1
|
Lu Y, Fang YX, Ou-Yang ZM, Wu T, Zhang Q, Zou YW, Zheng HW, Jing J, Lin LH, Ma JD, Liang Z, Dai L. Deficiency of FUN14 domain-containing 1 enhances the migration and invasion of fibroblast-like synoviocytes in rheumatoid arthritis through mitochondrial dysregulation. Cell Signal 2025; 132:111829. [PMID: 40274085 DOI: 10.1016/j.cellsig.2025.111829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/01/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Fibroblast-like synoviocytes (FLS) display aggressive phenotypes contributing to synovitis and joint destruction in rheumatoid arthritis (RA). Disrupted mitochondrial homeostasis has been proposed to aggravate the RA pathogenesis, however, the underlying mechanism remains to be elucidated. This study aimed to elucidate the role of mitophagy receptor FUN14 domain-containing 1 (FUNDC1) on RA-FLS migration and invasion. METHODS We analyzed the correlation of synovial FUNDC1 expression with joint destruction and disease activity in RA patients. Single cell sequencing data analysis combined with immunofluorescence indicated the specific expression and localization of FUNDC1 in synovial tissue and RA-FLS. The roles of FUNDC1 in the migration, invasion, and cytokine secretion of RA-FLS were examined by patient-derived primary culture as well as SCID mouse models. We investigated the effects and mechanism of FUNDC1 on mitophagy and mitochondrial quality control network in primary RA-FLS. RESULTS We found that the FUNDC1 was mainly expressed in FLS and exhibited a decreased level in RA synovium, which was correlated with severe joint destruction. Deficiency of FUNDC1 enhanced migration, invasion as well as secretion of matrix metalloproteinases in RA-FLS. On the contrary, overexpression of FUNDC1 in RA-FLS with low FUNDC1 inhibited the migration, invasion and secretion capacity of RA-FLS. Mechanistically, repressed FUNDC1 level in RA-FLS impaired mitophagy, imbalanced mitochondrial quality control, and increased mitochondrial reactive oxygen species (mtROS) production, leading to the overactivation of the MAPK pathway. Treatment with mtROS scavenger mtTEMPO can reverse this process and diminish the invasiveness of RA-FLS. CONCLUSIONS Deficiency of FUNDC1 dysregulates mitochondrial quality-control system and induces aggressive phenotype of RA-FLS, resulting in joint destruction during RA progression.
Collapse
Affiliation(s)
- Ye Lu
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Ya-Xiong Fang
- Bioscience and Biomedical Engineering Thrust, Brain and Intelligence Research Institute, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511453, PR China
| | - Zhi-Ming Ou-Yang
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Tao Wu
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Qian Zhang
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yao-Wei Zou
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Hu-Wei Zheng
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Jun Jing
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Le-Hang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Jian-Da Ma
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Zhuoyi Liang
- Bioscience and Biomedical Engineering Thrust, Brain and Intelligence Research Institute, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511453, PR China.
| | - Lie Dai
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| |
Collapse
|
2
|
Ungvari A, Gulej R, Patai R, Papp Z, Toth A, Szabó AÁ, Podesser BK, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Maier AB, Csiszar A, Ungvari Z. Sex-specific mechanisms in vascular aging: exploring cellular and molecular pathways in the pathogenesis of age-related cardiovascular and cerebrovascular diseases. GeroScience 2025; 47:301-337. [PMID: 39754010 PMCID: PMC11872871 DOI: 10.1007/s11357-024-01489-2] [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: 11/04/2024] [Accepted: 12/17/2024] [Indexed: 03/04/2025] Open
Abstract
Aging remains the foremost risk factor for cardiovascular and cerebrovascular diseases, surpassing traditional factors in epidemiological significance. This review elucidates the cellular and molecular mechanisms underlying vascular aging, with an emphasis on sex differences that influence disease progression and clinical outcomes in older adults. We discuss the convergence of aging processes at the macro- and microvascular levels and their contributions to the pathogenesis of vascular diseases. Critical analysis of both preclinical and clinical studies reveals significant sex-specific variations in these mechanisms, which could be pivotal in understanding the disparity in disease morbidity and mortality between sexes. The review highlights key molecular pathways, including oxidative stress, inflammation, and autophagy, and their differential roles in the vascular aging of males and females. We argue that recognizing these sex-specific differences is crucial for developing targeted therapeutic strategies aimed at preventing and managing age-related vascular pathologies. The implications for personalized medicine and potential areas for future research are also explored, emphasizing the need for a nuanced approach to the study and treatment of vascular aging.
Collapse
Affiliation(s)
- Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Attila Á Szabó
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health and Exercise Science, University of Oklahoma, Norman, OK, USA
- Reynolds Section of Geriatrics and Palliative Medicine, Department of Medicine, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andrea B Maier
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
- @AgeSingapore, Healthy Longevity Program, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
3
|
Li M, Zhuo X, Liu Y, You J, Lin J. PPARγ activation attenuates neonatal CRD-induced visceral pain sensitization and anxiety in male rats by alleviating oxidative stress. BMC Gastroenterol 2025; 25:22. [PMID: 39833676 PMCID: PMC11749074 DOI: 10.1186/s12876-025-03618-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Visceral pain sensitization and emotional reactions due to irritable bowel syndrome (IBS) occur frequently in the general population. Oxidative stress plays a crucial role in the pathogenesis of IBS. Previous studies have demonstrated that activation of peroxisome proliferator-activated receptor gamma (PPARγ) has analgesic effects. Therefore, we aimed to determine whether PPARγ activation ameliorates oxidative stress and affects thus nociceptive sensitization and emotional responses in IBS. METHODS The study utilized male Sprague-Dawley (SD) rats, that suffered from neonatal colorectal distension (CRD), to assess the effects of various doses of rosiglitazone on visceral hyperalgesia and anxiety. Electromyography (EMG) of the external abdominal oblique muscles was used to evaluate visceral hypersensitivity, and Open Field Test (OFT) and Elevated Plus Maze (EPM) were used to evaluate anxiety. Superoxide dismutase (SOD) and malondialdehyde (MDA) in the spinal cord were analyzed by water-soluble tetrazolium-1 (WST-1) and thiobarbituric acid (TBA) methods, respectively, the expression levels of PPARγ in the spinal cord were assessed by qRT-PCR and Western blotting. RESULTS Neonatal CRD-induced rats showed visceral pain sensitization and anxiety in adulthood, with down-regulated expression of PPARγ and SOD and elevated MDA levels in the spinal cord. Rosiglitazone alleviated visceral hypersensitivity and anxiety by activating PPARγ protein expression and promoting MDA up-regulation and SOD down-regulation in the spinal cord, which were reversed by GW9662, an antagonist of PPARγ. CONCLUSION This study demonstrated that rosiglitazone alleviated visceral pain sensitization and anxiety in male IBS rats by alleviating oxidative stress through activation of PPARγ.
Collapse
Affiliation(s)
- Minjie Li
- Department of Anesthesiology, First Affiliated Hospital, Fujian Medical University, No. 20, Cha Zhong Road, Fuzhou, Fujian Province, People's Republic of China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiyu Zhuo
- Department of Anesthesiology, Fujian Provincial Hospital, Fuzhou, China
| | - Yongxiao Liu
- Department of Anesthesiology, First Affiliated Hospital, Fujian Medical University, No. 20, Cha Zhong Road, Fuzhou, Fujian Province, People's Republic of China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jinchao You
- Department of Anesthesiology, First Affiliated Hospital, Fujian Medical University, No. 20, Cha Zhong Road, Fuzhou, Fujian Province, People's Republic of China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianqing Lin
- Department of Anesthesiology, First Affiliated Hospital, Fujian Medical University, No. 20, Cha Zhong Road, Fuzhou, Fujian Province, People's Republic of China.
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Xu S, Wang Z, Guo F, Zhang Y, Peng H, Zhang H, Liu Z, Cao C, Xin G, Chen YY, Fu J. Mitophagy in ischemic heart disease: molecular mechanisms and clinical management. Cell Death Dis 2024; 15:934. [PMID: 39737905 DOI: 10.1038/s41419-024-07303-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025]
Abstract
The influence of the mitochondrial control system on ischemic heart disease has become a major focus of current research. Mitophagy, as a very crucial part of the mitochondrial control system, plays a special role in ischemic heart disease, unlike mitochondrial dynamics. The published reviews have not explored in detail the unique function of mitophagy in ischemic heart disease, therefore, the aim of this paper is to summarize how mitophagy regulates the progression of ischemic heart disease. We conclude that mitophagy affects ischemic heart disease by promoting cardiomyocyte hypertrophy and fibrosis, the progression of oxidative stress, the development of inflammation, and cardiomyocyte death, and that the specific mechanisms of mitophagy are worthy of further investigation.
Collapse
Affiliation(s)
- Shujuan Xu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Zihan Wang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110122, China
| | - Fan Guo
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yehao Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Han Peng
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Huiyu Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Zixin Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Ce Cao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Gaojie Xin
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yuan Yuan Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jianhua Fu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
5
|
Kunutsor SK, Kaminsky LA, Lehoczki A, Laukkanen JA. Unraveling the link between cardiorespiratory fitness and cancer: a state-of-the-art review. GeroScience 2024; 46:5559-5585. [PMID: 38831183 PMCID: PMC11493895 DOI: 10.1007/s11357-024-01222-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024] Open
Abstract
Cardiorespiratory fitness (CRF) not only reflects an individual's capacity to perform physical activities but also encapsulates broader effects on the basic biology of aging. This review aims to summarize the evidence on the influence of CRF on overall and site-specific cancer risks. It delves into the biological mechanisms through which CRF may exert its effects, explores the clinical implications of these findings, identifies gaps in the current evidence base, and suggests directions for future research. The synthesis of findings reveals that higher CRF levels (general threshold of > 7 METs) are consistently associated with a reduced risk of a range of cancers, including head and neck, lung, breast, gastrointestinal, particularly pancreatic and colorectal, bladder, overall cancer incidence and mortality, and potentially stomach and liver, bile duct, and gall bladder cancers. These inverse associations between CRF and cancer risk do not generally differ across age groups, sex, race, or adiposity, suggesting a universal protective effect of CRF. Nonetheless, evidence linking CRF with skin, mouth and pharynx, kidney, and endometrial cancers is limited and inconclusive. Conversely, higher CRF levels may be potentially linked to an increased risk of prostate cancer and hematological malignancies, such as leukemia and myeloma, although the evidence is still not conclusive. CRF appears to play a significant role in reducing the risk of several cancers through various biological mechanisms, including inflammation reduction, immune system enhancement, hormonal regulation, and metabolic improvements. Overall, enhancing CRF through regular physical activity offers a vital, accessible strategy for reducing cancer risk and extending the health span. Future research should aim to fill the existing evidence gaps regarding specific cancers and elucidate the detailed dose-response relationships between CRF levels and cancer risk. Studies are also needed to elucidate the causal relationships and mechanistic pathways linking CRF to cancer outcomes.
Collapse
Affiliation(s)
- Setor K Kunutsor
- Diabetes Research Centre, Leicester General Hospital, University of Leicester, Leicester, LE5 4WP, UK.
| | - Leonard A Kaminsky
- Clinical Exercise Physiology, College of Health, Ball State University, Muncie, IN, USA
| | - Andrea Lehoczki
- Department of Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Haematology and Stem Cell Transplantation, National Institute for Haematology and Infectious Diseases, South Pest Central Hospital, 1097, Budapest, Hungary
| | - Jari A Laukkanen
- Institute of Clinical Medicine, Department of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Wellbeing Services County of Central Finland, Jyväskylä, Finland
| |
Collapse
|
6
|
He H, Huang W, Xiong L, Ma C, Wang Y, Sun P, Shi D, Li L, Yan H, Wu Y. FUNDC1-mediated mitophagy regulates photodamage independently of the PINK1/Parkin-dependent pathway. Free Radic Biol Med 2024; 225:630-640. [PMID: 39389212 DOI: 10.1016/j.freeradbiomed.2024.10.272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Ultraviolet B(UVB) triggers a pro-survival response through mitophagy, but the role of FUNDC1-mediated mitophagy in photodamaged skin remains unexplored. OBJECTIVES To clarify the function of mitophagy in UVB-induced photodamaged skin. METHODS To investigate the role of FUNDC1-mediated mitophagy in UVB-induced mitochondrial damage and cell apoptosis, FUNDC1 knockdown in C57BL/6 mice was performed using adeno-associated virus. Additionally, FUNDC1 overexpression and knockdown in HaCaT cells were conducted using lentivirus. A comprehensive analysis was conducted on a panel of human sun-exposed skin samples, alongside control samples, to assess the expression levels of FUNDC1. RESULTS In UVB-induced C57BL/6 mice, the dorsal skin showed photodamage including erythema, scaling, erosion, and scabs. The expression levels of PINK1, Parkin, and BNIP3 did not show significant changes, while FUNDC1 expression consistently declined along with LC3B. Cytochrome C, Bax, and cleaved-caspase3 were upregulated, while Bcl2 was downregulated. UVB-induced HaCaT cells showed mitochondrial damage, accompanied by FUNDC1 downregulation and BNIP3 upregulation, while PINK1 and Parkin showed no significant changes. FUNDC1 overexpression led to an increase in mtROS and a decrease in mitochondrial membrane potential and ATP levels, indicating complete mitochondrial clearance and exacerbated cell death. FUNDC1 knockdown protected against UVB-induced photodamage in mice and mitigated mitochondrial damage and apoptosis in HaCaT cells by activating compensatory PINK1/Parkin-dependent mitophagy, which was evidenced by upregulation of PINK1 and Bcl2 and downregulation of Bax. In human sun-exposed skin samples, there was a decrease in the number of FUNDC1+ cells compared with non-sun-exposed controls. CONCLUSIONS FUNDC1-mediated mitophagy regulates skin photodamage and provides a novel mechanism for resisting photodamage, presenting a potential target for future therapeutic interventions.
Collapse
Affiliation(s)
- Hailun He
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China; Department of Medical Aesthetics, The Third People's Hospital of Chengdu, Southwest Jiaotong University, Chengdu, China
| | - Wenyue Huang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Lidan Xiong
- Evaluation Center for Cosmetics Safety and Efficacy, West China Hospital, Sichuan University, Chengdu, China
| | - Cong Ma
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China; Department of Dermatology, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, China
| | - Yichong Wang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Peihong Sun
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Dongxin Shi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Liangman Li
- Orthopedics Department, The First Hospital of China Medical University, Shenyang, China
| | - Hongwei Yan
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China
| | - Yan Wu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China; National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang, China; Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Craige SM, Mammel RK, Amiri N, Willoughby OS, Drake JC. Interplay of ROS, mitochondrial quality, and exercise in aging: Potential role of spatially discrete signaling. Redox Biol 2024; 77:103371. [PMID: 39357424 PMCID: PMC11474192 DOI: 10.1016/j.redox.2024.103371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Affiliation(s)
- Siobhan M Craige
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA.
| | - Rebecca K Mammel
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA
| | - Niloufar Amiri
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA; Institute for Critical Technology and Applied Science, Virginia Tech, Blacksburg, 24061, USA
| | - Orion S Willoughby
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA
| | - Joshua C Drake
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA.
| |
Collapse
|
8
|
Zhang H, Zhang Y, Zhang J, Jia D. Exercise Alleviates Cardiovascular Diseases by Improving Mitochondrial Homeostasis. J Am Heart Assoc 2024; 13:e036555. [PMID: 39291488 DOI: 10.1161/jaha.124.036555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Engaging in regular exercise and physical activity contributes to delaying the onset of cardiovascular diseases (CVDs). However, the physiological mechanisms underlying the benefits of regular exercise or physical activity in CVDs remain unclear. The disruption of mitochondrial homeostasis is implicated in the pathological process of CVDs. Exercise training effectively delays the onset and progression of CVDs by significantly ameliorating the disruption of mitochondrial homeostasis. This includes improving mitochondrial biogenesis, increasing mitochondrial fusion, decreasing mitochondrial fission, promoting mitophagy, and mitigating mitochondrial morphology and function. This review provides a comprehensive overview of the benefits of physical exercise in the context of CVDs, establishing a connection between the disruption of mitochondrial homeostasis and the onset of these conditions. Through a detailed examination of the underlying molecular mechanisms within mitochondria, the study illuminates how exercise can provide innovative perspectives for future therapies for CVDs.
Collapse
Affiliation(s)
- Huijie Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Yuxuan Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Jiaqiao Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Dandan Jia
- School of Exercise and health Shanghai University of Sport Shanghai China
| |
Collapse
|
9
|
Wang X, He B. Endothelial dysfunction: molecular mechanisms and clinical implications. MedComm (Beijing) 2024; 5:e651. [PMID: 39040847 PMCID: PMC11261813 DOI: 10.1002/mco2.651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024] Open
Abstract
Cardiovascular disease (CVD) and its complications are a leading cause of death worldwide. Endothelial dysfunction plays a crucial role in the initiation and progression of CVD, serving as a pivotal factor in the pathogenesis of cardiovascular, metabolic, and other related diseases. The regulation of endothelial dysfunction is influenced by various risk factors and intricate signaling pathways, which vary depending on the specific disease context. Despite numerous research efforts aimed at elucidating the mechanisms underlying endothelial dysfunction, the precise molecular pathways involved remain incompletely understood. This review elucidates recent research findings on the pathophysiological mechanisms involved in endothelial dysfunction, including nitric oxide availability, oxidative stress, and inflammation-mediated pathways. We also discuss the impact of endothelial dysfunction on various pathological conditions, including atherosclerosis, heart failure, diabetes, hypertension, chronic kidney disease, and neurodegenerative diseases. Furthermore, we summarize the traditional and novel potential biomarkers of endothelial dysfunction as well as pharmacological and nonpharmacological therapeutic strategies for endothelial protection and treatment for CVD and related complications. Consequently, this review is to improve understanding of emerging biomarkers and therapeutic approaches aimed at reducing the risk of developing CVD and associated complications, as well as mitigating endothelial dysfunction.
Collapse
Affiliation(s)
- Xia Wang
- Department of CardiologyShanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ben He
- Department of CardiologyShanghai Chest Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
10
|
Han Q, Yu Y, Liu X, Guo Y, Shi J, Xue Y, Li Y. The Role of Endothelial Cell Mitophagy in Age-Related Cardiovascular Diseases. Aging Dis 2024:AD.2024.0788. [PMID: 39122456 DOI: 10.14336/ad.2024.0788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Aging is a major risk factor for cardiovascular diseases (CVD), and mitochondrial autophagy impairment is considered a significant physiological change associated with aging. Endothelial cells play a crucial role in maintaining vascular homeostasis and function, participating in various physiological processes such as regulating vascular tone, coagulation, angiogenesis, and inflammatory responses. As aging progresses, mitochondrial autophagy impairment in endothelial cells worsens, leading to the development of numerous cardiovascular diseases. Therefore, regulating mitochondrial autophagy in endothelial cells is vital for preventing and treating age-related cardiovascular diseases. However, there is currently a lack of systematic reviews in this area. To address this gap, we have written this review to provide new research and therapeutic strategies for managing aging and age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Quancheng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiujuan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yonghong Guo
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
11
|
Tan S, Ge Y, Bi J. Methylation regulation for FUNDC1 stability in childhood leukemia was up-regulated and facilitates metastasis and reduces ferroptosis of leukemia through mitochondrial damage by FBXL2. Open Med (Wars) 2024; 19:20230810. [PMID: 38947217 PMCID: PMC11211875 DOI: 10.1515/med-2023-0810] [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: 06/18/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 07/02/2024] Open
Abstract
Leukemia, the most common malignant tumor in childhood, can be categorized into acute leukemia and chronic leukemia. However, the role of FUNDC1 in childhood leukemia (CL) remains unknown. This study aims to investigate the effects of FUNDC1 on patients with CL and its underlying mechanism both in vivo and in vitro. The mRNA expression levels of FUNDC1 were found to be up-regulated in serum samples from CL patients as well as in leukemia cell lines. Furthermore, it was observed that the mRNA expression of FUNDC1 was lower in stage I-II CL patients compared to stage III-IV patients. The up-regulation of FUNDC1 was found to promote leukemia metastasis. Additionally, it was discovered that FUNDC1 up-regulation reduces ferroptosis by inhibiting mitochondrial damage. In a leukemia model, FUNDC1 up-regulation induces the expression of FBXL2. Moreover, FUNDC1 up-regulation reduces FBXL2 ubiquitination, thus maintaining FBXL2 protein expression in leukemia. By inducing FBXL2, FUNDC1 reduces ferroptosis in leukemia through the inhibition of mitochondrial damage. The stability of FUNDC1 is controlled by METTL3 methylation. Overall, this study sheds light on the role of FUNDC1 in CL and provides insights into its underlying mechanisms.
Collapse
Affiliation(s)
- Sihai Tan
- Department of Pediatric, Hubei Enshi Tujia and Miao Autonomous Prefecture Central Hospital, Hubei Province, Enshi445000, China
| | - Yirong Ge
- Department of Pediatric, Hubei Enshi Tujia and Miao Autonomous Prefecture Central Hospital, Hubei Province, Enshi445000, China
| | - Jing Bi
- Department of Pediatric, Hubei Enshi Tujia and Miao Autonomous Prefecture Central Hospital, Hubei Province, Enshi445000, China
| |
Collapse
|
12
|
Lv Y, Yu Z, Zhang P, Zhang X, Li H, Liang T, Guo Y, Cheng L, Peng F. The structure and function of FUN14 domain-containing protein 1 and its contribution to cardioprotection by mediating mitophagy. Front Pharmacol 2024; 15:1389953. [PMID: 38828457 PMCID: PMC11140143 DOI: 10.3389/fphar.2024.1389953] [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: 02/22/2024] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Cardiovascular disease (CVD) is a serious public health risk, and prevention and treatment efforts are urgently needed. Effective preventive and therapeutic programs for cardiovascular disease are still lacking, as the causes of CVD are varied and may be the result of a multifactorial combination. Mitophagy is a form of cell-selective autophagy, and there is increasing evidence that mitophagy is involved in cardioprotective processes. Recently, many studies have shown that FUN14 domain-containing protein 1 (FUNDC1) levels and phosphorylation status are highly associated with many diseases, including heart disease. Here, we review the structure and functions of FUNDC1 and the path-ways of its mediated mitophagy, and show that mitophagy can be effectively activated by dephosphorylation of Ser13 and Tyr18 sites, phosphorylation of Ser17 site and ubiquitination of Lys119 site in FUNDC1. By effectively activating or inhibiting excessive mitophagy, the quality of mitochondria can be effectively controlled. The main reason is that, on the one hand, improper clearance of mitochondria and accumulation of damaged mitochondria are avoided, and on the other hand, excessive mitophagy causing apoptosis is avoided, both serving to protect the heart. In addition, we explore the possible mechanisms by which FUNDC1-mediated mitophagy is involved in exercise preconditioning (EP) for cardioprotection. Finally, we also point out unresolved issues in FUNDC1 and its mediated mitophagy and give directions where further research may be needed.
Collapse
Affiliation(s)
- Yuhu Lv
- College of Physical Education, Guangdong University of Education, Guangzhou, China
- Research Center for Adolescent Sports and Health Promotion of Guangdong Province, Guangzhou, China
| | - Zhengze Yu
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Peiwen Zhang
- College of Nursing and Rehabilitation, Xi an FanYi University, Xi’an, China
| | - Xiqian Zhang
- College of Physical Education, Guangdong University of Education, Guangzhou, China
- Research Center for Adolescent Sports and Health Promotion of Guangdong Province, Guangzhou, China
| | - Huarui Li
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Ting Liang
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Yanju Guo
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Lin Cheng
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Fenglin Peng
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| |
Collapse
|
13
|
Li T, Yang K, Gao W, Peng F, Zou X. Cellular senescence in acute kidney injury: Target and opportunity. Biochem Biophys Res Commun 2024; 706:149744. [PMID: 38479244 DOI: 10.1016/j.bbrc.2024.149744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/24/2024]
Abstract
Acute kidney injury (AKI) is a common clinical disease with a high incidence and mortality rate. It typically arises from hemodynamic alterations, sepsis, contrast agents, and toxic drugs, instigating a series of events that culminate in tissue and renal damage. This sequence of processes often leads to acute renal impairment, prompting the initiation of a repair response. Cellular senescence is an irreversible arrest of the cell cycle. Studies have shown that renal cellular senescence is closely associated with AKI through several mechanisms, including the promotion of oxidative stress and inflammatory response, telomere shortening, and the down-regulation of klotho expression. Exploring the role of cellular senescence in AKI provides innovative therapeutic ideas for both the prevention and treatment of AKI. Furthermore, it has been observed that targeted removal of senescent cells in vivo can efficiently postpone senescence, resulting in an enhanced prognosis for diseases associated with senescence. This article explores the effects of common anti-senescence drugs senolytics and senostatic and lifestyle interventions on renal diseases, and mentions the rapid development of mesenchymal stem cells (MSCs). These studies have taken senescence-related research to a new level. Overall, this article comprehensively summarizes the studies on cellular senescence in AKI, aiming is to elucidate the relationship between cellular senescence and AKI, and explore treatment strategies to improve the prognosis of AKI.
Collapse
Affiliation(s)
- Ting Li
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, China.
| | - Kexin Yang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, China
| | - Fujun Peng
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, China
| | - Xiangyu Zou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, China.
| |
Collapse
|
14
|
Ying X, Zheng X, Zhang X, Yin Y, Wang X. Kynurenine in IDO1 high cancer cell-derived extracellular vesicles promotes angiogenesis by inducing endothelial mitophagy in ovarian cancer. J Transl Med 2024; 22:267. [PMID: 38468343 PMCID: PMC10929174 DOI: 10.1186/s12967-024-05054-5] [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: 12/04/2023] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Mitophagy, a prominent cellular homeostasis process, has been implicated in modulating endothelial cell function. Emerging evidence suggests that extracellular vesicles (EVs) participate in intercellular communication, which could modulate tumor angiogenesis, a hallmark of ovarian cancer (OC) progression. However, the underlying mechanisms through how EVs regulate endothelial mitophagy associated with tumor angiogenesis during OC development remain obscure. METHODS The effect of cancer cell-derived EVs on endothelial mitophagy and its correlation with tumor angiogenesis and OC development were explored by in vitro and in vivo experiments. Multi-omics integration analysis was employed to identify potential regulatory mechanisms of cancer cell-derived EVs on endothelial mitophagy, which is involved in tumor angiogenesis associated with OC development. These insights were then further corroborated through additional experiments. An orthotopic OC mouse model was constructed to assess the antiangiogenic and therapeutic potential of the Indoleamine 2,3 dioxygenase-1 (IDO1) inhibitor. RESULTS Cancer cell-derived EVs promoted tumor angiogenesis via the activation of endothelial mitophagy, contributing to the growth and metastasis of OC. The aberrantly high expression of IDO1 mediated abnormal tryptophan metabolism in cancer cells and promoted the secretion of L-kynurenine (L-kyn)-enriched EVs, with associated high levels of L-kyn in EVs isolated from both the tumor tissues and patient plasma in OC. EVs derived from IDO1high ovarian cancer cells elevated nicotinamide adenine dinucleotide (NAD +) levels in endothelial cells via delivering L-kyn. Besides, IDO1high ovarian cancer cell-derived EVs upregulated sirt3 expression in endothelial cells by increasing acetylation modification. These findings are crucial for promoting endothelial mitophagy correlated with tumor angiogenesis. Notably, both endothelial mitophagy and tumor angiogenesis could be suppressed by the IDO1 inhibitor in the orthotopic OC mouse model. CONCLUSIONS Together, our findings unveil a mechanism of mitophagy in OC angiogenesis and indicate the clinical relevance of EV enriched L-kyn as a potential biomarker for tumorigenesis and progression. Additionally, IDO1 inhibitors might become an alternative option for OC adjuvant therapy.
Collapse
Affiliation(s)
- Xiang Ying
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xiaoqian Zhang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
15
|
Guo Y, Guan T, Yu Q, Sanghai N, Shafiq K, Li M, Jiao X, Na D, Zhang G, Kong J. ALS-linked SOD1 mutations impair mitochondrial-derived vesicle formation and accelerate aging. Redox Biol 2024; 69:102972. [PMID: 38056310 PMCID: PMC10746562 DOI: 10.1016/j.redox.2023.102972] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
Oxidative stress (OS) is regarded as the dominant theory for aging. While compelling correlative data have been generated to support the OS theory, a direct cause-and-effect relationship between the accumulation of oxidation-mediated damage and aging has not been firmly established. Superoxide dismutase 1 (SOD1) is a primary antioxidant in all cells. It is, however, susceptible to oxidation due to OS and gains toxic properties to cells. This study investigates the role of oxidized SOD1 derived from amyotrophic lateral sclerosis (ALS) linked SOD1 mutations in cell senescence and aging. Herein, we have shown that the cell line NSC34 expressing the G93A mutation of human SOD1 (hSOD1G93A) entered premature senescence as evidenced by a decreased number of the 5-ethynyl-2'-deoxyuridine (EdU)-positive cells. There was an upregulation of cellular senescence markers compared to cells expressing the wild-type human SOD1 (hSOD1WT). Transgenic mice carrying the hSOD1G93A gene showed aging phenotypes at an early age (135 days) with high levels of P53 and P16 but low levels of SIRT1 and SIRT6 compared with age-matched hSOD1WT transgenic mice. Notably, the levels of oxidized SOD1 were significantly elevated in both the senescent NSC34 cells and 135-day hSOD1G93A mice. Selective removal of oxidized SOD1 by our CT4-directed autophagy significantly decelerated aging, indicating that oxidized SOD1 is a causal factor of aging. Intriguingly, mitochondria malfunctioned in both senescent NSC34 cells and middle-aged hSODG93A transgenic mice. They exhibited increased production of mitochondrial-derived vesicles (MDVs) in response to mild OS in mutant humanSOD1 (hSOD1) transgenic mice at a younger age; however, the mitochondrial response gradually declined with aging. In conclusion, our data show that oxidized SOD1 derived from ALS-linked SOD1 mutants is a causal factor for cellular senescence and aging. Compromised mitochondrial responsiveness to OS may serve as an indicator of premature aging.
Collapse
Affiliation(s)
- Ying Guo
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada; Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Qiang Yu
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Nitesh Sanghai
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Canada
| | - Kashfia Shafiq
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Meiyu Li
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Xin Jiao
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Donghui Na
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China.
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada.
| |
Collapse
|
16
|
Chen Z, Li S, Liu M, Yin M, Chen J, Li Y, Li Q, Zhou Y, Xia Y, Chen A, Lu D, Li C, Chen Y, Qian J, Ge J. Nicorandil alleviates cardiac microvascular ferroptosis in diabetic cardiomyopathy: Role of the mitochondria-localized AMPK-Parkin-ACSL4 signaling pathway. Pharmacol Res 2024; 200:107057. [PMID: 38218357 DOI: 10.1016/j.phrs.2024.107057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
Mitochondria-associated ferroptosis exacerbates cardiac microvascular dysfunction in diabetic cardiomyopathy (DCM). Nicorandil, an ATP-sensitive K+ channel opener, protects against endothelial dysfunction, mitochondrial dysfunction, and DCM; however, its effects on ferroptosis and mitophagy remain unexplored. The present study aimed to assess the beneficial effects of nicorandil against endothelial ferroptosis in DCM and the underlying mechanisms. Cardiac microvascular perfusion was assessed using a lectin perfusion assay, while mitophagy was assessed via mt-Keima transfection and transmission electron microscopy. Ferroptosis was examined using mRNA sequencing, fluorescence staining, and western blotting. The mitochondrial localization of Parkin, ACSL4, and AMPK was determined via immunofluorescence staining. Following long-term diabetes, nicorandil treatment improved cardiac function and remodeling by alleviating cardiac microvascular injuries, as evidenced by the improved microvascular perfusion and structural integrity. mRNA-sequencing and biochemical analyses showed that ferroptosis occurred and Pink1/Parkin-dependent mitophagy was suppressed in cardiac microvascular endothelial cells after diabetes. Nicorandil treatment suppressed mitochondria-associated ferroptosis by promoting the Pink1/Parkin-dependent mitophagy. Moreover, nicorandil treatment increased the phosphorylation level of AMPKα1 and promoted its mitochondrial translocation, which further inhibited the mitochondrial translocation of ACSL4 via mitophagy and ultimately suppressed mitochondria-associated ferroptosis. Importantly, overexpression of mitochondria-localized AMPKα1 (mitoAα1) shared similar benefits with nicorandil on mitophagy, ferroptosis and cardiovascular protection against diabetic injury. In conclusion, the present study demonstrated the therapeutic effects of nicorandil against cardiac microvascular ferroptosis in DCM and revealed that the mitochondria-localized AMPK-Parkin-ACSL4 signaling pathway mediates mitochondria-associated ferroptosis and the development of cardiac microvascular dysfunction.
Collapse
Affiliation(s)
- Zhangwei Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Muyin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ming Yin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jinxiang Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Youran Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Qiyu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yan Xia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ao Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Danbo Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Chenguang Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Yuqiong Chen
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
17
|
Tang S, Geng Y, Lin Q. The role of mitophagy in metabolic diseases and its exercise intervention. Front Physiol 2024; 15:1339128. [PMID: 38348222 PMCID: PMC10859464 DOI: 10.3389/fphys.2024.1339128] [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: 11/17/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
Mitochondria are energy factories that sustain life activities in the body, and their dysfunction can cause various metabolic diseases that threaten human health. Mitophagy, an essential intracellular mitochondrial quality control mechanism, can maintain cellular and metabolic homeostasis by removing damaged mitochondria and participating in developing metabolic diseases. Research has confirmed that exercise can regulate mitophagy levels, thereby exerting protective metabolic effects in metabolic diseases. This article reviews the role of mitophagy in metabolic diseases, the effects of exercise on mitophagy, and the potential mechanisms of exercise-regulated mitophagy intervention in metabolic diseases, providing new insights for future basic and clinical research on exercise interventions to prevent and treat metabolic diseases.
Collapse
Affiliation(s)
| | | | - Qinqin Lin
- School of Physical Education, Yanshan University, Qinhuangdao, China
| |
Collapse
|
18
|
Jia D, Tian Z, Wang R. Exercise mitigates age-related metabolic diseases by improving mitochondrial dysfunction. Ageing Res Rev 2023; 91:102087. [PMID: 37832607 DOI: 10.1016/j.arr.2023.102087] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
The benefits of regular physical activity are related to delaying and reversing the onset of ageing and age-related disorders, including cardiomyopathy, neurodegenerative diseases, cancer, obesity, diabetes, and fatty liver diseases. However, the molecular mechanisms of the benefits of exercise or physical activity on ageing and age-related disorders remain poorly understood. Mitochondrial dysfunction is implicated in the pathogenesis of ageing and age-related metabolic diseases. Mitochondrial health is an important mediator of cellular function. Therefore, exercise alleviates metabolic diseases in individuals with advancing ageing and age-related diseases by the remarkable promotion of mitochondrial biogenesis and function. Exerkines are identified as signaling moieties released in response to exercise. Exerkines released by exercise have potential roles in improving mitochondrial dysfunction in response to age-related disorders. This review comprehensive summarizes the benefits of exercise in metabolic diseases, linking mitochondrial dysfunction to the onset of age-related diseases. Using relevant examples utilizing this approach, the possibility of designing therapeutic interventions based on these molecular mechanisms is addressed.
Collapse
Affiliation(s)
- Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
19
|
Chu C, Wang X, Yang C, Chen F, Shi L, Xu W, Wang K, Liu B, Wang C, Sun D, Ding W. Neutrophil extracellular traps drive intestinal microvascular endothelial ferroptosis by impairing Fundc1-dependent mitophagy. Redox Biol 2023; 67:102906. [PMID: 37812880 PMCID: PMC10579540 DOI: 10.1016/j.redox.2023.102906] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023] Open
Abstract
Microvascular endothelial damage caused by intestinal ischemia‒reperfusion (II/R) is a primary catalyst for microcirculation dysfunction and enterogenous infection. Previous studies have mainly focused on how neutrophil extracellular traps (NETs) and ferroptosis cause intestinal epithelial injury, and little attention has been given to how NETs, mainly from circulatory neutrophils, affect intestinal endothelial cells during II/R. This study aimed to unravel the mechanisms through which NETs cause intestinal microvascular dysfunction. We first detected heightened local NET infiltration around the intestinal microvasculature, accompanied by increased endothelial cell ferroptosis, resulting in microcirculation dysfunction in both human and animal II/R models. However, the administration of the ferroptosis inhibitor ferrostatin-1 or the inhibition of NETs via neutrophil-specific peptidylarginine deiminase 4 (Pad4) deficiency led to positive outcomes, with reduced intestinal endothelial ferroptosis and microvascular function recovery. Moreover, RNA-seq analysis revealed a significant enrichment of mitophagy- and ferroptosis-related signaling pathways in HUVECs incubated with NETs. Mechanistically, elevated NET formation induced Fundc1 phosphorylation at Tyr18 in intestinal endothelial cells, which led to mitophagy inhibition, mitochondrial quality control imbalance, and excessive mitochondrial ROS generation and lipid peroxidation, resulting in endothelial ferroptosis and microvascular dysfunction. Nevertheless, using the mitophagy activator urolithin A or AAV-Fundc1 transfection could reverse this process and ameliorate microvascular damage. We first demonstrate that increased NETosis could result in intestinal microcirculatory dysfunction and conclude that suppressed NET formation can mitigate intestinal endothelial ferroptosis by improving Fundc1-dependent mitophagy. Targeting NETs could be a promising approach for treating II/R-induced intestinal microcirculatory dysfunction.
Collapse
Affiliation(s)
- Chengnan Chu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xinyu Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chao Yang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Fang Chen
- School of Medicine, Southeast University, Nanjing, 210002, Jiangsu Province, China
| | - Lin Shi
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Weiqi Xu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Kai Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Baochen Liu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chenyang Wang
- Key Laboratory of Intestinal Injury, Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, PR China
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Weiwei Ding
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
20
|
Guo Y, Guan T, Shafiq K, Yu Q, Jiao X, Na D, Li M, Zhang G, Kong J. Mitochondrial dysfunction in aging. Ageing Res Rev 2023; 88:101955. [PMID: 37196864 DOI: 10.1016/j.arr.2023.101955] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/27/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Aging is a complex process that features a functional decline in many organelles. Although mitochondrial dysfunction is suggested as one of the determining factors of aging, the role of mitochondrial quality control (MQC) in aging is still poorly understood. A growing body of evidence points out that reactive oxygen species (ROS) stimulates mitochondrial dynamic changes and accelerates the accumulation of oxidized by-products through mitochondrial proteases and mitochondrial unfolded protein response (UPRmt). Mitochondrial-derived vesicles (MDVs) are the frontline of MQC to dispose of oxidized derivatives. Besides, mitophagy helps remove partially damaged mitochondria to ensure that mitochondria are healthy and functional. Although abundant interventions on MQC have been explored, over-activation or inhibition of any type of MQC may even accelerate abnormal energy metabolism and mitochondrial dysfunction-induced senescence. This review summarizes mechanisms essential for maintaining mitochondrial homeostasis and emphasizes that imbalanced MQC may accelerate cellular senescence and aging. Thus, appropriate interventions on MQC may delay the aging process and extend lifespan.
Collapse
Affiliation(s)
- Ying Guo
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kashfia Shafiq
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Qiang Yu
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xin Jiao
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Donghui Na
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Meiyu Li
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China.
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
21
|
Sagar S, Gustafsson AB. Cardiovascular aging: the mitochondrial influence. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:33. [PMID: 37583788 PMCID: PMC10426788 DOI: 10.20517/jca.2023.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Age-associated cardiovascular disease is becoming progressively prevalent due to the increased lifespan of the population. However, the fundamental mechanisms underlying the aging process and the corresponding decline in tissue functions are still poorly understood. The heart has a very high energy demand and the cellular energy needed to sustain contraction is primarily generated by mitochondrial oxidative phosphorylation. Mitochondria are also involved in supporting various metabolic processes, as well as activation of the innate immune response and cell death pathways. Given the central role of mitochondria in energy metabolism and cell survival, the heart is highly susceptible to the effects of mitochondrial dysfunction. These key organelles have been implicated as underlying drivers of cardiac aging. Here, we review the evidence demonstrating the mitochondrial contribution to the cardiac aging process and disease susceptibility. We also discuss the potential mechanisms responsible for the age-related decline in mitochondrial function.
Collapse
Affiliation(s)
- Shakti Sagar
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Asa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|