1
|
Zhou S, Peng L, Wang Y, Cheng D, Li Z, Xiong H, Wang T, Liu Y, Jia Z, Sun W, Ni C. Bazibushen attenuates fibroblast senescence in silica-induced pulmonary fibrosis via FOXO1/PINK1/Parkin Axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156714. [PMID: 40215811 DOI: 10.1016/j.phymed.2025.156714] [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: 11/22/2024] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Silicosis, an age-related disease, is still a heavy burden on global occupational health. Emerging evidence has revealed that targeting senescent cells may be a promising therapeutic strategy for silicosis. This study was designed to investigate the novel function of Bazibushen (BZBS), a known anti-aging drug, in improving silica-induced lung fibrosis. We first confirmed the accumulation of senescent fibroblasts in the fibrotic regions of silicotic lungs. In both young (6-8 weeks) and aged (12 months) silicotic mice, BZBS exhibited anti-fibrosis and anti-senescence effects. Results of in vitro experiments showed the ability of BZBS to block the expression of p21, fibrotic markers, and senescence-associated secretory phenotype factors. Furthermore, BZBS was observed to attenuate mitochondrial dysfunction in senescent fibroblasts through FOXO1/PINK1/Parkin signaling. Collectively, these results indicated BZBS as a potential anti-fibrosis agent, which exerted its role through maintaining mitochondrial homeostasis in senescent fibroblasts.
Collapse
Affiliation(s)
- Siyun Zhou
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Lan Peng
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Yue Wang
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Demin Cheng
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Ziwei Li
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Haojie Xiong
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Ting Wang
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Yi Liu
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Zhenhua Jia
- Hebei Yiling Hospital, High-level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, 050091, Hebei, China; National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, Hebei, China.
| | - Wenqing Sun
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi Medical Center, Nanjing medical university, Wuxi, China.
| | - Chunhui Ni
- Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
2
|
Li S, Xu H, Liu S, Hou J, Han Y, Li C, Li Y, Zheng G, Wei Z, Yang F, Gao S, Wang S, Geng J, Dai H, Wang C. Targeting Lp-PLA2 inhibits profibrotic monocyte-derived macrophages in silicosis through restoring cardiolipin-mediated mitophagy. Cell Mol Immunol 2025:10.1038/s41423-025-01288-5. [PMID: 40389600 DOI: 10.1038/s41423-025-01288-5] [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: 06/16/2024] [Accepted: 04/08/2025] [Indexed: 05/21/2025] Open
Abstract
Monocyte-derived macrophages (MoMacs) are the most important effector cells that cause pulmonary fibrosis. However, the characteristics of MoMac differentiation in silicosis and the mechanisms by which MoMacs affect the progression of pulmonary fibrosis remain unclear. Integration of single-cell and spatial transcriptomic analyses revealed that the silicosis niche was occupied by a subset of MoMacs, identified as Spp1hiMacs, which remain in an immature transitional state of differentiation during silicosis. This study investigated the mechanistic foundations of mitochondrial damage induced by the lipoprotein-associated phospholipase A2 (Lp-PLA2, encoded by Pla2g7)-acyl-CoA:lysocardiolipin acyltransferase-1 (ALCAT1)-cardiolipin (CL) signaling pathway, which interferes with Spp1hiMac differentiation. We demonstrated that in SiO2-induced MoMacs, Lp-PLA2 induces abnormal CL acylation through the activation of ALCAT1, resulting in impaired mitochondrial localization of PINK1 and LC3B and mitochondrial autophagy defects. Simultaneously, lysosomal dysfunction causes the release of the lysosomal protein cathepsin B into the cytoplasm, which involves M1 and M2 macrophage polarization and the activation of proinflammatory and profibrotic pathways. Furthermore, we assessed the efficacy of the Lp-PLA2 inhibitor darapladib in ameliorating silica-induced pulmonary fibrosis in a murine model. Our findings enhance our understanding of silicosis pathogenesis and offer promising opportunities for developing targeted therapies to mitigate fibrotic progression and maintain lung function in affected individuals.
Collapse
Affiliation(s)
- Shifeng Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Hong Xu
- Health Science Center, Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei, China
| | - Shupeng Liu
- School of Public Health, Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei, China
| | - Jinkun Hou
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking Union Medical College, Beijing, China
- Immune Dysfunction and Pulmonary Fibrosis Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yueyin Han
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking Union Medical College, Beijing, China
| | - Chen Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
- Immune Dysfunction and Pulmonary Fibrosis Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yupeng Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
- Second Affiliated Hospital of Harbin Medical University, Respiratory and Critical Care Medicine, Harbin, Heilongjiang, China
| | - Gaigai Zheng
- School of Public Health, Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei, China
| | - Zhongqiu Wei
- Basic Medical College, Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei, China
| | - Fang Yang
- School of Public Health, Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei, China
| | - Shuwei Gao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
- Immune Dysfunction and Pulmonary Fibrosis Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Shiyao Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
- Immune Dysfunction and Pulmonary Fibrosis Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Jing Geng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China.
- Immune Dysfunction and Pulmonary Fibrosis Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China.
| | - Huaping Dai
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China.
- Peking Union Medical College, Beijing, China.
- Immune Dysfunction and Pulmonary Fibrosis Joint Laboratory for Clinical Medicine, Capital Medical University, Beijing, China.
| | - Chen Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China.
- Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Wang S, He S, Hu X, Liu F, Fang X, Huang P. Nrf2 mediated signaling axis in sepsis-induced cardiomyopathy: potential Pharmacological receptor. Inflamm Res 2025; 74:76. [PMID: 40299042 DOI: 10.1007/s00011-025-02037-0] [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: 10/18/2024] [Revised: 03/30/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Sepsis has emerged as the most pressing health concerns globally in emergency and intensive care unit. Sepsis-Induced Cardiomyopathy (SIC) represents an acute cardiac insufficiency syndrome secondary to sepsis, characterized by a high incidence and a significant increase in mortality among sepsis patients. To date, no specific treatment exists for this condition. In recent years, mounting evidence has indicated that Nrf2 plays a critical protective role in SIC and may represent a potential therapeutic target. METHODS Pubmed database literature was searched for studies pertaining to the role of Nrf2 in sepsis, from the inception of the database to October 1, 2024. Biorender software was performed to draw the corresponding mechanism diagram. RESULTS Using the keywords "Nrf2 and Sepsis", we initially identified 454 articles. To refine our search, we employed "Nrf2 and Sepsis and Cardiac" as keywords, yielding 63 articles. Upon reviewing the full texts, we selected 26 studies for inclusion in our review. Nrf2 is implicated in various protective aspects against cardiomyocyte injury stemming from sepsis, including its inhibitory effects on inflammation, apoptosis, mitochondrial dysfunction, pyroptosis, and ferroptosis. 23 natural compounds under investigation for this application were identified. CONCLUSION The Nrf2-mediated signaling pathway plays a critical role in sepsis-induced myocardial injury. Given the complex, systemic, and multifactorial nature of sepsis, these natural compounds should be regarded as adjunctive therapeutic options for scholarly investigation rather than standalone therapeutic interventions. Substantial future research will still be required to validate their clinical efficacy and mechanistic roles.
Collapse
Affiliation(s)
- Sumei Wang
- Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Affiliated with Capital Medical University, Beijing, China
- Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Xiao Hu
- Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Fusheng Liu
- Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Xiaolei Fang
- Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China.
| | - Po Huang
- Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
4
|
Peng M, Jiang C, Dai Z, Xie B, Chen Q, Lin J. Identification and verification of mitochondria-related genes biomarkers associated with immune infiltration for COPD using WGCNA and machine learning algorithms. Sci Rep 2025; 15:14347. [PMID: 40274954 PMCID: PMC12022275 DOI: 10.1038/s41598-025-99002-y] [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: 10/12/2024] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Mitochondrial dysfunction plays a pivotal role in the pathogenesis of chronic obstructive pulmonary disease (COPD). This study combines bioinformatics analysis with machine learning to elucidate potential key mitochondrial-related genes associated with COPD and its immune microenvironment. We utilized the limma package and Weighted Gene Co-expression Network Analysis (WGCNA) to analyze datasets from the Gene Expression Omnibus (GEO) database (GSE57148), identifying 12 key differentially expressed mitochondrial genes (MitoDEGs). Using 12 distinct machine learning algorithms (comprising 143 predictive models), we identified the optimal diagnostic model, which includes five pivotal MitoDEGs: ERN1, FASTK, HIGD1B, NDUFA7 and NDUFB7. The diagnostic specificity and sensitivity of each gene, as well as the diagnostic model itself, were evaluated using Receiver operating characteristic (ROC) curves. This model demonstrated high specificity in the validation cohorts (GSE76925, GSE151052, GSE239897). Expression analysis revealed upregulation of ERN1 and downregulation of FASTK, HIGD1B, NDUFA7 and NDUFB7 in COPD patients. Spearman's correlation analysis indicated a significant association between MitoDEGs and immune cell infiltration, with ERN1 expression positively correlated with neutrophil infiltration and the other genes negatively correlated. The GABA receptor modulator androstenol was identified as a potential therapeutic candidate. In vivo studies confirmed reduced mRNA expression of HIGD1B and NDUFB7 in COPD mice. These findings elucidate mitochondrial-immune interactions in COPD and highlight novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Meijuan Peng
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Chen Jiang
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Ziyu Dai
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Bin Xie
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jianing Lin
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.
| |
Collapse
|
5
|
Zhang Y, Lou Q, Lian H, Yang R, Cui R, Wang L, Ma B, Hou L, Jin L, Teng W. sLithospermic acid etched ZIF-8 nanoparticles delays osteoarthritis progression by inhibiting inflammatory signaling pathways and rescuing mitochondrial damage. Mater Today Bio 2025; 31:101589. [PMID: 40104643 PMCID: PMC11919455 DOI: 10.1016/j.mtbio.2025.101589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/27/2025] [Accepted: 02/16/2025] [Indexed: 03/20/2025] Open
Abstract
Osteoarthritis (OA) is the most common chronic inflammatory joint disease. Improving the joint inflammatory microenvironment is expected to promote early intervention and delay the progression of OA. However, effective strategies for inhibiting OA-related joint inflammation are still lacking. Lithospermic acid (LA), a polycyclic phenol carboxylic acid extracted from salvia miltiorrhiza, has strong anti-inflammatory and antioxidant effects. However, its role in the treatment of OA and the underlying mechanisms are unclear. To improve the bioavailability of LA, an LA synergistic protects etched zeolitic imidazolate framework (ZIF)-8 nanoparticles (LA@ZIF-8) was designed and developed for targeted delivery to modulate the inflammatory microenvironment in OA. This study confirmed that LA@ZIF-8 inhibits the pro-inflammatory phenotype of RAW264.7 macrophages through the NF-ĸB signaling pathway, effectively alleviates mitochondrial dysfunction, and delays articular cartilage degeneration caused by the joint inflammatory microenvironment mediated by synoval macrophages. In summary, LA@ZIF-8 delays the progression of OA by inhibiting synovial macrophage-mediated inflammatory responses, highlighting its clinical application potential.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Qiqi Lou
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Hao Lian
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Ran Yang
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Ruolin Cui
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Leyang Wang
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Bitao Ma
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Lingli Hou
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Lilun Jin
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Weiran Teng
- Department of Traditional Chinese Medicine, School of Medicine, Xinhua Hospital, Afffliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| |
Collapse
|
6
|
Zhang D, Li F, Sun C, Chen C, Qin H, Wu X, Jiang M, Zhou K, Yao C, Hu Y. Inhibition of PGAM5 hyperactivation reduces neuronal apoptosis in PC12 cells and experimental vascular dementia rats. Arch Gerontol Geriatr 2025; 131:105732. [PMID: 39754994 DOI: 10.1016/j.archger.2024.105732] [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: 08/12/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
PURPOSE The incidence of vascular dementia (VaD), as one of the main types of dementia in old age, has been increasing year by year, and exploring its pathogenesis and seeking practical and effective treatment methods are undoubtedly the key to solving this problem. Phosphoglycerate translocase 5 (PGAM5), as a crossroads of multiple signaling pathways, can lead to mitochondrial fission, which in turn triggers the onset and development of necroptosis, and thus PGAM5 may be a novel target for the prevention and treatment of vascular dementia. METHODS Animal model of vascular dementia was established by Two-vessel occlusion (2-VO) method, and cellular model of vascular dementia was established by oxygen glucose deprivation (OGD) method. Neuronal damage was detected in vivo and in vitro in different groups using different concentrations of the PGAM5-specific inhibitor LFHP-1c, and necroptosis and mitochondrial dynamics-related factors were determined. RESULTS In vivo experiments, 10 mg/kg-1 and 20 mg/kg-1 LFHP-1c improved cognitive deficits, reduced neuronal edema and vacuoles, increased the number of nissl bodies, and it could modulate the expression of Caspase family and Bcl-2 family related proteins and mRNAs and ameliorate neuronal damage. Simultaneously, in vitro experiments, 5 μM, 10 μM and 20 μM LFHP-1c increased the activity and migration number of model cells, reduced the number of apoptotic cells, ameliorated the excessive accumulation of intracellular reactive oxygen species, inhibited the over-activation of caspase-family and Bcl-2-family related proteins and mRNAs, and improved the mitochondrial dynamics of the fission and fusion states. Moreover, in vivo and in vitro experiments have shown that LFHP-1c can also upregulate the expression level of BDNF, inhibit the expression content of TNF-α and ROS, regulate the expression of proteins and mRNAs related to the RIPK1/RIPK3/MLKL pathway and mitochondrial dynamics, and reduce neuronal apoptosis. CONCLUSIONS Inhibition of PGAM5 expression level can reduce neuronal damage caused by chronic cerebral ischemia and hypoxia, which mainly prevents necroptosis by targeting the RIPK1/RIPK3/MLKL signaling pathway and regulates the downstream mitochondrial dynamics homeostasis system to prevent excessive mitochondrial fission, thus improving cognition and exerting cerebroprotective effects.
Collapse
Affiliation(s)
- Ding Zhang
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China; Neurology Ward 1, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Fangcun Li
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China; Neurology Ward 1, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Chunying Sun
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Canrong Chen
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Hongling Qin
- Neurology Ward 1, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Xuzhou Wu
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Minghe Jiang
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Keqing Zhou
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China
| | - Chun Yao
- School of Doctoral Studies, Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China.
| | - Yueqiang Hu
- Neurology Ward 1, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Qingxiu District, Nanning, 530001, China.
| |
Collapse
|
7
|
Dong Z, Wang X, Wang P, Bai M, Wang T, Chu Y, Qin Y. Idiopathic Pulmonary Fibrosis Caused by Damaged Mitochondria and Imbalanced Protein Homeostasis in Alveolar Epithelial Type II Cell. Adv Biol (Weinh) 2025; 9:e2400297. [PMID: 39390651 PMCID: PMC12001015 DOI: 10.1002/adbi.202400297] [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: 05/30/2024] [Revised: 08/21/2024] [Indexed: 10/12/2024]
Abstract
Alveolar epithelial Type II (ATII) cells are closely associated with early events of Idiopathic pulmonary fibrosis (IPF). Proteostasis dysfunction, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction are known causes of decreased proliferation of alveolar epithelial cells and the secretion of pro-fibrotic mediators. Here, a large body of evidence is systematized and a cascade relationship between protein homeostasis, endoplasmic reticulum stress, mitochondrial dysfunction, and fibrotropic cytokines is proposed, providing a theoretical basis for ATII cells dysfunction as a possible pathophysiological initiating event for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhaoxiong Dong
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Xiaolong Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
| | - Peiwen Wang
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Mingjian Bai
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Tianyu Wang
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Yanhui Chu
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Yan Qin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
| |
Collapse
|
8
|
Liu S, Xi Q, Li X, Liu H. Mitochondrial dysfunction and alveolar type II epithelial cell senescence: The destroyer and rescuer of idiopathic pulmonary fibrosis. Front Cell Dev Biol 2025; 13:1535601. [PMID: 40230412 PMCID: PMC11994736 DOI: 10.3389/fcell.2025.1535601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic respiratory disease with an unknown origin and complex pathogenic mechanisms. A deeper understanding of these mechanisms is essential for effective treatment. Pulmonary fibrosis is associated with the senescence of alveolar type II epithelial (ATⅡ) cells. Additionally, ATⅡ senescence can lead to a senescence-associated secretory phenotype, which affects cellular communication and disrupts lung tissue repair, contributing to the development of IPF. The role of mitochondrial dysfunction in senescence-related diseases is increasingly recognized. It can induce ATⅡ senescence through apoptosis, impaired autophagy, and disrupted energy metabolism, potentially playing a key role in IPF progression. This article explores the therapeutic potential of targeting cellular senescence and mitochondrial dysfunction, emphasizing their significant roles in IPF pathogenesis.
Collapse
Affiliation(s)
- Suqi Liu
- The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qian Xi
- Six Sections of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xuannian Li
- The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huaman Liu
- Six Sections of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
9
|
Li X, Nie M, Yang K, Qi X, Yang L. Association between exposure to a mixture of dichlorophenol and parabens and lung function in adults from NHANES, 2007-2012. Heart Lung 2025; 72:32-41. [PMID: 40121927 DOI: 10.1016/j.hrtlng.2025.03.006] [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: 12/17/2024] [Revised: 02/02/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Few studies have examined the effects of dichlorophenol and parabens on lung function, with most focusing on individual chemicals. OBJECTIVES This study analyzes the effects of these chemicals on single or mixed exposures on lung function in adults. METHODS We utilized data from the 2007-2012 National Health and Nutrition Examination Survey (NHANES), focusing on urinary levels of dichlorophenol, parabens, and lung function measures. The generalized linear regression (GLM), weighted quantile sum (WQS) regression, and Bayesian kernel machine regression (BKMR) models were applied to assess the impact of chemical exposure on lung function. RESULTS A total of 2,599 adults with complete data were included in the analysis. Detection rates exceeding 75 % for 2,5-Dichlorophenol (2,5-DCP), 2,4-Dichlorophenol (2,4-DCP), methyl paraben, and propyl paraben were observed. In GLMs, 2,5-DCP was significantly negatively associated with forced vital capacity (FVC) and forced expiratory volume in 1 s (FEV1), either as a continuous (FVC: β = -0.012, 95 % CI = -0.018, -0.006; FEV1: β = -0.010, 95 % CI = -0.017, -0.004) or quartile variable (FVC: β = -0.027, 95 % CI = -0.039, -0.016; FEV1: β = -0.024, 95 % CI = -0.036, -0.011). The WQS regression confirmed a negative effect of the chemical mixture on lung function, with significant associations for both FVC (β = -0.013, 95 % CI = -0.018, -0.008) and FEV1 (β = -0.013, 95 % CI = -0.017, -0.007). These results were consistent in BKMR models. CONCLUSION Exposure to dichlorophenol and parabens, either individually or in combination, is associated with decreased lung function in humans.
Collapse
Affiliation(s)
- Xinwei Li
- Department of Geriatrics, Shanghai Fourth People's Hospital affiliated to Tongji University, Shanghai, China
| | - Meiyun Nie
- Department of Geriatrics, Shanghai Fourth People's Hospital affiliated to Tongji University, Shanghai, China
| | - Keke Yang
- Department of Geriatrics, Shanghai Fourth People's Hospital affiliated to Tongji University, Shanghai, China
| | - Xiaodong Qi
- Department of Geriatrics, Shanghai Fourth People's Hospital affiliated to Tongji University, Shanghai, China
| | - Ling Yang
- Department of Geriatrics, Shanghai Fourth People's Hospital affiliated to Tongji University, Shanghai, China.
| |
Collapse
|
10
|
Jia G, Song E, Huang Q, Chen M, Liu G. Mitochondrial fusion protein: a new therapeutic target for lung injury diseases. Front Physiol 2025; 16:1500247. [PMID: 40177356 PMCID: PMC11962016 DOI: 10.3389/fphys.2025.1500247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Mitochondria are essential organelles responsible for cellular energy supply. The maintenance of mitochondrial structure and function relies heavily on quality control systems, including biogenesis, fission, and fusion. Mitochondrial fusion refers to the interconnection of two similar mitochondria, facilitating the exchange of mitochondrial DNA, metabolic substrates, proteins, and other components. This process is crucial for rescuing damaged mitochondria and maintaining their normal function. In mammals, mitochondrial fusion involves two sequential steps: outer membrane fusion, regulated by mitofusin 1 and 2 (MFN1/2), and inner membrane fusion, mediated by optic atrophy 1 (OPA1). Dysfunction in mitochondrial fusion has been implicated in the development of various acute and chronic lung injuries. Regulating mitochondrial fusion, maintaining mitochondrial dynamics, and improving mitochondrial function are effective strategies for mitigating lung tissue and cellular damage. This study reviews the expression and regulatory mechanisms of mitochondrial fusion proteins in lung injuries of different etiologies, explores their relationship with lung injury diseases, and offers a theoretical foundation for developing novel therapeutic approaches targeting mitochondrial fusion proteins in lung injury.
Collapse
Affiliation(s)
- Guiyang Jia
- Department of Critical Care Medicine, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Erqin Song
- Department of Critical Care Medicine, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qianxia Huang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Miao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guoyue Liu
- Department of Critical Care Medicine, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
11
|
Li J, Cheng XY, Ma RX, Zou B, Zhang Y, Wu MM, Yao Y, Li J. Nicotinamide mononucleotide combined with PJ-34 protects microglial cells from lipopolysaccharide-induced mitochondrial impairment through NMNAT3-PARP1 axis. J Transl Med 2025; 23:279. [PMID: 40050860 PMCID: PMC11884077 DOI: 10.1186/s12967-025-06280-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/20/2025] [Indexed: 03/10/2025] Open
Abstract
Lipopolysaccharide (LPS) is known to induce cell injury and mitochondrial dysfunction, which are pivotal in neuroinflammation and related disorders. Recent studies have demonstrated the potential of nicotinamide mononucleotide (NMN) and poly(ADP-ribose) polymerase-1 (PARP1) inhibitors to enhance mitochondrial function. However, the underlying mechanisms have not been fully elucidated. This study investigates the impact of NMN in conjunction with PJ-34, a PARP1 inhibitor, on LPS-induced mitochondrial damage, focusing on nicotinamide mononucleotide adenylyl transferase 3 (NMNAT3) -PARP1 axis. The results showed that LPS treatment led to down-regulation of NMNAT3 (decreased 58.72% at 1 µM), up-regulation of PARP1 (enhanced 22.78% at 1 µM), thereby impairing mitophagy and mitochondrial function. The negative effects can be mitigated through supplementation with NMN and PJ-34. Specifically, compared to the LPS group, the expression of NMNAT3 increased by 63.29% and PARP1 decreased by 27.94% at a concentration of 400 µM NMN. Additionally, when 400 µM NMN was combined with 5 µM PJ-34, PARP1 expression decreased by 21.99%. Mechanistic studies reveal that NMN and PJ-34 counteracted the detrimental effects by promoting the binding of FoxO1 to the PINK1 promoter to activate the PINK1/Parkin mediated mitophagy pathway. Further experimental results demonstrate that the down-regulation of NMNAT3 can activate PARP1 and inhibit the initiation of autophagic processes. Consequently, targeting the NMNAT3-PARP1 signaling pathway holds promise for the development of novel therapeutic strategies to alleviate mitochondrial damage-related disorders.
Collapse
Affiliation(s)
- Jia Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Xiao-Yu Cheng
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People's Republic of China
| | - Rui-Xia Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Bin Zou
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Yue Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Miao-Miao Wu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Yao Yao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, People's Republic of China.
| | - Juan Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People's Republic of China.
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ningxia Engineering and Technology Research Center for Modernization of Characteristic Chinese Medicine, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, People's Republic of China.
| |
Collapse
|
12
|
Dai Z, Xie B, Jiang C, Peng Y, Lin J, Chen Q, Sun J. Aerosolized nicotine-free e-liquid base constituents exacerbates mitochondrial dysfunction and endothelial glycocalyx shedding via the AKT/GSK3β-mPTP pathway in lung injury models. Respir Res 2025; 26:82. [PMID: 40025534 PMCID: PMC11871641 DOI: 10.1186/s12931-025-03155-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/13/2025] [Indexed: 03/04/2025] Open
Abstract
Smoking has been recognized as a risk factor of cancer, heart disease, stroke, diabetes, and lung diseases such as chronic obstructive pulmonary disease, and nicotine appears to be the responsible component of tobacco smoke that affects lung development. While nicotine-free electronic cigarettes (e-cigarettes) are often promoted as a safer alternative to traditional smoking, recent evidence suggests that they might pose significant health risks. This study investigates the effects of nicotine-free e-cigarette vapor (ECV) on lung tissue and endothelial function. A mouse model of ECV-induced lung injury and human pulmonary microvascular endothelial cells (HPMVECs) were utilized to evaluate the impact of ECV exposure on mitochondrial function, endothelial cell viability, and glycocalyx shedding. ECV exposure significantly damages lung tissue, characterized by alveolar enlargement, inflammation, and vascular remodeling, indicative of emphysematous changes. In vitro, HPMVECs exposed to nicotine-free e-cigarette extract (ECE) demonstrated dose-dependent increases in mitochondrial reactive oxygen species (ROS), mitochondrial membrane depolarization, mPTP opening, and reduced ATP production, leading to enhanced endothelial permeability and glycocalyx degradation. The inhibition of mPTP opening with Cyclosporin A (CsA) was found to mitigate the mitochondrial dysfunction and glycocalyx damage induced by ECE, indicating a protective role of mPTP inhibition in preserving endothelial integrity. The AKT/GSK3β signaling pathway was identified as a key regulator of these processes, with ECE exposure downregulating p-AKT and p-GSK3β, thereby promoting mPTP opening. Activation of AKT signaling partially reversed these effects, highlighting the potential of targeting the AKT/GSK3β-mPTP axis to mitigate the adverse effects of e-cigarette exposure on lung and endothelial function. These findings underscore the potential risks associated with nicotine-free e-cigarettes and suggest novel therapeutic targets for preventing lung injury progression.
Collapse
Affiliation(s)
- Ziyu Dai
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bin Xie
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chen Jiang
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yun Peng
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jianing Lin
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jingyi Sun
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
13
|
Zheng Z, Qiao X, Yin J, Kong J, Han W, Qin J, Meng F, Tian G, Feng X. Advancements in omics technologies: Molecular mechanisms of acute lung injury and acute respiratory distress syndrome (Review). Int J Mol Med 2025; 55:38. [PMID: 39749711 PMCID: PMC11722059 DOI: 10.3892/ijmm.2024.5479] [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: 09/06/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an inflammatory response arising from lung and systemic injury with diverse causes and associated with high rates of morbidity and mortality. To date, no fully effective pharmacological therapies have been established and the relevant underlying mechanisms warrant elucidation, which may be facilitated by multi‑omics technology. The present review summarizes the application of multi‑omics technology in identifying novel diagnostic markers and therapeutic strategies of ALI/ARDS as well as its pathogenesis.
Collapse
Affiliation(s)
- Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junjie Kong
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Wanqing Han
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jing Qin
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Fanda Meng
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, P.R. China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
14
|
Yang ZC, Lu WY, Geng ZY, Zhao Y, Chen XM, Zheng T, Wu JZ, Huang KJ, Yuan HX, Yang Y. Identification and validation of biomarkers related to mitochondria during ex vivo lung perfusion for lung transplants based on machine learning algorithm. Gene 2025; 936:149097. [PMID: 39549776 DOI: 10.1016/j.gene.2024.149097] [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: 07/04/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Ex vivo lung perfusion (EVLP) is a critical strategy to rehabilitate marginal donor lungs, thereby increasing lung transplantation (LTx) rates. Ischemia-reperfusion (I/R) injury inevitably occurs during LTx. Exploring the common mechanisms between EVLP and I/R may unveil new treatment targets to enhance LTx outcomes. METHODS We obtained datasets from the public Gene Expression Omnibus (GEO) for EVLP (GSE127055 and GSE127057) and I/R (GSE145989) processes. We performed analysis of differentially expressed genes (DEGs) and Gene Set Enrichment Analysis (GSEA). Mitochondrial genes were sourced from the MitoCarta 3.0 database. Hub mitochondrial-related DEGs (MitoDEGs) were identified using a combination of protein-protein interaction networks and machine learning methods, which were further validated in cell and mice models of EVLP. RESULTS GSEA analysis of DEGs following EVLP and I/R revealed significant inhibition of mitochondrial function pathways, encompassing mitochondrial central dogma, mtRNA metabolism, OXPHOS assembly factors, metals and cofactors, and heme synthesis and processing processes. Machine learning algorithms including Random Forest, LASSO, and XGBoost identified five downregulated genes (MTERF1, ACACA, COX15, OSGEPL1, and COQ9) as hub MitoDEGs for both EVLP and I/R processes. We confirmed the reduced expression of these hub MitoDEGs in endothelial cells during EVLP and observed mitochondrial damage in endothelial cells characterized by swollen morphology and cristae disappearance. CONCLUSIONS Imbalance in mitochondrial homeostasis is a shared pathological process during EVLP and I/R-induced lung injury. The identified hub mitochondrial-related genes (MTERF1, ACACA, COX15, OSGEPL1, and COQ9) suggest promising therapeutic targets for lung injury during LTx. The downregulation of these genes indicates a significant disruption in mitochondrial function. This study provides potential mitochondrial-related therapeutic targets for I/R-induced lung injury and for donor lung repair during EVLP procedure in LTx.
Collapse
Affiliation(s)
- Zhi-Chang Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Wen-Yuan Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Zhen-Yang Geng
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Yang Zhao
- Department of Thoracic Surgery, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Xiao-Ming Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Tong Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Ji-Ze Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Kai-Jun Huang
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Hao-Xiang Yuan
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China.
| | - Yang Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
15
|
Torres-Machorro AL, García-Vicente Á, Espina-Ordoñez M, Luis-García E, Negreros M, Herrera I, Becerril C, Toscano F, Cisneros J, Maldonado M. Update of Aging Hallmarks in Idiopathic Pulmonary Fibrosis. Cells 2025; 14:222. [PMID: 39937013 PMCID: PMC11817138 DOI: 10.3390/cells14030222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/19/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is an epithelial-driven interstitial lung disease of unknown etiology characterized by the excessive proliferation of fibroblast populations that synthesize large amounts of extracellular matrix. In this devastating disorder, all aging hallmarks appear prematurely or are altered. This review highlights key findings about IPF characteristics recently recognized as hallmarks of aging, including mechanical alterations, inflammaging, dysbiosis, alternative splicing, and disabled macroautophagy. It also revisits the classic hallmarks of aging, which encompass stem cell exhaustion, cellular senescence, and altered intercellular communication. Enhancing our understanding of the fundamental processes that underlie the altered hallmarks of aging in IPF may facilitate the development of innovative experimental strategies to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Ana Lilia Torres-Machorro
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico; (A.L.T.-M.)
| | - Ángeles García-Vicente
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
- Posgrado en Ciencias Biomédicas, Unidad de Posgrado, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Marco Espina-Ordoñez
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico; (M.E.-O.); (J.C.)
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Erika Luis-García
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico; (A.L.T.-M.)
| | - Miguel Negreros
- Clínica de Vasculitis Sistémicas Primarias, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico;
| | - Iliana Herrera
- Laboratorio de Biopatología Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico
| | - Carina Becerril
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico; (A.L.T.-M.)
| | - Fernanda Toscano
- Laboratorio de Biopatología Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico
| | - Jose Cisneros
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico; (M.E.-O.); (J.C.)
| | - Mariel Maldonado
- Laboratorio de Biopatología Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico
| |
Collapse
|
16
|
Li Q, Chang X, Han Y, Guo Z, Liu Y, Guo B, Liu C, Yang B, Fan Z, Jiang H, Chang X. Consumption of Endogenous Caspase-3 Activates Molecular Theranostic Nanoplatform against Inflammation-Induced Profibrotic Positive Feedback in Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412303. [PMID: 39686776 PMCID: PMC11809389 DOI: 10.1002/advs.202412303] [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: 10/03/2024] [Revised: 11/30/2024] [Indexed: 12/18/2024]
Abstract
The limited and backward diagnostic approaches elicit high mortality associated with pulmonary fibrosis (PF) because they fail to identify injury phase of PF. Developing a precisely theranostic nanoplatform presents a promising shortcut to reverse PF. Herein, a specific molecular nanotheranostic (Casp-GNMT), which is triggered by endogenous cysteinyl aspartate specific proteinase-3 (caspase-3), boosts antifibrotic efficacy through bioimaging synergistic with chemotherapy at molecular level, facilitating by ionizable lipid and reactive oxygen species sensitive lipid for precise and manageable therapy. The activation of molecular imaging probe (pCY-pairs) by consumption of endogenous caspase-3 initiates fluorescence resonance energy transfer-guided theranostic pattern, aiming to restore mitochondrial dysfunction-induced oxidative stress and inflammatory responses in alveolar epithelial cells II (AECs II). This process sequentially resists the expression of interleukin-1β and vascular endothelial growth factor receptor through combined with nintedanib, further suppressing abnormal injury of AECs II and persistent migration and proliferation of inflammatory cells. Especially, the homeostasis of injured AECs II diminishes excessive accumulation of transforming growth factor-β to restrain myofibroblasts proliferation and collagen deposition, thereby amplifying the possibility of reversing PF. This theranostic nanoplatform is proposed to provide a prompt and exact approach to enhance diagnostic authenticity and treating efficiency through harnessing endogenous indicator for PF reversal.
Collapse
Affiliation(s)
- Qiu‐Ling Li
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Xin Chang
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Yu‐Mo Han
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Zi‐Chao Guo
- The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Yi‐Na Liu
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Bin Guo
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Chang Liu
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Bin‐Rong Yang
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Zhong‐Kai Fan
- The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou Medical UniversityJinzhouLiaoning121001China
| | - Hu‐Lin Jiang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Jiangsu Key Laboratory of Druggability of BiopharmaceuticalsChina Pharmaceutical UniversityNanjingJiangsu210009China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and ExcipientsChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Xin Chang
- School of PharmacyJinzhou Medical UniversityJinzhouLiaoning121001China
- Liaoning Provincial Key Laboratory of Marine Bioactive SubstancesJinzhou Medical UniversityJinzhouLiaoning121001China
| |
Collapse
|
17
|
Zhou S, Hu Y, Liu L, Li L, Deng F, Mo L, Huang H, Liang Q. Extract of Nanhaia speciosa J. Compton & Schrire alleviates LPS-induced acute lung injury via the NF-κB/Nrf2/AQPs pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118831. [PMID: 39278292 DOI: 10.1016/j.jep.2024.118831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 09/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nanhaia speciosas J. Compton & Schrire (the name Nanhaia speciosas J. Compton & Schrire has been accepted by the World Checklist of Vascular Plants https://www.worldfloraonline.org/taxon/wfo-0001444004) is a traditional Zhuang medicine that have been widely used for centuries. It has been used in the treatment of lung inflammation, tuberculosis, rheumatic pain, lumbar muscle strain, and various other ailments, such as chronic hepatitis, menoxenia, leukorrhea, and injuries. In addition, N. speciosa has also been used to treat acute lung injury (ALI). AIM OF THE STUDY The objective of this study was to conduct a comparative analysis of the effects of various constituents present in N. speciosas extract (NSE) on ALI and the related mechanisms while also elucidating the potential active monomeric components. MATERIALS AND METHODS NSE was extracted using an AB-8 macroporous resin column, and five fractions (Fr. 0%, 25%, 50%, 75% and 95%) were obtained. The anti-inflammatory and antioxidant capacities of the five fractions were evaluated in an A549 cell-based in vitro model, with the aim of evaluating their potential therapeutic effects. The anti-inflammatory and antioxidant capacities of NSE were assessed in a murine model of ALI induced by intratracheal injection of LPS. We utilized an in vitro model to analyse the critical molecular mechanisms through which NSE ameliorates ALI. The chemical composition of the optimal fraction was analysed and confirmed using UHPLC/MS. RESULTS Different fractions (especially Fr. 75%) significantly reduced inflammation and oxidative stress in A549 cells. Fr.75% abrogated LPS-induced pathological alterations and decreased the lung W/D ratio, total protein concentration in BALF, and the levels of the proinflammatory factors TNF-α, IL-6, and IL-1β. Moreover, Fr.75% reduced MPO and MDA concentrations and elevated SOD and GSH concentrations in pulmonary tissues. Additionally, it decreased the pulmonary tissue inflammation caused by LPS by downregulating the expression of p-NF-κB p65 and upregulating the expression of Nrf2, AQP1 and AQP5. Fr. 75% decreased p-NF-κB p65 protein levels; increased Keap1, Nrf2, HO-1, NQO1, AQP1 and AQP5 protein levels; and promoted the entry of Nrf2 into the nucleus. After UHPLC/MS analysis was conducted, the flavonoid Maackiain was determined to potentially play a pivotal role in this process. CONCLUSION Fr.75% alleviates ALI by regulating the NF-κB/Nrf2/AQPs signalling pathway. The flavonoid Maackiain may also play an important role in this process. Overall, N. speciosas may be a potential therapeutic agent for the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Shiyao Zhou
- Guilin Medical University, Guilin, 541199, China
| | - Yuting Hu
- Guilin Medical University, Guilin, 541199, China
| | - Lihua Liu
- Guilin Medical University, Guilin, 541199, China
| | - Lilan Li
- Guilin Medical University, Guilin, 541199, China
| | - Fang Deng
- Guilin Medical University, Guilin, 541199, China
| | - Luhe Mo
- Guilin Medical University, Guilin, 541199, China
| | - Huixue Huang
- Guilin Medical University, Guilin, 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Guilin, 541199, China; Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, 530200, China.
| | - Qiuyun Liang
- Guilin Medical University, Guilin, 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, Guilin, 541199, China.
| |
Collapse
|
18
|
Zou X, Huang Q, Kang T, Shen S, Cao C, Wu J. An integrated investigation of mitochondrial genes in COPD reveals the causal effect of NDUFS2 by regulating pulmonary macrophages. Biol Direct 2025; 20:4. [PMID: 39789601 PMCID: PMC11715544 DOI: 10.1186/s13062-025-00593-3] [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: 09/03/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Despite the increasing body of evidence that mitochondrial activities implicate in chronic obstructive pulmonary disease (COPD), we are still far from a causal-logical and mechanistic understanding of the mitochondrial malfunctions in COPD pathogenesis. RESULTS Differential expression genes (DEGs) from six publicly available bulk human lung tissue transcriptomic datasets of COPD patients were intersected with the known mitochondria-related genes from MitoCarta3.0 to obtain mitochondria-related DEGs associated with COPD (MitoDEGs). The 32 hub MitoDEGs identified from protein-protein interaction (PPI) networks demonstrated superior overall diagnostic efficacy to non-hub MitoDEGs. Random forest (RF) analysis, least absolute shrinkage and selection operator (LASSO) regression, and Mendelian Randomization (MR) analysis of hub MitoDEGs further nominated NDUFS2, CAT, and MRPL2 as causal MitoDEGs for COPD, whose predominate expressions in pulmonary macrophages were revealed by an independent single-cell transcriptomic dataset of COPD human lungs. Finally, NDUFS2 was evaluated as the top-ranked contributor to COPD in the nomogram model and its downregulation in pulmonary macrophages could result in pro-inflammatory secretion, enhanced intercellular communications, whereas depressed phagocytosis of macrophages as revealed by gene set variation analysis (GSVA) and cell-cell interaction (CCI) analysis of single-cell transcriptomic dataset of COPD human lungs, which was later confirmed in COPD mouse model and macrophage cell lines. CONCLUSIONS Our study established the causal linkage between mitochondrial malfunctions and COPD, providing a potential therapeutic avenue to alleviate pulmonary inflammation accounting for COPD by targeting mitochondria-related genes. NDUFS2, a canonical component of mitochondrial electron respiratory chain, was highlighted instrumental for the susceptibility of risk-exposed individuals to COPD.
Collapse
Affiliation(s)
- Xiaoli Zou
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Qiqing Huang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Tutu Kang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Shaoran Shen
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Chenxi Cao
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jianqing Wu
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
19
|
Pokharel MD, Feng A, Liang Y, Ma W, Aggarwal S, Unwalla H, Black SM, Wang T. Drp1-associated genes implicated in sepsis survival. Front Immunol 2025; 15:1516145. [PMID: 39845954 PMCID: PMC11750657 DOI: 10.3389/fimmu.2024.1516145] [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: 10/23/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025] Open
Abstract
Sepsis is a severe and life-threatening medical syndrome that can lead to organ failure and death. Despite advances in medical treatment, current therapies are often inadequate, with high septic mortality rates. Therefore, there is a critical need for reliable prognostic markers to be used in clinical settings to improve the management and outcomes of patients with sepsis. Recent studies have suggested that mitochondrial dynamics, including the processes of mitochondrial fission and fusion, are closely related to the severity of sepsis and the status of inflammation. By monitoring transcriptomic signals related to mitochondrial dynamics, new and reliable biomarkers can be engineered to more accurately predict sepsis survival risk. Such biomarkers would be invaluable in clinical settings, aiding healthcare providers in the early identification of high-risk patients and improving treatment strategies. To achieve this goal, we utilized the major mitochondrial fission regulatory protein dynamin-related protein 1 (Drp1, gene code DNM1L) and identified Drp1-associated genes that are enriched with sepsis survival genes. A 12-gene signature (GS) was established as a differentially expressed gene (DEG)-based GS. Next, we compared genes of proteins that interact with Drp1 to sepsis survival genes and identified 7 common genes, establishing a GS we term as protein-protein interaction (PPI)-based GS. To evaluate if these GSs can predict sepsis survival, we used publicly available human blood transcriptomic datasets from sepsis patients. We confirmed that both GSs can successfully predict sepsis survival in both discovery and validation cohorts with high sensitivity and specificity, with the PPI-based GS showing enhanced prognostic performance. Together, this study successfully engineers a new and validated blood-borne biomarker (PPI-based 7-gene GS) for sepsis survival risk prediction. This biomarker holds the potential for improving the early identification of high-risk sepsis patients and optimizing personalized treatment strategies to reduce sepsis mortality.
Collapse
Affiliation(s)
- Marissa D. Pokharel
- Center for Translational Science, Florida International University, Port Saint Lucie, FL, United States
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL, United States
| | - Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL, United States
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| | - Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL, United States
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| | - Wenli Ma
- Center for Translational Science, Florida International University, Port Saint Lucie, FL, United States
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL, United States
| | - Hoshang Unwalla
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL, United States
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL, United States
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL, United States
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL, United States
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL, United States
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| |
Collapse
|
20
|
Zhou J, Xi Y, Wu T, Zeng X, Yuan J, Peng L, Fu H, Zhou C. A potential therapeutic approach for ulcerative colitis: targeted regulation of mitochondrial dynamics and mitophagy through phytochemicals. Front Immunol 2025; 15:1506292. [PMID: 39840057 PMCID: PMC11747708 DOI: 10.3389/fimmu.2024.1506292] [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: 10/04/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Mitochondria are important organelles that regulate cellular energy and biosynthesis, as well as maintain the body's response to environmental stress. Their dynamics and autophagy influence occurrence of cellular function, particularly under stressful conditions. They can generate reactive oxygen species (ROS) which is a major contributor to inflammatory diseases such as ulcerative colitis (UC). In this review, we discuss the key effects of mitochondrial dynamics and mitophagy on the pathogenesis of UC, with a particular focus on the cellular energy metabolism, oxidative stress, apoptosis, and immunoinflammatory activities. The therapeutic efficacy of existing drugs and phytochemicals targeting the mitochondrial pathway are discussed to reveal important insights for developing therapeutic strategies for treating UC. In addition, new molecular checkpoints with therapeutic potential are identified. We show that the integration of mitochondrial biology with the clinical aspects of UC may generate ideas for enhancing the clinical management of UC.
Collapse
Affiliation(s)
- Jianping Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuting Xi
- Zigong Hospital of Traditional Chinese Medicine, Zigong, China
| | - Ting Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei Peng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hao Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ce Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
21
|
Beegam S, Zaaba NE, Elzaki O, Nemmar A. α-Bisabolol alleviates diesel exhaust particle-induced lung injury and mitochondrial dysfunction by regulating inflammatory, oxidative stress, and apoptotic biomarkers through the c-Jun N-terminal kinase signaling pathway. Front Pharmacol 2025; 15:1485101. [PMID: 39830335 PMCID: PMC11738621 DOI: 10.3389/fphar.2024.1485101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/11/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Exposure to particulate matter ≤2.5 μm in diameter (PM2.5) is associated with adverse respiratory outcomes, including alterations to lung morphology and function. These associations were reported even at concentrations lower than the current annual limit of PM2.5. Inhalation of PM2.5, of which diesel exhaust particles (DEPs) is a major component, induces lung inflammation and oxidative stress. α-Bisabolol (BIS) is a bioactive dietary phytochemical with various pharmacological properties, including anti-inflammatory and antioxidant actions. Here, we evaluated the possible protective effects of BIS on DEP-induced lung injury. Methods Mice were exposed to DEPs (20 µg/mouse) or saline (control) by intratracheal instillation. BIS was administered orally at two doses (25 and 50 mg/kg) approximately 1 h before DEP exposure. Twenty-four hours after DEP administration, multiple respiratory endpoints were evaluated. Results BIS administration was observed to prevent DEP-induced airway hyperreactivity to methacholine; influx of macrophages, neutrophils, and lymphocytes in the bronchoalveolar lavage fluid; and increases in epithelial and endothelial permeabilities. DEP exposure caused increases in the levels of myeloperoxidase, proinflammatory cytokines, and oxidative stress markers in lung tissue homogenates, and all these effects were abated by BIS treatment. The activities of mitochondrial complexes I, II, III, and IV were markedly increased in the lungs of mice exposed to DEPs, and these effects were significantly reduced in the BIS-treated group. Intratracheal instillation of DEPs induced DNA damage and increase in the apoptotic marker cleaved caspase-3. The latter effects were prevented in mice treated with BIS and exposed to DEPs. Moreover, BIS mitigated DEP-induced increase in the expression of phospho-c-Jun N-terminal kinase (JNK) in a dose-dependent manner. Discussion BIS markedly alleviated DEP-induced lung injury by regulating the inflammatory, oxidative stress, and apoptotic biomarkers through the JNK signaling pathway. Following additional studies, BIS may be considered as a plausible protective agent against inhaled-particle-induced pulmonary adverse effects.
Collapse
Affiliation(s)
| | | | | | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
22
|
Pei L, Yao Z, Liang D, Yang K, Tao L. Mitochondria in skeletal system-related diseases. Biomed Pharmacother 2024; 181:117505. [PMID: 39499974 DOI: 10.1016/j.biopha.2024.117505] [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: 06/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 12/21/2024] Open
Abstract
Skeletal system-related diseases, such as osteoporosis, arthritis, osteosarcoma and sarcopenia, are becoming major public health concerns. These diseases are characterized by insidious progression, which seriously threatens patients' health and quality of life. Early diagnosis and prevention in high-risk populations can effectively prevent the deterioration of these patients. Mitochondria are essential organelles for maintaining the physiological activity of the skeletal system. Mitochondrial functions include contributing to the energy supply, modulating the Ca2+ concentration, maintaining redox balance and resisting the inflammatory response. They participate in the regulation of cellular behaviors and the responses of osteoblasts, osteoclasts, chondrocytes and myocytes to external stimuli. In this review, we describe the pathogenesis of skeletal system diseases, focusing on mitochondrial function. In addition to osteosarcoma, a characteristic of which is active mitochondrial metabolism, mitochondrial damage occurs during the development of other diseases. Impairment of mitochondria leads to an imbalance in osteogenesis and osteoclastogenesis in osteoporosis, cartilage degeneration and inflammatory infiltration in arthritis, and muscle atrophy and excitationcontraction coupling blockade in sarcopenia. Overactive mitochondrial metabolism promotes the proliferation and migration of osteosarcoma cells. The copy number of mitochondrial DNA and mitochondria-derived peptides can be potential biomarkers for the diagnosis of these disorders. High-risk factor detection combined with mitochondrial component detection contributes to the early detection of these diseases. Targeted mitochondrial intervention is an effective method for treating these patients. We analyzed skeletal system-related diseases from the perspective of mitochondria and provided new insights for their diagnosis, prevention and treatment by demonstrating the relationship between mitochondria and the skeletal system.
Collapse
Affiliation(s)
- Liang Pei
- Department of Pediatrics, Shengjing Hospital of China Medical University, China
| | - Zhuo Yao
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Dong Liang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China..
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China..
| |
Collapse
|
23
|
Yang L, Ren Q, Wang Y, Zheng Y, Du F, Wang F, Zhou J, Gui L, Chen S, Chen X, Zhang W, Sun Y, Zhong X, Liu H, Jiang X, Zhang Z. Research progress of mitochondrial dysfunction induced pyroptosis in acute lung injury. Respir Res 2024; 25:398. [PMID: 39511593 PMCID: PMC11545853 DOI: 10.1186/s12931-024-03028-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) is a common critical respiratory disease in clinical practice, especially in the ICU, with a high mortality rate. The pathogenesis of ALI is relatively complex, mainly involving inflammatory response imbalance, oxidative stress, cell apoptosis, and other aspects. However, currently, the treatment measures taken based on the above mechanisms have not had significant effects. Recent research shows that mitochondrial dysfunction and pyroptosis play an important role in ALI, but there is not much analysis on the relationship between mitochondrial dysfunction and pyroptosis at present. This article reviews the situation of mitochondrial dysfunction in ALI, pyroptosis in ALI, whether mitochondrial dysfunction is related to pyroptosis in ALI, and how to do so, and further analyzes the relationship between them in ALI. This review describes how to alleviate mitochondrial dysfunction, and then suppress the associated immunological pyroptosis, providing new ideas for the clinical treatment of ALI.
Collapse
Affiliation(s)
- Luhan Yang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qingyi Ren
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yaohui Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yucheng Zheng
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fei Du
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fang Wang
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jie Zhou
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Linxi Gui
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shengdong Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Wanting Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yuhong Sun
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaolin Zhong
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Xian Jiang
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, 646000, China.
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
24
|
Lin Q, Lin Y, Liao X, Chen Z, Deng M, Zhong Z. ACSL1 improves pulmonary fibrosis by reducing mitochondrial damage and activating PINK1/Parkin mediated mitophagy. Sci Rep 2024; 14:26504. [PMID: 39489819 PMCID: PMC11532343 DOI: 10.1038/s41598-024-78136-5] [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: 07/17/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
Pulmonary fibrosis is a chronic interstitial lung disease with no curative therapeutic treatment, leading to significant mortality. The aims of this study were to investigate the regulatory mechanisms of mitophagy in the progression of pulmonary fibrosis. Through bioinformatics analysis, we identified the downregulation of long-chain fatty acyl-CoA synthetase 1 (ACSL1) as being associated with the severity of pulmonary fibrosis. A pulmonary fibrosis model was established through bleomycin (BLM) exposure both in vivo and in vitro. Mitoquinone (MitoQ) pretreatment significantly decreased redox damage, stabilized mitochondrial membrane potential (MMP), improved mitochondrial dynamics, and activated PINK1/Parkin-mediated mitophagy, thereby alleviating pulmonary fibrosis. In vitro, overexpression of ACSL1 mitigated mitochondrial damage and restored PINK1/Parkin-mediated mitophagy under BLM exposure. In contrast, ACSL1 inhibition exacerbated pulmonary fibrosis, and these adverse effects could not be reversed by MitoQ treatment. Taken together, our study reveals a novel mechanism underlying the pathogenesis of pulmonary fibrosis and suggests a potential therapeutic target for its treatment.
Collapse
Affiliation(s)
- Qi Lin
- Department of Pharmacy, The Affiliated Hospital of Putian University, Putian, China.
- Pharmaceutical and Medical Technology College, Putian university, Putian, China.
- Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China.
| | - Yating Lin
- Pharmaceutical and Medical Technology College, Putian university, Putian, China
| | - Xinyan Liao
- Pharmaceutical and Medical Technology College, Putian university, Putian, China
| | - Ziyi Chen
- Pharmaceutical and Medical Technology College, Putian university, Putian, China
| | - Mengmeng Deng
- Pharmaceutical and Medical Technology College, Putian university, Putian, China
| | - Zhihao Zhong
- Pharmaceutical and Medical Technology College, Putian university, Putian, China
| |
Collapse
|
25
|
Tian L, Jin J, Lu Q, Zhang H, Tian S, Lai F, Liu C, Liang Y, Lu Y, Zhao Y, Yao S, Ren W. Bidirectional modulation of extracellular vesicle-autophagy axis in acute lung injury: Molecular mechanisms and therapeutic implications. Biomed Pharmacother 2024; 180:117566. [PMID: 39423751 DOI: 10.1016/j.biopha.2024.117566] [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: 07/24/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Acute lung injury (ALI), a multifactorial pathological condition, manifests through heightened inflammatory responses, compromised lung epithelial-endothelial barrier function, and oxidative stress, potentially culminating in respiratory failure and mortality. This study explores the intricate interplay between two crucial cellular mechanisms-extracellular vesicles (EVs) and autophagy-in the context of ALI pathogenesis and potential therapeutic interventions.EVs, bioactive membrane-bound structures secreted by cells, serve as versatile carriers of molecular cargo, facilitating intercellular communication and significantly influencing disease progression. Concurrently, autophagy, an essential intracellular degradation process, maintains cellular homeostasis and has emerged as a promising therapeutic target in ALI and acute respiratory distress syndrome.Our research unveils a fascinating "EV-Autophagy dual-drive pathway," characterized by reciprocal regulation between these two processes. EVs modulate autophagy activation and inhibition, while autophagy influences EV production, creating a dynamic feedback loop. This study posits that precise manipulation of this pathway could revolutionize ALI treatment strategies.By elucidating the mechanisms underlying this cellular crosstalk, we open new avenues for targeted therapies. The potential for engineered EVs to fine-tune autophagy in ALI treatment is explored, alongside innovative concepts such as EV-based vaccines for ALI prevention and management. This research not only deepens our understanding of ALI pathophysiology but also paves the way for novel, more effective therapeutic approaches in critical care medicine.
Collapse
Affiliation(s)
- Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Clinical Medical Center of Tissue Egineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Jin
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China
| | - Qianying Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China
| | - Huajing Zhang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China
| | - Sijia Tian
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Chuanchuan Liu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Yangfan Liang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Yujia Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
| | - Yanmei Zhao
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China.
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China; School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Clinical Medical Center of Tissue Egineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| |
Collapse
|
26
|
Sun X, Moreno Caceres S, Yegambaram M, Lu Q, Pokharel MD, Boehme JT, Datar SA, Aggarwal S, Wang T, Fineman JR, Black SM. The mitochondrial redistribution of ENOS is regulated by AKT1 and dimer status. Nitric Oxide 2024; 152:90-100. [PMID: 39332480 PMCID: PMC12068231 DOI: 10.1016/j.niox.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Previously, we have shown that endothelial nitric-oxide synthase (eNOS) dimer levels directly correlate with the interaction of eNOS with hsp90 (heat shock protein 90). Further, the disruption of eNOS dimerization correlates with its redistribution to the mitochondria. However, the causal link between these events has yet to be investigated and was the focus of this study. Our data demonstrates that simvastatin, which decreases the mitochondrial redistribution of eNOS, increased eNOS-hsp90 interactions and enhanced eNOS dimerization in cultured pulmonary arterial endothelial cells (PAEC) from a lamb model of pulmonary hypertension (PH). Our data also show that the dimerization of a monomeric fraction of human recombinant eNOS was stimulated in the presence of hsp90 and ATP. The over-expression of a dominant negative mutant of hsp90 (DNHsp90) decreased eNOS dimer levels and enhanced its mitochondrial redistribution. We also found that the peroxynitrite donor3-morpholinosydnonimine (SIN-1) increased the mitochondrial redistribution of eNOS in PAEC and this was again associated with decreased eNOS dimer levels. Our data also show in COS-7 cells, the SIN-1 mediated mitochondrial redistribution of wildtype eNOS (WT-eNOS) is significantly higher than a dimer stable eNOS mutant protein (C94R/C99R-eNOS). Conversely, the mitochondrial redistribution of a monomeric eNOS mutant protein (C96A-eNOS) was enhanced. Finally, we linked the SIN-1-mediated mitochondrial redistribution of eNOS to the Akt1-mediated phosphorylation of eNOS at Serine(S)617 and showed that the accessibility of this residue to phosphorylation is regulated by dimerization status. Thus, our data reveal a novel mechanism of pulmonary endothelial dysfunction mediated by mitochondrial redistribution of eNOS, regulated by dimerization status and the phosphorylation of S617.
Collapse
Affiliation(s)
- Xutong Sun
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Santiago Moreno Caceres
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Marissa D Pokharel
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA
| | - Jason T Boehme
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Sanjeev A Datar
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA
| | - Jeffrey R Fineman
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; The Department of Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA; Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33174, USA; Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA.
| |
Collapse
|
27
|
Zhang J, Wang S, Zhang H, Yang X, Ren X, Wang L, Yang Y, Yang Y, Wen Y. Drp1 acetylation mediated by CDK5-AMPK-GCN5L1 axis promotes cerebral ischemic injury via facilitating mitochondrial fission. Mol Med 2024; 30:173. [PMID: 39390372 PMCID: PMC11468353 DOI: 10.1186/s10020-024-00948-y] [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: 07/27/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
The aberrant acetylation of mitochondrial proteins is involved in the pathogenesis of multiple diseases including neurodegenerative diseases and cerebral ischemic injury. Previous studies have shown that depletion of mitochondrial NAD+, which is necessary for mitochondrial deacetylase activity, leads to decreased activity of mitochondrial deacetylase and thus causes hyperacetylation of mitochondrial proteins in ischemic brain tissues, which results in altered mitochondrial dynamics. However, it remains largely unknown about how mitochondrial dynamics-related protein Drp1 is acetylated in ischemic neuronal cells and brain tissues. Here, we showed that Drp1 and GCN5L1 expression was up-regulated in OGD-treated neuronal cells and ischemic brain tissues induced by dMCAO, accompanied by the increased mitochondrial fission, mtROS accumulation, and cell apoptosis. Further, we confirmed that ischemia/hypoxia promoted Drp1 interaction with GCN5L1 in neuronal cells and brain tissues. GCN5L1 knockdown attenuated, while its overexpression enhanced Drp1 acetylation and mitochondrial fission, indicating that GCN5L1 plays a crucial role in ischemia/hypoxia-induced mitochondrial fission by acetylating Drp1. Mechanistically, ischemia/hypoxia induced Drp1 phosphorylation by CDK5 upregulation-mediated activation of AMPK in neuronal cells, which in turn facilitated the interaction of GCN5L1 with Drp1, thus enhancing Drp1 acetylation and mitochondrial fission. Accordingly, inhibition of AMPK alleviated ischemia/hypoxia- induced Drp1 acetylation and mitochondrial fission and protected brain tissues from ischemic damage. These findings provide a novel insight into the functional roles of GCN5L1 in regulating Drp1 acetylation and identify a previously unrecognized CDK5-AMPK-GCN5L1 pathway that mediates the acetylation of Drp1 in ischemic brain tissues.
Collapse
Affiliation(s)
- Jiejie Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Shan Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Haitao Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Xiaotong Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Xin Ren
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Lei Wang
- Department of Human Anatomy, Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yihan Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Yi Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| | - Ya Wen
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
28
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [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: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
29
|
Liu R, Zhou T, Li X, Zou Q, Yu J, Ye J, Wang W, Zhou Y, Sun SK. A Non-Metallic Nanozyme Ameliorates Pulmonary Hypertension Through Inhibiting ROS/TGF-β1 Signaling. Adv Healthc Mater 2024:e2401909. [PMID: 39155419 DOI: 10.1002/adhm.202401909] [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: 05/23/2024] [Revised: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Pulmonary hypertension (PH) is a life-threatening cardiovascular disease with a lack of effective treatment options. Nanozymes, though promising for PH therapy, pose safety risks due to their metallic nature. Here, a non-metallic nanozyme is reported for the treatment of monocrotaline (MCT)-induced PH with a therapeutic mechanism involving the ROS/TGF-β1 signaling. The synthesized melanin-polyvinylpyrrolidone-polyethylene glycol (MPP) nanoparticles showcase ultra-small size, excellent water solubility, high biocompatibility, and remarkable antioxidant capacity. The MPP nanoparticles are capable of effectively eliminating ROS in isolated pulmonary artery smooth muscle cells (PASMCs) from PH rats, and significantly reduce PASMC proliferation and migration. In vivo results from a PH model demonstrate that MPP nanoparticles significantly increase pulmonary artery acceleration time, decrease wall thickening and PCNA expression in lung tissues, as evidenced by echocardiograpy, histology and immunoblot analysis. Additionally, MPP nanoparticles treatment improve running capacity, decrease Fulton index, and attenuate right ventricular fibrosis in MCT-PH rats by using treadmill test, picrosirius red, and trichrome Masson staining. Further transcriptomic and biochemical analyses reveal that inhibiting ROS-driven activation of TGF-β1 in the PA is the mechanism by which MPP nanoparticles exert their therapeutic effect. This study provides a novel approach for treating PH with non-metallic nanozymes based on a well-understood mechanism.
Collapse
Affiliation(s)
- Ruxia Liu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Ting Zhou
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
- Department of CT, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Xinsheng Li
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Quan Zou
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, 300203, China
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jiaojiao Yu
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Jingjing Ye
- Trauma Treatment Center, Peking University People's Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine, Beijing, 100044, China
| | - Wenhui Wang
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yan Zhou
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| |
Collapse
|
30
|
Li CL, Liu SF. Cellular and Molecular Biology of Mitochondria in Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2024; 25:7780. [PMID: 39063022 PMCID: PMC11276859 DOI: 10.3390/ijms25147780] [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: 06/28/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive respiratory disorder characterized by enduring airflow limitation and chronic inflammation. Growing evidence highlights mitochondrial dysfunction as a critical factor in COPD development and progression. This review explores the cellular and molecular biology of mitochondria in COPD, focusing on structural and functional changes, including alterations in mitochondrial shape, behavior, and respiratory chain complexes. We discuss the impact on cellular signaling pathways, apoptosis, and cellular aging. Therapeutic strategies targeting mitochondrial dysfunction, such as antioxidants and mitochondrial biogenesis inducers, are examined for their potential to manage COPD. Additionally, we consider the role of mitochondrial biomarkers in diagnosis, evaluating disease progression, and monitoring treatment efficacy. Understanding the interplay between mitochondrial biology and COPD is crucial for developing targeted therapies to slow disease progression and improve patient outcomes. Despite advances, further research is needed to fully elucidate mitochondrial dysfunction mechanisms, discover new biomarkers, and develop targeted therapies, aiming for comprehensive disease management that preserves lung function and enhances the quality of life for COPD patients.
Collapse
Affiliation(s)
- Chin-Ling Li
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
| | - Shih-Feng Liu
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, #123, Ta-Pei Road, Niaosong District, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
31
|
Zhu M, Wang Y, Han J, Sun Y, Wang S, Yang B, Wang Q, Kuang H. Artesunate Exerts Organ- and Tissue-Protective Effects by Regulating Oxidative Stress, Inflammation, Autophagy, Apoptosis, and Fibrosis: A Review of Evidence and Mechanisms. Antioxidants (Basel) 2024; 13:686. [PMID: 38929125 PMCID: PMC11200509 DOI: 10.3390/antiox13060686] [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: 04/12/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The human body comprises numerous organs and tissues operating in synchrony, it facilitates metabolism, circulation, and overall organismal function. Consequently, the well-being of our organs and tissues significantly influences our overall health. In recent years, research on the protective effects of artesunate (AS) on various organ functions, including the heart, liver, brain, lungs, kidneys, gastrointestinal tract, bones, and others has witnessed significant advancements. Findings from in vivo and in vitro studies suggest that AS may emerge as a newfound guardian against organ damage. Its protective mechanisms primarily entail the inhibition of inflammatory factors and affect anti-fibrotic, anti-aging, immune-enhancing, modulation of stem cells, apoptosis, metabolic homeostasis, and autophagy properties. Moreover, AS is attracting a high level of interest because of its obvious antioxidant activities, including the activation of Nrf2 and HO-1 signaling pathways, inhibiting the release of reactive oxygen species, and interfering with the expression of genes and proteins associated with oxidative stress. This review comprehensively outlines the recent strides made by AS in alleviating organismal injuries stemming from various causes and protecting organs, aiming to serve as a reference for further in-depth research and utilization of AS.
Collapse
Affiliation(s)
- Mingtao Zhu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; (M.Z.); (Y.W.); (J.H.); (Y.S.); (S.W.); (B.Y.)
| | - Yu Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; (M.Z.); (Y.W.); (J.H.); (Y.S.); (S.W.); (B.Y.)
| | - Jianwei Han
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; (M.Z.); (Y.W.); (J.H.); (Y.S.); (S.W.); (B.Y.)
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; (M.Z.); (Y.W.); (J.H.); (Y.S.); (S.W.); (B.Y.)
| | - Shuang Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; (M.Z.); (Y.W.); (J.H.); (Y.S.); (S.W.); (B.Y.)
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; (M.Z.); (Y.W.); (J.H.); (Y.S.); (S.W.); (B.Y.)
| | - Qiuhong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510024, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; (M.Z.); (Y.W.); (J.H.); (Y.S.); (S.W.); (B.Y.)
| |
Collapse
|
32
|
Peña MJ, De Sanctis CV, De Sanctis JB, Garmendia JV. Frequency of Gene Polymorphisms in Admixed Venezuelan Women with Recurrent Pregnancy Loss: Microsomal Epoxy Hydroxylase (rs1051740) and Enos (rs1799983). Curr Issues Mol Biol 2024; 46:3460-3469. [PMID: 38666947 PMCID: PMC11049659 DOI: 10.3390/cimb46040217] [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: 03/29/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Recurrent pregnancy loss (RPL) affects around 2% of women of reproductive age. Primary RPL is defined by ≥2 pregnancy losses and no normal birth delivery. In secondary RPL, the losses are after a normal pregnancy and delivery. Most cases have no clear aetiology, although primary cases are the most complex. Several gene single nucleotide polymorphisms (SNPs) have been associated with RPL. The frequency of some SNPs is increased in women suffering from RLP from Asian or Caucasian races; however, in admixed populations, the information on possible genetic links is scarce and contradictory. This study aimed to assess the frequency of two SNPs present in two different enzymes involved in medical conditions observed during pregnancy. It is a case-control study. Microsomal epoxy hydrolase (mEPH) is involved in detoxifying xenobiotics, is present in the ovaries, and is hormonally regulated. The endothelial nitric oxide synthase (NOS3) that forms nitric is involved in vascular tone. Two SNPs, rs1051740 (mEPH) and rs1799983 (NOS3), were assessed. The study included 50 controls and 63 primary RPL patients. The frequency of mutated alleles in both SNPs was significantly higher in patients (p < 0.05). Double-mutated homozygotes were encountered only in RPL patients (p < 0.05). Genetic polymorphisms rs1051740 and rs1799983 may be involved in primary RPL in the Venezuelan admix population. Genetic studies could provide crucial information on the aetiology of primary RPL.
Collapse
Affiliation(s)
- María Johanna Peña
- Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (M.J.P.); (C.V.D.S.)
| | - Claudia Valentina De Sanctis
- Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (M.J.P.); (C.V.D.S.)
| | - Juan Bautista De Sanctis
- Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (M.J.P.); (C.V.D.S.)
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Jenny Valentina Garmendia
- Institute of Immunology, Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (M.J.P.); (C.V.D.S.)
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| |
Collapse
|
33
|
Li CL, Liu JF, Liu SF. Mitochondrial Dysfunction in Chronic Obstructive Pulmonary Disease: Unraveling the Molecular Nexus. Biomedicines 2024; 12:814. [PMID: 38672169 PMCID: PMC11048013 DOI: 10.3390/biomedicines12040814] [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: 02/17/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent and debilitating respiratory disorder characterized by persistent airflow limitation and chronic inflammation. In recent years, the role of mitochondrial dysfunction in COPD pathogenesis has emerged as a focal point of investigation. This review endeavors to unravel the molecular nexus between mitochondrial dysfunction and COPD, delving into the intricate interplay of oxidative stress, bioenergetic impairment, mitochondrial genetics, and downstream cellular consequences. Oxidative stress, a consequence of mitochondrial dysfunction, is explored as a driving force behind inflammation, exacerbating the intricate cascade of events leading to COPD progression. Bioenergetic impairment sheds light on the systemic consequences of mitochondrial dysfunction, impacting cellular functions and contributing to the overall energy imbalance observed in COPD patients. This review navigates through the genetic landscape, elucidating the role of mitochondrial DNA mutations, variations, and haplogroups in COPD susceptibility and severity. Cellular consequences, including apoptosis, autophagy, and cellular senescence, are examined, providing insights into the intricate mechanisms by which mitochondrial dysfunction influences COPD pathology. Therapeutic implications, spanning antioxidant strategies, mitochondria-targeted compounds, and lifestyle modifications, are discussed in the context of translational research. Important future directions include identifying novel biomarkers, advancing mitochondria-targeted therapies, and embracing patient-centric approaches to redefine COPD management. This abstract provides a comprehensive overview of our review, offering a roadmap for understanding and addressing the molecular nexus between mitochondrial dysfunction and COPD, with potential implications for precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- Chin-Ling Li
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Jui-Fang Liu
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
| | - Shih-Feng Liu
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|