1
|
Guo H, Chen J, Jiang L. The antioxidant stress effect of granulin precursor in vitiligo. Sci Rep 2025; 15:18189. [PMID: 40415096 DOI: 10.1038/s41598-025-03486-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 05/20/2025] [Indexed: 05/27/2025] Open
Abstract
The imbalance of the skin redox system is regarded as a crucial factor contributing to the loss of melanocytes in vitiligo. However, it remains unclear whether alterations in signal transmission between melanocytes and other cells impact the homeostasis of the skin microenvironment. Hence, leveraging single-cell sequencing and microarray data, we investigated the role of cell-cell and ligand receptor interactions in the pathogenesis of vitiligo. We discovered that the granulin-sortilin 1 ligand-receptor serves as an essential bridge for communication between melanocytes and other skin cells in normal skin, yet it is significantly downregulated in vitiligo lesions. Enrichment analysis indicates that the activation of granulin-sortilin 1 ligand-receptor is closely associated with the regulation of oxidative stress. In vitro experiments have verified that progranulin, the protein encoded by the granulin gene, enhances the ability of melanocytes to resist cell death induced by reactive oxygen species and markedly upregulates the expression of nuclear factor erythroid 2-related factor 2 and Heme Oxygenase-1. Notably, this process can be impeded by the interaction inhibitor. Moreover, the expression of nuclear factor erythroid 2-related factor 2 might be linked to the transcription of transcription factor EB activated by progranulin. In conclusion, the granulin-sortilin 1 ligand-receptor can activate the intracellular antioxidant system to counteract melanocyte death. The impairment of the granulin-sortilin 1 ligand-receptor may be implicated in melanocyte loss in vitiligo.
Collapse
Affiliation(s)
- Haoran Guo
- Department of Dermatology, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Yuelu, Changsha, 410013, China
| | - Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Yuelu, Changsha, 410013, China
| | - Ling Jiang
- Department of Dermatology, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Yuelu, Changsha, 410013, China.
| |
Collapse
|
2
|
Wang J, Song X, Hui Y, Dong B, Gong J, Zhao Y, Ji H, Qiu Y, Jiang S, Guo D, Gao X. TFEB orchestrates ferritinophagy and ferroptosis in ionophore drug-induced hepatotoxicity: unveiling a novel therapeutic avenue. Free Radic Biol Med 2025; 236:116-133. [PMID: 40409694 DOI: 10.1016/j.freeradbiomed.2025.05.401] [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: 03/19/2025] [Revised: 04/29/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Ionophore polyether antibiotics (IPAs) exhibit remarkable therapeutic potential in combating parasitic diseases and cancer, yet their clinical utility is significantly hampered by severe hepatotoxicity. Despite widespread documentation of IPAs-induced hepatotoxicity, the precise molecular mechanisms underlying this phenomenon remain elusive. This study elucidates the role of ferroptosis in IPAs-induced liver injury and delineates the associated regulatory pathways. Through comprehensive in vitro (HepG2 cells) and in vivo (mice) investigations, we demonstrate that IPAs, particularly the highly toxic maduramicin (Mad), induce hepatocyte ferroptosis. Mechanistic studies employing lipid reactive oxygen species (ROS) quantification, intracellular Fe2+ assays, and Western blot analysis revealed that IPAs-induced ferroptosis occurs through an autophagy-dependent pathway. Surface plasmon resonance (SPR) and molecular docking analyses confirmed direct binding and regulation of transcription factor EB (TFEB) by maduramicin. This interaction activates TFEB, subsequently mediating nuclear receptor coactivator 4 (NCOA4)-regulated lysosomal degradation processes that culminate in ferroptosis-mediated hepatotoxicity. Importantly, our findings extend beyond maduramicin, as other IPAs including monensin and salinomycin similarly targeted TFEB, triggering hepatocyte ferroptosis. Crucially, adeno-associated virus serotype 8 (AAV8)-mediated TFEB knockdown in mice conferred protection against IPAs-induced liver injury and attenuated hepatocyte ferroptosis. These findings establish TFEB-mediated NCOA4-dependent ferritinophagy and ferroptosis as central mechanisms in IPAs-induced hepatotoxicity, thereby identifying TFEB as a promising therapeutic target for mitigating IPAs-induced liver damage. This study provides critical insights into the molecular mechanisms of IPAs-induced liver injury and offers a novel strategy for therapeutic intervention.
Collapse
Affiliation(s)
- Junqi Wang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Xinhao Song
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yitong Hui
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Bin Dong
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Jiahao Gong
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Yuan Zhao
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Hui Ji
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Jiangsu Jiyu Biopharm Technology Co., Ltd, No. 8 Xingzhi Road, Nanjing, 211899, PR China
| | - Yawei Qiu
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Shanxiang Jiang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Dawei Guo
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Xiuge Gao
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China; Center for Veterinary Drug Research and Evaluation, Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China.
| |
Collapse
|
3
|
Cao P, Jaeschke H, Ni HM, Ding WX. The Ways to Die: Cell Death in Liver Pathophysiology. Semin Liver Dis 2025. [PMID: 40199509 DOI: 10.1055/a-2576-4332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Liver diseases are closely associated with various cell death mechanisms, including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis. Each process contributes uniquely to the pathophysiology of liver injury and repair. Importantly, these mechanisms are not limited to hepatocytes; they also significantly involve nonparenchymal cells. This review examines the molecular pathways and regulatory mechanisms underlying these forms of cell death in hepatocytes, emphasizing their roles in several liver diseases, such as ischemia-reperfusion injury, metabolic dysfunction-associated steatotic liver disease, drug-induced liver injury, and alcohol-associated liver disease. Recent insights into ferroptosis and pyroptosis may reveal novel therapeutic targets for managing liver diseases. This review aims to provide a comprehensive overview of these cell death mechanisms in the context of liver diseases, detailing their molecular signaling pathways and implications for potential treatment strategies.
Collapse
Affiliation(s)
- Peng Cao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
- Division of Gastroenterology, Hepatology and Mobility, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
4
|
Pfau DJ, Bryk R. High throughput screening assay for the identification of ATF4 and TFEB activating compounds. AUTOPHAGY REPORTS 2025; 4:2473765. [PMID: 40265045 PMCID: PMC11980509 DOI: 10.1080/27694127.2025.2473765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 04/24/2025]
Abstract
Macrophages act to defend against infection, but can fail to completely prevent bacterial replication and dissemination in an immunocompetent host. Recent studies have shown that activation of a host transcription factor, TFEB, a regulator of lysosomal biogenesis, could restrict intramacrophage replication of the human pathogen Mycobacterium tuberculosis and synergize with suboptimal levels of the antibiotic rifampin to reduce bacterial loads. Currently available small molecule TFEB activators lack selectivity and potency, but could be potentially useful in a variety of pathological conditions with suboptimal lysosomal activity. TFEB nuclear translocation and activation depend on its phosphorylation status which is controlled by multiple cellular pathways. We devised a whole cell, high throughput screening assay to identify small molecules that activate TFEB by establishing a stably transfected HEK293T reporter cell line for ATF4, a basic leucine zipper transcription factor induced by stress response and activated in parallel to TFEB. We optimized its use in vitro using compounds that target endoplasmic reticulum stress and intracellular calcium signaling. We report results from screening the commercially available LOPAC library and the Selleck Chemicals library modified to include only FDA-approved drugs and clinical research compounds. We identified twenty-one compounds across six clinical use categories that activate ATF4, and confirmed that two proteasome inhibitors promote TFEB activation. The results of this study provide an assay that could be used to screen for small molecules that activate ATF4 and TFEB and a potential list of compounds identified as activators of the ATF4 transcription factor in response to cellular stress.
Collapse
Affiliation(s)
- Daniel J. Pfau
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Ruslana Bryk
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
5
|
Wei Y, Zhang Y, Cao W, Cheng N, Xiao Y, Zhu Y, Xu Y, Zhang L, Guo L, Song J, Sha SH, Shao B, Ma F, Yang J, Ying Z, He Z, Chai R, Fang Q, Yang J. RONIN/HCF1-TFEB Axis Protects Against D-Galactose-Induced Cochlear Hair Cell Senescence Through Autophagy Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2407880. [PMID: 39985193 DOI: 10.1002/advs.202407880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/17/2025] [Indexed: 02/24/2025]
Abstract
Age-related hearing loss is characterized by senescent inner ear hair cells (HCs) and reduced autophagy. Despite the improved understanding of these processes, detailed molecular mechanisms underlying cochlear HC senescence remain unclear. Transcription Factor EB (TFEB), a key regulator of genes associated with autophagy and lysosomes, crucially affects aging-related illnesses. However, intricate regulatory networks that influence TFEB activity remain to be thoroughly elucidated. The findings revealed that RONIN (THAP11), through its interaction with host cell factor C1 (HCF1/HCFC1), modulated the transcriptional activity of Tfeb, thus contributing to the mitigation (D-galatactose [D-gal]) senescent HC loss. Specifically, RONIN overexpression improved autophagy levels and lysosomal activity and attenuated changes associated with the senescence of HCs triggered by D-gal. These findings highlight the possibility of using RONIN as a viable therapeutic target to ameliorate presbycusis by enhancing the TFEB function.
Collapse
Affiliation(s)
- Yongjie Wei
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yuhua Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Wei Cao
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Nan Cheng
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yun Xiao
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yongjun Zhu
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yan Xu
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Lei Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Lingna Guo
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jun Song
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Su-Hua Sha
- Department of Pathology and Laboratory Medicine, The Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Buwei Shao
- School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Fang Ma
- Center for Scientific Research of Anhui Medical University, Hefei, 230032, China
| | - Jingwen Yang
- International Department of Hefei 168 High School, Hefei, 230601, China
| | - Zheng Ying
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration Nantong University, Nantong, 226001, China
- Department of Neurology, Aerospace Center Hospital, School of Life Science Beijing Institute of Technology, Beijing, 100081, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China
| | - Qiaojun Fang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jianming Yang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| |
Collapse
|
6
|
Lin J, Yuan Y, Huang C, Zi J, Li L, Liu J, Wu X, Li W, Zhao Q, Li Y, Liu Z, Diao A. TFEB agonist clomiphene citrate activates the autophagy-lysosomal pathway and ameliorates Alzheimer's disease symptoms in mice. J Biol Chem 2024; 300:107929. [PMID: 39454957 PMCID: PMC11599454 DOI: 10.1016/j.jbc.2024.107929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Autophagy is a conserved eukaryotic cellular clearance and recycling process through the lysosome-mediated degradation of damaged organelles and protein aggregates to maintain homeostasis. Impairment of the autophagy-lysosomal pathway is implicated in the pathogenesis of Alzheimer's disease (AD). Transcription factor EB (TFEB) is a master regulator of autophagy and lysosomal biogenesis. Therefore, activating TFEB and autophagy provides a novel strategy for AD treatment. We previously described that clomiphene citrate (CC) promotes nuclear translocation of TFEB and increases autophagy and lysosomal biogenesis. In this study, 7- and 3-month-old APP/PS1 mice were treated with TFEB agonist CC and assessed. The behavioral tests were performed using Morris water maze and open field test. Additional changes in amyloid-β pathology, autophagy, and inflammatory response were determined. We found that CC activated TFEB and the autophagy-lysosomal pathway in neuronal cells. Moreover, using mouse model of Alzheimer's disease, CC treatment promoted clearance of amyloid-β plaques and ameliorated cognitive function in both 7- and 3-month-old APP/PS1 mice. The CC-induced activation of TFEB occurs by promoting acetylation of TFEB for nuclear translocation. These findings provide a molecular mechanism for the TFEB-mediated activation of the autophagy-lysosome pathway by CC, which has the potential to be repurposed and applied in the treatment or prevention of AD.
Collapse
Affiliation(s)
- Jieru Lin
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yi Yuan
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Chunhuan Huang
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Jiayu Zi
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Lu Li
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Jiamiao Liu
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xiaoting Wu
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Wei Li
- School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Qing Zhao
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yuyin Li
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Zhenxing Liu
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| | - Aipo Diao
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
7
|
Chen H, Hinz K, Zhang C, Rodriguez Y, Williams SN, Niu M, Ma X, Chao X, Frazier AL, McCarson KE, Wang X, Peng Z, Liu W, Ni HM, Zhang J, Swerdlow RH, Ding WX. Late-Life Alcohol Exposure Does Not Exacerbate Age-Dependent Reductions in Mouse Spatial Memory and Brain TFEB Activity. Biomolecules 2024; 14:1537. [PMID: 39766244 PMCID: PMC11673978 DOI: 10.3390/biom14121537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Alcohol consumption is believed to affect Alzheimer's disease (AD) risk, but the contributing mechanisms are not well understood. A potential mediator of the proposed alcohol-AD connection is autophagy, a degradation pathway that maintains organelle and protein homeostasis. Autophagy is regulated through the activity of Transcription factor EB (TFEB), which promotes lysosome and autophagy-related gene expression. The purpose of this study is to explore whether chronic alcohol consumption worsens the age-related decline in TFEB-mediated lysosomal biogenesis in the brain and exacerbates cognitive decline associated with aging. To explore the effect of alcohol on brain TFEB and autophagy, we exposed young (3-month-old) and aged (23-month-old) mice to two alcohol-feeding paradigms and assessed biochemical, transcriptome, histology, and behavioral endpoints. In young mice, alcohol decreased hippocampal nuclear TFEB staining but increased SQSTM1/p62, LC3-II, ubiquitinated proteins, and phosphorylated Tau. Hippocampal TFEB activity was lower in aged mice than it was in young mice, and Gao-binge alcohol feeding did not worsen the age-related reduction in TFEB activity. Morris Water and Barnes Maze spatial memory tasks were used to characterize the effects of aging and chronic alcohol exposure (mice fed alcohol for 4 weeks). The aged mice showed worse spatial memory acquisition in both tests. Alcohol feeding slightly impaired spatial memory in the young mice, but had little effect or even slightly improved spatial memory acquisition in the aged mice. In conclusion, aging produces greater reductions in brain autophagy flux and impairment of spatial memory than alcohol consumption.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Kaitlyn Hinz
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Chen Zhang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Yssa Rodriguez
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Sha Neisha Williams
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Mengwei Niu
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Alexandria L. Frazier
- R.L. Smith IDDRC Rodent Behavior Facility, Disease Model and Assessment Services, The University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Kenneth E. McCarson
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
- R.L. Smith IDDRC Rodent Behavior Facility, Disease Model and Assessment Services, The University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Xiaowan Wang
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (X.W.); (R.H.S.)
| | - Zheyun Peng
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, MI 48201, USA; (Z.P.); (W.L.)
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, MI 48201, USA; (Z.P.); (W.L.)
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Jianhua Zhang
- Department of Pathology, Division of Molecular Cellular Pathology, University of Alabama at Birmingham, 901 19th Street South, Birmingham, AL 35294, USA;
| | - Russell H. Swerdlow
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (X.W.); (R.H.S.)
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
- Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Chen YJ, Guo ZT, Chen HQ, Zhang SF, Bao YX, Xie Z, Ke JL, Ye WJ, Liang JC, Chen JC, Li N, Zheng FX, Liao H, Wu T, Pang JX. Salinomycin, a potent inhibitor of XOD and URAT1, ameliorates hyperuricemic nephropathy by activating NRF2, modulating the gut microbiota, and promoting SCFA production. Chem Biol Interact 2024; 403:111220. [PMID: 39222901 DOI: 10.1016/j.cbi.2024.111220] [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: 04/10/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Long-term hyperuricemia can induce kidney damage, clinically referred to as hyperuricemic nephropathy (HN), which is characterized by renal fibrosis, inflammation, and oxidative stress. However, currently used uric acid-lowering drugs are not capable of protecting the kidneys from damage. Therefore, uric acid-lowering drugs that can also protect the kidneys are urgently needed. In this study, we first discovered that salinomycin, an antibiotic, can regulate uric acid homeostasis and ameliorate kidney damage in mice with HN. Mechanistically, salinomycin inhibited serum and hepatic xanthine oxidase (XOD) activities and downregulated renal urate transporter 1 (URAT1) expression and transport activity, thus exerting uric acid-lowering effects in mice with HN. Furthermore, we found that salinomycin promoted p-NRF2 Ser40 expression, resulting in increased nuclear translocation of NRF2 and activation of NRF2. More importantly, salinomycin affected the gut microbiota and promoted the generation of short-chain fatty acids (SCFAs) in mice with HN. In conclusion, our results revealed that salinomycin maintains uric acid homeostasis and alleviates kidney injury in mice with HN by multiple mechanisms, suggesting that salinomycin might be a desirable candidate for HN treatment in the clinic.
Collapse
Affiliation(s)
- Yong-Jun Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zi-Tao Guo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hai-Qiao Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shi-Fan Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying-Xia Bao
- Baiyunshan Pharmaceutical General Factory, Guangzhou Baiyunshan Pharmaceutical Holdings Co., Ltd., Guangzhou, 510515, China; Key Laboratory of Key Technology Research on Chemical Raw Materials and Preparations of Guangdong Province, Guangzhou, 510515, China
| | - Zhoufan Xie
- Baiyunshan Pharmaceutical General Factory, Guangzhou Baiyunshan Pharmaceutical Holdings Co., Ltd., Guangzhou, 510515, China; Key Laboratory of Key Technology Research on Chemical Raw Materials and Preparations of Guangdong Province, Guangzhou, 510515, China
| | - Jia-le Ke
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Jie Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Cheng Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Chen Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ning Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Feng-Xin Zheng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hui Liao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ting Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Jian-Xin Pang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
9
|
Deng X, Li Y, Chen Y, Hu Q, Zhang W, Chen L, Lu X, Zeng J, Ma X, Efferth T. Paeoniflorin protects hepatocytes from APAP-induced damage through launching autophagy via the MAPK/mTOR signaling pathway. Cell Mol Biol Lett 2024; 29:119. [PMID: 39244559 PMCID: PMC11380789 DOI: 10.1186/s11658-024-00631-4] [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: 03/18/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Drug-induced liver injury (DILI) is gradually becoming a common global problem that causes acute liver failure, especially in acute hepatic damage caused by acetaminophen (APAP). Paeoniflorin (PF) has a wide range of therapeutic effects to alleviate a variety of hepatic diseases. However, the relationship between them is still poorly investigated in current studies. PURPOSE This work aimed to explore the protective effects of PF on APAP-induced hepatic damage and researched the potential molecular mechanisms. METHODS C57BL/6J male mice were injected with APAP to establish DILI model and were given PF for five consecutive days for treatment. Aiming to clarify the pharmacological effects, the molecular mechanisms of PF in APAP-induced DILI was elucidated by high-throughput and other techniques. RESULTS The results demonstrated that serum levels of ALP, γ-GT, AST, TBIL, and ALT were decreased in APAP mice by the preventive effects of PF. Moreover, PF notably alleviated hepatic tissue inflammation and edema. Meanwhile, the results of TUNEL staining and related apoptotic factors coincided with the results of transcriptomics, suggesting that PF inhibited hepatocyte apoptosis by regulated MAPK signaling. Besides, PF also acted on reactive oxygen species (ROS) to regulate the oxidative stress for recovery the damaged mitochondria. More importantly, transmission electron microscopy showed the generation of autophagosomes after PF treatment, and PF was also downregulated mTOR and upregulated the expression of autophagy markers such as ATG5, ATG7, and BECN1 at the mRNA level and LC3, p62, ATG5, and ATG7 at the protein level, implying that the process by which PF exerted its effects was accompanied by the occurrence of autophagy. In addition, combinined with molecular dynamics simulations and western blotting of MAPK, the results suggested p38 as a direct target for PF on APAP. Specifically, PF-activated autophagy through the downregulation of MAPK/mTOR signaling, which in turn reduced APAP injury. CONCLUSIONS Paeoniflorin mitigated liver injury by activating autophagy to suppress oxidative stress and apoptosis via the MAPK/mTOR signaling pathway. Taken together, our findings elucidate the role and mechanism of paeoniflorin in DILI, which is expected to provide a new therapeutic strategy for the development of paeoniflorin.
Collapse
Affiliation(s)
- Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lisheng Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, 55128, Germany.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, 55128, Germany.
| |
Collapse
|
10
|
Hinz K, Niu M, Ni HM, Ding WX. Targeting Autophagy for Acetaminophen-Induced Liver Injury: An Update. LIVERS 2024; 4:377-387. [PMID: 39301093 PMCID: PMC11412313 DOI: 10.3390/livers4030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Acetaminophen (APAP) overdose can induce hepatocyte necrosis and acute liver failure in experimental rodents and humans. APAP is mainly metabolized via hepatic cytochrome P450 enzymes to generate the highly reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI), which forms acetaminophen protein adducts (APAP-adducts) and damages mitochondria, triggering necrosis. APAP-adducts and damaged mitochondria can be selectively removed by autophagy. Increasing evidence implies that the activation of autophagy may be beneficial for APAP-induced liver injury (AILI). In this minireview, we briefly summarize recent progress on autophagy, in particular, the pharmacological targeting of SQSTM1/p62 and TFEB in AILI.
Collapse
Affiliation(s)
- Kaitlyn Hinz
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Mengwei Niu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
11
|
Chen H, Hinz K, Zhang C, Rodriguez Y, Williams SN, Niu M, Ma X, Chao X, Frazier AL, McCarson KE, Wang X, Peng Z, Liu W, Ni HM, Zhang J, Swerdlow RH, Ding WX. Late-Life Alcohol Exposure Does Not Exacerbate Age-Dependent Reductions in Mouse Spatial Memory and Brain TFEB Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581774. [PMID: 38464149 PMCID: PMC10925107 DOI: 10.1101/2024.02.23.581774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Alcohol consumption is believed to affect Alzheimer's disease (AD) risk, but the contributing mechanisms are not well understood. A potential mediator of the proposed alcohol-AD connection is autophagy, a degradation pathway that maintains organelle and protein homeostasis. Autophagy is in turn regulated through the activity of Transcription factor EB (TFEB), which promotes lysosome and autophagy-related gene expression. To explore the effect of alcohol on brain TFEB and autophagy, we exposed young (3-month old) and aged (23-month old) mice to two alcohol-feeding paradigms and assessed biochemical, transcriptome, histology, and behavioral endpoints. In young mice, alcohol decreased hippocampal nuclear TFEB staining but increased SQSTM1/p62, LC3-II, ubiquitinated proteins, and phosphorylated Tau. Hippocampal TFEB activity was lower in aged mice than it was in young mice, and Gao-binge alcohol feeding did not worsen the age-related reduction in TFEB activity. To better assess the impact of chronic alcohol exposure, we fed young and aged mice alcohol for four weeks before completing Morris Water and Barnes Maze spatial memory testing. The aged mice showed worse spatial memory on both tests. While alcohol feeding slightly impaired spatial memory in the young mice, it had little effect or even slightly improved spatial memory in the aged mice. These findings suggest that aging is a far more important driver of spatial memory impairment and reduced autophagy flux than alcohol consumption.
Collapse
|