.226] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/11/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) represents a global health concern. It is characterised by fatty liver, hepatocyte cell death and inflammation, which are associated with lipotoxicity, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, iron overload and oxidative stress. NF-E2 p45-related factor 2 (Nrf2) is a transcription factor that combats oxidative stress. Remarkably, Nrf2 is downregulated during the development of NASH, which probably accelerates disease, whereas in pre-clinical studies the upregulation of Nrf2 inhibits NASH. We now review the scientific literature that proposes Nrf2 downregulation during NASH involves its increased ubiquitylation and proteasomal degradation, mediated by Kelch-like ECH-associated protein 1 (Keap1) and/or β-transducin repeat-containing protein (β-TrCP) and/or HMG-CoA reductase degradation protein 1 (Hrd1, also called synoviolin (SYVN1)). Additionally, downregulation of Nrf2-mediated transcription during NASH may involve diminished recruitment of coactivators by Nrf2, due to increased levels of activating transcription factor 3 (ATF3) and nuclear factor-kappaB (NF-κB) p65, or competition for promoter binding due to upregulation of BTB and CNC homology 1 (Bach1). Many processes that downregulate Nrf2 are triggered by transforming growth factor-beta (TGF-β), with oxidative stress amplifying its signalling. Oxidative stress may also increase suppression of Nrf2 by β-TrCP through facilitating formation of the DSGIS-containing phosphodegron in Nrf2 by glycogen synthase kinase-3. In animal models, knockout of Nrf2 increases susceptibility to NASH, while pharmacological activation of Nrf2 by inducing agents that target Keap1 inhibits development of NASH. These inducing agents probably counter Nrf2 downregulation affected by β-TrCP, Hrd1/SYVN1, ATF3, NF-κB p65 and Bach1, by suppressing oxidative stress. Activation of Nrf2 is also likely to inhibit NASH by ameliorating lipotoxicity, inflammation, ER stress and iron overload. Crucially, pharmacological activation of Nrf2 in mice in which NASH has already been established supresses liver steatosis and inflammation. There is therefore compelling evidence that pharmacological activation of Nrf2 provides a comprehensive multipronged strategy to treat NASH.
Collapse
Affiliation(s)
- Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Holly Robertson
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK; Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - John F Dillon
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK.
| |
Collapse
|
48
|
Khodakarami A, Adibfar S, Karpisheh V, Abolhasani S, Jalali P, Mohammadi H, Gholizadeh Navashenaq J, Hojjat-Farsangi M, Jadidi-Niaragh F. The molecular biology and therapeutic potential of Nrf2 in leukemia. Cancer Cell Int 2022; 22:241. [PMID: 35906617 PMCID: PMC9336077 DOI: 10.1186/s12935-022-02660-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 07/19/2022] [Indexed: 02/07/2023] Open
Abstract
NF-E2-related factor 2 (Nrf2) transcription factor has contradictory roles in cancer, which can act as a tumor suppressor or a proto-oncogene in different cell conditions (depending on the cell type and the conditions of the cell environment). Nrf2 pathway regulates several cellular processes, including signaling, energy metabolism, autophagy, inflammation, redox homeostasis, and antioxidant regulation. As a result, it plays a crucial role in cell survival. Conversely, Nrf2 protects cancerous cells from apoptosis and increases proliferation, angiogenesis, and metastasis. It promotes resistance to chemotherapy and radiotherapy in various solid tumors and hematological malignancies, so we want to elucidate the role of Nrf2 in cancer and the positive point of its targeting. Also, in the past few years, many studies have shown that Nrf2 protects cancer cells, especially leukemic cells, from the effects of chemotherapeutic drugs. The present paper summarizes these studies to scrutinize whether targeting Nrf2 combined with chemotherapy would be a therapeutic approach for leukemia treatment. Also, we discussed how Nrf2 and NF-κB work together to control the cellular redox pathway. The role of these two factors in inflammation (antagonistic) and leukemia (synergistic) is also summarized.
Collapse
Affiliation(s)
- Atefeh Khodakarami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Adibfar
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Abolhasani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Kitamura H, Takeda H, Motohashi H. Genetic, Metabolic and Immunological Features of Cancers with NRF2 Addiction. FEBS Lett 2022; 596:1981-1993. [PMID: 35899372 DOI: 10.1002/1873-3468.14458] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/18/2022] [Indexed: 11/10/2022]
Abstract
Nuclear factor erythroid-derived 2-like 2 (NRF2) is a master transcription factor that coordinately regulates the expression of many cytoprotective genes and plays a central role in defense mechanisms against oxidative and electrophilic insults. Although increased NRF2 activity is principally beneficial for our health, NRF2 activation in cancer cells is detrimental. Many human cancers exhibit persistent NRF2 activation and such cancer cells rely on NRF2 for most of their malignant characteristics, such as therapeutic resistance and aggressive tumorigenesis, and thus fall into NRF2 addiction. The persistent activation of NRF2 confers great advantages on cancer cells, whereas it is not tolerated by normal cells, suggesting that certain requirements are necessary for a cell to exploit NRF2 and evolve into malignant a cancer cell. In this review, recent reports and data on the genetic, metabolic and immunological features of NRF2-activated cancer cells are summarized, and prerequisites for NRF2 addiction in cancer cells and their therapeutic applications are discussed.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Haruna Takeda
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|
50
|
Wang Z, Ma J, He Y, Miu KK, Yao S, Tang C, Ye Y, Lin G. Nrf2-mediated liver protection by 18β-glycyrrhetinic acid against pyrrolizidine alkaloid-induced toxicity through PI3K/Akt/GSK3β pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154162. [PMID: 35598524 DOI: 10.1016/j.phymed.2022.154162] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/22/2022] [Accepted: 05/08/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Misusage of pyrrolizidine alkaloid (PA)-containing plants or unaware intake of PA-contaminated foodstuffs causes thousands of PA poisoning cases in humans. PA intoxication is accompanied by oxidative stress and subsequent extensive hepatocellular damage. Our previous study has demonstrated that 18β-glycyrrhetinic acid (GA), a bioactive constituent of liquorice, prevented PA-induced hepatotoxicity in rats, however the underlying mechanisms remain unclear. OBJECTIVE This study aims to explore the mechanisms underlying the hepato-protective effect of GA in combating retrorsine (RTS, a representative toxic PA)-induced liver injury. METHODS Histological and biochemical assessments were employed to evaluate the protective effect of GA on RTS-induced hepatotoxicity in rats. Sulforhodamine B assay, real-time PCR, western blotting, and immunostaining were used to explore the underlying mechanisms in human hepatocytes and rats. RESULTS Our findings demonstrated that GA alleviated RTS-induced elevation of serum ALT and bilirubin levels, as well as hepatocytes necrosis and sinusoidal endothelial cells (SECs) damage in rats. GA also enhanced the activities and expressions of several antioxidant enzymes through upregulating nuclear factor-erythroid 2-related factor2 (Nrf2). Moreover, inhibition of Nrf2 blocked the hepatoprotective effect of GA against RTS intoxication. Mechanistically, GA increased the phosphorylation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and enhanced glycogen synthase kinase 3 beta (GSK3β) inhibitory phosphorylation at serine 9, thus promoting the nuclear accumulation of Nrf2 and activating its downstream targets. CONCLUSION This study for the first time demonstrated that GA exerted protective effects against RTS-induced liver injury by potentiating the Nrf2-mediated antioxidant system through PI3K/Akt/GSK3β pathway. The findings indicated that GA may serve as a potential candidate drug for the treatment of PA intoxication.
Collapse
Affiliation(s)
- Zhangting Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 505A, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Hong Kong SAR, China
| | - Jiang Ma
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 505A, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Hong Kong SAR, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yisheng He
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 505A, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Hong Kong SAR, China
| | - Kai-Kei Miu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 505A, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Hong Kong SAR, China
| | - Sheng Yao
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chunping Tang
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Ye
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 505A, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Hong Kong SAR, China.
| |
Collapse
|