1
|
Zhang Y, Rao X, Wang J, Liu H, Wang Q, Wang X, Hua F, Guan X, Lin Y. Mitochondria-Associated Membranes: A Key Point of Neurodegenerative Diseases. CNS Neurosci Ther 2025; 31:e70378. [PMID: 40406921 PMCID: PMC12099310 DOI: 10.1111/cns.70378] [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: 12/11/2024] [Revised: 03/12/2025] [Accepted: 03/29/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Neurodegenerative diseases pose significant health challenges in the 21st century, with increasing morbidity and mortality, particularly among the elderly population. One of the key factors contributing to the pathogenesis of these diseases is the disrupted crosstalk between mitochondria and the endoplasmic reticulum. Mitochondria-associated membranes (MAMs), which are regions where the ER interfaces with mitochondria, serve as crucial platforms facilitating communication between these organelles. OBJECTIVES This review focuses on the structural composition and functions of MAMs and highlights their roles. Additionally, in this review, we summarize the relationship between MAM dysfunction and various neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and others. The involvement of key proteins such as Sig-1R, IP3R, and VAPB in maintaining ER-mitochondrial communication and their dysfunction in neurodegenerative diseases is emphasized. CONCLUSION Through analyzing the effects of MAM on neurodegenerative diseases, we provide the newest insights and potential therapeutic targets for the treatment of these debilitating conditions.
Collapse
Affiliation(s)
- Yiwei Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxi ProvinceChina
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
- Queen Mary CollegeNanchang UniversityNanchangJiangxi ProvinceChina
| | - Xiuqin Rao
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxi ProvinceChina
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
| | - Jiayi Wang
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
- Queen Mary CollegeNanchang UniversityNanchangJiangxi ProvinceChina
| | - Hantian Liu
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
- Queen Mary CollegeNanchang UniversityNanchangJiangxi ProvinceChina
| | - Qixian Wang
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
- Queen Mary CollegeNanchang UniversityNanchangJiangxi ProvinceChina
| | - Xifeng Wang
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxi ProvinceChina
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxi ProvinceChina
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
| | - Xilong Guan
- Department of AnesthesiologyYingtan City People's HospitalYingtan CityJiangxi ProvinceChina
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxi ProvinceChina
- Jiangxi Provincial Key Laboratory of AnesthesiologyNanchangJiangxi ProvinceChina
| |
Collapse
|
2
|
Li J, Lin Q, Ren C, Li X, Li X, Li H, Li S. The perspective of modern transplant science - transplant arteriosclerosis: inspiration derived from mitochondria associated endoplasmic reticulum membrane dysfunction in arterial diseases. Int J Surg 2025; 111:3430-3440. [PMID: 40146783 DOI: 10.1097/js9.0000000000002362] [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: 09/03/2024] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
The mitochondria-associated endoplasmic reticulum membrane (MAM) is a crucial structure connecting mitochondria and the endoplasmic reticulum (ER), regulating intracellular calcium homeostasis, lipid metabolism, and various signaling pathways essential for arterial health. Recent studies highlight MAM's significant role in modulating vascular endothelial cells (EC) and vascular smooth muscle cells (VSMC), establishing it as a key regulator of arterial health and a contributor to vascular disease pathogenesis. Organ transplantation is the preferred treatment for end-stage organ failure, but transplant arteriosclerosis (TA) can lead to chronic transplant dysfunction, significantly impacting patient survival. TA, like other vascular diseases, features endothelial dysfunction and abnormal proliferation and migration of VSMC. Previous research on TA has focused on immune factors; the pathological and physiological changes in grafts following immune system attacks have garnered insufficient attention. For example, the potential roles of MAM in TA have not been thoroughly investigated. Investigating the relationship between MAM and TA, as well as the mechanisms behind TA progression, is essential. This review aims to outline the fundamental structure and the primary functions of MAM, summarize its key molecular regulators of vascular health, and explore future prospects for MAM in the context of TA research, providing insights for both basic research and clinical management of TA.
Collapse
Affiliation(s)
- Jingyi Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qian Lin
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Ren
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaodong Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaowei Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Haofeng Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Shadan Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Simmen T, Pellegrini L. A lipid in transit - the journey of cholesterol into the heart of mitochondrial research. J Cell Sci 2025; 138:jcs263907. [PMID: 40337919 DOI: 10.1242/jcs.263907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Abstract
Mitochondrial cholesterol biology in non-steroidogenic tissues remains understudied in cell science. Although detecting cholesterol in mitochondria is challenging due to isolation difficulties, studies using mitoplasts (mitochondria stripped of their outer membrane) and imaging approaches confirm its presence in the inner mitochondrial membrane. Through analysis of published evidence and first-principles reasoning, we advance a model of cholesterol trafficking into and out of mitochondria via phospholipids at mitochondria-associated membranes (MAMs), challenging the traditional view of protein-driven transport. In this model, cholesterol enters mitochondria alongside phosphatidylserine and exits with phosphatidylethanolamine - either unchanged or in a hydroxylated form after modification by the enzyme CYP27A1. Strong cholesterol-phospholipid binding energies, ∼17 kcal/mol (71.128 kJ/mol), support this lipid-mediated mechanism, suggesting it complements protein-based pathways. Future research should explore how these mechanisms collaborate to regulate mitochondrial cholesterol trafficking. By rethinking cholesterol dynamics, we raise the possibility that cholesterol plays a larger role in mitochondrial biology, influencing membrane-dependent functions like cristae structure, respiratory efficiency and inter-organelle communication. This Perspective also highlights the potential of mitochondria to regulate both dietary and endogenous cholesterol flux and homeostasis across the cell.
Collapse
Affiliation(s)
- Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Luca Pellegrini
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
4
|
Hong W, Zeng X, Ma R, Tian Y, Miu H, Ran X, Song R, Luo Z, Ju D, Ma D, Ashrafizadeh M, Bhutia SK, Conde J, Sethi G, Huang H, Duan C. Age-associated reduction in ER-Mitochondrial contacts impairs mitochondrial lipid metabolism and autophagosome formation in the heart. Cell Death Differ 2025:10.1038/s41418-025-01511-w. [PMID: 40254645 DOI: 10.1038/s41418-025-01511-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
The accumulation of dysfunctional giant mitochondria is a hallmark of aged cardiomyocytes. This study investigated the core mechanism underlying this phenomenon, focusing on the disruption of mitochondrial lipid metabolism and its effects on mitochondrial dynamics and autophagy, using both naturally aging mouse models and etoposide-induced cellular senescence models. In aged cardiomyocytes, a reduction in endoplasmic reticulum-mitochondrial (ER-Mito) contacts impairs lipid transport and leads to insufficient synthesis of mitochondrial phosphatidylethanolamine (PE). A deficiency in phosphatidylserine decarboxylase (PISD) further hinders the conversion of phosphatidylserine to PE within mitochondria, exacerbating the deficit of PE production. This PE shortage disrupts autophagosomal membrane formation, leading to impaired autophagic flux and the accumulation of damaged mitochondria. Modulating LACTB expression to enhance PISD activity and PE production helps maintain mitochondrial homeostasis and the integrity of aging cardiomyocytes. These findings highlight the disruption of mitochondrial lipid metabolism as a central mechanism driving the accumulation of dysfunctional giant mitochondria in aged cardiomyocytes and suggest that inhibiting LACTB expression could serve as a potential therapeutic strategy for mitigating cardiac aging and preserving mitochondrial function.
Collapse
Affiliation(s)
- Weilong Hong
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Xue Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, P.R. China
| | - Ruiyan Ma
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, P.R. China
| | - Yu Tian
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Huimin Miu
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Xiaoping Ran
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Rui Song
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Zhenchun Luo
- Intensive Care Unit, Chongqing Traditional Chinese Medicine Hospital, Chongqing, P.R. China
| | - Dapeng Ju
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Daqing Ma
- Perioperative and Systems Medicine Laboratory, Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, 769008, Odisha, India
| | - João Conde
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS | FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China.
| | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China.
| |
Collapse
|
5
|
Liao J, Shao M, Zhou Z, Wang S, Lv Y, Lu Y, Yao F, Li W, Yang L. Correlation of organelle interactions in the development of non-alcoholic fatty liver disease. Front Immunol 2025; 16:1567743. [PMID: 40308615 PMCID: PMC12040704 DOI: 10.3389/fimmu.2025.1567743] [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: 01/28/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Organelles, despite having distinct functions, interact with each other. Interactions between organelles typically occur at membrane contact sites (MCSs) to maintain cellular homeostasis, allowing the exchange of metabolites and other pieces of information required for normal cellular physiology. Imbalances in organelle interactions may lead to various pathological processes. Increasing evidence suggests that abnormalorganelle interactions contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). However, the key role of organelle interactions in NAFLD has not been fully evaluated and researched. In this review, we summarize the role of organelle interactions in NAFLD and emphasize their correlation with cellular calcium homeostasis, lipid transport, and mitochondrial dynamics.
Collapse
Affiliation(s)
- Jiabao Liao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Department of Endocrinology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Mengqiu Shao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Ze Zhou
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Si Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - You Lv
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yanming Lu
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Fang Yao
- Department of Endocrinology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Wenting Li
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Ling Yang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
6
|
Zanfardino P, Amati A, Perrone M, Petruzzella V. The Balance of MFN2 and OPA1 in Mitochondrial Dynamics, Cellular Homeostasis, and Disease. Biomolecules 2025; 15:433. [PMID: 40149969 PMCID: PMC11940761 DOI: 10.3390/biom15030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondrial dynamics, governed by fusion and fission, are crucial for maintaining cellular homeostasis, energy production, and stress adaptation. MFN2 and OPA1, key regulators of mitochondrial fusion, play essential roles beyond their structural functions, influencing bioenergetics, intracellular signaling, and quality control mechanisms such as mitophagy. Disruptions in these processes, often caused by MFN2 or OPA1 mutations, are linked to neurodegenerative diseases like Charcot-Marie-Tooth disease type 2A (CMT2A) and autosomal dominant optic atrophy (ADOA). This review explores the molecular mechanisms underlying mitochondrial fusion, the impact of MFN2 and OPA1 dysfunction on oxidative phosphorylation and autophagy, and their role in disease progression. Additionally, we discuss the divergent cellular responses to MFN2 and OPA1 mutations, particularly in terms of proliferation, senescence, and metabolic signaling. Finally, we highlight emerging therapeutic strategies to restore mitochondrial integrity, including mTOR modulation and autophagy-targeted approaches, with potential implications for neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Vittoria Petruzzella
- Department of Translational Biomedicine and Neurosciences (DiBraiN), University of Bari Aldo Moro, Piazza Giulio Cesare, 70124 Bari, Italy; (P.Z.); (A.A.); (M.P.)
| |
Collapse
|
7
|
Zhang X, Zhang J, Xun G, Gao Y, Zhao J, Fu Y, Su S, Kong D, Wang Q, Wang X. Alleviation effect of macrophage depletion on hepatotoxicity of triptolide: A new insight based on metabolomics and proteomics. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119485. [PMID: 39947369 DOI: 10.1016/j.jep.2025.119485] [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: 07/18/2024] [Revised: 12/14/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Triptolide (TP) is an abietane-type diterpenoid isolated from the traditional Chinese herb Tripterygium wilfordii Hook. F, which is used to relieve rheumatism, alleviate joint pain and swelling, and promote blood circulation for more than 600 years in China. The most common preparations containing TP from Tripterygium wilfordii Hook F, which are Tripterygium tablets and Tripterygium glycoside tablets, are widely used in clinical for treating rheumatoid arthritis and other autoimmune diseases at present. However, the clinical application is hindered by severe systemic toxicity induced by TP, especially hepatotoxicity. It is crucial to discover potent and specific detoxification strategy for TP. AIM OF STUDY According to our previous study, TP-induced hepatotoxicity is primarily related to macrophages. This study aimed to investigate the alleviation effects of macrophage depletion on the TP-induced liver injury in mice and to explore the related mechanisms by integration of metabolomics and proteomics. MATERIALS AND METHODS Mice were treated with clodronate liposomes to deplete macrophage before administration of triptolide. The alleviation effects were evaluated by biochemical analysis of serum and histopathology observation of the hepatic tissues. Metabolomics and proteomics were carried out to explore the mechanism of macrophage depletion on triptolide-induced liver injury. The levels of mRNA and protein of TLR4- MyD88-NF-κB axis were further detected. RESULTS The altered levels of biochemistry indicators, including aminotransferase (ALT) and aspartate aminotransferase (AST), albumin (ALB), and γ-glutamyltranspeptidase (GGT) were significantly recovered, and histopathological liver injury also showed restoring tendency in mice with macrophage depletion compared to mice with TP-treatment. The inflammation indicator interleukin-6 (IL-6) and interleukin-1β (IL-1β) were recovered significantly after depletion of macrophage. Results of metabolomics and proteomics demonstrated that macrophage depletion exerted protective effects on triptolide-induced liver injury by regulating 85 metabolites and 202 proteins. Joint analysis of multi-omics data suggested macrophage depletion could regulate lipid metabolism and maintain inflammatory homeostasis. The increased expression of NF-κB, TLR4, and MyD88 were decreased after depletion of macrophage. CONCLUSION TP-induced hepatotoxicity is mainly associated with dysfunction of macrophages and imbalance of inflammatory homeostasis. The findings of this study may help facilitate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoguang Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Jia Zhang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Ge Xun
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Yanhua Gao
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Jie Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China
| | - Yan Fu
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, Hebei, PR China; School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Suwen Su
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Qiao Wang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China.
| | - Xu Wang
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
8
|
Ono T, Taketomi Y, Higashi T, Sato H, Mochizuki-Ono C, Nagasaki Y, Ueta T, Miyai T, Tokuoka SM, Oda Y, Nishito Y, Ono T, Taya C, Arata S, Watanabe S, Soga T, Hirabayashi T, Aihara M, Murakami M. PNPLA6 regulates retinal homeostasis by choline through phospholipid turnover. Nat Commun 2025; 16:2221. [PMID: 40082403 PMCID: PMC11906636 DOI: 10.1038/s41467-025-57402-8] [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: 04/02/2023] [Accepted: 02/20/2025] [Indexed: 03/16/2025] Open
Abstract
Although mutations in human patatin-like phospholipase PNPLA6 are associated with hereditary retinal degenerative diseases, its mechanistic action in the retina is poorly understood. Here, we uncover the molecular mechanism by which PNPLA6 dysfunction disturbs retinal homeostasis and visual function. PNPLA6, by acting as a phospholipase B, regulates choline mobilization from phosphatidylcholine and subsequent choline turnover for phosphatidylcholine regeneration in retinal pigment epithelial cells. PNPLA6-driven choline is supplied from retinal pigment epithelial cells to adjacent photoreceptor cells to support their survival. Inhibition of this pathway results in abnormal morphology, proliferation, metabolism, and functions of retinal pigment epithelial and photoreceptor cells, and mice with retina-specific PNPLA6 deletion exhibit retinitis pigmentosa-like retinal degeneration. Notably, these abnormalities are entirely rescued by choline supplementation. Thus, PNPLA6 plays an essential role in retinal homeostasis by controlling choline availability for phospholipid recycling and provide a framework for the development of an ophthalmic drug target for retinal degeneration.
Collapse
Affiliation(s)
- Takashi Ono
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takayoshi Higashi
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroyasu Sato
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Chika Mochizuki-Ono
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuki Nagasaki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takashi Ueta
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takashi Miyai
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Suzumi M Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yoshiya Oda
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yasumasa Nishito
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Tomio Ono
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Choji Taya
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Satoru Arata
- Faculty of Arts and Sciences at Fujiyoshida, Showa University, Fuji-yoshida-shi, Yamanashi, Japan
| | - Sumiko Watanabe
- Department of Retinal Biology and Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Tetsuya Hirabayashi
- Biomembrane group, Technology Research Division, Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda City, Tokyo, Japan.
| |
Collapse
|
9
|
Qu Y, Liu ZX, Zheng XX, Wu SN, An JQ, Zou MH, Zhang ZR. MFN2-mediated decrease in mitochondria-associated endoplasmic reticulum membranes contributes to sunitinib-induced endothelial dysfunction and hypertension. J Mol Cell Cardiol 2025; 200:45-60. [PMID: 39848488 DOI: 10.1016/j.yjmcc.2025.01.005] [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: 07/30/2024] [Revised: 12/04/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Treatment of cancer patients with tyrosine kinase inhibitors (TKIs) often results in hypertension, but the underlying mechanism remains unclear. This study aimed to examine the role of mitochondrial morphology and function, particularly mitochondria-associated endoplasmic reticulum membranes (MAMs), in sunitinib-induced hypertension. METHODS Both in vitro and in vivo experiments performed to assesse reactive oxygen species (ROS), nitric oxide (NO), endothelium-dependent vasorelaxation, systemic blood pressure, and mitochondrial function in human umbilical vein endothelial cells (HUVECs) and C57BL/6 mouse aortic endothelial cells, under vehicle or sunitinib treatment condition. RESULTS Sunitinib increased mitochondrial ROS accumulation, decreased oxygen consumption rate, ATP production, and mitochondrial calcium ([Ca2+]M) levels, and impaired endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) signaling in HUVECs. In addition, sunitinib also decreased mitochondrial membrane potential, elongated mitochondria, and reduced MAMs. Remarkably, these effects were reversed by an adeno-virus linker (Ad-linker) that reinforces MAMs. Engineered augmentation of MAMs using AAV-FLT1-linker significantly mitigated sunitinib-induced hypertension, by restoring endothelium-dependent relaxation in mice, highlighting the crucial role of MAMs in this process. Further analyses revealed that sunitinib enhanced Akt-mediated expression of mitofusin 2 (MFN2), causing mitochondrial elongation, and induced dephosphorylation of inositol 1,4,5-trisphosphate receptor type 1 (IP3R1) at residues Y1737/Y1738, reducing [Ca2+]M. Our study suggests that increased MFN2 expression and IP3R1 dephosphorylation are critical in sunitinib-induced MAMs reduction and [Ca2+]M homeostasis. CONCLUSION Sunitinib induces mitochondrial dysfunction, Akt/MFN2-mediated decrease in MAMs and mitochondrial elongation, and IP3R1 dephosphorylation in endothelial cells, leading to endothelial dysfunction and hypertension. Our results provide the potential therapeutic targets for combating TKI-induced hypertension.
Collapse
Affiliation(s)
- Yao Qu
- Department of Cardiology, Harbin Medical University Cancer Hospital, NHC Key Laboratory of Cell Transplantation, Department of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Institute of Metabolic Disease, Heilongjiang Academy of Medical Sciences, Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin, China; Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Zhi-Xue Liu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Xiao-Xu Zheng
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Sheng-Nan Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Jun-Qing An
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Ming-Hui Zou
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China; Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA.
| | - Zhi-Ren Zhang
- Department of Cardiology, Harbin Medical University Cancer Hospital, NHC Key Laboratory of Cell Transplantation, Department of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Institute of Metabolic Disease, Heilongjiang Academy of Medical Sciences, Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin, China.
| |
Collapse
|
10
|
Wang Z, Sun W, Zhang K, Ke X, Wang Z. New insights into the relationship of mitochondrial metabolism and atherosclerosis. Cell Signal 2025; 127:111580. [PMID: 39732307 DOI: 10.1016/j.cellsig.2024.111580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Atherosclerotic cardiovascular and cerebrovascular diseases are the number one killer of human health. In view of the important role of mitochondria in the formation and evolution of atherosclerosis, our manuscript aims to comprehensively elaborate the relationship between mitochondria and the formation and evolution of atherosclerosis from the aspects of mitochondrial dynamics, mitochondria-organelle interaction (communication), mitochondria and cell death, mitochondria and vascular smooth muscle cell phenotypic switch, etc., which is combined with genome, transcriptome and proteome, in order to provide new ideas for the pathogenesis of atherosclerosis and the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Zexun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China
| | - Wangqing Sun
- Department of Radiology, Yixing Tumor Hospital, Yixing 214200, China
| | - Kai Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
11
|
Pérez-Sancho J, Smokvarska M, Dubois G, Glavier M, Sritharan S, Moraes TS, Moreau H, Dietrich V, Platre MP, Paterlini A, Li ZP, Fouillen L, Grison MS, Cana-Quijada P, Immel F, Wattelet V, Ducros M, Brocard L, Chambaud C, Luo Y, Ramakrishna P, Bayle V, Lefebvre-Legendre L, Claverol S, Zabrady M, Martin PGP, Busch W, Barberon M, Tilsner J, Helariutta Y, Russinova E, Taly A, Jaillais Y, Bayer EM. Plasmodesmata act as unconventional membrane contact sites regulating intercellular molecular exchange in plants. Cell 2025; 188:958-977.e23. [PMID: 39983675 DOI: 10.1016/j.cell.2024.11.034] [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: 12/15/2023] [Revised: 06/06/2024] [Accepted: 11/26/2024] [Indexed: 02/23/2025]
Abstract
Membrane contact sites (MCSs) are fundamental for intracellular communication, but their role in intercellular communication remains unexplored. We show that in plants, plasmodesmata communication bridges function as atypical endoplasmic reticulum (ER)-plasma membrane (PM) tubular MCSs, operating at cell-cell interfaces. Similar to other MCSs, ER-PM apposition is controlled by a protein-lipid tethering complex, but uniquely, this serves intercellular communication. Combining high-resolution microscopy, molecular dynamics, and pharmacological and genetic approaches, we show that cell-cell trafficking is modulated through the combined action of multiple C2 domains transmembrane domain proteins (MCTPs) 3, 4, and 6 ER-PM tethers and phosphatidylinositol-4-phosphate (PI4P) lipid. Graded PI4P amounts regulate MCTP docking to the PM, their plasmodesmata localization, and cell-cell permeability. SAC7, an ER-localized PI4P-phosphatase, regulates MCTP4 accumulation at plasmodesmata and modulates cell-cell trafficking capacity in a cell-type-specific manner. Our findings expand MCS functions in information transmission from intracellular to intercellular cellular activities.
Collapse
Affiliation(s)
- Jessica Pérez-Sancho
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Marija Smokvarska
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Gwennogan Dubois
- Laboratoire Reproduction et Développement des Plantes, ENS de Lyon, CNRS, INRA, 69342 Lyon, France
| | - Marie Glavier
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Sujith Sritharan
- Laboratoire de Biochimie Théorique, UPR9080, CNRS, Université Paris Cité, Paris, France
| | - Tatiana S Moraes
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Hortense Moreau
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Victor Dietrich
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Matthieu P Platre
- Salk Institute for Biological Studies, Plant Molecular and Cellular Biology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Andrea Paterlini
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France; The Sainsbury Laboratory, University of Cambridge, Cambridge, UK
| | - Ziqiang P Li
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Laetitia Fouillen
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Magali S Grison
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Pepe Cana-Quijada
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Françoise Immel
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Valerie Wattelet
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France
| | - Mathieu Ducros
- Bordeaux Imaging Center, Plant Imaging Platform, UAR3420, CNRS-INSERM-University of Bordeaux-INRAE, Bordeaux, France
| | - Lysiane Brocard
- Bordeaux Imaging Center, Plant Imaging Platform, UAR3420, CNRS-INSERM-University of Bordeaux-INRAE, Bordeaux, France
| | - Clément Chambaud
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France; Bordeaux Imaging Center, Plant Imaging Platform, UAR3420, CNRS-INSERM-University of Bordeaux-INRAE, Bordeaux, France
| | - Yongming Luo
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Priya Ramakrishna
- Department of Plant Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Vincent Bayle
- Laboratoire Reproduction et Développement des Plantes, ENS de Lyon, CNRS, INRA, 69342 Lyon, France
| | | | | | - Matej Zabrady
- Biomedical Sciences Research Complex, University of St Andrews, Fife KY16 9ST, UK; Cell and Molecular Sciences, The James Hutton Institute, Dundee DD2 5DA, UK
| | - Pascal G P Martin
- Université de Bordeaux, INRAE, UMR1332 Biologie du Fruit et Pathologie, 33882 Villenave d'Ornon, France
| | - Wolfgang Busch
- Salk Institute for Biological Studies, Plant Molecular and Cellular Biology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Marie Barberon
- Department of Plant Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Jens Tilsner
- Biomedical Sciences Research Complex, University of St Andrews, Fife KY16 9ST, UK; Cell and Molecular Sciences, The James Hutton Institute, Dundee DD2 5DA, UK
| | - Yrjö Helariutta
- The Sainsbury Laboratory, University of Cambridge, Cambridge, UK; Institute of Biotechnology, HiLIFE/Organismal and Evolutionary Biology Research Programme, Faculty of Biological and Environmental Sciences, Viikki Plant Science Centre, University of Helsinki, Helsinki, Finland
| | - Eugenia Russinova
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Antoine Taly
- Laboratoire de Biochimie Théorique, UPR9080, CNRS, Université Paris Cité, Paris, France
| | - Yvon Jaillais
- Laboratoire Reproduction et Développement des Plantes, ENS de Lyon, CNRS, INRA, 69342 Lyon, France.
| | - Emmanuelle M Bayer
- Laboratoire de Biogenèse Membranaire, UMR5200, CNRS, Université de Bordeaux, Villenave-d'Ornon, France.
| |
Collapse
|
12
|
Talari NK, Mattam U, Rahman AP, Hemmelgarn BK, Wyder MA, Sylvestre PB, Greis KD, Chella Krishnan K. Functional compartmentalization of hepatic mitochondrial subpopulations during MASH progression. Commun Biol 2025; 8:258. [PMID: 39966593 PMCID: PMC11836293 DOI: 10.1038/s42003-025-07713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
The role of peridroplet mitochondria (PDM) in diseased liver, such as during the progression of metabolic dysfunction-associated steatohepatitis (MASH), remains unknown. We isolated hepatic cytoplasmic mitochondria (CM) and PDM from a mouse model of diet-induced MASLD/MASH to characterize their functions from simple steatosis to advanced MASH, using chow-fed mice as controls. Our findings show an inverse relationship between hepatic CM and PDM levels from healthy to steatosis to advanced MASH. Proteomics analysis revealed these two mitochondrial populations are compositionally and functionally distinct. We found that hepatic PDM are more bioenergetically active than CM, with higher pyruvate oxidation capacity in both healthy and diseased liver. Higher respiration capacity of PDM was associated with elevated OXPHOS protein complexes and increased TCA cycle flux. In contrast, CM showed higher fatty acid oxidation capacity with MASH progression. Transmission electron microscopy revealed larger and elongated mitochondria during healthy and early steatosis, which appeared small and fragmented during MASH progression. These changes coincided with higher MFN2 protein levels in hepatic PDM and higher DRP1 protein levels in hepatic CM. These findings highlight the distinct roles of hepatic CM and PDM in MASLD progression towards MASH.
Collapse
Affiliation(s)
- Noble Kumar Talari
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ushodaya Mattam
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Afra P Rahman
- Medical Sciences Baccalaureate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brook K Hemmelgarn
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Wyder
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pamela B Sylvestre
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D Greis
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Karthickeyan Chella Krishnan
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
13
|
Wang J, Wang M, Zeng X, Li Y, Lei L, Chen C, Lin X, Fang P, Guo Y, Jiang X, Wang Y, Chen L, Long J. Targeting membrane contact sites to mediate lipid dynamics: innovative cancer therapies. Cell Commun Signal 2025; 23:89. [PMID: 39955542 PMCID: PMC11830217 DOI: 10.1186/s12964-025-02089-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
Membrane contact sites (MCS) are specialized regions where organelles are closely interconnected through membrane structures, facilitating the transfer and exchange of ions, lipids, and other molecules. This proximity enables a synergistic regulation of cellular homeostasis and functions. The formation and maintenance of these contact sites are governed by specific proteins that bring organelle membranes into close apposition, thereby enabling functional crosstalk between cellular compartments. In eukaryotic cells, lipids are primarily synthesized and metabolized within distinct organelles and must be transported through MCS to ensure proper cellular function. Consequently, MCS act as pivotal platforms for lipid synthesis and trafficking, particularly in cancer cells and immune cells within the tumor microenvironment, where dynamic alterations are critical for maintaining lipid homeostasis. This article provides a comprehensive analysis of how these cells exploit membrane contact sites to modulate lipid synthesis, metabolism, and transport, with a specific focus on how MCS-mediated lipid dynamics influence tumor progression. We also examine the differences in MCS and associated molecules across various cancer types, exploring novel therapeutic strategies targeting MCS-related lipid metabolism for the development of anticancer drugs, while also addressing the challenges involved.
Collapse
Affiliation(s)
- Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China.
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Xueni Zeng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Yanhan Li
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Lingzhi Lei
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Changan Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Xi Lin
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Peiyuan Fang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Yuxuan Guo
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Yian Wang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China
| | - Lihong Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China.
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China.
| | - Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
14
|
Ma X, Wei X, Niu M, Zhang C, Peng Z, Liu W, Yan J, Su X, Lu S, Cui W, Sesaki H, Zong WX, Ni HM, Ding WX. Disruption of Mitochondrial Dynamics and Stasis Leads to Liver Injury and Tumorigenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637688. [PMID: 39990472 PMCID: PMC11844448 DOI: 10.1101/2025.02.11.637688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Background & Aims Mitochondrial dysfunction has been implicated in aging and various cancer development. As highly dynamic organelles, mitochondria constantly undergo fission, mediated by dynamin-related protein 1 (DRP1, gene name Dnm1l ), and fusion, regulated by mitofusin 1 (MFN1), MFN2, and optic atrophy 1 (OPA1). However, whether and how dysregulation of mitochondria dynamics would be involved in liver pathogenesis and tumorigenesis is unknown. Methods Dnm1l Flox/Flox ( Dnm1l F/F ), Mfn1 F/F and Mfn2 F/F mice were crossed with albumin-Cre mice to generate liver-specific Dnm1l knockout (L- Dnm1l KO), L- Mfn1 KO, L- Mfn2 KO, L- Mfn1, Mfn2 double KO (DKO), and L- Mfn1, Mfn2, Dnm1l triple KO (TKO) mice. These mice were housed for various periods up to 18 months. Some mice also received hydrodynamic tail vein injections of a Sleeping Beauty transposon-transposase plasmid system with c-MYC and YAP . Blood and liver tissues were harvested for biochemical and histological analysis. Results L- Dnm1l KO mice had elevated serum alanine aminotransferase levels and increased hepatic fibrosis as early as two months of age. By 12 to 18 months, male L- Dnm1l KO mice developed spontaneous liver tumors, primarily hepatocellular adenomas. While female L- Dnm1l KO mice also developed liver tumors, their incidence was much lower. In contrast, neither L- Mfn1 KO nor L- Mfn2 KO mice had notable liver injury or tumorigenesis. However, a small portion of DKO mice developed tumors at 15-18 month-old. Increased DNA damage, senescence and compensatory proliferation were observed in L- Dnm1l KO mice but were less evident in L- Mfn1 KO, L- Mfn2 KO or DKO mice, indicating that mitochondrial fission is more important to maintain hepatocyte homeostasis and prevent liver tumorigenesis. Interestingly, further deletion of Mfn1 and Mfn2 in L- Dnm1l KO mice markedly abolished liver injury, fibrosis, and both spontaneous and oncogene-induced tumorigenesis. RNA sequencing and metabolomics analysis revealed significant activation of the cGAS-STING-interferon pathway and alterations in the tumor microenvironment pathways, alongside increased pyrimidine synthesis and metabolism in the livers of L- Dnm1l KO mice. Notably, the changes in gene expression and pyrimidine metabolism were considerably corrected in the TKO mice. Conclusions Mitochondrial dynamics and stability are essential for maintaining hepatic mitochondrial homeostasis and hepatocyte functions. Loss of hepatic DRP1 promotes liver tumorigenesis by increasing pyrimidine metabolism and activating the cGAS-STING-mediated innate immune response.
Collapse
|
15
|
Joaquim M, Altin S, Bulimaga MB, Simões T, Nolte H, Bader V, Franchino CA, Plouzennec S, Szczepanowska K, Marchesan E, Hofmann K, Krüger M, Ziviani E, Trifunovic A, Chevrollier A, Winklhofer KF, Motori E, Odenthal M, Escobar-Henriques M. Mitofusin 2 displays fusion-independent roles in proteostasis surveillance. Nat Commun 2025; 16:1501. [PMID: 39929801 PMCID: PMC11811173 DOI: 10.1038/s41467-025-56673-5] [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: 07/21/2023] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Mitochondria are essential organelles and their functional state dictates cellular proteostasis. However, little is known about the molecular gatekeepers involved, especially in absence of external stress. Here we identify a role of MFN2 in quality control independent of its function in organellar shape remodeling. MFN2 ablation alters the cellular proteome, marked for example by decreased levels of the import machinery and accumulation of the kinase PINK1. Moreover, MFN2 interacts with the proteasome and cytosolic chaperones, thereby preventing aggregation of newly translated proteins. Similarly to MFN2-KO cells, patient fibroblasts with MFN2-disease variants recapitulate excessive protein aggregation defects. Restoring MFN2 levels re-establishes proteostasis in MFN2-KO cells and rescues fusion defects of MFN1-KO cells. In contrast, MFN1 loss or mitochondrial shape alterations do not alter protein aggregation, consistent with a fusion-independent role of MFN2 in cellular homeostasis. In sum, our findings open new possibilities for therapeutic strategies by modulation of MFN2 levels.
Collapse
Affiliation(s)
- Mariana Joaquim
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Selver Altin
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Maria-Bianca Bulimaga
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute of Pathology, Medical Faculty of the University of Cologne and University Hospital of Cologne, Cologne, Germany
| | - Tânia Simões
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Hendrik Nolte
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- MPI for Biology of Ageing, 50931, Cologne, Germany
| | - Verian Bader
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany, and Cluster of Excellence RESOLV, Bochum, Germany
| | - Camilla Aurora Franchino
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Solenn Plouzennec
- University of Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| | - Karolina Szczepanowska
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- ReMedy International Research Agenda Unit, International Institute of Molecular Mechanisms and Machines (IMol), Polish Academy of Sciences, 00-783, Warsaw, Poland
| | | | - Kay Hofmann
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Marcus Krüger
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Elena Ziviani
- Deparment of Biology, University of Padova, Padova, Italy
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Arnaud Chevrollier
- University of Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| | - Konstanze F Winklhofer
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany, and Cluster of Excellence RESOLV, Bochum, Germany
| | - Elisa Motori
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Margarete Odenthal
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute of Pathology, Medical Faculty of the University of Cologne and University Hospital of Cologne, Cologne, Germany
| | - Mafalda Escobar-Henriques
- Institute for Genetics, University of Cologne, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
16
|
Tábara LC, Segawa M, Prudent J. Molecular mechanisms of mitochondrial dynamics. Nat Rev Mol Cell Biol 2025; 26:123-146. [PMID: 39420231 DOI: 10.1038/s41580-024-00785-1] [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] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Mitochondria not only synthesize energy required for cellular functions but are also involved in numerous cellular pathways including apoptosis, calcium homoeostasis, inflammation and immunity. Mitochondria are dynamic organelles that undergo cycles of fission and fusion, and these transitions between fragmented and hyperfused networks ensure mitochondrial function, enabling adaptations to metabolic changes or cellular stress. Defects in mitochondrial morphology have been associated with numerous diseases, highlighting the importance of elucidating the molecular mechanisms regulating mitochondrial morphology. Here, we discuss recent structural insights into the assembly and mechanism of action of the core mitochondrial dynamics proteins, such as the dynamin-related protein 1 (DRP1) that controls division, and the mitofusins (MFN1 and MFN2) and optic atrophy 1 (OPA1) driving membrane fusion. Furthermore, we provide an updated view of the complex interplay between different proteins, lipids and organelles during the processes of mitochondrial membrane fusion and fission. Overall, we aim to present a valuable framework reflecting current perspectives on how mitochondrial membrane remodelling is regulated.
Collapse
Affiliation(s)
- Luis-Carlos Tábara
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Mayuko Segawa
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
Kai F, Leidal AM, Weaver VM. Tension-induced organelle stress: an emerging target in fibrosis. Trends Pharmacol Sci 2025; 46:117-131. [PMID: 39818520 PMCID: PMC11805623 DOI: 10.1016/j.tips.2024.12.006] [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: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Fibrosis accounts for approximately one-third of disease-related deaths globally. Current therapies fail to cure fibrosis, emphasizing the need to identify new antifibrotic approaches. Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and resultant stiffening of tissue stroma. This stiffening appropriates actomyosin-mediated mechanical tension within cells to ultimately affect cell fate decisions and function. Recent studies demonstrate that subcellular organelles are physically connected to the actin cytoskeleton and sensitive to mechanoperturbations. These insights highlight mechanisms that may contribute to the chronic organelle stress in many fibrotic diseases, including those of the lung and liver. In this review, we discuss the hypothesis that a stiffened fibrotic ECM corrupts intracellular mechanical tension to compromise organelle homeostasis. We summarize potential therapeutics that could intervene in this mechanical dialog and that may have clinical benefit for resolving pathological organelle stress in fibrosis.
Collapse
Affiliation(s)
- FuiBoon Kai
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Biochemistry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Andrew M Leidal
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Valerie M Weaver
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA; UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Department of Radiation Oncology, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
18
|
Cartes-Saavedra B, Ghosh A, Hajnóczky G. The roles of mitochondria in global and local intracellular calcium signalling. Nat Rev Mol Cell Biol 2025:10.1038/s41580-024-00820-1. [PMID: 39870977 DOI: 10.1038/s41580-024-00820-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 01/29/2025]
Abstract
Activation of Ca2+ channels in Ca2+ stores in organelles and the plasma membrane generates cytoplasmic calcium ([Ca2+]c) signals that control almost every aspect of cell function, including metabolism, vesicle fusion and contraction. Mitochondria have a high capacity for Ca2+ uptake and chelation, alongside efficient Ca2+ release mechanisms. Still, mitochondria do not store Ca2+ in a prolonged manner under physiological conditions and lack the capacity to generate global [Ca2+]c signals. However, mitochondria take up Ca2+ at high local [Ca2+]c signals that originate from neighbouring organelles, and also during sustained global elevations of [Ca2+]c. Accumulated Ca2+ in the mitochondria stimulates oxidative metabolism and upon return to the cytoplasm, can produce spatially confined rises in [Ca2+]c to exert control over processes that are sensitive to Ca2+. Thus, the mitochondrial handling of [Ca2+]c is of physiological relevance. Furthermore, dysregulation of mitochondrial Ca2+ handling can contribute to debilitating diseases. We discuss the mechanisms and relevance of mitochondria in local and global calcium signals.
Collapse
Affiliation(s)
- Benjamín Cartes-Saavedra
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Arijita Ghosh
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Castañé H, Jiménez-Franco A, Hernández-Aguilera A, Martínez-Navidad C, Cambra-Cortés V, Onoiu AI, Jiménez-Aguilar JM, París M, Hernández M, Parada D, Guilarte C, Zorzano A, Hernández-Alvarez MI, Camps J, Joven J. Multi-omics profiling reveals altered mitochondrial metabolism in adipose tissue from patients with metabolic dysfunction-associated steatohepatitis. EBioMedicine 2025; 111:105532. [PMID: 39731853 PMCID: PMC11743550 DOI: 10.1016/j.ebiom.2024.105532] [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: 08/03/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) and its more severe form steatohepatitis (MASH) contribute to rising morbidity and mortality rates. The storage of fat in humans is closely associated with these diseases' progression. Thus, adipose tissue metabolic homeostasis could be key in both the onset and progression of MASH. METHODS We conducted a case-control observational research using a systems biology-based approach to analyse liver, abdominal subcutaneous adipose tissue (SAT), omental visceral adipose tissue (VAT), and blood of n = 100 patients undergoing bariatric surgery (NCT05554224). MASH was diagnosed through histologic assessment. Whole-slide image analysis, lipidomics, proteomics, and transcriptomics were performed on tissue samples. Lipidomics and proteomics profiles were determined on plasma samples. FINDINGS Liver transcriptomics, proteomics, and lipidomics revealed interconnected pathways associated with inflammation, mitochondrial dysfunction, and lipotoxicity in MASH. Paired adipose tissue biopsies had larger adipocyte areas in both fat depots in MASH. Enrichment analyses of proteomics and lipidomics data confirmed the association of liver lesions with mitochondrial dysfunction in VAT. Plasma lipidomics identified candidates with high diagnostic accuracy (AUC = 0.919, 95% CI 0.840-0.979) for screening MASH. INTERPRETATION Mitochondrial dysfunction is also present in VAT in patients with obesity-associated MASH. This may cause a disruption in the metabolic equilibrium of lipid processing and storage, which impacts the liver and accelerates detrimental adaptative responses. FUNDING The project leading to these results has received funding from 'la Caixa' Foundation (HR21-00430), and from the Instituto de Salud Carlos III (ISCIII) (PI21/00510) and co-funded by the European Union.
Collapse
Affiliation(s)
- Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain; Department of Medicine and Surgery, Faculty of Medicine, Universitat Rovira i Virgili, Reus, Spain.
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain; Department of Medicine and Surgery, Faculty of Medicine, Universitat Rovira i Virgili, Reus, Spain
| | | | - Cristian Martínez-Navidad
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain
| | - Vicente Cambra-Cortés
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain
| | - Alina-Iuliana Onoiu
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain
| | - Juan Manuel Jiménez-Aguilar
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain
| | - Marta París
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain; Department of Surgery, Hospital Universitari de Sant Joan, Reus, Spain
| | - Mercè Hernández
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain; Department of Surgery, Hospital Universitari de Sant Joan, Reus, Spain
| | - David Parada
- Department of Pathology, Hospital Universitari de Sant Joan, Reus, Spain
| | - Carmen Guilarte
- Department of Pathology, Hospital Universitari de Sant Joan, Reus, Spain
| | - Antonio Zorzano
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - María Isabel Hernández-Alvarez
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Institut de Biomedicina de la Universitat de Barcelona IBUB, Barcelona, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain; Department of Medicine and Surgery, Faculty of Medicine, Universitat Rovira i Virgili, Reus, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Universitat Rovira i Virgili, Institut d'Investigació Sanitària Pere Virgili, Reus, Spain; Department of Medicine and Surgery, Faculty of Medicine, Universitat Rovira i Virgili, Reus, Spain; The Campus of International Excellence Southern Catalonia, Tarragona, Spain.
| |
Collapse
|
20
|
Zeng W, Wang L, Wang C, Xiong X, Huang Q, Chen S, Liu C, Liu W, Wang Y, Huang Q. SENP1 prevents high fat diet-induced non-alcoholic fatty liver diseases by regulating mitochondrial dynamics. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167527. [PMID: 39332783 DOI: 10.1016/j.bbadis.2024.167527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
Mitochondrial dynamics plays a crucial role in the occurrence and development of non-alcoholic fatty liver diseases (NAFLD). SENP1, a SUMO-specific protease, catalyzes protein de-SUMOylation and involves in various physiological and pathological processes. However, the exact role of SENP1 in NAFLD remains unclear. Therefore, we investigated the regulatory role of SENP1 in mitochondrial dynamics during the progression of NAFLD. In the study, the NAFLD in vivo model induced by high fat diet (HFD) and in vitro model induced by free fatty acids (FFA) were established to investigate the role and underlying mechanism of SENP1 through detecting mitochondrial morphology and dynamics. Our results showed that the down-regulation of SENP1 expression and the mitochondrial dynamics dysregulation occurred in the NAFLD, evidenced as mitochondrial fragmentation, up-regulation of p-Drp1 ser616 and down-regulation of MFN2, OPA1. However, over-expression of SENP1 significantly alleviated the NAFLD, rectified the mitochondrial dynamics disorder, reduced Cyt-c release and ROS levels induced by FFA or HFD; moreover, the over-expression of SENP1 also reduced the SUMOylation levels of Drp1 and prevented the Drp1 translocation to mitochondria. Our findings suggest that the possible mechanisms of SENP1 were through rectifying the mitochondrial dynamics disorder, reducing Cyt-c release and ROS-mediated oxidative stress. The findings would provide a novel target for the prevention and treatment of NALFD.
Collapse
Affiliation(s)
- Wenjing Zeng
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Li Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Chaowen Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiaowei Xiong
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Qianqian Huang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Sheng Chen
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Chen Liu
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Wentao Liu
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Yuan Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Qiren Huang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
21
|
Sowton AP, Holzner LMW, Krause FN, Baxter R, Mocciaro G, Krzyzanska DK, Minnion M, O'Brien KA, Harrop MC, Darwin PM, Thackray BD, Vacca M, Feelisch M, Griffin JL, Murray AJ. Chronic inorganic nitrate supplementation does not improve metabolic health and worsens disease progression in mice with diet-induced obesity. Am J Physiol Endocrinol Metab 2025; 328:E69-E91. [PMID: 39653040 DOI: 10.1152/ajpendo.00256.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/16/2024] [Accepted: 11/07/2024] [Indexed: 01/11/2025]
Abstract
Inorganic nitrate (NO3-) has been proposed to be of therapeutic use as a dietary supplement in obesity and related conditions including the metabolic syndrome (MetS), type II diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD). Administration of NO3- to endothelial nitric oxide synthase-deficient mice reversed aspects of MetS; however, the impact of NO3- supplementation in diet-induced obesity is not well understood. Here we investigated the whole body metabolic phenotype and cardiac and hepatic metabolism in mice fed a high-fat, high-sucrose (HFHS) diet for up to 12 mo of age, supplemented with 1 mM NaNO3 (or NaCl) in their drinking water. HFHS feeding was associated with a progressive obesogenic and diabetogenic phenotype, which was not ameliorated by NO3-. Furthermore, HFHS-fed mice supplemented with NO3- showed elevated levels of cardiac fibrosis and accelerated progression of MASLD including development of hepatocellular carcinoma in comparison with NaCl-supplemented mice. NO3- did not enhance mitochondrial β-oxidation capacity in any tissue assayed and did not suppress hepatic lipid accumulation, suggesting it does not prevent lipotoxicity. We conclude that NO3- is ineffective in preventing the metabolic consequences of an obesogenic diet and may instead be detrimental to metabolic health against the background of HFHS feeding. This is the first report of an unfavorable effect of long-term nitrate supplementation in the context of the metabolic challenges of overfeeding, warranting urgent further investigation into the mechanism of this interaction.NEW & NOTEWORTHY Inorganic nitrate has been suggested to be of therapeutic benefit in obesity-related conditions, as it increases nitric oxide bioavailability, enhances mitochondrial β-oxidation, and reverses metabolic syndrome in eNOS-/- mice. However, we here show that over 12 months nitrate was ineffective in preventing metabolic consequences in high fat, high sucrose-fed mice and worsened aspects of metabolic health, impairing cholesterol handling, increasing cardiac fibrosis, and exacerbating steatotic liver disease progression, with acceleration to hepatocellular carcinoma.
Collapse
Affiliation(s)
- Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Lorenz M W Holzner
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Fynn N Krause
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ruby Baxter
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Gabriele Mocciaro
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Dominika K Krzyzanska
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Magdalena Minnion
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Matthew C Harrop
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Paula M Darwin
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Benjamin D Thackray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Michele Vacca
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Julian L Griffin
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
22
|
Anari M, Karimkhanloo H, Nie S, Dong L, Fidelito G, Bayliss J, Keenan SN, Slavin J, Lin S, Cheng Z, Lu J, Miotto PM, De Nardo W, Devereux CJ, Williamson NA, Watt MJ, Montgomery MK. Lipidome profiling in advanced metabolic liver disease identifies phosphatidylserine synthase 1 as a regulator of hepatic lipoprotein metabolism. Cell Rep 2024; 43:115007. [PMID: 39666456 DOI: 10.1016/j.celrep.2024.115007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 09/10/2024] [Accepted: 11/06/2024] [Indexed: 12/14/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is characterized by defective lipid metabolism, which causes disease progression. MASH is also linked to various cardiometabolic risk factors, including obesity and type 2 diabetes. The contribution of defective lipid metabolism in MASH to cardiometabolic comorbidities is incompletely understood. Using hepatic lipidome profiling in eight mouse strains that differ in MASH susceptibility and patients with MASH, we show that phosphatidylserine (PS) accumulation and preservation of PS synthase 1 (PSS1) expression is associated with resistance to MASH and hypertriglyceridemia. Mechanistically, hepatocyte-specific PSS1 overexpression remodels the hepatic and very-low-density lipoprotein (VLDL) lipidome in mice with MASH. Specifically, we show an increase in VLDL ceramide that suppresses the expression and activity of lipoprotein lipase in skeletal muscle, thereby reducing VLDL-triglyceride clearance, fatty acid uptake, and lipid accumulation in muscle, overall exacerbating hypertriglyceridemia. Together, the results of this study identify hepatic PSS1 as a regulator of systemic lipoprotein metabolism.
Collapse
Affiliation(s)
- Marziyeh Anari
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Hamzeh Karimkhanloo
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia; Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, and Department of Physiology, Monash University, Clayton, VIC 3800, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Li Dong
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gio Fidelito
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jacqueline Bayliss
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Stacey N Keenan
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - John Slavin
- St. Vincent's Pathology, St. Vincent's Hospital, Melbourne, VIC 3065, Australia
| | - Sihan Lin
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Zhili Cheng
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jie Lu
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Paula M Miotto
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - William De Nardo
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Camille J Devereux
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Nicholas A Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
23
|
Zhang M, Wei J, He C, Sui L, Jiao C, Zhu X, Pan X. Inter- and intracellular mitochondrial communication: signaling hubs in aging and age-related diseases. Cell Mol Biol Lett 2024; 29:153. [PMID: 39695918 DOI: 10.1186/s11658-024-00669-4] [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: 08/21/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
Mitochondria are versatile and complex organelles that can continuously communicate and interact with the cellular milieu. Deregulated communication between mitochondria and host cells/organelles has significant consequences and is an underlying factor of many pathophysiological conditions, including the process of aging. During aging, mitochondria lose function, and mitocellular communication pathways break down; mitochondrial dysfunction interacts with mitochondrial dyscommunication, forming a vicious circle. Therefore, strategies to protect mitochondrial function and promote effective communication of mitochondria can increase healthy lifespan and longevity, which might be a new treatment paradigm for age-related disorders. In this review, we comprehensively discuss the signal transduction mechanisms of inter- and intracellular mitochondrial communication, as well as the interactions between mitochondrial communication and the hallmarks of aging. This review emphasizes the indispensable position of inter- and intracellular mitochondrial communication in the aging process of organisms, which is crucial as the cellular signaling hubs. In addition, we also specifically focus on the status of mitochondria-targeted interventions to provide potential therapeutic targets for age-related diseases.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Liutao Sui
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chucheng Jiao
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
24
|
Mu C, Wang S, Wang Z, Tan J, Yin H, Wang Y, Dai Z, Ding D, Yang F. Mechanisms and therapeutic targets of mitochondria in the progression of metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2024; 30:101774. [PMID: 39701281 DOI: 10.1016/j.aohep.2024.101774] [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: 08/28/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) includes liver disease processes from simple fatty liver to nonalcoholic steatohepatitis, which may progress to liver fibrosis, cirrhosis, and even hepatocellular carcinoma (HCC). As the incidence of HCC derived from viral hepatitis decreases, MASLD has emerged as a significant health threat, driven by lifestyle changes and rising obesity rates among patients. The pathogenesis of MASLD is complex, involving factors such as insulin resistance, gut microbiota imbalance, and genetic and epigenetic factors. In recent years, the role of mitochondrial dysfunction in MASLD has gained significant attention, involving β-oxidation imbalance, oxidative stress increase, mitophagy defects, and mitochondrial DNA (mtDNA) mutations. This article reviews the pathophysiological mechanisms of mitochondrial dysfunction in MASLD, diagnostic methods, and potential therapeutic strategies. By synthesizing current research findings, the review aims to highlight the critical role of mitochondrial dysfunction as a target for future diagnostic and therapeutic interventions. This focus could pave the way for innovative clinical strategies, ultimately improving treatment options and patient prognosis in MASLD.
Collapse
Affiliation(s)
- Chenyang Mu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China; Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Sijie Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China; Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Zenghan Wang
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Jian Tan
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Haozan Yin
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Yuefan Wang
- Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhihui Dai
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Dongyang Ding
- Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Fu Yang
- Department of Medical Genetics, Naval Medical University, Shanghai, China; Shanghai Key Laboratory of Medical Bioprotection, Shanghai, China; Key Laboratory of Biological Defense, Ministry of Education, Shanghai, China.
| |
Collapse
|
25
|
Zhao S, Jiang X, Li N, Wang T. SLMO transfers phosphatidylserine between the outer and inner mitochondrial membrane in Drosophila. PLoS Biol 2024; 22:e3002941. [PMID: 39680501 DOI: 10.1371/journal.pbio.3002941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Phospholipids are critical building blocks of mitochondria, and proper mitochondrial function and architecture rely on phospholipids that are primarily transported from the endoplasmic reticulum (ER). Here, we show that mitochondrial form and function rely on synthesis of phosphatidylserine (PS) in the ER through phosphatidylserine synthase (PSS), trafficking of PS from ER to mitochondria (and within mitochondria), and the conversion of PS to phosphatidylethanolamine (PE) by phosphatidylserine decarboxylase (PISD) in the inner mitochondrial membrane (IMM). Using a forward genetic screen in Drosophila, we found that Slowmo (SLMO) specifically transfers PS from the outer mitochondrial membrane (OMM) to the IMM within the inner boundary membrane (IBM) domain. Thus, SLMO is required for shaping mitochondrial morphology, but its putative conserved binding partner, dTRIAP, is not. Importantly, SLMO's role in maintaining mitochondrial morphology is conserved in humans via the SLMO2 protein and is independent of mitochondrial dynamics. Our results highlight the importance of a conserved PSS-SLMO-PISD pathway in maintaining the structure and function of mitochondria.
Collapse
Affiliation(s)
- Siwen Zhao
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xuguang Jiang
- National Institute of Biological Sciences, Beijing, China
| | - Ning Li
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Tao Wang
- College of Biological Sciences, China Agricultural University, Beijing, China
- National Institute of Biological Sciences, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| |
Collapse
|
26
|
Yang Z, Wang J, Zhao T, Wang L, Liang T, Zheng Y. Mitochondrial structure and function: A new direction for the targeted treatment of chronic liver disease with Chinese herbal medicine. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118461. [PMID: 38908494 DOI: 10.1016/j.jep.2024.118461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Excessive fat accumulation, biological clock dysregulation, viral infections, and sustained inflammatory responses can lead to liver inflammation, fibrosis, and cancer, thus promoting the development of chronic liver disease. A comprehensive understanding of the etiological factors leading to chronic liver disease and the intrinsic mechanisms influencing its onset and progression can aid in identifying potential targets for targeted therapy. Mitochondria, as key organelles that maintain the metabolic homeostasis of the liver, provide an important foundation for exploring therapeutic targets for chronic liver disease. Recent studies have shown that active ingredients in herbal medicines and their natural products can modulate chronic liver disease by influencing the structure and function of mitochondria. Therefore, studying how Chinese herbs target mitochondrial structure and function to treat chronic liver diseases is of great significance. AIM OF THE STUDY Investigating the prospects of herbal medicine the Lens of chronic liver disease based on mitochondrial structure and function. MATERIALS AND METHODS A computerized search of PubMed was conducted using the keywords "mitochondrial structure", "mitochondrial function", "mitochondria and chronic liver disease", "botanicals, mitochondria and chronic liver disease".Data from the Web of Science and Science Direct databases were also included. The research findings regarding herbal medicines targeting mitochondrial structure and function for the treatment of chronic liver disease are summarized. RESULTS A computerized search of PubMed using the keywords "mitochondrial structure", "mitochondrial function", "mitochondria and chronic liver disease", "phytopharmaceuticals, mitochondria, and chronic liver disease", as well as the Web of Science and Science Direct databases was conducted to summarize information on studies of mitochondrial structure- and function-based Chinese herbal medicines for the treatment of chronic liver disease and to suggest that the effects of herbal medicines on mitochondrial division and fusion.The study suggested that there is much room for research on the influence of Chinese herbs on mitochondrial division and fusion. CONCLUSIONS Targeting mitochondrial structure and function is crucial for herbal medicine to combat chronic liver disease.
Collapse
Affiliation(s)
- Zhihui Yang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Jiahui Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Tiejian Zhao
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Lei Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Tianjian Liang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China.
| | - Yang Zheng
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China.
| |
Collapse
|
27
|
Zhang L, Lu T, Zhou B, Sun Y, Wang L, Qiao G, Yang T. Lipidomic analysis of serum exosomes identifies a novel diagnostic marker for type 2 diabetes mellitus. Lab Med 2024; 55:724-731. [PMID: 38809765 DOI: 10.1093/labmed/lmae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) intricately involves disrupted lipid metabolism. Exosomes emerge as carriers of biomarkers for early diagnosis and monitoring. This study aims to identify lipid metabolites in serum exosomes for T2DM diagnosis. METHODS Serum samples were collected from newly diagnosed T2DM patients and age and body mass index-matched healthy controls. Exosomes were isolated using exosome isolation reagent, and untargeted/targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify and validate altered lipid metabolites. Receiver operating characteristic curve analysis was used to evaluate the diagnostic value of candidate lipid metabolites. RESULTS Serum exosomes were successfully isolated from both groups, with untargeted LC-MS/MS revealing distinct lipid metabolite alterations. Notably, phosphatidylethanolamine (PE) (22:2(13Z,16Z)/14:0) showed stable elevation in T2DM-serum exosomes. Targeted LC-MS/MS confirmed significant increase of PE (22:2(13Z,16Z)/14:0) in T2DM exosomes but not in serum. PE (22:2(13Z,16Z)/14:0) levels not only positively correlated with hemoglobin A1C levels and blood glucose levels, but also effectively distinguished T2DM patients from healthy individuals (area under the curve = 0.9141). CONCLUSION Our research sheds light on the importance of serum exosome lipid metabolites in diagnosing T2DM, providing valuable insights into the complex lipid metabolism of diabetes.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Clinical Laboratory, Yixing People's Hospital, Yixing, China
| | - Ting Lu
- Department of Endocrinology, Yixing People's Hospital, Yixing, China
| | - Baocheng Zhou
- Department of Medical Laboratory, Lianyungang Maternal and Child Health Hospital, Lianyungang, China
| | - Yaoxiang Sun
- Department of Clinical Laboratory, Yixing People's Hospital, Yixing, China
| | - Liyun Wang
- Department of Endocrinology, Yixing People's Hospital, Yixing, China
| | - Guohong Qiao
- Department of Clinical Laboratory, Yixing People's Hospital, Yixing, China
| | - Tingting Yang
- Department of Clinical Laboratory, Yixing People's Hospital, Yixing, China
| |
Collapse
|
28
|
Zhang XY, Han C, Yao Y, Wei TT. Current insights on mitochondria-associated endoplasmic reticulum membranes (MAMs) and their significance in the pathophysiology of ocular disorders. Exp Eye Res 2024; 248:110110. [PMID: 39326773 DOI: 10.1016/j.exer.2024.110110] [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: 06/20/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
The intricate interaction network necessary for essential physiological functions underscores the interdependence among eukaryotic cells. Mitochondria-Associated Endoplasmic Reticulum Membranes (MAMs), specialized junctions between mitochondria and the ER, were recently discovered. These junctions participate in various cellular processes, including calcium level regulation, lipid metabolism, mitochondrial integrity maintenance, autophagy, and inflammatory responses via modulating the structure and molecular composition of various cellular components. Therefore, MAMs contribute to the pathophysiology of numerous ocular disorders, including Diabetic Retinopathy (DR), Age-related Macular Degeneration (AMD) and glaucoma. In addition to providing a concise overview of the architectural and functional aspects of MAMs, this review explores the key pathogenetic pathways involving MAMs in the development of several ocular disorders.
Collapse
Affiliation(s)
- Xin-Yu Zhang
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Cheng Han
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Yong Yao
- Department of Ophthalmology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| | - Ting-Ting Wei
- Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
29
|
Reid MV, Fredickson G, Mashek DG. Mechanisms coupling lipid droplets to MASLD pathophysiology. Hepatology 2024:01515467-990000000-01067. [PMID: 39475114 DOI: 10.1097/hep.0000000000001141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/17/2024] [Indexed: 01/03/2025]
Abstract
Hepatic steatosis, the buildup of neutral lipids in lipid droplets (LDs), is commonly referred to as metabolic dysfunction-associated steatotic liver disease when alcohol or viral infections are not involved. Metabolic dysfunction-associated steatotic liver disease encompasses simple steatosis and the more severe metabolic dysfunction-associated steatohepatitis, characterized by inflammation, hepatocyte injury, and fibrosis. Previously viewed as inert markers of disease, LDs are now understood to play active roles in disease etiology and have significant nonpathological and pathological functions in cell signaling and function. These dynamic properties of LDs are tightly regulated by hundreds of proteins that coat the LD surface, controlling lipid metabolism, trafficking, and signaling. The following review highlights various facets of LD biology with the primary goal of discussing key mechanisms through which LDs promote the development of advanced liver diseases, including metabolic dysfunction-associated steatohepatitis.
Collapse
Affiliation(s)
- Mari V Reid
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gavin Fredickson
- Department of Integrated Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
30
|
Hatawsh A, Al-Haddad RH, Okafor UG, Diab LM, Dekanoidze N, Abdulwahab AA, Mohammed OA, Doghish AS, Moussa R, Elimam H. Mitoepigenetics pathways and natural compounds: a dual approach to combatting hepatocellular carcinoma. Med Oncol 2024; 41:302. [PMID: 39465473 DOI: 10.1007/s12032-024-02538-8] [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: 08/27/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024]
Abstract
Hepatocellular carcinoma (HCC) is a leading liver cancer that significantly impacts global life expectancy and remains challenging to treat due to often late diagnoses. Despite advances in treatment, the prognosis is still poor, especially in advanced stages. Studies have pointed out that investigations into the molecular mechanisms underlying HCC, including mitochondrial dysfunction and epigenetic regulators, are potentially important targets for diagnosis and therapy. Mitoepigenetics, or the epigenetic modifications of mitochondrial DNA, have drawn wide attention for their role in HCC progression. Besides, molecular biomarkers such as mitochondrial DNA alterations and non-coding RNAs showed early diagnosis and prognosis potential. Additionally, natural compounds like alkaloids, resveratrol, curcumin, and flavonoids show promise in HCC show promise in modulating mitochondrial and epigenetic pathways involved in cancer-related processes. This review discusses how mitochondrial dysfunction and epigenetic modifications, especially mitoepigenetics, influence HCC and delves into the potential of natural products as new adjuvant treatments against HCC.
Collapse
Affiliation(s)
- Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26th of July Corridor, Sheikh Zayed City, Giza, 12588, Egypt
| | - Roya Hadi Al-Haddad
- Research and Technology Center of Environment, Water and Renewable Energy, Scientific Research Commission, Baghdad, Iraq
| | | | - Lamis M Diab
- Department of Medical Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | | | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Helwan, Cairo, 11795, Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sādāt, 32897, Egypt.
| |
Collapse
|
31
|
Chen S, Sun Y, Qin Y, Yang L, Hao Z, Xu Z, Björklund M, Liu W, Hong Z. Dynamic interaction of REEP5-MFN1/2 enables mitochondrial hitchhiking on tubular ER. J Cell Biol 2024; 223:e202304031. [PMID: 39133213 PMCID: PMC11318672 DOI: 10.1083/jcb.202304031] [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: 04/08/2023] [Revised: 02/15/2024] [Accepted: 06/12/2024] [Indexed: 08/13/2024] Open
Abstract
Mitochondrial functions can be regulated by membrane contact sites with the endoplasmic reticulum (ER). These mitochondria-ER contact sites (MERCs) are functionally heterogeneous and maintained by various tethers. Here, we found that REEP5, an ER tubule-shaping protein, interacts with Mitofusins 1/2 to mediate mitochondrial distribution throughout the cytosol by a new transport mechanism, mitochondrial "hitchhiking" with tubular ER on microtubules. REEP5 depletion led to reduced tethering and increased perinuclear localization of mitochondria. Conversely, increasing REEP5 expression facilitated mitochondrial distribution throughout the cytoplasm. Rapamycin-induced irreversible REEP5-MFN1/2 interaction led to mitochondrial hyperfusion, implying that the dynamic release of mitochondria from tethering is necessary for normal mitochondrial distribution and dynamics. Functionally, disruption of MFN2-REEP5 interaction dynamics by forced dimerization or silencing REEP5 modulated the production of mitochondrial reactive oxygen species (ROS). Overall, our results indicate that dynamic REEP5-MFN1/2 interaction mediates cytosolic distribution and connectivity of the mitochondrial network by "hitchhiking" and this process regulates mitochondrial ROS, which is vital for multiple physiological functions.
Collapse
Affiliation(s)
- Shue Chen
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
- Nuclear Organization and Gene Expression Section, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yang Sun
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Yuling Qin
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Lan Yang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Zhenhua Hao
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Zhihao Xu
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Mikael Björklund
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
- University of Edinburgh Medical School, Biomedical Sciences, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Wei Liu
- Metabolic Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Zhi Hong
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
- University of Edinburgh Medical School, Biomedical Sciences, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
32
|
Khatun J, Gelles JD, Chipuk JE. Dynamic death decisions: How mitochondrial dynamics shape cellular commitment to apoptosis and ferroptosis. Dev Cell 2024; 59:2549-2565. [PMID: 39378840 PMCID: PMC11469553 DOI: 10.1016/j.devcel.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024]
Abstract
The incorporation of mitochondria into early eukaryotes established organelle-based biochemistry and enabled metazoan development. Diverse mitochondrial biochemistry is essential for life, and its homeostatic control via mitochondrial dynamics supports organelle quality and function. Mitochondrial crosstalk with numerous regulated cell death (RCD) pathways controls the decision to die. In this review, we will focus on apoptosis and ferroptosis, two distinct forms of RCD that utilize divergent signaling to kill a targeted cell. We will highlight how proteins and processes involved in mitochondrial dynamics maintain biochemically diverse subcellular compartments to support apoptosis and ferroptosis machinery, as well as unite disparate RCD pathways through dual control of organelle biochemistry and the decision to die.
Collapse
Affiliation(s)
- Jesminara Khatun
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jesse D Gelles
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
33
|
Wang Y, Han D, Chai L, Qiu Y, Liu J, Li D, Zhang Q, Shen N, Chen Y, Chen H, Zhang J, Wang Q, Wang J, Li S, Xie X, Li M. MFN2-dependent mitochondrial dysfunction contributes to Relm-β-induced pulmonary arterial hypertension via USP18/Twist1/miR-214 pathway. Eur J Pharmacol 2024; 980:176828. [PMID: 39094924 DOI: 10.1016/j.ejphar.2024.176828] [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: 03/21/2024] [Revised: 07/03/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Induction of resistin-like molecule β (Relm-β) and mitofusin 2 (MFN2) mediated aberrant mitochondrial fission have been found to be involved in the pathogenesis of pulmonary arterial hypertension (PAH). However, the molecular mechanisms underlying Relm-β regulation of MFN2 therefore mitochondrial fission remain unclear. This study aims to address these issues. Primary cultured PASMCs and monocrotaline (MCT)-induced PAH rats were applied in this study. The results showed that Relm-β promoted cells proliferation in PASMCs, this was accompanied with the upregulation of USP18, Twist1 and miR-214, and downregulation of MFN2. We found that Relm-β increased USP18 expression which in turn raised Twist1 by suppressing its proteasome degradation. Elevation of Twist1 increased miR-214 expression and then reduced MFN2 expression and mitochondrial fragmentation leading to PASMCs proliferation. In vivo study, we confirmed that Relm-β was elevated in MCT-induced PAH rat model, and USP18/Twist1/miR-214/MFN2 axis was altered similar as in vitro. Targeting this cascade by Relm-β receptor inhibitor Calhex231, proteasome inhibitor MG-132, Twist1 inhibitor Harmine or miR-214 antagomiR prevented the development of pulmonary vascular remodeling and therefore PAH in MCT-treated rats. In conclusion, we demonstrate that Relm-β promotes PASMCs proliferation and vascular remodeling by activating USP18/Twist1/miR-214 dependent MFN2 reduction and mitochondrial fission, suggesting that this signaling pathway might be a promising target for management of PAH.
Collapse
Affiliation(s)
- Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Dong Han
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, PR China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jia Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
34
|
Wai T. Is mitochondrial morphology important for cellular physiology? Trends Endocrinol Metab 2024; 35:854-871. [PMID: 38866638 DOI: 10.1016/j.tem.2024.05.005] [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: 03/28/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
Mitochondria are double membrane-bound organelles the network morphology of which in cells is shaped by opposing events of fusion and fission executed by dynamin-like GTPases. Mutations in these genes can perturb the form and functions of mitochondria in cell and animal models of mitochondrial diseases. An expanding array of chemical, mechanical, and genetic stressors can converge on mitochondrial-shaping proteins and disrupt mitochondrial morphology. In recent years, studies aimed at disentangling the multiple roles of mitochondrial-shaping proteins beyond fission or fusion have provided insights into the homeostatic relevance of mitochondrial morphology. Here, I review the pleiotropy of mitochondrial fusion and fission proteins with the aim of understanding whether mitochondrial morphology is important for cell and tissue physiology.
Collapse
Affiliation(s)
- Timothy Wai
- Institut Pasteur, Mitochondrial Biology, CNRS UMR 3691, Université Paris Cité, Paris, France.
| |
Collapse
|
35
|
Thomas D, Yang J, Cho SJ, Stout-Delgado H. Role of Mitofusin 1 in mediating reactive oxygen species in alveolar macrophages during Streptococcuspneumoniae. Redox Biol 2024; 76:103329. [PMID: 39197317 PMCID: PMC11400614 DOI: 10.1016/j.redox.2024.103329] [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: 08/13/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024] Open
Abstract
Alveolar macrophages (AM) are key effectors of the immune response and are essential for host responses to S. pneumoniae. Mitochondria are highly dynamic organelles whose function aids in regulating the cell cycle, innate immunity, autophagy, redox signaling, calcium homeostasis, and mitochondrial quality control in AM. In response to cellular stress, mitochondria can engage in stress-induced mitochondrial hyperfusion (SIMH). The current study aimed to investigate the role of Mfn1 on mitochondrial control of reactive oxygen species (ROS) in AMs and the role of Mfn1 deficiency on immune responses to S. pneumoniae. Compared to Mfn1FloxCre- controls, there were distinct histological differences in lung tissue collected from Mfn1Floxed; CreLysM mice, with less injury and inflammation observed in mice with Mfn1 deficient myeloid cells. There was a significant decrease in lipid peroxidation and ROS production in Mfn1 deficient AM that was associated with increased superoxide dismutase (SOD) and antioxidant activity. Our findings demonstrate that Mfn1 deficiency in myeloid cells decreased inflammation and lung tissue injury during S. pneumoniae infection.
Collapse
Affiliation(s)
- David Thomas
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, USA; New York-Presbyterian Hospital, New York, NY, USA
| | - Jianjun Yang
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, USA
| | - Soo Jung Cho
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, USA; New York-Presbyterian Hospital, New York, NY, USA
| | - Heather Stout-Delgado
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, USA.
| |
Collapse
|
36
|
Zhou Y, Ling D, Wang L, Xu Z, You W, Chen W, Nong Q, Valencak TG, Shan T. Dietary "Beigeing" Fat Contains More Phosphatidylserine and Enhances Mitochondrial Function while Counteracting Obesity. RESEARCH (WASHINGTON, D.C.) 2024; 7:0492. [PMID: 39329159 PMCID: PMC11425158 DOI: 10.34133/research.0492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
Activation of mitochondrial function and heat production in adipose tissue by the modification of dietary fat is a promising strategy against obesity. However, as an important source of lipids for ketogenic and daily diets, the function of fats extracted from different adipose tissue sites was largely unknown. In this study, we illustrated the function of fats extracted from adipose tissues with different "beigeing" properties in the ketogenic diet and identified lipid profiles of fats that facilitate energy expenditure. We found that the anti-obesity effect of ketogenic diets was potentiated by using "beigeing" fat [porcine subcutaneous adipose tissue (SAT)] as a major energy-providing ingredient. Through lipidomic analyses, phosphatidylserine (PS) was identified as a functional lipid activating thermogenesis in adipose tissue. Moreover, in vivo studies showed that PS induces adipose tissue thermogenesis and alleviates diet-induced obesity in mice. In vitro studies showed that PS promotes UCP1 expression and lipolysis of adipocytes. Mechanistically, PS promoted mitochondrial function in adipocytes via the ADCY3-cAMP-PKA-PGC1α pathway. In addition, PS-PGC1a binding may affect the stability of the PGC1α protein, which further augments PS-induced thermogenesis. These results demonstrated the efficacy of dietary SAT fats in diminishing lipid accumulation and the underlying molecular mechanism of PS in enhancing UCP1 expression and mitochondrial function. Thus, our findings suggest that as dietary fat, "beigeing" fat provides more beneficial lipids that contribute to the improvement of mitochondrial function, including PS, which may become a novel, nonpharmacological therapy to increase energy expenditure and counteract obesity and its related diseases.
Collapse
Affiliation(s)
- Yanbing Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| | - Defeng Ling
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| | - Ziye Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| | - Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| | - Qiuyun Nong
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| | - Teresa G Valencak
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
37
|
Guo M, Liu R, Zhang F, Qu J, Yang Y, Li X. A new perspective on liver diseases: Focusing on the mitochondria-associated endoplasmic reticulum membranes. Pharmacol Res 2024; 208:107409. [PMID: 39284429 DOI: 10.1016/j.phrs.2024.107409] [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: 06/30/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The pathogenesis of liver diseases is multifaceted and intricate, posing a persistent global public health challenge with limited therapeutic options. Therefore, further research into liver diseases is imperative for better comprehension and advancement in treatment strategies. Numerous studies have confirmed the endoplasmic reticulum (ER) and mitochondria as key organelles driving liver diseases. Notably, the mitochondrial-associated ER membranes (MAMs) establish a physical and functional connection between the ER and mitochondria, highlighting the importance of inter-organelle communication in maintaining their functional homeostasis. This review delves into the intricate architecture and regulative mechanism of the integrated MAM that facilitate the physiological transfer of signals and substances between organelles. Additionally, we also provide a detailed overview regarding the varied pathogenic roles of malfunctioning MAM in liver diseases, focusing on its involvement in the progression of ER stress and mitochondrial dysfunction, the regulation of mitochondrial dynamics and Ca2+ transfer, as well as the disruption of lipid and glucose homeostasis. Furthermore, the current challenges and prospects associated with MAM in liver disease research are thoroughly discussed. In conclusion, elucidating the specific structure and function of MAM in different liver diseases may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Mengyu Guo
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 100029, China
| | - Fukun Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Jiaorong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Yun Yang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China.
| |
Collapse
|
38
|
Nian X, Lin P, Bai Y, Yu D, Yang X, Zhou B, Gao J, Zhao Y. Osr1-mediated mesothelial transition of liver mesenchymal cells exacerbates fibrotic liver damage. Mol Ther 2024; 32:2984-2991. [PMID: 38414241 PMCID: PMC11403217 DOI: 10.1016/j.ymthe.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/13/2024] [Accepted: 02/24/2024] [Indexed: 02/29/2024] Open
Abstract
In chronic liver diseases, hepatic stellate cells (HSCs) are induced to form the myofibroblasts responsible for scar formation, leading to liver fibrosis and cirrhosis. Here, single-cell RNA sequencing with in vivo lineage tracing in nonalcoholic steatohepatitis (NASH) model mice reveals a subpopulation of HSCs transitioning back to a state resembling their developmental precursors, mesothelial cells (MCs), after liver injury. These damage-associated intermediates between HSCs and MCs (DIHMs) can be traced with a dual recombinase system by labeling Krt19-expressing cells within prelabeled Pdgfrb+ HSCs, and DIHMs highly express inflammation- and fibrosis-associated genes. Cre and Dre-inducible depletion of DIHMs by administering diphtheria toxin reduces liver fibrosis and alleviates liver damage in NASH model mice. Importantly, knockdown of Osr1, a zinc finger transcription factor of the OSR gene family, can block DIHM induction in vitro. Conditional knockout Osr1 in Pdgfrb-expressing mesenchymal cells in NASH model mice can reduce liver fibrosis in vivo. Our study collectively uncovers an injury-induced developmental reversion process wherein HSCs undergo what we call a mesenchymal-to-mesothelial transition, which can be targeted to develop interventions to treat chronic liver diseases.
Collapse
Affiliation(s)
- Xinxin Nian
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Science, Peking University, Beijing 100871, China
| | - Pengyan Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yunfei Bai
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Donglin Yu
- Department of Biochemistry and Biophysics, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Xinyan Yang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Bin Zhou
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Beijing 100044, China
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Science, Peking University, Beijing 100871, China.
| |
Collapse
|
39
|
Deng Y, Dong Y, Zhang S, Feng Y. Targeting mitochondrial homeostasis in the treatment of non-alcoholic fatty liver disease: a review. Front Pharmacol 2024; 15:1463187. [PMID: 39290869 PMCID: PMC11405192 DOI: 10.3389/fphar.2024.1463187] [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: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, and its prevalence is rapidly increasing. Antioxidants, lipid-lowering medications, and lifestyle interventions are the most commonly used treatment options for NAFLD, but their efficacy in inhibiting steatosis progression is limited and their long-term ineffectiveness and adverse effects have been widely reported. Therefore, it is important to gain a deeper understanding of the pathogenesis of NAFLD and to identify more effective therapeutic approaches. Mitochondrial homeostasis governs cellular redox biology, lipid metabolism, and cell death, all of which are crucial to control hepatic function. Recent findings have indicated that disruption of mitochondrial homeostasis occurs in the early stage of NAFLD and mitochondrial dysfunction reinforces disease progression. In this review, we summarize the physical roles of the mitochondria and describe their response and dysfunction in the context of NAFLD. We also discuss the drug targets associated with the mitochondria that are currently in the clinical trial phase of exploration. From our findings, we hope that the mitochondria may be a promising therapeutic target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Yalan Deng
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yuan Dong
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Sitian Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yingmei Feng
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Lacombe A, Scorrano L. The interplay between mitochondrial dynamics and autophagy: From a key homeostatic mechanism to a driver of pathology. Semin Cell Dev Biol 2024; 161-162:1-19. [PMID: 38430721 DOI: 10.1016/j.semcdb.2024.02.001] [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: 11/06/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
The complex relationship between mitochondrial dynamics and autophagy illustrates how two cellular housekeeping processes are intimately linked, illuminating fundamental principles of cellular homeostasis and shedding light on disparate pathological conditions including several neurodegenerative disorders. Here we review the basic tenets of mitochondrial dynamics i.e., the concerted balance between fusion and fission of the organelle, and its interplay with macroautophagy and selective mitochondrial autophagy, also dubbed mitophagy, in the maintenance of mitochondrial quality control and ultimately in cell viability. We illustrate how conditions of altered mitochondrial dynamics reverberate on autophagy and vice versa. Finally, we illustrate how altered interplay between these two key cellular processes participates in the pathogenesis of human disorders affecting multiple organs and systems.
Collapse
Affiliation(s)
- Alice Lacombe
- Dept. of Biology, University of Padova, Padova, Italy
| | - Luca Scorrano
- Dept. of Biology, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
41
|
Lin S, Xiao Y, Lin J, Yuan Y, Shi H, Hong M, Ding L. Chromium Affects Mitochondrial Function, Leading to Apoptosis and Autophagy in Turtle Primary Hepatocytes. Animals (Basel) 2024; 14:2403. [PMID: 39199937 PMCID: PMC11350686 DOI: 10.3390/ani14162403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Hexavalent chromium (Cr(VI)), a pervasive industrial contaminant, is highly toxic to both humans and animals. However, its effects on turtles are largely unexplored. Our study aimed to investigate the toxic effects of Cr(VI) on the Reeves' turtles (Mauremys reevesii) primary hepatocytes. We exposed hepatocytes to two concentrations (25 μM and 50 μM) of Cr(VI) for 24 h. The results showed that compared to controls, Cr(VI)-treated cells showed elevated antioxidant enzyme activity (catalase (CAT) and superoxide dismutase (SOD)) and increased reactive oxygen species (ROS) levels. Adenosine triphosphatae (ATP) levels decreased, indicating mitochondrial dysfunction. Additionally, we found significant changes in mitochondrial dynamics related genes, with downregulation of mitofusin 2 (Mfn2) and silent information regulator 1 (SIRT1) and a decrease in sirtuin 3 (SIRT3) and tumor protein 53 (p53) mRNA levels. Annexin V-FITC fluorescence staining-positive cells increased with higher Cr(VI) concentrations, marked by elevated bcl-2-associated X protein (Bax) and cysteinyl aspartate specific proteinase (Caspase3) mRNA levels and reduced B-cell lymphoma-2 (Bcl2) expression. Autophagy-related genes were also affected, with increased microtubule-associated protein 1 light chain 3 (LC3-I), microtubule-associated protein light chain 3II (LC3-II), unc-51-like autophagy-activating kinase 1 (ULK1), and sequestosome 1 (p62/SQSTM1) mRNA levels and decreased mammalian target of rapamycin (mTOR) and Beclin1 expression. Taken together, Cr(VI) promotes cell apoptosis and autophagy in turtle hepatocytes by inducing oxidative stress and disrupting mitochondrial function. These findings highlight the serious health risks posed by Cr(VI) pollution and emphasize the need for protecting wild turtle populations.
Collapse
Affiliation(s)
| | | | | | | | | | - Meiling Hong
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Li Ding
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| |
Collapse
|
42
|
Meng D, Yin G, Chen S, Zhang X, Yu W, Wang L, Liu H, Jiang W, Sun Y, Zhang F. Diosgenin attenuates nonalcoholic hepatic steatosis through the hepatic SIRT1/PGC-1α pathway. Eur J Pharmacol 2024; 977:176737. [PMID: 38866362 DOI: 10.1016/j.ejphar.2024.176737] [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: 03/23/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has been increasing worldwide in recent years, causing severe economic and social burdens. Therefore, the lack of currently approved drugs for anti-NAFLD has gradually gained attention. SIRT1, as a member of the sirtuins family, is now the most widely studied in the pathophysiology of many metabolic diseases, and has great potential for preventing and treating NAFLD. Natural products such as Diosgenin (DG) have the potential to be developed as clinical drugs for the treatment of NAFLD due to their excellent multi-target therapeutic effects. In this study, we found that DG can activate the SIRT1/PGC-1α pathway and upregulate the expression of its downstream targets nuclear respiratory factor 1 (NRF1), complex IV (COX IV), mitofusin-2 (MFN2), and PPARα (perox-isome proliferator-activated receptor α) in SD rats induced by high-fat diet (HFD) and HepG2 cells caused by free fatty acids (FFAs, sodium oleate: sodium palmitate = 2:1). Conversely, the levels of dynamin-related protein 1 (DRP1) and inflammatory factors, including NF-κB p65, IL6, and TNFα, were downregulated both in vitro and in vivo. This improved mitochondrial dysfunction, fatty acid oxidation (FAO), lipid accumulation, steatosis, oxidative stress, and hepatocyte inflammation. Subsequently, we applied SIRT1 inhibitor EX527 and SIRT1 agonist SRT1720 to confirm further the necessity of activating SIRT1 for DG to exert therapeutic effects on NAFLD. In summary, these results further demonstrate the potential therapeutic role of DG as a SIRT1 natural agonist for NAFLD. (Graphical Abstracts).
Collapse
Affiliation(s)
- Decheng Meng
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Guoliang Yin
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Suwen Chen
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Xin Zhang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Wenfei Yu
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Linya Wang
- Traditional Chinese Medicine College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Hongshuai Liu
- Traditional Chinese Medicine College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Wenying Jiang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Yuqing Sun
- Traditional Chinese Medicine College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China.
| |
Collapse
|
43
|
Kolitsida P, Saha A, Caliri A, Assali E, Riera AM, Itskanov S, Magana CS, Stork B, Shirihai O, Sekler I, Koehler CM, van der Bliek AM. Mfn2 induces NCLX-mediated calcium release from mitochondria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606704. [PMID: 39149365 PMCID: PMC11326197 DOI: 10.1101/2024.08.05.606704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Mfn2 is a mitochondrial outer membrane fusion protein with the additional role of tethering mitochondria to the ER. Here, we describe a novel connection between Mfn2 and calcium release from mitochondria. We show that Mfn2 controls the mitochondrial inner membrane sodium-calcium exchange protein NCLX, which is a major source for calcium release from mitochondria. This discovery was made with the fungal toxin Phomoxanthone (PXA), which induces calcium release from mitochondria. PXA-induced calcium release is blocked by a chemical inhibitor of NCLX, while NCLX and Mfn2 deletions both also prevent PXA-induced calcium release. CETSA experiments show that PXA directly targets Mfn2, which likely controls NCLX through physical interactions since co-immunoprecipitation and proximity ligation assays show increased association between Mfn2 and NCLX upon treatment with PXA. Interactions between Mfn2 and NCLX also increase when cells are treated with mitochondrial ROS-inducing conditions, such as oligomycin treatment of respiring cells, while the interactions do not increase in Oma1 -/- cells. It seems likely that opening of cristae by Oma1-mediated cleavage of Opa1 promotes translocation of NCLX from cristae to the rim where it can come into contact with Mfn2 thus promoting PXA-induced calcium release from mitochondria. These results therefore delineate a pathway that connects ROS produced inside mitochondria with calcium release and signaling in the cytosol.
Collapse
Affiliation(s)
| | - Akash Saha
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
| | - Andrew Caliri
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
| | - Essam Assali
- Department of Physiology and Cell Biology, Ben Gurion University, Israel
- Current: Yale School of Medicine, New haven CT
| | - Alejandro Martorell Riera
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
- Current: Kite Pharmaceutical, Santa Monica CA
| | - Samuel Itskanov
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
- Current: Gilead Sciences, San Francisco CA
| | - Catalina S Magana
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
| | - Björn Stork
- Institute of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Orian Shirihai
- Department of Medicine, David Geffen School of Medicine at UCLA
| | - Israel Sekler
- Department of Physiology and Cell Biology, Ben Gurion University, Israel
| | | | | |
Collapse
|
44
|
Radosavljevic T, Brankovic M, Samardzic J, Djuretić J, Vukicevic D, Vucevic D, Jakovljevic V. Altered Mitochondrial Function in MASLD: Key Features and Promising Therapeutic Approaches. Antioxidants (Basel) 2024; 13:906. [PMID: 39199152 PMCID: PMC11351122 DOI: 10.3390/antiox13080906] [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: 06/18/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease (NAFLD), encompasses a range of liver conditions from steatosis to nonalcoholic steatohepatitis (NASH). Its prevalence, especially among patients with metabolic syndrome, highlights its growing global impact. The pathogenesis of MASLD involves metabolic dysregulation, inflammation, oxidative stress, genetic factors and, notably, mitochondrial dysfunction. Recent studies underscore the critical role of mitochondrial dysfunction in MASLD's progression. Therapeutically, enhancing mitochondrial function has gained interest, along with lifestyle changes and pharmacological interventions targeting mitochondrial processes. The FDA's approval of resmetirom for metabolic-associated steatohepatitis (MASH) with fibrosis marks a significant step. While resmetirom represents progress, further research is essential to understand MASLD-related mitochondrial dysfunction fully. Innovative strategies like gene editing and small-molecule modulators, alongside lifestyle interventions, can potentially improve MASLD treatment. Drug repurposing and new targets will advance MASLD therapy, addressing its increasing global burden. Therefore, this review aims to provide a better understanding of the role of mitochondrial dysfunction in MASLD and identify more effective preventive and treatment strategies.
Collapse
Affiliation(s)
- Tatjana Radosavljevic
- Institute of Pathophysiology “Ljubodrag Buba Mihailovic”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milica Brankovic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.B.); (J.S.)
| | - Janko Samardzic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.B.); (J.S.)
| | - Jasmina Djuretić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia;
| | - Dusan Vukicevic
- Uniklinik Mannheim, Theodor-Kutyer-Ufer 1-3, 68167 Mannheim, Germany;
| | - Danijela Vucevic
- Institute of Pathophysiology “Ljubodrag Buba Mihailovic”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia;
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia
| |
Collapse
|
45
|
Sun Y, Zhou W, Zhu M. Serum Metabolomics Uncovers the Mechanisms of Inulin in Preventing Non-Alcoholic Fatty Liver Disease. Pharmaceuticals (Basel) 2024; 17:895. [PMID: 39065745 PMCID: PMC11279973 DOI: 10.3390/ph17070895] [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: 05/16/2024] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Inulin may be a promising therapeutic molecule for treating non-alcoholic fatty liver disease (NAFLD). However, the underlying mechanisms of its therapeutic activity remain unclear. To address this issue, a high-fat-diet-induced NAFLD mouse model was developed and treated with inulin. The NAFLD phenotype was evaluated via histopathological analysis and biochemical parameters, including serum levels of alanine aminotransferase, aspartate aminotransferase, liver triglycerides, etc. A serum metabolomics study was conducted using ultra-performance liquid chromatography coupled with tandem mass spectrometry. The results revealed that inulin mitigated NAFLD symptoms such as histopathological changes and liver cholesterol levels. Through the serum metabolomics study, 347 differential metabolites were identified between the model and control groups, and 139 differential metabolites were identified between the inulin and model groups. Additionally, 48 differential metabolites (such as phosphatidylserine, dihomo-γ-linolenic acid, L-carnitine, and 13-HODE) were identified as candidate targets of inulin and subjected to pathway enrichment analysis. The results revealed that these 48 differential metabolites were enriched in several metabolic pathways such as fatty acid biosynthesis and cardiolipin biosynthesis. Taken together, our results suggest that inulin might attenuate NAFLD partially by modulating 48 differential metabolites and their correlated metabolic pathways, constituting information that might help us find novel therapies for NAFLD.
Collapse
Affiliation(s)
- Yunhong Sun
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Mingzhe Zhu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| |
Collapse
|
46
|
Vilas-Boas EA, Kowaltowski AJ. Mitochondrial redox state, bioenergetics, and calcium transport in caloric restriction: A metabolic nexus. Free Radic Biol Med 2024; 219:195-214. [PMID: 38677486 DOI: 10.1016/j.freeradbiomed.2024.04.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Mitochondria congregate central reactions in energy metabolism, many of which involve electron transfer. As such, they are expected to both respond to changes in nutrient supply and demand and also provide signals that integrate energy metabolism intracellularly. In this review, we discuss how mitochondrial bioenergetics and reactive oxygen species production is impacted by dietary interventions that change nutrient availability and impact on aging, such as calorie restriction. We also discuss how dietary interventions alter mitochondrial Ca2+ transport, regulating both mitochondrial and cytosolic processes modulated by this ion. Overall, a plethora of literature data support the idea that mitochondrial oxidants and calcium transport act as integrating signals coordinating the response to changes in nutritional supply and demand in cells, tissues, and animals.
Collapse
Affiliation(s)
- Eloisa A Vilas-Boas
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Brazil.
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil.
| |
Collapse
|
47
|
Liu R, Hong W, Hou D, Huang H, Duan C. Decoding Organelle Interactions: Unveiling Molecular Mechanisms and Disease Therapies. Adv Biol (Weinh) 2024; 8:e2300288. [PMID: 38717793 DOI: 10.1002/adbi.202300288] [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: 07/06/2023] [Revised: 01/05/2024] [Indexed: 07/13/2024]
Abstract
Organelles, substructures in the cytoplasm with specific morphological structures and functions, interact with each other via membrane fusion, membrane transport, and protein interactions, collectively termed organelle interaction. Organelle interaction is a complex biological process involving the interaction and regulation of several organelles, including the interaction between mitochondria-endoplasmic reticulum, endoplasmic reticulum-Golgi, mitochondria-lysosomes, and endoplasmic reticulum-peroxisomes. This interaction enables intracellular substance transport, metabolism, and signal transmission, and is closely related to the occurrence, development, and treatment of many diseases, such as cancer, neurodegenerative diseases, and metabolic diseases. Herein, the mechanisms and regulation of organelle interactions are reviewed, which are critical for understanding basic principles of cell biology and disease development mechanisms. The findings will help to facilitate the development of novel strategies for disease prevention, diagnosis, and treatment opportunities.
Collapse
Affiliation(s)
- Ruixue Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Weilong Hong
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Dongyao Hou
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| |
Collapse
|
48
|
Xu X, Feng J, Wang X, Zeng X, Luo Y, He X, Yang M, Lv T, Feng Z, Bao L, Zhao L, Huang D, Huang Y. Mitochondrial GRIM19 Loss Induces Liver Fibrosis through NLRP3/IL33 Activation via Reactive Oxygen Species/NF-кB Signaling. J Clin Transl Hepatol 2024; 12:539-550. [PMID: 38974954 PMCID: PMC11224902 DOI: 10.14218/jcth.2023.00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 07/09/2024] Open
Abstract
Background and Aims Hepatic fibrosis (HF) is a critical step in the progression of hepatocellular carcinoma (HCC). Gene associated with retinoid-IFN-induced mortality 19 (GRIM19), an essential component of mitochondrial respiratory chain complex I, is frequently attenuated in various human cancers, including HCC. Here, we aimed to investigate the potential relationship and underlying mechanism between GRIM19 loss and HF pathogenesis. Methods GRIM19 expression was evaluated in normal liver tissues, hepatitis, hepatic cirrhosis, and HCC using human liver disease spectrum tissue microarrays. We studied hepatocyte-specific GRIM19 knockout mice and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 (Cas9) lentivirus-mediated GRIM19 gene-editing in murine hepatocyte AML12 cells in vitro and in vivo. We performed flow cytometry, immunofluorescence, immunohistochemistry, western blotting, and pharmacological intervention to uncover the potential mechanisms underlying GRIM19 loss-induced HF. Results Mitochondrial GRIM19 was progressively downregulated in chronic liver disease tissues, including hepatitis, cirrhosis, and HCC tissues. Hepatocyte-specific GRIM19 heterozygous deletion induced spontaneous hepatitis and subsequent liver fibrogenesis in mice. In addition, GRIM19 loss caused chronic liver injury through reactive oxygen species (ROS)-mediated oxidative stress, resulting in aberrant NF-кB activation via an IKK/IкB partner in hepatocytes. Furthermore, GRIM19 loss activated NLRP3-mediated IL33 signaling via the ROS/NF-кB pathway in hepatocytes. Intraperitoneal administration of the NLRP3 inhibitor MCC950 dramatically alleviated GRIM19 loss-driven HF in vivo. Conclusions The mitochondrial GRIM19 loss facilitates liver fibrosis through NLRP3/IL33 activation via ROS/NF-кB signaling, providing potential therapeutic approaches for earlier HF prevention.
Collapse
Affiliation(s)
- Xiaohui Xu
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
| | - Jinmei Feng
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
- Department of Laboratory Medicine, Chongqing Western Hospital, Chongqing, China
| | - Xin Wang
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, China
| | - Xin Zeng
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ying Luo
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Xinyu He
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Meihua Yang
- Departments of Neurology, Epilepsy Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Tiewei Lv
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
| | - Zijuan Feng
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Liming Bao
- Department of Clinical Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Li Zhao
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Daochao Huang
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Yi Huang
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
- Departments of Medicine (Oncology), Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
49
|
Mathyk BA, Tabetah M, Karim R, Zaksas V, Kim J, Anu RI, Muratani M, Tasoula A, Singh RS, Chen YK, Overbey E, Park J, Cope H, Fazelinia H, Povero D, Borg J, Klotz RV, Yu M, Young SL, Mason CE, Szewczyk N, St Clair RM, Karouia F, Beheshti A. Spaceflight induces changes in gene expression profiles linked to insulin and estrogen. Commun Biol 2024; 7:692. [PMID: 38862620 PMCID: PMC11166981 DOI: 10.1038/s42003-023-05213-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/03/2023] [Indexed: 06/13/2024] Open
Abstract
Organismal adaptations to spaceflight have been characterized at the molecular level in model organisms, including Drosophila and C. elegans. Here, we extend molecular work to energy metabolism and sex hormone signaling in mice and humans. We found spaceflight induced changes in insulin and estrogen signaling in rodents and humans. Murine changes were most prominent in the liver, where we observed inhibition of insulin and estrogen receptor signaling with concomitant hepatic insulin resistance and steatosis. Based on the metabolic demand, metabolic pathways mediated by insulin and estrogen vary among muscles, specifically between the soleus and extensor digitorum longus. In humans, spaceflight induced changes in insulin and estrogen related genes and pathways. Pathway analysis demonstrated spaceflight induced changes in insulin resistance, estrogen signaling, stress response, and viral infection. These data strongly suggest the need for further research on the metabolic and reproductive endocrinologic effects of space travel, if we are to become a successful interplanetary species.
Collapse
Affiliation(s)
- Begum Aydogan Mathyk
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Marshall Tabetah
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Rashid Karim
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45220, USA
- Novartis Institutes for Biomedical Research, 181 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Victoria Zaksas
- Center for Translational Data Science, University of Chicago, Chicago, IL, 60637, USA
- Clever Research Lab, Springfield, IL, 62704, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - R I Anu
- Department of Cancer Biology & Therapeutics, Precision Oncology and Multi-omics clinic, Genetic counseling clinic. Department of Clinical Biochemistry, MVR Cancer Centre and Research Institute, Calicut, India
| | - Masafumi Muratani
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, 305-8575, Japan
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Alexia Tasoula
- Department of Life Science Engineering, FH Technikum, Vienna, Austria
| | | | - Yen-Kai Chen
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Eliah Overbey
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jiwoon Park
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Henry Cope
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
| | - Hossein Fazelinia
- Department of Biomedical and Health Informatics and Proteomics Core Facility, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Davide Povero
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences, Msida, MSD2090, Malta
| | - Remi V Klotz
- Department of Stem Cell Biology & Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Min Yu
- Department of Stem Cell Biology & Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Steven L Young
- Division of Reproductive Endocrinology and Infertility, Duke School of Medicine, Durham, NC, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Nathaniel Szewczyk
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Riley M St Clair
- Department of Life Sciences, Quest University, Squamish, BC, Canada
| | - Fathi Karouia
- Blue Marble Space Institute of Science, Exobiology Branch, NASA Ames Research Center, Moffett Field, CA, USA
- Space Research Within Reach, San Francisco, CA, USA; Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Afshin Beheshti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
50
|
Morcillo P, Kabra K, Velasco K, Cordero H, Jennings S, Yun TD, Larrea D, Akman HO, Schon EA. Aberrant ER-mitochondria communication is a common pathomechanism in mitochondrial disease. Cell Death Dis 2024; 15:405. [PMID: 38858390 PMCID: PMC11164949 DOI: 10.1038/s41419-024-06781-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024]
Abstract
Genetic mutations causing primary mitochondrial disease (i.e those compromising oxidative phosphorylation [OxPhos]) resulting in reduced bioenergetic output display great variability in their clinical features, but the reason for this is unknown. We hypothesized that disruption of the communication between endoplasmic reticulum (ER) and mitochondria at mitochondria-associated ER membranes (MAM) might play a role in this variability. To test this, we assayed MAM function and ER-mitochondrial communication in OxPhos-deficient cells, including cybrids from patients with selected pathogenic mtDNA mutations. Our results show that each of the various mutations studied indeed altered MAM functions, but notably, each disorder presented with a different MAM "signature". We also found that mitochondrial membrane potential is a key driver of ER-mitochondrial connectivity. Moreover, our findings demonstrate that disruption in ER-mitochondrial communication has consequences for cell survivability that go well beyond that of reduced ATP output. The findings of a "MAM-OxPhos" axis, the role of mitochondrial membrane potential in controlling this process, and the contribution of MAM dysfunction to cell death, reveal a new relationship between mitochondria and the rest of the cell, as well as providing new insights into the diagnosis and treatment of these devastating disorders.
Collapse
Affiliation(s)
- Patricia Morcillo
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA.
| | - Khushbu Kabra
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kevin Velasco
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Hector Cordero
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Immunology Group, Department of Physiology, Faculty of Veterinary, University of Extremadura, Caceres, 10003, Spain
| | - Sarah Jennings
- Stony Brook University, Stony Brook, New York, NY, 11794, USA
| | - Taekyung D Yun
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Delfina Larrea
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - H Orhan Akman
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Eric A Schon
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA.
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|