1
|
He M, Wang H, Fu J, Ruan J, Li F, Liang X, Wei L. Oxidative stress and mitochondrial dysfunctions induced by cyanobacterial microcystin-LR in primary grass carp hepatocytes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 282:107327. [PMID: 40121739 DOI: 10.1016/j.aquatox.2025.107327] [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: 12/25/2024] [Revised: 03/03/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Microcystin-LR (MC-LR), a cyclic heptapeptide produced by freshwater cyanobacteria, induces a range of liver injuries. However, the mechanisms underlying MC-LR toxicity in primary hepatocytes of aquatic organisms remains poorly understood. In this study, we investigated the effects of MC-LR on oxidative stress and mitochondrial function using primarily cultured grass carp hepatocytes. The results revealed that IC50 of MC-LR on grass carp primary liver cells for 24 hours was 2.40 μmol/L. Based on 24h-IC50, concentrations of 0, 0.30, 0.60, and 1.20 μmol/L were used in subsequent experiments. MC-LR exposure led to a significant reduction in cell viability, induced abnormal cell morphology, and caused plasma membrane rupture, as indicated by elevated LDH activity in a concentration-dependent manner. Additionally, MC-LR exposure induced oxidative stress, resulting in increased ROS levels and downregulation of genes associated with oxidative stress, including keap1, nrf2, cat, sod1, gpx, gst, and gr (P<0.05). Furthermore, the electron microscopy results showed that MC-LR caused damage to the ultrastructure of primary hepatocytes, including mitochondrial membrane rupture, vacuolation, and induction of mitochondrial autophagy. Moreover, MC-LR exposure elevated intracellular Ca2+ concentration, reduced MMP and ATP levels, and inhibited mitochondrial respiratory chain complex I activity (P<0.05). qRT-PCR analysis demonstrated that MC-LR treatment significantly decreased the transcriptional levels of genes related to mitochondrial quality control including pgc-1α, tfam, nrf1, drp1, opa1, mfn1, and mfn2 (P<0.05). Collectively, our findings highlight that MC-LR causes oxidative stress and impairs mitochondrial function, leading to further hepatocyte damage, which provides insights into the mechanisms of MC-LR-induced hepatotoxicity and offers valuable references for further investigations.
Collapse
Affiliation(s)
- Miao He
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province 330045, PR China.
| | - Hui Wang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province 330045, PR China
| | - Jianping Fu
- College of life sciences, Jiangxi Normal University, Nanchang, Jiangxi Province 330022, PR China
| | - Jiming Ruan
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province 330045, PR China
| | - Fugui Li
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province 330045, PR China
| | - Ximei Liang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province 330045, PR China
| | - Lili Wei
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province 330045, PR China.
| |
Collapse
|
2
|
Manavi MA, Mohammad Jafari R, Shafaroodi H, Dehpour AR. The Keap1/Nrf2/ARE/HO-1 axis in epilepsy: Crosstalk between oxidative stress and neuroinflammation. Int Immunopharmacol 2025; 153:114304. [PMID: 40117806 DOI: 10.1016/j.intimp.2025.114304] [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: 10/08/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Epilepsy is a complex neurological disorder characterized by recurrent seizures, which are driven by multifaceted pathophysiological mechanisms, including oxidative stress and neuroinflammation. Despite advancements in anti-seizure medications (ASMs), a significant proportion of patients remain resistant to treatment, highlighting the need for novel therapeutic strategies. This review focuses on the Kelch-like ECH-associated protein 1 (Keap1) / Nuclear factor erythroid 2-related factor 2 (Nrf2) / Antioxidant Response Element (ARE) / Heme Oxygenase-1 (HO-1) axis as a promising target for neuroprotection in epilepsy. We explored the intricate interactions between Keap1 and Nrf2 under homeostatic conditions and how oxidative stress disrupts this balance, triggering Nrf2 activation. This review details the subsequent process of Nrf2 nuclear translocation, its binding to AREs, and the induction of cytoprotective gene expression, which collectively orchestrate a robust cellular defense response. Special emphasis is placed on HO-1, a key effector of Nrf2-mediated neuroprotection, highlighting its enzymatic function and protective mechanisms, including antioxidant, anti-inflammatory, and anti-apoptotic effects. Additionally, the review examines HO-1's role in mitigating seizure-induced neuronal damage. However, challenges remain, including variability in therapeutic responses, gaps in long-term clinical validation, and the need for standardized protocols. Future research should focus on biomarkers for personalized treatment, advanced imaging, and genetic tools to explore the Keap1/Nrf2/ARE/HO-1 axis in greater depth. Future studies should focus on overcoming the challenges of translating preclinical findings into clinical applications and exploring the long-term effects of targeting this pathway in epilepsy treatment.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran university of medical sciences, Tehran, Iran; Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Razieh Mohammad Jafari
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran
| | - Hamed Shafaroodi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran.
| |
Collapse
|
3
|
Zhao Z, Lu H, Wang J, Wu T, Xu S, Ge Y, You Q, Jiang Z, Lu M. Discovery of β-amino acid substituted naphthalene sulfonamide derivatives as potent Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2) protein-protein interaction inhibitors for ulcerative colitis management. Eur J Med Chem 2025; 288:117384. [PMID: 39965408 DOI: 10.1016/j.ejmech.2025.117384] [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: 12/20/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 02/20/2025]
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a key regulator of cellular defense system against oxidative insults. Directly inhibiting the Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 protein-protein interaction (PPI) has emerged as a promising approach to activate Nrf2 for the treatment of diseases associated with oxidative stress. Herein, we identified β-amino acids as privileged structural fragments for designing novel naphthalene sulfonamide-based Keap1-Nrf2 PPI inhibitors. Comprehensive structure-activity relationship (SAR) exploration identified compound 19 as the optimal inhibitor with an IC50 of 0.55 μM for disrupting the Keap1-Nrf2 interaction and a Kd of 0.50 μM for binding to Keap1. Further studies demonstrated that 19 effectively activated the Nrf2-regulated cytoprotective system and provided protective effects against dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in both in vitro and in vivo models. These findings highlight the potential of β-amino acid substituted naphthalene sulfonamide Keap1-Nrf2 inhibitor 19 as a prospective therapeutic agent for UC via Keap1 targeting.
Collapse
Affiliation(s)
- Ziquan Zhao
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hongjin Lu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Junjie Wang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tingting Wu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shicheng Xu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuxin Ge
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| | - Zhengyu Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Mengchen Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| |
Collapse
|
4
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [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: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
5
|
El-Mahrouk SR, El-Ghiaty MA, El-Kadi AOS. The role of nuclear factor erythroid 2-related factor 2 (NRF2) in arsenic toxicity. J Environ Sci (China) 2025; 150:632-644. [PMID: 39306435 DOI: 10.1016/j.jes.2024.02.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 09/25/2024]
Abstract
Arsenic, a naturally occurring toxic element, manifests in various chemical forms and is widespread in the environment. Exposure to arsenic is a well-established risk factor for an elevated incidence of various cancers and chronic diseases. The crux of arsenic-mediated toxicity lies in its ability to induce oxidative stress, characterized by an unsettling imbalance between oxidants and antioxidants, accompanied by the rampant generation of reactive oxygen species and free radicals. In response to this oxidative turmoil, cells deploy their defense mechanisms, prominently featuring the redox-sensitive transcription factor known as nuclear factor erythroid 2-related factor 2 (NRF2). NRF2 stands as a primary guardian against the oxidative harm wrought by arsenic. When oxidative stress activates NRF2, it orchestrates a symphony of downstream antioxidant genes, leading to the activation of pivotal antioxidant enzymes like glutathione-S-transferase, heme oxygenase-1, and NAD(P)H: quinone oxidoreductase 1. This comprehensive review embarks on the intricate and diverse ways by which various arsenicals influence the NRF2 antioxidant pathway and its downstream targets, shedding light on their roles in defending against arsenic exposure toxic effects. It offers valuable insights into targeting NRF2 as a strategy for safeguarding against or treating the harmful and carcinogenic consequences of arsenic exposure.
Collapse
Affiliation(s)
- Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; Faculty of Pharmacy, Tanta University, Tanta, Gharbia, Egypt
| | - Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada.
| |
Collapse
|
6
|
El-Mahrouk SR, El-Kadi AOS. Dimethylmonothioarsinic acid (DMMTA V) induces NQO1 expression through coordinated activation of NRF2 and AHR pathways. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 115:104674. [PMID: 40058744 DOI: 10.1016/j.etap.2025.104674] [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: 12/19/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Dimethylmonothioarsinic acid (DMMTAV), a potent toxic metabolite of arsenic, exhibits higher cytotoxicity than other arsenicals. This study investigates its influence on NAD(P)H:quinone oxidoreductase (NQO1) regulation in C57BL/6 mice and Hepa-1c1c7 cells. Mice were administered DMMTAV (6 mg/kg, IP) with or without TCDD (15 µg/kg, IP), and hepatic and extrahepatic tissues were analyzed for NQO1 expression. In vitro, Hepa-1c1c7 cells were treated with 0-2 µM DMMTAV in the presence and absence of TCDD (1 nM), and NQO1 levels were assessed over time. Western blot, real-time PCR, and ARE-luciferase assays determined protein and transcriptional regulation. DMMTAV upregulated NQO1 in liver tissues and induced a time-dependent increase in vitro, peaking at 12 h. It enhanced TCDD-induced NQO1 expression and increased nuclear NRF2 and AHR levels, with peak accumulation at two hours. ARE-luciferase activity confirmed transcriptional activation. These findings reveal DMMTAV enhances NQO1 primarily via NRF2/AHR pathway activation, providing insight into cellular responses to thioarsenicals.
Collapse
Affiliation(s)
- Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Faculty of Pharmacy, Tanta University, Tanta, Gharbia, Egypt
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
7
|
Ray SK, Mukherjee S. New insights into reductive stress responses and its clinical relation in cancer. Tissue Cell 2025; 93:102736. [PMID: 39826384 DOI: 10.1016/j.tice.2025.102736] [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: 08/22/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Cells are susceptible to both oxidative and reductive stresses, with reductive stress being less studied and potentially therapeutic in cancer. Reductive stress, characterized by an excess of reducing equivalents exceeding the activity of endogenous oxidoreductases, can lead to an imbalance in homeostasis, causing an increase in reactive oxygen species induction, affecting cellular antioxidant load and flux. Unlike oxidative stress, reductive stress has been understudied and poorly understood, and there is still much to learn about its mechanisms in cancer, its therapeutic potential, and how cancer cells react to it. Changes in redox balance and interference with redox signaling are linked to cancer cell growth, metastasis, and resistance to chemotherapy and radiation. Overconsumption of reducing equivalents can reduce metabolism, alter protein disulfide bond formation, disrupt mitochondrial homeostasis, and disrupt cancer cell signaling pathways. Novel approaches to delivering or using cancer medicines and techniques to influence redox biology have been discovered. Under reductive stress, cancer cells may coordinate separate pools of redox pairs, potentially impacting biology.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent Researcher, Bhopal, Madhya Pradesh 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh 462020, India.
| |
Collapse
|
8
|
Vali R, Shirvanian K, Farkhondeh T, Aschner M, Samini F, Samarghandian S. A review study on the effect of zinc on oxidative stress-related neurological disorders. J Trace Elem Med Biol 2025; 88:127618. [PMID: 39978164 DOI: 10.1016/j.jtemb.2025.127618] [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: 12/05/2023] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
Zinc plays a main role in maintaining homeostasis and neuronal function. Disorders in zinc homeostasis are connected to several neurological disorders due to inflammation and oxidative stress. This review explores the effect of zinc on neurological disorders through the Nrf2 signaling pathway. The Nrf2 pathway modulates oxidative stress and regulates antioxidant defenses, which is critical in the pathogenesis of neurological diseases. We provide an overview of in vivo and in vitro studies illustrating zinc's neuroprotective effects in conditions such as Alzheimer's disease, spinal cord injury, and stroke. The dual role of zinc, where both excess and deficiency can be detrimental, is highlighted, emphasizing the need for optimal zinc levels. Limitations of current research and future perspectives are also discussed.
Collapse
Affiliation(s)
- Reyhaneh Vali
- Department of Biology, Faculty of Modern Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran.
| | - Kasra Shirvanian
- Department of Biology, Faculty of Sciences, University of Tehran, Iran.
| | - Tahereh Farkhondeh
- Geriatric Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Fariborz Samini
- Department of Neurosurgery, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
9
|
Kopacz A, Kloska D, Bar A, Targosz-Korecka M, Cysewski D, Awsiuk K, Piechota-Polanczyk A, Cichon M, Chlopicki S, Jozkowicz A, Grochot-Przeczek A. Endothelial miR-34a deletion guards against aneurysm development despite endothelial dysfunction. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167812. [PMID: 40139409 DOI: 10.1016/j.bbadis.2025.167812] [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: 11/08/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
We previously reported a link between NRF2, a cytoprotective transcription factor, and the ageing of endothelial cells (ECs) and aorta. We also found that NRF2 KO mice are more susceptible to the development of abdominal aortic aneurysm (AAA), which is an age-associated condition. Since miR-34a is a marker of ageing, we explored its relationship with NRF2 and its role in vascular function and AAA formation. Here, we demonstrate that premature NRF2-dependent ageing of ECs is mediated by miR-34a. Infusion of hypertensive angiotensin II (Ang II) in mice increases miR-34a in the aortic endothelial layer and serum, particularly in mice developing AAA. Mice lacking endothelial miR-34a exhibit severe EC dysfunction. Despite that, they are protected from AAA, also on the NRF2 KO background. This protective effect is reversed by rapamycin, which suppresses Ang II-induced EC proliferation. We identified MTA2, but not SIRT1, as a target of miR-34a that inhibits EC proliferation stimulated by Ang II. These findings suggest that fine-tuning of EC proliferation could have potential therapeutic implications for the treatment of aneurysms.
Collapse
Affiliation(s)
- Aleksandra Kopacz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Damian Kloska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Marta Targosz-Korecka
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Kamil Awsiuk
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Aleksandra Piechota-Polanczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Milena Cichon
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
10
|
Wang K, Baird L, Yamamoto M. The clinical-grade CBP/ p300 inhibitor CCS1477 represses the global NRF2-dependent cytoprotective transcription program and re-sensitizes cancer cells to chemotherapeutic drugs. Free Radic Biol Med 2025; 233:102-117. [PMID: 40127850 DOI: 10.1016/j.freeradbiomed.2025.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 03/26/2025]
Abstract
Constitutive activation of NRF2 provides a selective advantage to malignant tumour clones through the hijacking of the NRF2-dependent cytoprotective transcriptional program, which allows the cancer cells to survive and thrive in the chemically stressful tumour niche, whilst also providing resistance to anti-cancer drugs due to the upregulation of xenobiotic metabolizing enzymes and drug efflux pumps. Through a small-molecule epigenetic screen carried out in KEAP1 mutant lung cancer cells, in this study, we identified CCS1477 (Inobrodib) to be an inhibitor of the global NRF2-dependent transcription program. Mechanistically, CCS1477 is able to repress NRF2's cytoprotective response through the inhibition of its obligate transcriptional activator partner CBP/p300. Importantly, in addition to repressing NRF2-dependent anti-oxidative stress and xenobiotic metabolizing enzyme gene expression, CCS1477 treatment is also able to reverse the chemoresistance phenotype and re-sensitize NRF2-activated tumour cells to anti-cancer drugs. Furthermore, in co-culture experiments of KEAP1 mutant cancer cells with primary human T cells, CCS1477 treatment suppressed the acquisition of the T cell exhaustion transcriptional state, which should function to augment the anti-cancer immune response. Thus, CCS1477-mediated inhibition of CBP/p300 represents a novel therapeutic strategy with which to target the currently untreatable tumours with aberrant NRF2 activation.
Collapse
Affiliation(s)
- Ke Wang
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Liam Baird
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, 980-8575, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Miyagi, Japan.
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, 980-8575, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Miyagi, Japan.
| |
Collapse
|
11
|
Froyen EB, Barrantes GP. A Review of the Effects of Flavonoids on NAD(P)H Quinone Oxidoreductase 1 Expression and Activity. J Med Food 2025. [PMID: 40097203 DOI: 10.1089/jmf.2023.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
Cancer is a significant cause of death worldwide. It has been suggested that the consumption of flavonoids decreases the risk for cancer by increasing phase II enzymes, such as Nicotinamide Adenine Dinucleotide Phosphate Hydrogen (NAD(P)H) quinone oxidoreductase 1 (NQO1), glutathione S-transferases, and Uridine 5'-diphospho- (UDP)-glucuronosyltransferases that assist in removing carcinogens from the human body. Flavonoids are bioactive compounds found in a variety of dietary sources, including fruits, vegetables, legumes, nuts, and teas. As such, it is important to investigate which flavonoids are involved in the metabolism of carcinogens to help reduce the risk of cancer. Therefore, the objective of this narrative review was to investigate the effects of commonly consumed flavonoids on NQO1 mRNA expression, protein, and activity in human cell and murine models. PubMed was used to search for peer-reviewed journal articles, which demonstrated that selected flavonoids (e.g., quercetin, apigenin, luteolin, genistein, and daidzein) increase NQO1, and therefore, increase the excretion of carcinogens. However, more research is needed regarding the mechanisms by which flavonoids induce NQO1. Furthermore, it is suggested that future efforts focus on providing precise flavonoid recommendations to decrease the risk factors for chronic diseases.
Collapse
Affiliation(s)
- Erik B Froyen
- Department of Nutrition and Food Science, Huntley College of Agriculture, California State Polytechnic University, Pomona, California, USA
| | - Gianluis Pimentel Barrantes
- Department of Nutrition and Food Science, Huntley College of Agriculture, California State Polytechnic University, Pomona, California, USA
| |
Collapse
|
12
|
Negrao MV, Paula AG, Molkentine D, Hover L, Nilsson M, Vokes N, Engstrom L, Calinisan A, Briere DM, Waters L, Hallin J, Diao L, Altan M, Blumenschein GR, Skoulidis F, Wang J, Kopetz SE, Hong DS, Gibbons DL, Olson P, Christensen JG, Heymach JV. Impact of Co-mutations and Transcriptional Signatures in Non-Small Cell Lung Cancer Patients Treated with Adagrasib in the KRYSTAL-1 Trial. Clin Cancer Res 2025; 31:1069-1081. [PMID: 39804166 PMCID: PMC11911804 DOI: 10.1158/1078-0432.ccr-24-2310] [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: 07/24/2024] [Revised: 10/01/2024] [Accepted: 01/09/2025] [Indexed: 03/18/2025]
Abstract
PURPOSE KRAS inhibitors are revolutionizing the treatment of non-small cell lung cancer (NSCLC), but clinico-genomic determinants of treatment efficacy warrant continued exploration. EXPERIMENTAL DESIGN Patients with advanced KRASG12C-mutant NSCLC treated with adagrasib [KRYSTAL-1 (NCT03785249)] were included in the analysis. Pretreatment next-generation sequencing data were collected per protocol. HTG EdgeSeq Transcriptome Panel was used for gene expression profiling. Clinical endpoints included objective response, progression-free survival (PFS), and overall survival (OS). KRASG12C-mutant NSCLC cell lines and xenograft models were used for sensitivity analyses and combination drug screens. RESULTS KEAP1 MUT and STK11MUT were associated with shorter survival to adagrasib [KEAP1: PFS 4.1 vs. 9.9 months, HR 2.7, P < 0.01; OS 5.4 vs. 19.0 months, HR 3.6, P < 0.01; STK11: PFS 4.2 vs. 11.0 months, HR 2.2, P < 0.01; OS 9.8 months vs. not reached (NR), HR 2.6, P < 0.01]. KEAP1WT/STK11WT status identified adagrasib-treated patients with significantly longer PFS (16.9 months) and OS (NR). Preclinical analyses further validate the association between KEAP1 loss of function and adagrasib resistance. Adagrasib and mTOR inhibitor combinations produced higher treatment efficacy in NSCLC models harboring STK11 and KEAP1 co-mutations. NRF2HIGH signaling was associated with shorter survival to adagrasib (PFS: 4.2 vs. 8.4 months, HR 2.0, P = 0.02; OS: 6.5 vs. 19.0 months, HR 2.8, P < 0.01) even in patients with KEAP1WT NSCLC. KEAP1WT/STK11WT/NRF2LOW status identified patients-32%-with longer survival to adagrasib (PFS 12.0 vs. 4.2 months, HR 0.2, P < 0.01; OS NR vs. 8.0 months, HR 0.1, P < 0.01). CONCLUSIONS KEAP1, STK11, and NRF2 status define patients with KRASG12C-mutant NSCLC with markedly distinct outcomes to adagrasib. These results further support the use of genomic features-mutational and nonmutational-for the treatment selection of patients with KRASG12C-mutant NSCLC.
Collapse
Affiliation(s)
- Marcelo V. Negrao
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Alvaro G. Paula
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - David Molkentine
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | | | - Monique Nilsson
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Natalie Vokes
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Lars Engstrom
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Andrew Calinisan
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - David M. Briere
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Laura Waters
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Jill Hallin
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Lixia Diao
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - George R. Blumenschein
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Ferdinandos Skoulidis
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Jing Wang
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott E. Kopetz
- Department of Gastro-Intestinal Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - David S. Hong
- Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Peter Olson
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - James G. Christensen
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| |
Collapse
|
13
|
Sahu M, Jain U. Activation, interaction and intimation of Nrf2 pathway and their mutational studies causing Nrf2 associated cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167764. [PMID: 40088576 DOI: 10.1016/j.bbadis.2025.167764] [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: 09/09/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025]
Abstract
Responses against infection trigger several signaling pathways that lead to the production of cytokines, these cytokines release ROS and RNS, damaging DNA and proteins turn into various diseases including cancer. To combat these harmful cytokines, the Nrf2 pathway is activated. The gene NFE2L2 encodes Nrf2, which is divided into seven conserved domains (Neh1-7). The DLG and ETGE motifs, conserved sequences of amino acid in the Neh2 domain of Nrf2, bind to the BTB domain of Keap1. BTB domain promotes Keap1's homodimerization resulting in Cul3 recruitment providing scaffold formation to E2 ubiquitin ligase to form ubiquitin complex. Under normal conditions, this complex regularly degrades Nrf2. However, once the cell is exposed to oxidative stress by ROS interaction with Keap1 resulting in conformational changes that stabilize the Nrf2. Nrf2 further concentrates on the nucleus where it binds with the transcriptional factor to perform the desired genes transcription for synthesizing SOD, GSH, CAT, and various other proteins which reduce the ROS levels preventing certain diseases. To prevent cells from oxidative stress, molecular hydrogen activates the Nrf2 pathway. To activate the Nrf2 pathway, molecular hydrogen oxidizes the iron porphyrin which acts as an electrophile and interacts with Keap1's cysteine residue.
Collapse
Affiliation(s)
- Mridul Sahu
- School of Health Sciences and Technology (SoHST), UPES, Bidholi, Dehradun - 248007, India
| | - Utkarsh Jain
- School of Health Sciences and Technology (SoHST), UPES, Bidholi, Dehradun - 248007, India.
| |
Collapse
|
14
|
Ozleyen A, Duran GN, Donmez S, Ozbil M, Doveston RG, Tumer TB. Identification and inhibition of PIN1-NRF2 protein-protein interactions through computational and biophysical approaches. Sci Rep 2025; 15:8907. [PMID: 40087364 PMCID: PMC11909128 DOI: 10.1038/s41598-025-89342-0] [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: 06/30/2024] [Accepted: 02/04/2025] [Indexed: 03/17/2025] Open
Abstract
NRF2 is a transcription factor responsible for coordinating the expression of over a thousand cytoprotective genes. Although NRF2 is constitutively expressed, its stability is modulated by the redox-sensitive protein KEAP1 and other conditional binding partner regulators. The new era of NRF2 research has highlighted the cooperation between NRF2 and PIN1 in modifying its cytoprotective effect. Despite numerous studies, the understanding of the PIN1-NRF2 interaction remains limited. Herein, we described the binding interaction of PIN1 and three different 14-mer long phospho-peptides mimicking NRF2 protein using computer-based, biophysical, and biochemical approaches. According to our computational analyses, the residues positioned in the WW domain of PIN1 (Ser16, Arg17, Ser18, Tyr23, Ser32, Gln33, and Trp34) were found to be crucial for PIN1-NRF2 interactions. Biophysical FP assays were used to verify the computational prediction. The data demonstrated that Pintide, a peptide predominantly interacting with the PIN1 WW-domain, led to a significant reduction in the binding affinity of the NRF2 mimicking peptides. Moreover, we evaluated the impact of known PIN1 inhibitors (juglone, KPT-6566, and EGCG) on the PIN1-NRF2 interaction. Among the inhibitors, KPT-6566 showed the most potent inhibitory effect on PIN1-NRF2 interaction within an IC50 range of 0.3-1.4 µM. Furthermore, our mass spectrometry analyses showed that KPT-6566 appeared to covalently modify PIN1 via conjugate addition, rather than disulfide exchange of the sulfonyl-acetate moiety. Altogether, such inhibitors would also be highly valuable molecular probes for further investigation of PIN1 regulation of NRF2 in the cellular context and potentially pave the way for drug molecules that specifically inhibit the cytoprotective effects of NRF2 in cancer.
Collapse
Affiliation(s)
- Adem Ozleyen
- Leicester Institute for Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, UK
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK
- Health Institutes of Türkiye, Türkiye Biotechnology Institute, 06270, Ankara, Turkey
| | - Gizem Nur Duran
- Institute of Biotechnology, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Serhat Donmez
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey
- Institute of Science and Technology Austria (ISTA), 3400, Klosterneuburg, Austria
| | - Mehmet Ozbil
- Institute of Biotechnology, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Richard G Doveston
- Leicester Institute for Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, UK.
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey.
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
15
|
Zhang DD. Thirty years of NRF2: advances and therapeutic challenges. Nat Rev Drug Discov 2025:10.1038/s41573-025-01145-0. [PMID: 40038406 DOI: 10.1038/s41573-025-01145-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2025] [Indexed: 03/06/2025]
Abstract
Over the last 30 years, NRF2 has evolved from being recognized as a transcription factor primarily involved in redox balance and detoxification to a well-appreciated master regulator of cellular proteostasis, metabolism and iron homeostasis. NRF2 plays a pivotal role in diverse pathologies, including cancer, and metabolic, inflammatory and neurodegenerative disorders. It exhibits a Janus-faced duality, safeguarding cellular integrity in normal cells against environmental insults to prevent disease onset, whereas in certain cancers, constitutively elevated NRF2 levels provide a tumour survival advantage, promoting progression, therapy resistance and metastasis. Advances in understanding the mechanistic regulation of NRF2 and its roles in human pathology have propelled the investigation of NRF2-targeted therapeutic strategies. This Review dissects the mechanistic intricacies of NRF2 signalling, its cross-talk with biological processes and its far-reaching implications for health and disease, highlighting key discoveries that have shaped innovative therapeutic approaches targeting NRF2.
Collapse
Affiliation(s)
- Donna D Zhang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Hayes JD, Dayalan Naidu S, Dinkova-Kostova AT. Regulating Nrf2 activity: ubiquitin ligases and signaling molecules in redox homeostasis. Trends Biochem Sci 2025; 50:179-205. [PMID: 39875264 DOI: 10.1016/j.tibs.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/29/2024] [Accepted: 12/13/2024] [Indexed: 01/30/2025]
Abstract
Transcription factor NF-E2 p45-related factor 2 (Nrf2) orchestrates defenses against oxidants and thiol-reactive electrophiles. It is controlled at the protein stability level by several E3 ubiquitin ligases (CRL3Keap1, CRL4DCAF11, SCFβ-TrCP, and Hrd1). CRL3Keap1 is of the greatest importance because it constitutively targets Nrf2 for proteasomal degradation under homeostatic conditions but is prevented from doing so by oxidative stressors. Repression of Nrf2 by CRL3Keap1 is attenuated by SQSTM1/p62, and this is reinforced by phosphorylation of SQSTM1/p62. Repression by SCFβ-TrCP requires phosphorylation of Nrf2 by GSK3, the activity of which is inhibited by PKB/Akt and other kinases. We discuss how Nrf2 activity is controlled by the ubiquitin ligases under different circumstances. We also describe endogenous signaling molecules that inactivate CRL3Keap1 to alleviate stress and restore homeostasis.
Collapse
Affiliation(s)
- John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK.
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK.
| |
Collapse
|
17
|
Tao T, Xu Y, Zhang CH, Zhang X, Chen J, Liu J. Single-cell transcriptomic analysis and luteolin treatment reveal three adipogenic genes, including Aspn, Htra1 and Efemp1. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159585. [PMID: 39662603 DOI: 10.1016/j.bbalip.2024.159585] [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: 05/07/2024] [Revised: 12/01/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
A comparative transcriptomic analysis in adipose stem and progenitor cells (ASPCs) between obese and lean mice might facilitate the identification of novel adipogenic genes. Here, we compare transcriptomic differences in the ASPCs of subcutaneous adipose tissue (SAT) between the mice fed on a high-fat-diet (HFD) and the chow diet (CD)-fed mice by analyzing three independent single-cell RNA sequencing datasets. Six differential genes, including three up-regulated genes Aspn, Rrbp1, Fbln2 and three down-regulated genes Htra1, Plpp3, Efemp1, are identified and confirmed in HFD-fed mice. Further, the expression of these genes is found to be significantly diminished in the differentiated 3T3-L1 cells. Treatment with luteolin, a dietary flavonoid known to inhibit 3T3-L1 adipogenesis, reverses the decreased expression of Aspn, Htra1 and Efemp1. Furthermore, knockdown of Aspn, Htra1 and Efemp1 significantly facilitates 3T3-L1 adipogenesis. Together, these genes not only are differential in ASPCs between obese and lean mice, but also are the adipogenic inhibitory genes that can be up-regulated by luteolin treatment.
Collapse
Affiliation(s)
- Tao Tao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yanting Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Cheng-Hui Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230009, China.
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230009, China.
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230009, China.
| |
Collapse
|
18
|
Wei X, He Y, Yu Y, Tang S, Liu R, Guo J, Jiang Q, Zhi X, Wang X, Meng D. The Multifaceted Roles of BACH1 in Disease: Implications for Biological Functions and Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412850. [PMID: 39887888 PMCID: PMC11905017 DOI: 10.1002/advs.202412850] [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: 10/13/2024] [Revised: 12/22/2024] [Indexed: 02/01/2025]
Abstract
BTB domain and CNC homolog 1 (BACH1) belongs to the family of basic leucine zipper proteins and is expressed in most mammalian tissues. It can regulate its own expression and play a role in transcriptionally activating or inhibiting downstream target genes. It has a crucial role in various biological processes, such as oxidative stress, cell cycle, heme homeostasis, and immune regulation. Recent research highlights BACH1's significant regulatory roles in a series of conditions, including stem cell pluripotency maintenance and differentiation, growth, senescence, and apoptosis. BACH1 is closely associated with cardiovascular diseases and contributes to angiogenesis, atherosclerosis, restenosis, pathological cardiac hypertrophy, myocardial infarction, and ischemia/reperfusion (I/R) injury. BACH1 promotes tumor cell proliferation and metastasis by altering tumor metabolism and the epithelial-mesenchymal transition phenotype. Moreover, BACH1 appears to show an adverse role in diseases such as neurodegenerative diseases, gastrointestinal disorders, leukemia, pulmonary fibrosis, and skin diseases. Inhibiting BACH1 may be beneficial for treating these diseases. This review summarizes the role of BACH1 and its regulatory mechanism in different cell types and diseases, proposing that precise targeted intervention of BACH1 may provide new strategies for human disease prevention and treatment.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yunquan He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Yueyang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Sichong Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Ruiwen Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Jieyu Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Qingjun Jiang
- Department of Vascular & Endovascular Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Zhongshan Hospital Immunotherapy Translational Research Center, Fudan University, Shanghai, 200032, China
| |
Collapse
|
19
|
Ng C, Kim M, Yanti, Kwak MK. Oxidative stress and NRF2 signaling in kidney injury. Toxicol Res 2025; 41:131-147. [PMID: 40013079 PMCID: PMC11850685 DOI: 10.1007/s43188-024-00272-x] [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: 09/10/2024] [Revised: 11/24/2024] [Accepted: 11/30/2024] [Indexed: 02/28/2025] Open
Abstract
Oxidative stress plays a crucial role in the pathogenesis of acute kidney injury (AKI), chronic kidney disease (CKD), and the AKI-to-CKD transition. This review examines the intricate relationship between oxidative stress and kidney pathophysiology, emphasizing the potential therapeutic role of nuclear factor erythroid 2-related factor 2 (NRF2), a master regulator of cellular redox homeostasis. In diverse AKI and CKD models, diminished NRF2 activity exacerbates oxidative stress, whereas genetic and pharmacological NRF2 activation alleviates kidney damage induced by nephrotoxic agents, ischemia-reperfusion injury, fibrotic stimuli, and diabetic nephropathy. The renoprotective effects of NRF2 extend beyond antioxidant defense, encompassing its anti-inflammatory and anti-fibrotic properties. The significance of NRF2 in renal fibrosis is further underscored by its interaction with the transforming growth factor-β signaling cascade. Clinical trials using bardoxolone methyl, a potent NRF2 activator, have yielded both encouraging and challenging outcomes, illustrating the intricacy of modulating NRF2 in human subjects. In summary, this overview suggests the therapeutic potential of targeting NRF2 in kidney disorders and highlights the necessity for continued research to refine treatment approaches.
Collapse
Affiliation(s)
- Cherry Ng
- Department of Pharmacy and BK21FOUR Advanced Program for Smart Pharma Leaders, Graduate School of The Catholic University of Korea, Gyeonggi-do, 14662 Republic of Korea
| | - Maxine Kim
- Department of Pharmacy and BK21FOUR Advanced Program for Smart Pharma Leaders, Graduate School of The Catholic University of Korea, Gyeonggi-do, 14662 Republic of Korea
| | - Yanti
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta, 12930 Indonesia
| | - Mi-Kyoung Kwak
- Department of Pharmacy and BK21FOUR Advanced Program for Smart Pharma Leaders, Graduate School of The Catholic University of Korea, Gyeonggi-do, 14662 Republic of Korea
- College of Pharmacy, The Catholic University of Korea, 43 Jibong-Ro, Bucheon, Gyeonggi-do 14662 Republic of Korea
| |
Collapse
|
20
|
Wuputra K, Hsu WH, Ku CC, Yang YH, Kuo KK, Yu FJ, Yu HS, Nagata K, Wu DC, Kuo CH, Yokoyama KK. The AHR-NRF2-JDP2 gene battery: Ligand-induced AHR transcriptional activation. Biochem Pharmacol 2025; 233:116761. [PMID: 39855429 DOI: 10.1016/j.bcp.2025.116761] [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: 11/02/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Aryl hydrocarbon receptor (AHR) and nuclear factor-erythroid 2-related factor 2 (NRF2) can regulate a series of genes encoding the detoxifying phase I and II enzymes, via a signaling crosstalk known as the "AHR-NRF2 gene battery". The chromatin transcriptional regulator Jun dimerization protein 2 (JDP2) plays a central role in thetranscription of AHR gene in response to the phase I enzyme ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin. It forms a transcriptional complex with AHR-AHR nuclear translocator (ARNT) and NRF2-small musculoaponeurotic fibrosarcoma proteins (sMAF), which are then recruited to the respective cis-elements, such as dioxin response elements and antioxidant response elements, respectively, in the AHR promoter. Here, we present a revised description of the AHR-NRF2 gene battery as the AHR-NRF2-JDP2 gene battery for transactivating the AHR promoter by phase I enzyme ligands. The chromatin regulator JDP2 was found to be involved in the movement of AHR-NRF2 complexes from the dioxin response element to the antioxidant response element in the AHR promoter, during its activation in a spatiotemporal manner. This new epigenetic and chromatin remodeling role of AHR-NRF2-JDP2 axis is useful for identifying new therapeutic targets for various diseases, including immunological response, detoxification, development, and cancer-related diseases.
Collapse
Affiliation(s)
- Kenly Wuputra
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Wen-Hung Hsu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chia-Chen Ku
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ya-Han Yang
- Division of General Surgery, E-DA Dachang Hospital, Kaohsiung 80706, Taiwan.
| | - Kung-Kai Kuo
- Division of General Surgery, E-DA Dachang Hospital, Kaohsiung 80706, Taiwan.
| | - Fang-Jung Yu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan.
| | - Hsin-Su Yu
- Emeritus Professor in College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Kyosuke Nagata
- Professor, Insitutte of Medicine, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chao-Hung Kuo
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Superintendant in Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan.
| | - Kazunari K Yokoyama
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
21
|
Clemente-Suárez VJ, Redondo-Flórez L, Martín-Rodríguez A, Curiel-Regueros A, Rubio-Zarapuz A, Tornero-Aguilera JF. Impact of Vegan and Vegetarian Diets on Neurological Health: A Critical Review. Nutrients 2025; 17:884. [PMID: 40077754 PMCID: PMC11901473 DOI: 10.3390/nu17050884] [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: 02/13/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES The global shift towards vegan and vegetarian diets has garnered attention for their ethical, environmental, and potential health benefits. These diets are often rich in phytonutrients and antioxidants, which have been associated with lower levels of inflammatory markers, such as C-reactive protein (CRP) and interleukin-6 (IL-6), suggesting a potential protective effect against systemic inflammation and oxidative stress. However, despite these benefits, concerns remain regarding their impact on neurological health due to the possible deficiencies of critical nutrients such as vitamin B12, DHA, EPA, and iron. This review critically evaluates the influence of these dietary patterns on neurological outcomes, emphasizing their nutritional composition, potential deficiencies, and their interplay with inflammation and oxidative stress. METHODS A systematic review of the literature published between 2010 and 2023 was conducted, focusing on studies that explore the relationship between vegan and vegetarian diets and neurological health. Key nutrients such as vitamin B12, omega-3 fatty acids, iron, and zinc were analyzed alongside antinutritional factors and their effects on the nervous system. RESULTS Evidence suggests that vegan and vegetarian diets, when well planned, can be rich in phytonutrients and antioxidants, which have been associated with lower levels of inflammatory markers, such as C-reactive protein (CRP) and interleukin-6 (IL-6). These findings indicate a potential role in reducing systemic inflammation and oxidative stress, both of which are linked to neurodegenerative diseases. However, deficiencies in critical nutrients such as vitamin B12, DHA, EPA, and iron have been consistently associated with an increased risk of cognitive decline, mood disturbances, and neurodegenerative disorders. Additionally, the presence of antinutritional factors like phytates and oxalates may further impair nutrient absorption, necessitating careful dietary planning and supplementation. CONCLUSIONS While plant-based diets provide anti-inflammatory and antioxidant benefits, their neurological implications depend on nutrient adequacy. Proper planning, supplementation, and food preparation techniques are essential to mitigate risks and enhance cognitive health. Further research is needed to explore long-term neurological outcomes and optimize dietary strategies.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Medicine, Health and Sports, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (V.J.C.-S.); (A.M.-R.); (A.C.-R.)
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Tajo Street s/n, 28670 Villaviciosa de Odon, Spain;
| | - Alexandra Martín-Rodríguez
- Faculty of Medicine, Health and Sports, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (V.J.C.-S.); (A.M.-R.); (A.C.-R.)
- Faculty of Applied Social Sciences and Communications, UNIE University, 28015 Madrid, Spain
| | - Agustín Curiel-Regueros
- Faculty of Medicine, Health and Sports, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (V.J.C.-S.); (A.M.-R.); (A.C.-R.)
| | - Alejandro Rubio-Zarapuz
- Faculty of Medicine, Health and Sports, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (V.J.C.-S.); (A.M.-R.); (A.C.-R.)
| | | |
Collapse
|
22
|
Althagafy HS, Hassanein EHM. Morin Mitigates 5-Fluorouracil-Induced Nephrotoxicity by Activating Nrf2/HO-1 and FXR, and Suppressing ERK/VCAM-1 and NF-κB Pathways. Int Immunopharmacol 2025; 148:114092. [PMID: 39823795 DOI: 10.1016/j.intimp.2025.114092] [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: 07/19/2024] [Revised: 10/06/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
5-Fluorouracil (5-FU) is a DNA analog used in chemotherapy to treat various tumors. However, the clinical use of 5-FU is limited due to its severe adverse effects, particularly its nephrotoxicity. Morin (MRN) is a flavanol found in many different plants, including those in the Moraceae family, and has anti-inflammatory and antioxidant bioactivities. The protective effects of MRN against experimental 5-FU-induced kidney injury were investigated in this work. The rats were assigned to four groups in our study: control, MRN (50 mg/kg), 5-FU (30 mg/kg), and 5-FU + MRN. The administration of MRN caused a significant (P < 0.05) decrease in the serum urea and creatinine levels and a reduction in the histopathological changes induced by 5-FU, as shown by H&E, PAS, and Sirius red staining. IHC shows that MRN attenuates renal oxidative stress induced by 5-FU via co-activation of Nrf2, HO-1, and FXR. MRN protects against renal inflammation induced by 5-FU, as evidenced by decreased TNF-α and IL-6 levels in the rat kidney mediated by the downregulation of the ERK1/2 and VCAM-1 proteins and decreased NF-κB phosphorylation as shown by Western blotting. These findings support using MRN as a novel and promising treatment for 5-FU-induced nephrotoxicity.
Collapse
Affiliation(s)
- Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Saudi Arabia.
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| |
Collapse
|
23
|
Sekiguchi A, Ishikawa M, Uchiyama A, Yokoyama Y, Ogino S, Torii R, Akai R, Iwawaki T, Motegi SI. Rapid thawing enhances tissue destruction in a mouse model of cutaneous cryoablation: Insights into oxidative stress and neutrophil activation. J Dermatol Sci 2025:S0923-1811(25)00019-2. [PMID: 40000304 DOI: 10.1016/j.jdermsci.2025.02.003] [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: 10/16/2024] [Revised: 01/09/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Cryoablation is an integral therapeutic approach in dermatology for eliminating viral warts and benign tumors by damaging tissue through freeze-thaw cycles. Rapid thawing of the frozen area by warming it with fingertips during cryoablation is a common technique in Japan; however, its efficacy has not been elucidated. OBJECTIVE This study aimed to evaluate the effect of rapid thawing on cryoablation-treated skin and clarify the underlying mechanisms using cryoablation model mice. METHODS Cryoablation was performed on the dorsal skin of mice using a liquid nitrogen-soaked cotton swab, followed by rapid thawing by warming with fingertips or natural thawing without treatment. The effects on skin ulcers, immune cell infiltration, and oxidative stress were assessed. RESULTS Rapid thawing enlarged cryoablation-induced skin ulcers. The numbers of cryoablation-induced CD3+ T cells, neutrophils, neutrophil extracellular traps (NETs), and TUNEL+ cells increased with rapid thawing. Visualization of oxidative stress in OKD48 transgenic mice showed that oxidative stress signals in the cryoablation-treated area were enhanced with rapid thawing. Real-time PCR analysis of mouse skin demonstrated that cryoblation-induced levels of NOX2 and HO-1 were significantly elevated with rapid thawing. In mouse melanoma tumors treated with cryoablation, rapid thawing significantly inhibited tumor growth and increased the infiltration of neutrophils, NETs, and TUNEL+ cells compared to the group without rapid thawing. CONCLUSION Rapid thawing during cryoablation enhances neutrophil and lymphocyte infiltration, increases oxidative stress, and induces cell death, leading to greater tissue destruction in mice. Dermatologists should consider employing rapid thawing techniques during cryoablation when higher therapeutic intensities are required.
Collapse
Affiliation(s)
- Akiko Sekiguchi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Mai Ishikawa
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihiko Uchiyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoko Yokoyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sachiko Ogino
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Torii
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Akai
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kanazawa, Japan
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kanazawa, Japan
| | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan.
| |
Collapse
|
24
|
Chembo J, Bowman BM, Lapak K, Wilkerson E, Paiboonrungruang C, Cho K, Medcalf MR, Patti GJ, Dolle RE, Chen X, Zolkind P, Major MB. Pyrimethamine and a potent analogue WCDD115 inhibit NRF2 by suppressing DHFR and one-carbon metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.637433. [PMID: 40027760 PMCID: PMC11870417 DOI: 10.1101/2025.02.13.637433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Nuclear factor erythroid 2-related factor 2 (NFE2L2/NRF2) is a critical mediator of the cellular oxidative stress response. Aberrant activation of NRF2 is common in lung and upper aerodigestive cancers, where it promotes tumor initiation and progression and confers resistance to chemotherapy, radiation therapy, and immune checkpoint inhibitors. As such, NRF2 therapeutic inhibitors are actively being sought. We previously reported that the antiparasitic drug Pyrimethamine (PYR) inhibits NRF2 in cell lines and in a NRF2-inducible genetically engineered mouse model. Here we design, synthesize, and define structure-activity relationships across a series of 25 PYR-based derivatives to reveal WCDD115 as a 22-fold more potent inhibitor of NRF2 (57nM versus 1.2µM). PYR is known to inhibit plasmodial and human dihydrofolate reductase (DHFR). We found that WCDD115 inhibits hDHFR with 31-fold greater potency than PYR (144nM versus 4.49µM). Metabolomics showed strong similarities between PYR, WCDD115 and methotrexate. Genetic, pharmacological and metabolic epistasis studies reveal that DHFR inactivation is required for NRF2 suppression by WCDD115 and PYR. Global and targeted proteomics revealed overlapping profiles for WCDD115, PYR and methotrexate, including suppression of NRF2 oxidative stress response and activation of TP53 and the DNA damage response. Therefore, PYR and a novel potent derivative WCDD115 are effective, indirect inhibitors of NRF2 and its antioxidant functions. These data underscore the importance of one- carbon metabolism for the NRF2 signaling pathway and support a new therapeutic strategy to suppress NRF2-driven cancer biology.
Collapse
Affiliation(s)
- Julius Chembo
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO 63110
| | - Brittany M. Bowman
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO 63110
| | - Kyle Lapak
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO 63110
| | - Emily Wilkerson
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO 63110
| | - Chorlada Paiboonrungruang
- Coriell Institute for Medical Research, Camden, NJ 08103
- Surgical Research Lab, Department of Surgery and MD Anderson Cancer Center at Cooper, Cooper University Health Care and Cooper Medical School of Rowan University, Camden, NJ 08103
| | - Kevin Cho
- Department of Chemistry, Medicine, and Center for Mass Spectrometry and Metabolic Tracing, Washington University in St. Louis, St. Louis, MO 63110
| | - Matthew R. Medcalf
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO 63110
| | - Gary J. Patti
- Department of Chemistry, Medicine, and Center for Mass Spectrometry and Metabolic Tracing, Washington University in St. Louis, St. Louis, MO 63110
| | - Roland E. Dolle
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO 63110
| | - Xiaoxin Chen
- Coriell Institute for Medical Research, Camden, NJ 08103
- Surgical Research Lab, Department of Surgery and MD Anderson Cancer Center at Cooper, Cooper University Health Care and Cooper Medical School of Rowan University, Camden, NJ 08103
| | - Paul Zolkind
- Department of Otolaryngology, Washington University in St. Louis, St. Louis, MO 63110
| | - Michael B. Major
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO 63110
- Department of Otolaryngology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
25
|
Duarte P, Sanchez-Porro FJ, Crisman E, Cores Á, Jiménez I, Cuadrado A, Menéndez JC, León R. Network-Based Drug Optimization toward the Treatment of Parkinson's Disease: NRF2, MAO-B, Oxidative Stress, and Chronic Neuroinflammation. J Med Chem 2025; 68:3495-3517. [PMID: 39818855 PMCID: PMC11881042 DOI: 10.1021/acs.jmedchem.4c02659] [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: 10/31/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, affects around 10 million people worldwide. It is a multifactorial disease marked by dopaminergic neuron loss with oxidative stress (OS) and neuroinflammation as key pathological drivers. Current treatments focus on dopamine replacement and are symptomatic, underscoring the urgent need for disease-modifying therapies. Here, we present a novel class of dual MAO-B inhibitors and NRF2 inducers with neuroprotective properties in in vitro PD models. Through an optimization program, we enhanced their MAO-B inhibitory potency, selectivity, and NRF2 induction capacity while achieving favorable pharmacokinetic profiles. Virtual library screening identified two core derivatives, leading to the development of compound 11, which exhibited potent anti-inflammatory and neuroprotective activity in OS-related in vitro models. Compound 11 also demonstrated high liver microsomal stability and favorable pharmacokinetics in mice, making it a promising candidate for further investigation as a potential PD therapy.
Collapse
Affiliation(s)
- Pablo Duarte
- Consejo
Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
| | - Francisco J. Sanchez-Porro
- Consejo
Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
| | - Enrique Crisman
- Consejo
Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
- Fundación
Teófilo Hernando para la I+D del Medicamento, Las Rozas, 28290 Madrid, Spain
| | - Ángel Cores
- Unidad
de Química Orgánica y Farmacéutica, Departamento
de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Irene Jiménez
- Consejo
Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
- Fundación
Teófilo Hernando para la I+D del Medicamento, Las Rozas, 28290 Madrid, Spain
| | - Antonio Cuadrado
- Instituto
de Investigación Sanitaria La Paz (IdiPaz) and Departamento
de Bioquímica, Facultad de Medicina, UAM, Instituto de Investigaciones Biomédicas “Alberto Sols”
UAM-CSIC, 28029 Madrid, Spain
- Centro
de Investigación Biomédica en Red Sobre Enfermedades
Neurodegenerativas (CIBERNED), ISCIII, 28029 Madrid, Spain
| | - J. Carlos Menéndez
- Unidad
de Química Orgánica y Farmacéutica, Departamento
de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Rafael León
- Consejo
Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
| |
Collapse
|
26
|
Xu H, Brown JL, Bhaskaran S, Van Remmen H. Reactive oxygen species in the pathogenesis of sarcopenia. Free Radic Biol Med 2025; 227:446-458. [PMID: 39613046 PMCID: PMC11816180 DOI: 10.1016/j.freeradbiomed.2024.11.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
One of the most critical factors impacting healthspan in the elderly is the loss of muscle mass and function, clinically referred to as sarcopenia. Muscle atrophy and weakness lead to loss of mobility, increased risk of injury, metabolic changes and loss of independence. Thus, defining the underlying mechanisms of sarcopenia is imperative to enable the development of effective interventions to preserve muscle function and quality in the elderly and improve healthspan. Over the past few decades, understanding the roles of mitochondrial dysfunction and oxidative stress has been a major focus of studies seeking to reveal critical molecular pathways impacted during aging. In this review, we will highlight how oxidative stress might contribute to sarcopenia by discussing the impact of oxidative stress on the loss of innervation and alteration in the neuromuscular junction (NMJ), on muscle mitochondrial function and atrophy pathways, and finally on muscle contractile function.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States
| | - Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
27
|
Tian F, Sun S, Ge Z, Ge Y, Ge X, Shi Z, Qian X. Understanding the Anticancer Effects of Phytochemicals: From Molecular Docking to Anticarcinogenic Signaling. J Nutr 2025; 155:431-444. [PMID: 39581266 DOI: 10.1016/j.tjnut.2024.11.015] [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: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
As nontraditional nutrients, the biological activity of phytochemicals have been extensively studied for their antioxidant, anti-inflammatory, and apoptosis-promoting effects in various diseases. The general anticancer benefits of phytochemicals have been demonstrated in both basic researches and clinical trials. However, researchers understanding of how phytochemicals target cancer-related signaling pathways is still in its infancy. Molecular docking simulation analyses have yielded a large amount of cellular target molecules of phytochemicals. Herein, we review the potential signaling pathways that may be involved in the phytochemical-driven cancer benefits. We expect these findings to help in the design of potential cancer treatments designed by manipulating the binding modes and sites of these plant chemicals.
Collapse
Affiliation(s)
- Fuwei Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuhong Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zehe Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqian Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Qian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Rojo AI, Buttari B, Cadenas S, Carlos AR, Cuadrado A, Falcão AS, López MG, Georgiev MI, Grochot-Przeczek A, Gumeni S, Jimenez-Villegas J, Horbanczuk JO, Konu O, Lastres-Becker I, Levonen AL, Maksimova V, Michaeloudes C, Mihaylova LV, Mickael ME, Milisav I, Miova B, Rada P, Santos M, Seabra MC, Strac DS, Tenreiro S, Trougakos IP, Dinkova-Kostova AT. Model organisms for investigating the functional involvement of NRF2 in non-communicable diseases. Redox Biol 2025; 79:103464. [PMID: 39709790 PMCID: PMC11733061 DOI: 10.1016/j.redox.2024.103464] [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: 10/25/2024] [Revised: 11/26/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024] Open
Abstract
Non-communicable chronic diseases (NCDs) are most commonly characterized by age-related loss of homeostasis and/or by cumulative exposures to environmental factors, which lead to low-grade sustained generation of reactive oxygen species (ROS), chronic inflammation and metabolic imbalance. Nuclear factor erythroid 2-like 2 (NRF2) is a basic leucine-zipper transcription factor that regulates the cellular redox homeostasis. NRF2 controls the expression of more than 250 human genes that share in their regulatory regions a cis-acting enhancer termed the antioxidant response element (ARE). The products of these genes participate in numerous functions including biotransformation and redox homeostasis, lipid and iron metabolism, inflammation, proteostasis, as well as mitochondrial dynamics and energetics. Thus, it is possible that a single pharmacological NRF2 modulator might mitigate the effect of the main hallmarks of NCDs, including oxidative, proteostatic, inflammatory and/or metabolic stress. Research on model organisms has provided tremendous knowledge of the molecular mechanisms by which NRF2 affects NCDs pathogenesis. This review is a comprehensive summary of the most commonly used model organisms of NCDs in which NRF2 has been genetically or pharmacologically modulated, paving the way for drug development to combat NCDs. We discuss the validity and use of these models and identify future challenges.
Collapse
Affiliation(s)
- Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain.
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161, Rome, Italy
| | - Susana Cadenas
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), Cantoblanco, Madrid, Spain
| | - Ana Rita Carlos
- CE3C-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016, Lisbon, Portugal
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Ana Sofia Falcão
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Hospital Universitario de la Princesa, Madrid, Spain
| | - Milen I Georgiev
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - José Jimenez-Villegas
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Jarosław Olav Horbanczuk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; Department of Neuroscience, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Isabel Lastres-Becker
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute Teófilo Hernando for Drug Discovery, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Viktorija Maksimova
- Department of Applied Pharmacy, Division of Pharmacy, Faculty of Medical Sciences, Goce Delcev University, Stip, Krste Misirkov Str., No. 10-A, P.O. Box 201, 2000, Stip, Macedonia
| | | | - Liliya V Mihaylova
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Michel Edwar Mickael
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, 1000, Ljubljana, Slovenia; Laboratory of oxidative stress research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, 1000, Ljubljana, Slovenia
| | - Biljana Miova
- Department of Experimental Physiology and Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University "St Cyril and Methodius", Skopje, Macedonia
| | - Patricia Rada
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Marlene Santos
- REQUIMTE/LAQV, Escola Superior de Saúde (E2S), Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 400, 4200-072, Porto, Portugal; Molecular Oncology & Viral Pathology, IPO-Porto Research Center (CI-IPOP), Portuguese Institute of Oncology, 4200-072, Porto, Portugal
| | - Miguel C Seabra
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10 000, Zagreb, Croatia
| | - Sandra Tenreiro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, UK; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Champsi S, Hood DA. Sulforaphane treatment mimics contractile activity-induced mitochondrial adaptations in muscle myotubes. Am J Physiol Cell Physiol 2025; 328:C335-C354. [PMID: 39672545 DOI: 10.1152/ajpcell.00669.2024] [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: 09/12/2024] [Revised: 11/15/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Mitochondria are metabolic hubs that govern skeletal muscle health. Although exercise has been established as a powerful inducer of quality control processes that ultimately enhance mitochondrial function, there are currently limited pharmaceutical interventions available that emulate exercise-induced mitochondrial adaptations. To investigate a novel candidate for this role, we examined sulforaphane (SFN), a naturally occurring compound found in cruciferous vegetables. SFN has been documented as a potent antioxidant inducer through its activation of the nuclear factor erythroid 2-related factor 2 (Nrf-2) antioxidant response pathway. However, its effects on muscle health have been underexplored. To investigate the interplay between chronic exercise and SFN, C2C12 myotubes were electrically stimulated to model chronic contractile activity (CCA) in the presence or absence of SFN. SFN promoted Nrf-2 nuclear translocation, enhanced mitochondrial respiration, and upregulated key antioxidant proteins including catalase and glutathione reductase. These adaptations were accompanied by reductions in cellular and mitochondrial reactive oxygen species (ROS) emission. Signaling toward biogenesis was enhanced, demonstrated by increases in mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α nuclear translocation, PGC-1α promoter activity, mitochondrial content, and organelle branching, suggestive of a larger, more interconnected mitochondrial pool. These mitochondrial adaptations were accompanied by an increase in lysosomal proteins, suggesting coordinated regulation. There was no difference in mitochondrial and antioxidant-related proteins between CCA and non-CCA SFN-treated cells. Our data suggest that SFN activates signaling cascades that are common to those produced by contractile activity, indicating that SFN-centered therapeutic strategies may improve the mitochondrial phenotype in skeletal muscle.NEW & NOTEWORTHY Nrf-2 is a transcription factor that has been implicated in mitigating oxidative stress and regulating mitochondrial homeostasis. However, limited research has demonstrated how Nrf-2-mediated adaptations compare with those produced by exercise. To investigate this, we treated myotubes with Sulforaphane, a well-established Nrf-2 activator, and combined this with stimulation-induced chronic contractile activity to model exercise training. Our work is the first to establish that sulforaphane mimics training-induced mitochondrial adaptations, including enhancements in respiration, biogenesis, and dynamics.
Collapse
Affiliation(s)
- Sabrina Champsi
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - David A Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
30
|
la Torre A, Lo Vecchio F, Angelillis VS, Gravina C, D’Onofrio G, Greco A. Reinforcing Nrf2 Signaling: Help in the Alzheimer's Disease Context. Int J Mol Sci 2025; 26:1130. [PMID: 39940900 PMCID: PMC11818887 DOI: 10.3390/ijms26031130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Oxidative stress plays a role in various pathophysiological diseases, including neurogenerative diseases, such as Alzheimer's disease (AD), which is the most prevalent neuro-pathology in the aging population. Oxidative stress has been reported to be one of the earliest pathological alterations in AD. Additionally, it was demonstrated that in older adults, there is a loss of free radical scavenging ability. The Nrf2 transcription factor is a key regulator in antioxidant defense systems, but, with aging, both the amount and the transcriptional activity of Nrf2 decrease. With the available treatments for AD being poorly effective, reinforcing the antioxidant defense systems via the Nrf2 pathway may be a way to prevent and treat AD. To highlight the predominant role of Nrf2 signaling in defending against oxidative stress and, therefore, against neurotoxicity, we present an overview of the natural compounds that exert their own neuroprotective roles through the activation of the Nrf2 pathway. This review is an opportunity to promote a holistic approach in the treatment of AD and to highlight the need to further refine the development of new potential Nrf2-targeting drugs.
Collapse
Affiliation(s)
- Annamaria la Torre
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Filomena Lo Vecchio
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Valentina Soccorsa Angelillis
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (V.S.A.); (A.G.)
| | - Carolina Gravina
- Laboratory of Gerontology and Geriatrics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.L.V.); (C.G.)
| | - Grazia D’Onofrio
- Clinical Psychology Service, Health Department, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Antonio Greco
- Complex Unit of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (V.S.A.); (A.G.)
| |
Collapse
|
31
|
Garg A, On KF, Xiao Y, Elkayam E, Cifani P, David Y, Joshua-Tor L. The molecular basis of Human FN3K mediated phosphorylation of glycated substrates. Nat Commun 2025; 16:941. [PMID: 39843453 PMCID: PMC11754801 DOI: 10.1038/s41467-025-56207-z] [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: 08/02/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025] Open
Abstract
Glycation, a non-enzymatic post-translational modification occurring on proteins, can be actively reversed via site-specific phosphorylation of the fructose-lysine moiety by FN3K kinase, to impact the cellular function of the target protein. A regulatory axis between FN3K and glycated protein targets has been associated with conditions like diabetes and cancer. However, the molecular basis of this relationship has not been explored so far. Here, we determined a series of crystal structures of HsFN3K in the apo-state, and in complex with different nucleotide analogs together with a sugar substrate mimic to reveal the features important for its kinase activity and substrate recognition. Additionally, the dynamics in sugar substrate binding during the kinase catalytic cycle provide important mechanistic insights into HsFN3K function. Our structural work provides the molecular basis for rational small molecule design targeting FN3K.
Collapse
Affiliation(s)
- Ankur Garg
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA
- Howard Hughes Medical Institute, Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA
| | - Kin Fan On
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA
- Howard Hughes Medical Institute, Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA
| | - Yang Xiao
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Elad Elkayam
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA
- Ventus Therapeutics, Waltham, Massachusetts, 02453, USA
| | - Paolo Cifani
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA
| | - Yael David
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Leemor Joshua-Tor
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA.
- Howard Hughes Medical Institute, Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, New York, 11724, USA.
| |
Collapse
|
32
|
Jiang B, Liu J, Qu Z, Wang Y, Wang Y, Li Z, Jin X, Lao Y, He R, Yang S. Mechanism of dihydroartemisinin in the treatment of ischaemia/reperfusion-induced acute kidney injury via network pharmacology, molecular dynamics simulation and experiments. Int Immunopharmacol 2025; 144:113705. [PMID: 39626534 DOI: 10.1016/j.intimp.2024.113705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE To investigate whether dihydroartemisinin (DHA) attenuates ischaemia-reperfusion injury (IRI)-induced acute kidney injury (AKI) in mice by inhibiting oxidative stress and inflammation and to explore its potential molecular mechanisms. MATERIALS AND METHODS Network pharmacology analysis was used to screen relevant targets, and molecular docking of DHA with core targets was performed. Molecular dynamics simulation of the target with the lowest binding free energy, NQO1-DHA.The renal protective effect of DHA on the IRI-induced AKI mouse model was evaluated. The expression levels of NQO1, Nrf2 and other proteins were detected by Western blotting. The expression levels of Nrf2 and others were detected by immunohistochemistry (IHC) and immunofluorescence (IF). RESULTS Through network pharmacological analysis, we obtained that PI3K/AKT and MAPK signaling pathway may be related to DHA in the treatment of AKI.Molecular dynamics simulation indicated that NQO1 is an important target protein for DHA to exert nephroprotective effects.Moreover, the potential molecular mechanisms were verified by experiments.DHA reduced the serum creatinine (Scr) and urea nitrogen (BUN) levels in AKI mice, significantly improved AKI pathology, alleviated oxidative stress and inflammatory injury, which may be related to its activation of the Nrf2 pathway and regulation of macrophage polarization. CONCLUSIONS Through network pharmacology, molecular dynamics simulation and experimental validation, we initially investigated that DHA alleviate AKI by ameliorating oxidative stress and inflammatory damage, which may be related to its activation of the Nrf2 pathway and the regulation of macrophage polarisation, which lays the foundation for subsequent in-depth study of the specific mechanism of action.
Collapse
Affiliation(s)
- Beibei Jiang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jiahui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Ziyi Qu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yanqing Wang
- Department of Anatomy,School of Basic Medical Sciences,Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yuzhi Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Zhongtang Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaoming Jin
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yunlan Lao
- Central Hospital of Guangdong Provincial Nongken, Guangdong, China
| | - Riming He
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
33
|
Serrano JJ, Medina MÁ. Metabolic Reprogramming at the Edge of Redox: Connections Between Metabolic Reprogramming and Cancer Redox State. Int J Mol Sci 2025; 26:498. [PMID: 39859211 PMCID: PMC11765076 DOI: 10.3390/ijms26020498] [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: 11/19/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
The importance of redox systems as fundamental elements in biology is now widely recognized across diverse fields, from ecology to cellular biology. Their connection to metabolism is particularly significant, as it plays a critical role in energy regulation and distribution within organisms. Over recent decades, metabolism has emerged as a relevant focus in studies of biological regulation, especially following its recognition as a hallmark of cancer. This shift has broadened cancer research beyond strictly genetic perspectives. The interaction between metabolism and redox systems in carcinogenesis involves the regulation of essential metabolic pathways, such as glycolysis and the Krebs cycle, as well as the involvement of redox-active components like specific amino acids and cofactors. The feedback mechanisms linking redox systems and metabolism in cancer highlight the development of redox patterns that enhance the flexibility and adaptability of tumor processes, influencing larger-scale biological phenomena such as circadian rhythms and epigenetics.
Collapse
Affiliation(s)
- José J. Serrano
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain;
| | - Miguel Ángel Medina
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain;
- Instituto de Investigación Biomédica y Plataforma en Nanomedicina IBIMA Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER, Spanish Network of Research Center in Rare Diseases), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
34
|
Sethi P, Mehan S, Khan Z, Maurya PK, Kumar N, Kumar A, Tiwari A, Sharma T, Das Gupta G, Narula AS, Kalfin R. The SIRT-1/Nrf2/HO-1 axis: Guardians of neuronal health in neurological disorders. Behav Brain Res 2025; 476:115280. [PMID: 39368713 DOI: 10.1016/j.bbr.2024.115280] [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: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
SIRT1 (Sirtuin 1) is a NAD+-dependent deacetylase that functions through nucleoplasmic transfer and is present in nearly all mammalian tissues. SIRT1 is believed to deacetylate its protein substrates, resulting in neuroprotective actions, including reduced oxidative stress and inflammation, increased autophagy, increased nerve growth factors, and preserved neuronal integrity in aging or neurological disease. Nrf2 is a transcription factor that regulates the genes responsible for oxidative stress response and substance detoxification. The activation of Nrf2 guards cells against oxidative damage, inflammation, and carcinogenic stimuli. Several neurological abnormalities and inflammatory disorders have been associated with variations in Nrf2 activation caused by either pharmacological or genetic factors. Recent evidence indicates that Nrf2 is at the center of a complex cellular regulatory network, establishing it as a transcription factor with genuine pleiotropy. HO-1 is most likely a component of a defense mechanism in cells under stress, as it provides negative feedback for cell activation and mediator synthesis. This mediator is upregulated by Nrf2, nitric oxide (NO), and other factors in various inflammatory states. HO-1 or its metabolites, such as CO, may mitigate inflammation by modulating signal transduction pathways. Neurological diseases may be effectively treated by modulating the activity of HO-1. Multiple studies have demonstrated that SIRT1 and Nrf2 share an important connection. SIRT1 enhances Nrf2, activates HO-1, protects against oxidative injury, and decreases neuronal death. This has been associated with numerous neurodegenerative and neuropsychiatric disorders. Therefore, activating the SIRT1/Nrf2/HO-1 pathway may help treat various neurological disorders. This review focuses on the current understanding of the SIRT1 and Nrf2/HO-1 neuroprotective processes and the potential therapeutic applications of their target activators in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Pranshul Sethi
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Pankaj Kumar Maurya
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, Sofia 1113, Bulgaria; Department of Healthcare, South-West University "NeofitRilski", Ivan Mihailov St. 66, Blagoevgrad 2700, Bulgaria
| |
Collapse
|
35
|
Alves F, Lane D, Nguyen TPM, Bush AI, Ayton S. In defence of ferroptosis. Signal Transduct Target Ther 2025; 10:2. [PMID: 39746918 PMCID: PMC11696223 DOI: 10.1038/s41392-024-02088-5] [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: 06/24/2024] [Revised: 10/10/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Rampant phospholipid peroxidation initiated by iron causes ferroptosis unless this is restrained by cellular defences. Ferroptosis is increasingly implicated in a host of diseases, and unlike other cell death programs the physiological initiation of ferroptosis is conceived to occur not by an endogenous executioner, but by the withdrawal of cellular guardians that otherwise constantly oppose ferroptosis induction. Here, we profile key ferroptotic defence strategies including iron regulation, phospholipid modulation and enzymes and metabolite systems: glutathione reductase (GR), Ferroptosis suppressor protein 1 (FSP1), NAD(P)H Quinone Dehydrogenase 1 (NQO1), Dihydrofolate reductase (DHFR), retinal reductases and retinal dehydrogenases (RDH) and thioredoxin reductases (TR). A common thread uniting all key enzymes and metabolites that combat lipid peroxidation during ferroptosis is a dependence on a key cellular reductant, nicotinamide adenine dinucleotide phosphate (NADPH). We will outline how cells control central carbon metabolism to produce NADPH and necessary precursors to defend against ferroptosis. Subsequently we will discuss evidence for ferroptosis and NADPH dysregulation in different disease contexts including glucose-6-phosphate dehydrogenase deficiency, cancer and neurodegeneration. Finally, we discuss several anti-ferroptosis therapeutic strategies spanning the use of radical trapping agents, iron modulation and glutathione dependent redox support and highlight the current landscape of clinical trials focusing on ferroptosis.
Collapse
Affiliation(s)
- Francesca Alves
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Darius Lane
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
36
|
Ashcroft FJ, Bourboula A, Mahammad N, Barbayianni E, Feuerherm AJ, Nguyen TT, Hayashi D, Kokotou MG, Alevizopoulos K, Dennis EA, Kokotos G, Johansen B. Next generation thiazolyl ketone inhibitors of cytosolic phospholipase A 2 α for targeted cancer therapy. Nat Commun 2025; 16:164. [PMID: 39747052 PMCID: PMC11696576 DOI: 10.1038/s41467-024-55536-9] [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: 10/24/2023] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Eicosanoids are key players in inflammatory diseases and cancer. Targeting their production by inhibiting Group IVA cytosolic phospholipase A2 (cPLA2α) offers a promising approach for cancer therapy. In this study, we synthesize a second generation of thiazolyl ketone inhibitors of cPLA2α starting with compound GK470 (AVX235) and test their in vitro and cellular activities. We identify a more potent and selective lead molecule, GK420 (AVX420), which we test in parallel with AVX235 and a structurally unrelated compound, AVX002 for inhibition of cell viability across a panel of cancer cell lines. From this, we show that activity of polycomb group repressive complex 2 is a key molecular determinant of sensitivity to cPLA2α inhibition, while resistance depends on antioxidant response pathways. Consistent with these results, we show that elevated intracellular reactive oxygen species and activating transcription factor 4 target gene expression precede cell death in AVX420-sensitive T-cell acute lymphoblastic leukemia cells. Our findings imply cPLA2α may support cancer by mitigating oxidative stress and inhibiting tumor suppressor expression and suggest that AVX420 has potential for treating acute leukemias and other cancers that are susceptible to oxidative cell death.
Collapse
Affiliation(s)
- Felicity J Ashcroft
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Asimina Bourboula
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Nur Mahammad
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Efrosini Barbayianni
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Astrid J Feuerherm
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Thanh Thuy Nguyen
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Daiki Hayashi
- Department of Applied Chemistry in Bioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Maroula G Kokotou
- Laboratory of Chemistry, Department of Food Science and Human Nutrition, Agricultural University of Athens, Athens, Greece
| | | | - Edward A Dennis
- Department of Chemistry and Biochemistry, University of California at San Diego, La Jolla, CA, USA
- Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece.
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece.
| | - Berit Johansen
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
37
|
Tian R, Guo S, Chen S, Wu J, Long A, Cheng R, Wang X, Huang L, Li C, Mao W, Xu P, Yu L, Pan H, Liu L. Natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy: Recent progress and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156342. [PMID: 39742572 DOI: 10.1016/j.phymed.2024.156342] [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: 09/30/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2, NFE2L2) is a pivotal regulator of redox balance, metabolism, protein homeostasis and inflammation. Nrf2 is critically involved in both ferroptosis and renal diseases, and may serve as a significant target for many natural products in the treatment of renal diseases. However, a comprehensive overview on this topic is still lacking. PURPOSE To review the protective or therapeutic effects of natural products regulating Nrf2-related ferroptosis against various renal diseases. METHODS We systematically searched the electronic databases involving PubMed, Web of Science, Google Scholar, China National Knowledge Internet (CNKI), Wanfang Database and VIP Database. To ensure a comprehensive exploration, keywords including Nrf2, ferroptosis, natural products, phytochemicals, renal disease, kidney disease, kidney injury and nephropathy were employed. RESULTS Ferroptosis is deeply implicated in various kinds of renal diseases, notably including cisplatin-induced acute kidney injury, sepsis-associated acute kidney injury, renal ischemia/reperfusion injury, diabetic nephropathy, kidney stones and renal fibrosis. Nrf2 plays a regulatory role on many important genes related to iron metabolism, antioxidant system and lipid metabolism, thereby modulating ferroptosis. More than twenty natural products exert renoprotective effects by inhibiting ferroptosis via the regulation of Nrf2. This review presents a comprehensive overview of recent advancements in elucidating the ferroptosis involvement in renal diseases, the role of Nrf2 in regulating ferroptosis, and summarizes the renoprotective natural products as Nrf2 modulators for ferroptosis inhibition. CONCLUSION Through the comprehensive insights, this review clarifies the protective or therapeutic effects of natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy, in the pursuit of providing new research ideas and directions for the treatment of renal diseases. Further drug development aimed at discovering more natural products and optimizing their utilization for disease treatment is necessary.
Collapse
Affiliation(s)
- Ruimin Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Shan Guo
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shudong Chen
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiaqi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Aoyang Long
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ran Cheng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaowan Wang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Lihua Huang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Chuang Li
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Wei Mao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Peng Xu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Lili Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Hudan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| |
Collapse
|
38
|
Xie Z, Hou Q, He Y, Xie Y, Mo Q, Wang Z, Zhao Z, Chen X, Peng T, Li L, Xie W. Ferritin Hinders Ferroptosis in Non-Tumorous Diseases: Regulatory Mechanisms and Potential Consequences. Curr Protein Pept Sci 2025; 26:89-104. [PMID: 39225224 DOI: 10.2174/0113892037315874240826112422] [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: 03/30/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Ferritin, as an iron storage protein, has the potential to inhibit ferroptosis by reducing excess intracellular free iron concentrations and lipid reactive oxygen species (ROS). An insufficient amount of ferritin is one of the conditions that can lead to ferroptosis through the Fenton reaction mediated by ferrous iron. Consequently, upregulation of ferritin at the transcriptional or posttranscriptional level may inhibit ferroptosis. In this review, we have discussed the essential role of ferritin in ferroptosis and the regulatory mechanism of ferroptosis in ferritin-deficient individuals. The description of the regulatory factors governing ferritin and its properties in regulating ferroptosis as underlying mechanisms for the pathologies of diseases will allow potential therapeutic approaches to be developed.
Collapse
Affiliation(s)
- Zhongcheng Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Qin Hou
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yinling He
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yushu Xie
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Qinger Mo
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziyi Wang
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziye Zhao
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Liang Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
39
|
Zhang S, Xing N, Jiao Y, Li J, Wang T, Zhang Q, Hu X, Li C, Kuang W. An arabinan from Citrus grandis fruits alleviates ischemia/reperfusion-induced myocardial cell apoptosis via the Nrf2/Keap1 and IRE1/GRP78 signaling pathways. Carbohydr Polym 2025; 347:122728. [PMID: 39486958 DOI: 10.1016/j.carbpol.2024.122728] [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: 04/02/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 11/04/2024]
Abstract
Citrus grandis fruit is a famous traditional Chinese medicine with various bioactivities, including cardioprotective effects. Polysaccharides are one of the key active ingredients responsible for its cardioprotective effects. This study aimed to investigate the structure and cardioprotective effect of a homogeneous polysaccharide from C. grandis fruit (CGP80-1) and explore its mechanism against myocardial ischemia-reperfusion (MI/R) injury. Structure analysis showed that CGP80-1 (11,917 Da) is an arabinan with compact coil chain conformation, containing →5)-α-L-Araf-(1→, →3,5)-α-L-Araf-(1→, and →2,3,5)-α-L-Araf-(1→ as the backbone, as well as →5)-α-L-Araf-(1→ and t-α-L-Araf as side-chains substituted at the C2 and C3 positions. Pharmacological experiments showed that pre-treatment with CGP80-1 could effectively alleviate MI/R injury by improving endogenous antioxidant enzymes and cardiac enzymes, reducing reactive oxygen species levels, and regulating apoptosis-related proteins such as caspase-3, Bax, and Bcl-2. The protective effects were correlated with the Nrf2/Keap1 and IRE1/GRP78 signaling pathways. Further analysis of structure-activity relationships revealed that the myocardial protection effects of CGP80-1 might be attributed to its appropriate molecular weight, high arabinose content, and unique compact coil chain conformation. Overall, our results provide insight into the chemical structure of CGP80-1 and its mechanism of action, suggesting that CGP80-1 could be a candidate drug for myocardial protection.
Collapse
Affiliation(s)
- Shaojie Zhang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Na Xing
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yukun Jiao
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, Jiangxi Health Industry Institute of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Nanchang 330115, China
| | - Junhao Li
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Tanggan Wang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Qian Zhang
- School of Pharmacy, Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xianjing Hu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Chong Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China; College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong 271018, China.
| | - Weihong Kuang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
40
|
Kulow VA, Roegner K, Labes R, Kasim M, Mathia S, Czopek CS, Berndt N, Becker PN, Ter-Avetisyan G, Luft FC, Enghard P, Hinze C, Klocke J, Eckardt KU, Schmidt-Ott KM, Persson PB, Rosenberger C, Fähling M. Beyond hemoglobin: Critical role of 2,3-bisphosphoglycerate mutase in kidney function and injury. Acta Physiol (Oxf) 2025; 241:e14242. [PMID: 39422260 DOI: 10.1111/apha.14242] [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: 08/14/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
AIM 2,3-bisphosphoglycerate mutase (BPGM) is traditionally recognized for its role in modulating oxygen affinity to hemoglobin in erythrocytes. Recent transcriptomic analyses, however, have indicated a significant upregulation of BPGM in acutely injured murine and human kidneys, suggesting a potential renal function for this enzyme. Here we aim to explore the physiological role of BPGM in the kidney. METHODS A tubular-specific, doxycycline-inducible Bpgm-knockout mouse model was generated. Histological, immunofluorescence, and proteomic analyses were conducted to examine the localization of BPGM expression and the impact of its knockout on kidney structure and function. In vitro studies were performed to investigate the metabolic consequences of Bpgm knockdown under osmotic stress. RESULTS BPGM expression was localized to the distal nephron and was absent in proximal tubules. Inducible knockout of Bpgm resulted in rapid kidney injury within 4 days, characterized by proximal tubular damage and tubulointerstitial fibrosis. Proteomic analyses revealed involvement of BPGM in key metabolic pathways, including glycolysis, oxidative stress response, and inflammation. In vitro, Bpgm knockdown led to enhanced glycolysis, decreased reactive oxygen species elimination capacity under osmotic stress, and increased apoptosis. Furthermore, interactions between nephron segments and immune cells in the kidney suggested a mechanism for propagating stress signals from distal to proximal tubules. CONCLUSION BPGM fulfills critical functions beyond the erythrocyte in maintaining glucose metabolism in the distal nephron. Its absence leads to metabolic imbalances, increased oxidative stress, inflammation, and ultimately kidney injury.
Collapse
Affiliation(s)
- Vera A Kulow
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Kameliya Roegner
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Robert Labes
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Mumtaz Kasim
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Susanne Mathia
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Claudia S Czopek
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Nikolaus Berndt
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp N Becker
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Gohar Ter-Avetisyan
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Enghard
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Christian Hinze
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jan Klocke
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Kai-Uwe Eckardt
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Kai M Schmidt-Ott
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Pontus B Persson
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Christian Rosenberger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Michael Fähling
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| |
Collapse
|
41
|
Oshikiri H, Taguchi K, Hirose W, Taniyama Y, Kamei T, Siegel D, Ross D, Kitson RRA, Baird L, Yamamoto M. Anticancer Effect of C19-Position Substituted Geldanamycin Derivatives Targeting NRF2-NQO1-activated Esophageal Squamous Cell Carcinoma. Mol Cell Biol 2024; 45:79-97. [PMID: 39717011 DOI: 10.1080/10985549.2024.2438817] [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: 10/22/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
In esophageal squamous cell carcinoma, genetic activation of NRF2 increases resistance to chemotherapy and radiotherapy, which results in a significantly worse prognosis for patients. Therefore NRF2-activated cancers create an urgent clinical need to identify new therapeutic options. In this context, we previously identified the geldanamycin family of HSP90 inhibitors, which includes 17DMAG, to be synthetic lethal with NRF2 activity. As the first-generation of geldanamycin-derivative drugs were withdrawn from clinical trials due to hepatotoxicity, we designed second-generation compounds with C19-substituted structures in order to inhibit glutathione conjugation-mediated hepatotoxicity. In this study, using a variety of in vitro and in vivo cancer models, we found that C19-substituted 17DMAG compounds maintain their enhanced toxicity profile and synthetic lethal interaction with NRF2-NQO1-activated cancer cells. Importantly, using a xenograft mouse tumor model, we found that C19-substituted 17DMAG displayed significant anticancer efficacy against NRF2-NQO1-activated cancer cells without causing hepatotoxicity. These results clearly demonstrate the improved clinical potential for this new class of HSP90 inhibitor anticancer drugs, and suggest that patients with NRF2-NQO1-activated esophageal carcinoma may benefit from this novel therapeutic approach.
Collapse
Affiliation(s)
- Hiroyuki Oshikiri
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keiko Taguchi
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Wataru Hirose
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yusuke Taniyama
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - David Siegel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David Ross
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Russell R A Kitson
- Department of Organic and Bioorganic Chemistry, Charles University, Hradec Králové, Czech Republic
| | - Liam Baird
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| |
Collapse
|
42
|
Sakai E, Tsukuba T. Transcriptomic Characterization Reveals Mitochondrial Involvement in Nrf2/Keap1-Mediated Osteoclastogenesis. Antioxidants (Basel) 2024; 13:1575. [PMID: 39765903 PMCID: PMC11673794 DOI: 10.3390/antiox13121575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Although osteoclasts play crucial roles in the skeletal system, the mechanisms that underlie oxidative stress during osteoclastogenesis remain unclear. The transcription factor Nrf2 and its suppressor, Keap1, function as central mediators of oxidative stress. To further elucidate the function of Nrf2/Keap1-mediated oxidative stress regulation in osteoclastogenesis, DNA microarray analysis was conducted in this study using wild-type (WT), Keap1 knockout (Keap1 KO), and Nrf2 knockout (Nrf2 KO) osteoclasts. Principal component analysis showed that 403 genes, including Nqo1, Il1f9, and Mmp12, were upregulated in Keap1 KO compared with WT osteoclasts, whereas 24 genes, including Snhg6, Ccdc109b, and Wfdc17, were upregulated in Nrf2 KO compared with WT osteoclasts. Moreover, 683 genes, including Car2, Calcr, and Pate4, were upregulated in Nrf2 KO cells compared to Keap1 KO cells. Functional analysis by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis showed upregulated genes in Nrf2 KO osteoclasts were mostly enriched in oxidative phosphorylation. Furthermore, GeneMANIA predicted the protein-protein interaction network of novel molecules such as Rufy4 from genes upregulated in Nrf2 KO osteoclasts. Understanding the complex interactions between these molecules may pave the way for developing promising therapeutic strategies against bone metabolic diseases caused by increased osteoclast differentiation under oxidative stress.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1, Sakamoto, Nagasaki 852-8588, Japan;
| | | |
Collapse
|
43
|
Tamaru E, Kokubu D, Ushida Y, Itoh K. Nrf2 induction potency of plant-derived compounds determined using an antioxidant response element luciferase reporter and conventional NAD(P)H-quinone acceptor oxidoreductase 1 activity assay. BMC Res Notes 2024; 17:373. [PMID: 39707451 DOI: 10.1186/s13104-024-07038-6] [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: 04/02/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Abstract
OBJECTIVE Various plants have been reported to contain compounds that promote the transcriptional activity of Nuclear factor erythroid 2-related factor 2 (Nrf2) to induce a set of xenobiotic detoxifying enzymes, such as NAD(P)H-quinone acceptor oxidoreductase 1 (NQO1), via the antioxidant response element (ARE). While conventional methods for evaluating Nrf2 induction potency include measurement of NQO1 activity, an ARE luciferase reporter assay was recently developed to specifically assess Nrf2 induction potency of compounds of interest. In this study, we compared the abilities of these two assays to evaluate and determine Nrf2 induction potency of plant-derived compounds. RESULTS Although the compounds exhibited a high degree of consistency between assays, several compounds did not. These results suggest that although the NQO1 assay can be used as an evaluation method to estimate Nrf2 induction potency of a compound, an ARE luciferase reporter approach may offer greater precision. In summary, inconsistency in Nrf2 induction potency evaluated by the reporter and NQO1 assays of plant-derived compounds, including resveratrol, may be due to a variety of factors that may regulate NQO1 gene expression other than Nrf2 and/or directly modulate NQO1 enzymatic activity or protein levels, with each compound having a different degree of effect on these factors.
Collapse
Affiliation(s)
- Erina Tamaru
- Diet & Well-being Research Institute, KAGOME CO, LTD. 17 Nishitomiyama, Nasushiobara, 329- 2762, Tochigi, Japan
| | - Daichi Kokubu
- Diet & Well-being Research Institute, KAGOME CO, LTD. 17 Nishitomiyama, Nasushiobara, 329- 2762, Tochigi, Japan.
- Department of Vegetable Life Science, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, 036-8562, Aomori, Japan.
| | - Yusuke Ushida
- Diet & Well-being Research Institute, KAGOME CO, LTD. 17 Nishitomiyama, Nasushiobara, 329- 2762, Tochigi, Japan
| | - Ken Itoh
- Department of Vegetable Life Science, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, 036-8562, Aomori, Japan
- Department of Stress Response Science, Biomedical Research Center, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Aomori, Japan
| |
Collapse
|
44
|
El-Mahrouk SR, El-Ghiaty MA, Alqahtani MA, El-Kadi AOS. Arsenic Trioxide (ATO III) Induces NAD(P)H Quinone Oxidoreductase 1 (NQO1) Expression in Hepatic and Extrahepatic Tissues of C57BL/6 Mice. Chem Res Toxicol 2024; 37:2040-2051. [PMID: 39630573 DOI: 10.1021/acs.chemrestox.4c00422] [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: 12/07/2024]
Abstract
Arsenic trioxide (ATOIII) has emerged as a potent therapeutic agent for acute promyelocytic leukemia (APL), yet its clinical application is often limited by significant adverse effects. This study investigates the molecular mechanisms underlying ATOIII's impact on cellular detoxification pathways, focusing on the regulation of NAD(P)H/quinone oxidoreductase (NQO1), a crucial enzyme in maintaining cellular homeostasis and cancer prevention. We explored ATOIII's effects on NQO1 expression in C57BL/6 mice and Hepa-1c1c7 cells, both independently and in combination with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a known NQO1 inducer. Our findings revealed that ATOIII significantly increased NQO1 expression in hepatic and extrahepatic tissues, as well as in Hepa-1c1c7 cells, at mRNA, protein, and activity levels. This upregulation occurred both in the presence and absence of TCDD. Mechanistically, we demonstrated that ATOIII promotes the nuclear translocation of both nuclear factor erythroid 2-related factor-2 (NRF2) and aryl hydrocarbon receptor (AHR) transcription factors. Furthermore, ATOIII exposure increased antioxidant response element (ARE)-driven reporter gene activity, indicating a transcriptional mechanism of NQO1 induction. Notably, gene silencing experiments confirmed the critical roles of both NRF2 and AHR in mediating ATOIII-induced NQO1 expression. In conclusion, ATOIII exposure is found to upregulate the NQO1 enzyme through a transcriptional mechanism via AHR- and NRF2- dependent mechanisms, offering valuable insights into its therapeutic mechanisms.
Collapse
Affiliation(s)
- Sara R El-Mahrouk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
- Faculty of Pharmacy, Tanta University, Gharbia, Tanta 31111, Egypt
| | - Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
| | - Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
| |
Collapse
|
45
|
Chu CT, Uruno A, Katsuoka F, Yamamoto M. Role of NRF2 in Pathogenesis of Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1529. [PMID: 39765857 PMCID: PMC11727090 DOI: 10.3390/antiox13121529] [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: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
Alzheimer's disease (AD) is a polygenic, multifactorial neurodegenerative disorder and remains the most prevalent form of dementia, globally. Despite decades of research efforts, there is still no effective cure for this debilitating condition. AD research has increasingly focused on transcription factor NRF2 (nuclear factor erythroid 2-related factor 2) as a potential therapeutic target. NRF2 plays a crucial role in protecting cells and tissues from environmental stressors, such as electrophiles and reactive oxygen species. Recently, an increasing number of studies have demonstrated that NRF2 is a key regulator in AD pathology. NRF2 is highly expressed in microglia, resident macrophages in the central nervous system, and contributes to neuroinflammation, phagocytosis and neurodegeneration in AD. NRF2 has been reported to modulate microglia-induced inflammation and facilitate the transition from homeostatic microglia to a disease-associated microglia subset. Genetic and pharmacological activation of NRF2 has been demonstrated to improve cognitive function. Here, we review the current understanding of the involvement of NRF2 in AD and the critical role that NRF2 plays in microglia in the context of AD. Our aim is to highlight the potential of targeting NRF2 in the microglia as a promising therapeutic strategy for mitigating the progression of AD.
Collapse
Affiliation(s)
- Ching-Tung Chu
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| | - Akira Uruno
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| | - Fumiki Katsuoka
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan;
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Japan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| |
Collapse
|
46
|
Fejes I, Markacz P, Tatai J, Rudas M, Dunkel P, Gyuris M, Nyerges M, Provost N, Duvivier V, Delerive P, Martiny V, Bristiel A, Vidal B, Richardson W, Rothweiler EM, Tranberg-Jensen J, Manning CE, Sweeney MN, Chalk R, Huber KVM, Bullock AN, Herner A, Seedorf K, Vinson C, Weber C, Kotschy A. Covalent Inhibitors of KEAP1 with Exquisite Selectivity. J Med Chem 2024; 67:21208-21222. [PMID: 39572012 DOI: 10.1021/acs.jmedchem.4c02019] [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: 12/13/2024]
Abstract
The NRF2-KEAP1 interaction is central for cytoprotection against stresses, giving it high clinical significance. Covalent modification of KEAP1 is an efficient approach, but the covalent inhibitors used in the clinic carry undesired side effects originating in their moderate selectivity. Starting with a phenotypic screen, we identified a new covalent inhibitor chemotype that was optimized to deliver a series of potent and highly selective KEAP1 binders. While the developed compounds showed both cellular and in vivo activity, upregulating antioxidant response element-dependent target genes, they showed no genotoxicity in vitro. The lead compound exhibited broad selectivity in activity-based protein profiling and showed no significant interaction with a panel of commonly studied receptors nor with a broad panel of kinases. The nature of its interaction with KEAP1 and the origin of its selectivity were revealed by X-ray crystallography.
Collapse
Affiliation(s)
- Imre Fejes
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Piroska Markacz
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Janos Tatai
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Monika Rudas
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Petra Dunkel
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Mario Gyuris
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Miklos Nyerges
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Nicolas Provost
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - Valérie Duvivier
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - Philippe Delerive
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - Virginie Martiny
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - Alexandra Bristiel
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - Brice Vidal
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - William Richardson
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Elisabeth M Rothweiler
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Jeppe Tranberg-Jensen
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Charlotte E Manning
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Melissa N Sweeney
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Rod Chalk
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Kilian V M Huber
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Alex N Bullock
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, OX3 7FZ Oxford, U.K
| | - Andras Herner
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Klaus Seedorf
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - Cedric Vinson
- Institute de Recherche Servier, 22 Route 128, 91190 Gif-sur-Yvette, France
| | - Csaba Weber
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| | - Andras Kotschy
- Servier Research Institute of Medicinal Chemistry, Zahony u. 7., H-1031 Budapest, Hungary
| |
Collapse
|
47
|
Xie K, Wang F, Yang Y, Pan S, Wang J, Xiao N, Wang X, Ma Z, Xu X, Dong Z. Monotropein alleviates septic acute liver injury by restricting oxidative stress, inflammation, and apoptosis via the AKT (Ser473)/GSK3β (Ser9)/Fyn/NRF2 pathway. Int Immunopharmacol 2024; 142:113178. [PMID: 39305888 DOI: 10.1016/j.intimp.2024.113178] [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: 06/30/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
Sepsis-associated acute liver injury (ALI) is a deadly condition resulting from a systemic inflammatory response to liver cell damage and malfunction. Monotropein (MON) belongs to the iris group of compounds extracted from the natural product Mollen dae officinalis radix, which has strong anti-inflammatory and antioxidant pharmacological effects. The purpose of this study was to elucidate the underlying mechanism of MON in the treatment of sepsis ALI. In this study, an in vivo caecal ligation puncture (CLP)-induced ALI model and in vitro LPS-stimulated AML12 cells and RAW264.7 cells model were established. Additionally, a variety of experimental techniques, including CCK8, H&E staining, DHE probe labelling, biochemical, QPCR, and Western blotting and blocking tests, were used to explore the role of MON in ALI. The results showed that MON improved liver morphological abnormalities, oedema, histopathological injury, and elevated ALT and AST, providing a protective effect against ALI. MON reduced CYP2E1 expression, alleviated oxidative stress (downregulation of MDA levels and upregulation of GSH, CAT, and T-AOC levels) and ROS accumulation with the involvement of the NRF2-Keap-1 pathway. MON inhibited inflammation via the TLR4/NF-κB/NLRP3 inflammasome pathway. In addition, it activated the Akt (Ser473)/GSK3β (Ser9)/Fyn pathway and accelerated NRF2 nuclear accumulation; MK-2206 blockade reversed the NRF2 nuclear accumulation and anti-inflammatory function of MON. MON also restricted the mitochondrial apoptosis pathway, a process specifically blocked by MK-2206. In summary, we concluded that MON alleviated septic ALI by restricting oxidative stress, inflammation, and apoptosis via the AKT (Ser473)/GSK3β (Ser9)/Fyn/NRF2 pathway.
Collapse
Affiliation(s)
- Kunmei Xie
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Feibiao Wang
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yue Yang
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Shoujie Pan
- Department of Pharmacy, Department of oncology, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang 222004, China
| | - Junyao Wang
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Nan Xiao
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xinyan Wang
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhihao Ma
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiaolong Xu
- Department of Pharmacy, Department of oncology, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang 222004, China.
| | - Zibo Dong
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
48
|
Ayala-Cosme EG, Yang D, Vences K, Davis LO, Borgini M. State-of-the-Art Nrf2 Inhibitors: Therapeutic Opportunities in Non-Cancer Diseases. ChemMedChem 2024; 19:e202400377. [PMID: 39083752 DOI: 10.1002/cmdc.202400377] [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: 05/16/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
Nuclear factor erythroid 2-related factor (Nrf2) is a cytoprotective transcription factor that induces the transcription of genes responsible for the cell's response to oxidative stress. While Nrf2 activation has led to the development of clinically relevant therapeutics, the oncogenic role of Nrf2 in the proliferation of cancer cells has underscored the complex nature of Nrf2 and the necessity for the development of Nrf2 inhibitors. Although the application of Nrf2 inhibitors appears limited as anticancer agents, recent studies have begun to pinpoint the impairment of autophagy in diseases as a cellular marker that shifts Nrf2 from a protective to a deleterious state. Therefore, the cytoplasmic accumulation of Nrf2 can lead to the accumulation of lipid hydroperoxides and, ultimately, to ferroptosis. However, some studies aimed at elucidating the role of Nrf2 in non-cancer diseases have yielded conflicting results, attributed to differences in approaches used to inhibit or activate Nrf2, as well as variations in in vitro and/or in vivo disease models. Overall, these results highlight the necessity for a deeper evaluation of Nrf2's role in diseases, especially chronic diseases. In this review, we discuss diseases where Nrf2 inhibition holds potential for beneficial therapeutic effects and summarize recently reported Nrf2 inhibitors exploiting medicinal chemistry approaches suitable for targeting transcription factors like Nrf2.
Collapse
Affiliation(s)
- Emil G Ayala-Cosme
- Department of Chemistry and Biochemistry, Augusta University, Augusta, 30912, GA, U.S.A
| | - Deborah Yang
- Department of Chemistry and Biochemistry, Augusta University, Augusta, 30912, GA, U.S.A
| | - Kyara Vences
- Department of Chemistry and Biochemistry, Augusta University, Augusta, 30912, GA, U.S.A
| | - Lindsey O Davis
- Department of Chemistry and Biochemistry, Augusta University, Augusta, 30912, GA, U.S.A
| | - Matteo Borgini
- Department of Chemistry and Biochemistry, Augusta University, Augusta, 30912, GA, U.S.A
| |
Collapse
|
49
|
Eroglu B, Isales C, Eroglu A. Age and duration of obesity modulate the inflammatory response and expression of neuroprotective factors in mammalian female brain. Aging Cell 2024; 23:e14313. [PMID: 39230054 PMCID: PMC11634740 DOI: 10.1111/acel.14313] [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: 02/20/2024] [Revised: 07/09/2024] [Accepted: 07/27/2024] [Indexed: 09/05/2024] Open
Abstract
Obesity has become a global epidemic and is associated with comorbidities, including diabetes, cardiovascular, and neurodegenerative diseases, among others. While appreciable insight has been gained into the mechanisms of obesity-associated comorbidities, effects of age, and duration of obesity on the female brain remain obscure. To address this gap, adolescent and mature adult female mice were subjected to a high-fat diet (HFD) for 13 or 26 weeks, whereas age-matched controls were fed a standard diet. Subsequently, the expression of inflammatory cytokines, neurotrophic/neuroprotective factors, and markers of microgliosis and astrogliosis were analyzed in the hypothalamus, hippocampus, and cerebral cortex, along with inflammation in visceral adipose tissue. HFD led to a typical obese phenotype in all groups independent of age and duration of HFD. However, the intermediate duration of obesity induced a limited inflammatory response in adolescent females' hypothalamus while the hippocampus, cerebral cortex, and visceral adipose tissue remained unaffected. In contrast, the prolonged duration of obesity resulted in inflammation in all three brain regions and visceral adipose tissue along with upregulation of microgliosis/astrogliosis and suppression of neurotrophic/neuroprotective factors in all brain regions, denoting the duration of obesity as a critical risk factor for neurodegenerative diseases. Importantly, when female mice were older (i.e., mature adult), even the intermediate duration of obesity induced similar adverse effects in all brain regions. Taken together, our findings suggest that (1) both age and duration of obesity have a significant impact on obesity-associated comorbidities and (2) early interventions to end obesity are critical to preserving brain health.
Collapse
Affiliation(s)
- Binnur Eroglu
- Department of Neuroscience and Regenerative MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
| | - Carlos Isales
- Department of Neuroscience and Regenerative MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
- Department of MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
| | - Ali Eroglu
- Department of Neuroscience and Regenerative MedicineMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
- Department of Obstetrics and GynecologyMedical College of Georgia, Augusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
50
|
Iwasaki T, Shirota H, Sasaki K, Ouchi K, Nakayama Y, Oshikiri H, Otsuki A, Suzuki T, Yamamoto M, Ishioka C. Specific cancer types and prognosis in patients with variations in the KEAP1-NRF2 system: A retrospective cohort study. Cancer Sci 2024; 115:4034-4044. [PMID: 39327066 PMCID: PMC11611756 DOI: 10.1111/cas.16355] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
The KEAP1-NRF2 system induces the expression of antioxidant genes in response to various types of oxidative stress. Some cancer cells activate this system, which increases their malignancy through genetic mutations. We performed a retrospective cohort study using the C-CAT database, which contains the gene-panel sequence data from 60,056 cases of diagnosed solid tumors. We analyzed somatic mutations in NRF2 and KEAP1 genes and their associations with clinical outcomes. Variants in the NRF2 gene were clustered in exon 2, which encodes the DLG and ETGE motifs essential for KEAP1 interaction. The NRF2 variants were frequently observed in esophageal and lung squamous cell carcinoma with frequencies of 35.9% and 19.6%, respectively. Among these mutations, the NRF2 variants in the ETGE motif were indicators of a worse prognosis. KEAP1 variants were found in 2.5% of all cases. The variants were frequent in lung cancer and showed a worse prognosis in lung and other types of adenocarcinomas. We then conducted gene expression analysis using TCGA data. While cancers with DLG and ETGE variants were similar in terms of gene expression profiles, there were significant differences between cancers with KEAP1 and NRF2 variants. Our results indicate that genetic alteration of the KEAP1-NRF2 pathway is a major factor in patient prognosis for each cancer type and its genetic variant. Variants in NRF2 and KEAP1 genes can characterize the biological basis of each cancer type and are involved in carcinogenesis, resistance to therapy, and other biological differences.
Collapse
Affiliation(s)
- Tomoyuki Iwasaki
- Department of Medical OncologyTohoku University HospitalSendaiJapan
| | - Hidekazu Shirota
- Department of Medical OncologyTohoku University HospitalSendaiJapan
| | - Keiju Sasaki
- Department of Medical OncologyTohoku University HospitalSendaiJapan
| | - Kota Ouchi
- Department of Medical OncologyTohoku University HospitalSendaiJapan
| | - Yuki Nakayama
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Hiroyuki Oshikiri
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Akihito Otsuki
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Takafumi Suzuki
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Chikashi Ishioka
- Department of Medical OncologyTohoku University HospitalSendaiJapan
| |
Collapse
|