1
|
Zhang HL, Pan ZN, Ju JQ, Ji YM, Wang Y, Sun SC. Formin INF2 supplementation alleviates cytoskeleton-based mitochondria defects for oocyte quality under obesity. Free Radic Biol Med 2025; 233:250-263. [PMID: 40180021 DOI: 10.1016/j.freeradbiomed.2025.04.003] [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: 01/15/2025] [Revised: 03/04/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
Obesity is one main cause of reproductive disorders in female, and oocytes show meiotic maturation defects under obesity, which leads to infertility. However, the molecular characterization for the obese oocytes remains largely unclear. Inverted-formin 2 (INF2) is a formin family member which is involved in actin-based multiple cellular events including vesicle transport and oxidative stress-induced apoptosis. In present study, we reported that INF2 deficiency linked with declined oocyte quality of obesity. Our results showed that INF2 expression decreased in the oocytes of obese mice. INF2 deficiency caused the failure of polar body extrusion and induced large polar bodies. We showed that INF2 depletion disturbed mitochondrial distribution and function, which might be due to the association with mitochondria fission factor DRP1. INF2 co-localized with cytoplasmic actin and its depletion reduced actin polymerization, which further caused the failure of spindle migration in both mouse and porcine oocytes. In addition, we also found that INF2 interacted with HDAC6 and further affected tubulin acetylation for microtubule stability, which disturbed mitochondrial transport. Exogenous INF2 mRNA supplement rescued the meiotic maturation defects of oocytes from obese mice. Thus, our study demonstrated that INF2 is responsible for both mouse and porcine oocyte maturation through its regulation on actin polymerization and tubulin acetylation for mitochondrial function, and its deficiency might be one cause for obesity-induced oocyte defects.
Collapse
Affiliation(s)
- Hao-Lin Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yi-Ming Ji
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yue Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Reproductive Medicine of Guangxi Medical and Health Key Discipline Construction Project, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
2
|
Jin J, Nolte PA. Mitochondrial Distribution and Osteocyte Mechanosensitivity. Curr Osteoporos Rep 2025; 23:22. [PMID: 40402395 PMCID: PMC12098195 DOI: 10.1007/s11914-025-00918-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE OF REVIEW Mechanical loading of bone is an important physical stimulus for bone tissue remodeling and adaptation. It is transmitted from the extracellular matrix all the way to the osteocyte nucleus via the extracellular matrix-integrin-cytoskeleton-nucleus system. Mitochondria are integral in sensing of mechanical loads to allow the cell to adapt to its environment. This review provides a background of mitochondrial distribution in osteocytes especially during mechanical loading, discussing the importance of mitochondrial distribution in osteocyte mechanosensitivity and mechanotransduction. RECENT FINDINGS Mitochondria throughout the osteocyte are highly dynamic and provide essential metabolic and signal functions to regulate osteocyte morphology and function. They undergo the processes of fission and fusion accompanied by mitochondrial DNA distribution. The mitochondrial network structure and function in osteocytes can be regulated by mechanical loading. Interestingly, mitochondria can be transmitted by osteocytes into adjacent cells to communicate with them via tunneling nanotubes, migrasomes, and blebbisomes, causing changes in cell morphology and/or function. Mitochondrial distribution in or out osteocytes can be rearranged by physical and (bio)chemical signals via fission and fusion, as well as tunneling nanotubes, migrasomes, and blebbisomes. Mechanical loading-induced changes in mitochondria may drive signaling pathways of cell function in aging and diseases. More insights into interactions between neighbouring osteocytes and between osteocytes and other cell types would facilitate the development of new strategies to apply mitochondrial therapy for bone-related diseases.
Collapse
Affiliation(s)
- Jianfeng Jin
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, Amsterdam, 1081 LA, The Netherlands
| | - Peter A Nolte
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, Amsterdam, 1081 LA, The Netherlands.
- Department of Orthopedic Surgery, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM, Hoofddorp, The Netherlands.
| |
Collapse
|
3
|
Kamerkar SC, Liu A, Higgs HN. Mitochondrial fission - changing perspectives for future progress. J Cell Sci 2025; 138:jcs263640. [PMID: 40104946 DOI: 10.1242/jcs.263640] [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: 03/20/2025] Open
Abstract
Mitochondrial fission is important for many aspects of cellular homeostasis, including mitochondrial distribution, stress response, mitophagy, mitochondrially derived vesicle production and metabolic regulation. Several decades of research has revealed much about fission, including identification of a key division protein - the dynamin Drp1 (also known as DNM1L) - receptors for Drp1 on the outer mitochondrial membrane (OMM), including Mff, MiD49 and MiD51 (also known as MIEF2 and MIEF1, respectively) and Fis1, and important Drp1 regulators, including post-translational modifications, actin filaments and the phospholipid cardiolipin. In addition, it is now appreciated that other organelles, including the endoplasmic reticulum, lysosomes and Golgi-derived vesicles, can participate in mitochondrial fission. However, a more holistic understanding of the process is lacking. In this Review, we address three questions that highlight knowledge gaps. First, how do we quantify mitochondrial fission? Second, how does the inner mitochondrial membrane (IMM) divide? Third, how many 'types' of fission exist? We also introduce a model that integrates multiple regulatory factors in mammalian mitochondrial fission. In this model, three possible pathways (cellular stimulation, metabolic switching or mitochondrial dysfunction) independently initiate Drp1 recruitment at the fission site, followed by a shared second step in which Mff mediates subsequent assembly of a contractile Drp1 ring. We conclude by discussing some perplexing issues in fission regulation, including the effects of Drp1 phosphorylation and the multiple Drp1 isoforms.
Collapse
Affiliation(s)
- Sukrut C Kamerkar
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| | - Ao Liu
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
4
|
Merta H, Gov K, Isogai T, Paul B, Sannigrahi A, Radhakrishnan A, Danuser G, Henne WM. Spatial proteomics of ER tubules reveals CLMN, an ER-actin tether at focal adhesions that promotes cell migration. Cell Rep 2025; 44:115502. [PMID: 40184252 DOI: 10.1016/j.celrep.2025.115502] [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: 02/14/2024] [Revised: 12/20/2024] [Accepted: 03/11/2025] [Indexed: 04/06/2025] Open
Abstract
The endoplasmic reticulum (ER) is structurally and functionally diverse, yet how its functions are organized within morphological subdomains is incompletely understood. Utilizing TurboID-based proximity labeling and CRISPR knockin technologies, we map the proteomic landscape of the human ER network. Sub-organelle proteomics reveals enrichments of proteins into ER tubules, sheets, and the nuclear envelope. We uncover an ER-enriched actin-binding protein, calmin/CLMN, and define it as an ER-actin tether that localizes to focal adhesions adjacent to ER tubules. Mechanistically, we find that CLMN depletion perturbs adhesion disassembly, actin dynamics, and cell movement. CLMN-depleted cells display decreased polarization of ER-plasma membrane contacts and calcium signaling factor STIM1 and altered calcium signaling near ER-actin interfaces, suggesting that CLMN influences calcium signaling to facilitate F-actin/adhesion dynamics. Collectively, we map the sub-organelle proteome landscape of the ER, identify CLMN as an ER-actin tether, and describe a non-canonical mechanism by which ER tubules engage actin to regulate cell migration.
Collapse
Affiliation(s)
- Holly Merta
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kaitlynn Gov
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tadamoto Isogai
- Lyda Hill Department of Bioinformatics and Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Blessy Paul
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Achinta Sannigrahi
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Arun Radhakrishnan
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gaudenz Danuser
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Lyda Hill Department of Bioinformatics and Cecil H. and Ida Green Center for Systems Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - W Mike Henne
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Wang N, Wang X, Lan B, Gao Y, Cai Y. DRP1, fission and apoptosis. Cell Death Discov 2025; 11:150. [PMID: 40195359 PMCID: PMC11977278 DOI: 10.1038/s41420-025-02458-0] [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: 07/20/2024] [Revised: 03/15/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
Mitochondrial fission is a critical physiological process in eukaryotic cells, participating in various vital activities such as mitosis, mitochondria quality control, and mitophagy. Recent studies have revealed a tight connection between mitochondrial fission and the mitochondrial metabolism, as well as apoptosis, which involves multiple cellular events and interactions between organelles. As a pivotal molecule in the process of mitochondrial fission, the function of DRP1 is regulated at multiple levels, including transcription, post-translational modifications. This review follows the guidelines for Human Gene Nomenclature and will focus on DRP1, discussing its activity regulation, its role in mitochondrial fission, and the relationship between mitochondrial fission and apoptosis.
Collapse
Affiliation(s)
- Nan Wang
- The Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xinwai Wang
- The Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Beiwu Lan
- The Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yufei Gao
- The Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Yuanyuan Cai
- The First Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Piñeiro-Llanes J, Suzuki-Hatano S, Jain A, Venigalla S, Kamat M, Basso KB, Cade WT, Simmons CS, Pacak CA. Rescue of mitochondrial dysfunction through alteration of extracellular matrix composition in barth syndrome cardiac fibroblasts. Biomaterials 2025; 315:122922. [PMID: 39509858 PMCID: PMC11625619 DOI: 10.1016/j.biomaterials.2024.122922] [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: 06/21/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Fibroblast-ECM (dys)regulation is associated with a plethora of diseases. The ECM acts as a reservoir of inflammatory factors and cytokines that mediate molecular mechanisms within cardiac cell populations. The role of ECM-mitochondria crosstalk in the development and progression of cardiac disorders remains uncertain. We evaluated the influence of ECM produced by stromal cells from patients with the mitochondrial cardiomyopathy (Barth syndrome, BTHS) and unaffected healthy controls on cardiac fibroblast (CF) metabolic function. To do this, cell-derived matrices CDMs were generated from BTHS and healthy human pluripotent stem cell-derived CFs (hPSC-CF) and used as cell culture substrates. BTHS CDMs negatively impacted the mitochondrial function of healthy hPSC-CFs while healthy CDMs improved mitochondrial function in BTHS hPSC-CFs. Mass spectrometry comparisons identified 5 matrisome proteins differentially expressed in BTHS compared to healthy CDM. Our results highlight a key role for the ECM in disease through its impact on mitochondrial function.
Collapse
Affiliation(s)
- Janny Piñeiro-Llanes
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA.
| | - Silveli Suzuki-Hatano
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| | - Ananya Jain
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA.
| | - Sree Venigalla
- Department of Neurology and Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, 55455, USA.
| | - Manasi Kamat
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA.
| | - Kari B Basso
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA.
| | - William T Cade
- Doctor of Physical Therapy Division, Duke University, Durham, NC, 27710, USA.
| | - Chelsey S Simmons
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA; Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32611, USA.
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA; Department of Neurology and Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, 55455, USA.
| |
Collapse
|
9
|
Wu H, Diao H, Zhang F, Jiang W, Pan T, Bian Y. Organelle interplay in cardiovascular diseases: Mechanisms, pathogenesis, and therapeutic perspectives. Biomed Pharmacother 2025; 185:117978. [PMID: 40073746 DOI: 10.1016/j.biopha.2025.117978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/16/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of rising morbidity and mortality among humans worldwide; however, our approach to the pathogenesis, exploration, and management of CVDs still remains limited. As the heart consists of cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, and several types of cells, different types of dysfunction in the interplay between organelles play an important damaging role, resulting in cardiac pathologies. The interplay between cellular organelles is intricate and vital for maintaining cellular homeostasis, as highlighted by multiple diseases linked to the dysfunction of both organelles. Many studies have revealed the potential mechanisms by which organelles communicate with each other and regulate the pathological processes of CVDs together. However, gaps remain in fully understanding the complexity of these interactions and translating these insights into therapeutic approaches. In this review, we summarized how the interplay between cellular organelles in the cardiomyocytes alters in various heart diseases. We find underexplored areas, such as the crucial signaling pathways governing organelle communication, and discuss their implications for disease future progression. Furthermore, we evaluate emerging potential medicines aimed at restoring organelle interactions. Finally, we propose future directions for researching to advance the development of novel medicines and therapies, addressing current gaps and providing a theoretical basis for improved clinical outcomes in CVDs.
Collapse
Affiliation(s)
- Han Wu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hongtao Diao
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Feng Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Weitao Jiang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tengfei Pan
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yu Bian
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
10
|
Li P, Fan Z, Huang Y, Luo L, Wu X. Mitochondrial dynamics at the intersection of macrophage polarization and metabolism. Front Immunol 2025; 16:1520814. [PMID: 40196123 PMCID: PMC11973336 DOI: 10.3389/fimmu.2025.1520814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Macrophages are vital sentinels in innate immunity, and their functions cannot be performed without internal metabolic reprogramming. Mitochondrial dynamics, especially mitochondrial fusion and fission, contributes to the maintenance of mitochondrial homeostasis. The link between mitochondrial dynamics and macrophages in the past has focused on the immune function of macrophages. We innovatively summarize and propose a link between mitochondrial dynamics and macrophage metabolism. Among them, fusion-related FAM73b, MTCH2, SLP-2 (Stomatin-like protein 2), and mtSIRT, and fission-related Fis1 and MTP18 may be the link between mitochondrial dynamics and macrophage metabolism association. Furthermore, post-translational modifications (PTMs) of mtSIRT play prominent roles in mitochondrial dynamics-macrophage metabolism connection, such as deacetylates and hypersuccinylation. MicroRNAs such as miR-150, miR-15b, and miR-125b are also possible entry points. The metabolic reprogramming of macrophages through the regulation of mitochondrial dynamics helps improve their adaptability and resistance to adverse environments and provides therapeutic possibilities for various diseases.
Collapse
Affiliation(s)
- Pan Li
- Department of Environment and Safety Engineering, Taiyuan Institute of Technology, Taiyuan, China
| | - Zhengbo Fan
- People’s Government of Huangshui Town, Shizhu Tujia Autonomous County, Chongqing, China
| | - Yanlan Huang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Liang Luo
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiaoyan Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
11
|
He P, Chang H, Qiu Y, Wang Z. Mitochondria associated membranes in dilated cardiomyopathy: connecting pathogenesis and cellular dysfunction. Front Cardiovasc Med 2025; 12:1571998. [PMID: 40166597 PMCID: PMC11955654 DOI: 10.3389/fcvm.2025.1571998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, yet therapeutic options remain limited. While traditional research has focused on mechanisms such as energy deficits and calcium dysregulation, increasing evidence suggests that mitochondria-associated membranes (MAMs) could provide new insights into understanding and treating DCM. In this narrative review, we summarize the key role of MAMs, crucial endoplasmic reticulum (ER)-mitochondria interfaces, in regulating cellular processes such as calcium homeostasis, lipid metabolism, and mitochondrial dynamics. Disruption of MAMs function may initiate pathological cascades, including ER stress, inflammation, and cell death. These disruptions in MAM function lead to further destabilization of cellular homeostasis. Identifying MAMs as key modulators of cardiac health may provide novel insights for early diagnosis and targeted therapies in DCM.
Collapse
Affiliation(s)
- Pingge He
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hongbo Chang
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yueqing Qiu
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhentao Wang
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
12
|
Chen C, Dai G, Fan M, Wang X, Niu K, Gao W. Mitochondria-associated endoplasmic reticulum membranes and myocardial ischemia: from molecular mechanisms to therapeutic strategies. J Transl Med 2025; 23:277. [PMID: 40050915 PMCID: PMC11884070 DOI: 10.1186/s12967-025-06262-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/17/2025] [Indexed: 03/10/2025] Open
Abstract
Myocardial ischemia has the highest disease burden among all cardiovascular diseases making it a significant challenge to the global public health. It can result in myocardial cell damage and death due to impaired mitochondrial and endoplasmic reticulum (ER) functions. These two organelles are important regulators of cell death. In recent years, research has shifted from isolated studies of individual organelles to a more integrative approach, with a particular focus on their membrane contact sites-Mitochondria-Associated Endoplasmic Reticulum Membranes (MAMs). These dynamic microdomains play a crucial role in regulating material exchange and signal transduction between the endoplasmic reticulum and mitochondria. This review comprehensively describes the intricate structure of MAMs and their multifaceted roles in cellular pathophysiological processes. Particular focus was directed at the far-reaching effects of MAMs in regulating key pathological events including calcium homeostasis, mitochondrial dysfunction, ER stress, oxidative stress, and autophagy in ischemic heart disease (IHD). The potential treatment targets and regulatory mechanisms of MAMs were discussed and summarized, providing novel research directions and treatment approaches for improving myocardial ischemia-related diseases.
Collapse
Affiliation(s)
- Chen Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guohua Dai
- Department of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Maoxia Fan
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xingmeng Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kaibin Niu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wulin Gao
- Department of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
13
|
Zhang J, Yan H, Wang Y, Yue X, Wang M, Liu L, Qiao P, Zhu Y, Li Z. Emerging insights into pulmonary hypertension: the potential role of mitochondrial dysfunction and redox homeostasis. Mol Cell Biochem 2025; 480:1407-1429. [PMID: 39254871 DOI: 10.1007/s11010-024-05096-9] [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: 05/16/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Pulmonary hypertension (PH) is heterogeneous diseases that can lead to death due to progressive right heart failure. Emerging evidence suggests that, in addition to its role in ATP production, changes in mitochondrial play a central role in their pathogenesis, regulating integrated metabolic and signal transduction pathways. This review focuses on the basic principles of mitochondrial redox status in pulmonary vascular and right ventricular disorders, a series of dysfunctional processes including mitochondrial quality control (mitochondrial biogenesis, mitophagy, mitochondrial dynamics, mitochondrial unfolded protein response) and mitochondrial redox homeostasis. In addition, we will summarize how mitochondrial renewal and dynamic changes provide innovative insights for studying and evaluating PH. This will provide us with a clearer understanding of the initial signal transmission of mitochondria in PH, which would further improve our understanding of the pathogenesis of PH.
Collapse
Affiliation(s)
- Junming Zhang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Huimin Yan
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yan Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Xian Yue
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Meng Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Limin Liu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Pengfei Qiao
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yixuan Zhu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Zhichao Li
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China.
| |
Collapse
|
14
|
Lindamood HL, Liu TM, Read TA, Vitriol EA. Using ALS to understand profilin 1's diverse roles in cellular physiology. Cytoskeleton (Hoboken) 2025; 82:111-129. [PMID: 39056295 PMCID: PMC11762371 DOI: 10.1002/cm.21896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Profilin is an actin monomer-binding protein whose role in actin polymerization has been studied for nearly 50 years. While its principal biochemical features are now well understood, many questions remain about how profilin controls diverse processes within the cell. Dysregulation of profilin has been implicated in a broad range of human diseases, including neurodegeneration, inflammatory disorders, cardiac disease, and cancer. For example, mutations in the profilin 1 gene (PFN1) can cause amyotrophic lateral sclerosis (ALS), although the precise mechanisms that drive neurodegeneration remain unclear. While initial work suggested proteostasis and actin cytoskeleton defects as the main pathological pathways, multiple novel functions for PFN1 have since been discovered that may also contribute to ALS, including the regulation of nucleocytoplasmic transport, stress granules, mitochondria, and microtubules. Here, we will review these newly discovered roles for PFN1, speculate on their contribution to ALS, and discuss how defects in actin can contribute to these processes. By understanding profilin 1's involvement in ALS pathogenesis, we hope to gain insight into this functionally complex protein with significant influence over cellular physiology.
Collapse
Affiliation(s)
- Halli L Lindamood
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Tatiana M Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Tracy-Ann Read
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Eric A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
15
|
Liu X, Li T, Tu X, Xu M, Wang J. Mitochondrial fission and fusion in neurodegenerative diseases:Ca 2+ signalling. Mol Cell Neurosci 2025; 132:103992. [PMID: 39863029 DOI: 10.1016/j.mcn.2025.103992] [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: 08/04/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Neurodegenerative diseases (NDs) are a group of disorders characterized by the progressive loss of neuronal structure and function. The pathogenesis is intricate and involves a network of interactions among multiple causes and systems. Mitochondria and Ca2+ signaling have long been considered to play important roles in the development of various NDs. Mitochondrial fission and fusion dynamics are important processes of mitochondrial quality control, ensuring the stability of mitochondrial structure and function. Mitochondrial fission and fusion imbalance and Ca2+ signaling disorders can aggravate the disease progression of NDs. In this review, we explore the relationship between mitochondrial dynamics and Ca2+ signaling in AD, PD, ALS, and HD, focusing on the roles of key regulatory proteins (Drp1, Fis1, Mfn1/2, and Opa1) and the association structures between mitochondria and the endoplasmic reticulum (MERCs/MAMs). We provide a detailed analysis of their involvement in the pathogenesis of these four NDs. By integrating these mechanisms, we aim to clarify their contributions to disease progression and offer insights into the development of therapeutic strategies that target mitochondrial dynamics and Ca2+ signaling. We also examine the progress in drug research targeting these pathways, highlighting their potential as therapeutic targets in the treatment of NDs.
Collapse
Affiliation(s)
- Xuan Liu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Tianjiao Li
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Xinya Tu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Mengying Xu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Jianwu Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| |
Collapse
|
16
|
Hemel IMGM, Steen C, Denil SLIJ, Ertaylan G, Kutmon M, Adriaens M, Gerards M. The unusual suspect: A novel role for intermediate filament proteins in mitochondrial morphology. Mitochondrion 2025; 81:102008. [PMID: 39909388 DOI: 10.1016/j.mito.2025.102008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Mitochondrial dynamics is crucial for cellular homeostasis. However, not all proteins involved are known. Using a protein-protein interaction (PPI) approach, we identified ITPRIPL2 for involvement in mitochondrial dynamics. ITPRIPL2 co-localizes with intermediate filament protein vimentin, supported by protein simulations. ITPRIPL2 knockdown reveals mitochondrial elongation, disrupts vimentin processing, intermediate filament formation, and alters vimentin-related pathways. Interestingly, vimentin knockdown also leads to mitochondrial elongation. These findings highlight ITPRIPL2 as vimentin-associated protein essential for intermediate filament structure and suggest a role for intermediate filaments in mitochondrial morphology. Our study demonstrates that PPI analysis is a powerful approach for identifying novel mitochondrial dynamics proteins.
Collapse
Affiliation(s)
- Irene M G M Hemel
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Carlijn Steen
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Simon L I J Denil
- Flemish Institute for Technological Research (VITO) 2400 Mol, Belgium
| | - Gökhan Ertaylan
- Flemish Institute for Technological Research (VITO) 2400 Mol, Belgium
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Michiel Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Mike Gerards
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands.
| |
Collapse
|
17
|
Greene ES, Chen PR, Walk C, Bedford M, Dridi S. Mitochondrial dysfunction is a hallmark of woody breast myopathy in broiler chickens. Front Physiol 2025; 16:1543788. [PMID: 40034536 PMCID: PMC11872917 DOI: 10.3389/fphys.2025.1543788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
The woody breast (WB) myopathy poses significant economic and welfare concerns to the poultry industry, however, there is no effective strategy to mitigate this pathology due to its unknown etiology. After showing previously that hypoxia is a key factor in WB progression, we used here various techniques demonstrating dysregulated mitochondria (morphology, biogenesis, tethering, function, and bioenergetics) in WB-affected muscles and in hypoxic myoblasts compared to healthy tissues and normoxic cells, respectively. The increased levels of calcium (Ca2+) in both WB-affected tissues and hypoxic myoblasts suggested that mitochondrial Ca2+ overload is likely a leading cause for mitochondrial dysfunction that merits further in-depth investigation. These findings are the first, to the best of our knowledge, to provide fundamental insights into the underlying molecular mechanisms of WB and open new vistas for understanding the interplay between calcium, mitochondrial (dys)function, and avian muscle health for subsequent development of effective preventative/corrective strategies.
Collapse
Affiliation(s)
- Elizabeth S. Greene
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, United States
| | - Paula R. Chen
- USDA-ARS, Plant Genetics Research Unit, Columbia, MO, United States
| | | | | | - Sami Dridi
- Center of Excellence for Poultry Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
18
|
Townson J, Progida C. The emerging roles of the endoplasmic reticulum in mechanosensing and mechanotransduction. J Cell Sci 2025; 138:JCS263503. [PMID: 39976266 DOI: 10.1242/jcs.263503] [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: 02/21/2025] Open
Abstract
Cells are continuously subjected to physical and chemical cues from the extracellular environment, and sense and respond to mechanical cues via mechanosensation and mechanotransduction. Although the role of the cytoskeleton in these processes is well known, the contribution of intracellular membranes has been long neglected. Recently, it has become evident that various organelles play active roles in both mechanosensing and mechanotransduction. In this Review, we focus on mechanosensitive roles of the endoplasmic reticulum (ER), the functions of which are crucial for maintaining cell homeostasis. We discuss the effects of mechanical stimuli on interactions between the ER, the cytoskeleton and other organelles; the role of the ER in intracellular Ca2+ signalling via mechanosensitive channels; and how the unfolded protein response and lipid homeostasis contribute to mechanosensing. The expansive structure of the ER positions it as a key intracellular communication hub, and we additionally explore how this may be leveraged to transduce mechanical signals around the cell. By synthesising current knowledge, we aim to shed light on the emerging roles of the ER in cellular mechanosensing and mechanotransduction.
Collapse
Affiliation(s)
- Jonathan Townson
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| |
Collapse
|
19
|
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
|
20
|
Cicero J, Manor U. Beyond static snapshots: Mitochondria in action. Curr Opin Cell Biol 2025; 92:102460. [PMID: 39736172 DOI: 10.1016/j.ceb.2024.102460] [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: 08/01/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/01/2025]
Abstract
Mitochondria are dynamic organelles essential for cellular homeostasis, undergoing continuous fission and fusion processes that regulate their morphology, distribution, and function. Disruptions in these dynamics are linked to numerous diseases, including neurodegenerative disorders and cancer. Understanding these processes is vital for developing therapeutic strategies aimed at mitigating mitochondrial dysfunction. This review provides an overview of recent perspectives on mitochondrial dynamics, focusing on the need for live video microscopy imaging in order to fully understand mitochondrial phenotypes and pathology. Advanced imaging tools, such as machine learning-based segmentation and label-free microscopy approaches, have the potential to transform our ability to study mitochondrial dynamics in live cells.
Collapse
Affiliation(s)
- Julien Cicero
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Uri Manor
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States.
| |
Collapse
|
21
|
Ji Y, Lin Y, He J, Xie Y, An W, Luo X, Qiao X, Li Z. Research progress of mitochondria and cytoskeleton crosstalk in tumour development. Biochim Biophys Acta Rev Cancer 2025; 1880:189254. [PMID: 39732178 DOI: 10.1016/j.bbcan.2024.189254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
During tumour progression, organelle function undergoes dramatic changes, and crosstalk among organelles plays a significant role. Crosstalk between mitochondria and other organelles such as the endoplasmic reticulum and cytoskeleton has focussed attention on the mechanisms of tumourigenesis. This review demonstrates an overview of the molecular structure of the mitochondrial-cytoskeletal junction and its biological interactions. It also presents a detailed and comprehensive description of mitochondrial-cytoskeletal crosstalk in tumour occurrence and development, including tumour cell proliferation, apoptosis, autophagy, metabolic rearrangement, and metastasis. Finally, the application of crosstalk in tumour therapy, including drug combinations and chemoresistance, is discussed. This review offers a theoretical basis for establishing mitochondrial-cytoskeletal junctions as therapeutic targets, and offers novel insights into the future management of malignant tumours.
Collapse
Affiliation(s)
- Yue Ji
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Yingchi Lin
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China; Provincial key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, China
| | - Jing He
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Diseases, Shenyang 110002, Liaoning Province, China
| | - Yuanyuan Xie
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Wenmin An
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Xinyu Luo
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China
| | - Xue Qiao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China.
| | - Zhenning Li
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang 110002, Liaoning Province, China.
| |
Collapse
|
22
|
Oliver-De La Cruz J, Roca-Cusachs P. Mitochondria mechanosensing: The powerhouse fueling cellular force signaling. Curr Biol 2025; 35:R76-R79. [PMID: 39837277 DOI: 10.1016/j.cub.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Mechanical forces influence mitochondrial dynamics through previously unexplored mechanisms. A new study demonstrates that actomyosin tension inhibits mitochondrial fission by phosphorylating a key component of the fission complex and that this event regulates the nuclear accumulation of critical transcription factors.
Collapse
Affiliation(s)
- Jorge Oliver-De La Cruz
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain.
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia, The Barcelona Institute for Science and Technology, Barcelona, Spain; Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
23
|
Huong QTT, Truc LTN, Ueda H, Fukui K, Higasa K, Sato Y, Takeda S, Hattori M, Tsukaguchi H. Nerve Enlargement in Patients with INF2 Variants Causing Peripheral Neuropathy and Focal Segmental Glomerulosclerosis. Biomedicines 2025; 13:127. [PMID: 39857711 PMCID: PMC11763285 DOI: 10.3390/biomedicines13010127] [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: 11/18/2024] [Revised: 12/23/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Charcot-Marie-Tooth (CMT) disease is an inherited peripheral neuropathy primarily involving motor and sensory neurons. Mutations in INF2, an actin assembly factor, cause two diseases: peripheral neuropathy CMT-DIE (MIM614455) and/or focal segmental glomerulosclerosis (FSGS). These two phenotypes arise from the progressive degeneration affecting podocytes and Schwann cells. In general, nerve enlargement has been reported in 25% of the demyelinating CMT subtype (CMT1), while little is known about the CMT-DIE caused by INF2 variants. Methods: To characterize the peripheral nerve phenotype of INF2-related CMT, we studied the clinical course, imaging, histology, and germline genetic variants in two unrelated CMT-DIE patients. Results: Patient 1 (INF2 p.Gly73Asp) and patient 2 (p.Val108Asp) first noticed walking difficulties at 10 to 12 years old. Both of them were electrophysiologically diagnosed with demyelinating neuropathy. In patient 2, the sural nerve biopsy revealed an onion bulb formation. Both patients developed nephrotic syndrome almost simultaneously with CMT and progressed into renal failure at the age of 16 to 17 years. Around the age of 30 years, both patients manifested multiple hypertrophy of the trunk, plexus, and root in the cervical, brachial, lumbosacral nerves, and cauda equina. The histology of the cervical mass in patient 2 revealed Schwannoma. Exome analysis showed that patient 2 harbors a germline LZTR1 p.Arg68Gly variant, while patient 1 has no schwannomatosis-related mutations. Conclusions: Peripheral neuropathy caused by INF2 variants may lead to the development of multifocal hypertrophy with age, likely due to the initial demyelination and subsequent Schwann cell proliferation. Schwannoma could co-occur when the tissues attain additional hits in schwannomatosis-related genes (e.g., LZTR1).
Collapse
Affiliation(s)
- Quynh Tran Thuy Huong
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
| | - Linh Tran Nguyen Truc
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
- Department of Internal Medicine, Pham Ngoc Thach University of Medicine, Ho Chi Minh 70000, Vietnam
| | - Hiroko Ueda
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
| | - Kenji Fukui
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Japan
| | - Koichiro Higasa
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Japan
| | - Yoshinori Sato
- Department of Medicine, Division of Nephrology, Showa University School of Medicine, Fujigaoka Hospital, Yokohama 227-8501, Japan
| | - Shinichi Takeda
- Internal Medicine, Kurobe City Hospital, Toyama 938-8502, Japan
| | - Motoshi Hattori
- Department of Pediatric Nephrology, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | - Hiroyasu Tsukaguchi
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
- Clinical Genetics Center, Kansai Medical University Hospital, Hirakata 573-1010, Japan
| |
Collapse
|
24
|
Lai Y, Zhang T, Huang L, Klymchenko AS, Lin W. An exchangeable SIM probe for monitoring organellar dynamics of necrosis cells and intracellular water heterogeneity in kidney repair. Proc Natl Acad Sci U S A 2025; 122:e2402348121. [PMID: 39752520 PMCID: PMC11725781 DOI: 10.1073/pnas.2402348121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/25/2024] [Indexed: 01/15/2025] Open
Abstract
Monitoring subcellular organelle dynamics in real time and precisely assessing membrane heterogeneity in living cells are very important for studying fundamental biological mechanisms and gaining a comprehensive understanding of cellular processes. However, there remains a shortage of effective tools for these purposes. Herein, we propose a strategy to develop the exchangeable water-sensing probeAPBD for time-lapse imaging of dynamics in cellular membrane-bound organelle morphology with structured illumination microscopy at the nanoscale. In this work, our results reveal mitochondria as the first organelle to undergo morphological changes through swelling, fission, and fusion in cell necrosis, leading to the rupture of the endoplasmic reticulum (ER) sheet adhered to the mitochondria. Meanwhile, the ER tubules are then reconstructed by stretching and fusion of autophagosomes. Moreover, APBD allows us to directly visualize spatially resolved distribution of biomembranes vs. water inside single mammalian cells. Our findings show that the renal ischemia-reperfusion injury (IRI) model results in the increased biomembrane to cytoplasmic water ratio in the tissue. This reveals intracellular water heterogeneity between the nucleus and the cytoplasm during the IRI process. Overall, this study presents a strategy for development of the molecular tools for cellular water heterogeneity and organelle dynamics.
Collapse
Affiliation(s)
- Youbo Lai
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning530004, Guangxi, People's Republic of China
| | - Tengteng Zhang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning530004, Guangxi, People's Republic of China
| | - Ling Huang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning530004, Guangxi, People's Republic of China
| | - Andrey S. Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch 67401, France
| | - Weiying Lin
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning530004, Guangxi, People's Republic of China
| |
Collapse
|
25
|
Gatti P, Schiavon C, Cicero J, Manor U, Germain M. Mitochondria- and ER-associated actin are required for mitochondrial fusion. Nat Commun 2025; 16:451. [PMID: 39774009 PMCID: PMC11707194 DOI: 10.1038/s41467-024-55758-x] [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: 08/15/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Mitochondria are crucial for cellular metabolism and signalling. Mitochondrial activity is modulated by mitochondrial fission and fusion, which are required to properly balance metabolic functions, transfer material between mitochondria, and remove defective mitochondria. Mitochondrial fission occurs at mitochondria-endoplasmic reticulum (ER) contact sites, and requires the formation of actin filaments that drive mitochondrial constriction and the recruitment of the fission protein DRP1. The role of actin in mitochondrial fusion remains entirely unexplored. Here we show that preventing actin polymerisation on either mitochondria or the ER disrupts both fission and fusion. We show that fusion but not fission is dependent on Arp2/3, whereas both fission and fusion require INF2 formin-dependent actin polymerization. We also show that mitochondria-associated actin marks fusion sites prior to the fusion protein MFN2. Together, our work introduces a method for perturbing organelle-associated actin and demonstrates a previously unknown role for actin in mitochondrial fusion.
Collapse
Affiliation(s)
- Priya Gatti
- Groupe de Recherche en Signalisation Cellulaire and Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada
- Réseau Intersectoriel de Recherche en Santé de l'Université du Québec (RISUQ), Québec, Canada
| | - Cara Schiavon
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Julien Cicero
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Uri Manor
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - Marc Germain
- Groupe de Recherche en Signalisation Cellulaire and Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada.
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada.
- Réseau Intersectoriel de Recherche en Santé de l'Université du Québec (RISUQ), Québec, Canada.
| |
Collapse
|
26
|
Roterman I, Slupina M, Konieczny L. Protein folding: Funnel model revised. Comput Struct Biotechnol J 2024; 23:3827-3838. [PMID: 39525086 PMCID: PMC11550765 DOI: 10.1016/j.csbj.2024.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
The spatial structure of proteins, largely determined by their amino acid sequences, is also dependent on the environmental conditions under which the folding process takes place. In aqueous environments, exposure of polar amino acids is the driving factor, whereas protein stabilization in amphipathic membranes requires exposure to hydrophobic residues. This observation can be extended to all other environmental conditions under which proteins exhibit biological activity and, most importantly, to the folding process. The fuzzy oil drop (FOD) model assumes a centric location of hydrophobic residues (hydrophobic core) with exposure of polar residues towards the aqueous environment, as the influence of the aqueous environment is extended to include the contribution of other non-aqueous factors, enabling the assessment of their influence on protein structuring. The application of the modified FOD model (FOD-M) we have developed allows the environment to be represented as an external force field in the form of a continuum. The role of environmental conditions allows modification of the funnel model expressing the localization of the energy minimum as dependent on external conditions expressed by the K scale, where K measures the degree of other than polar water factors participating in folding process.
Collapse
Affiliation(s)
- Irena Roterman
- Department of Bioinformatics and Telemedicine, Jagiellonian University Medical College, Medyczna 7, 30-688 Kraków, Poland
| | - Mateusz Slupina
- Department of Bioinformatics and Telemedicine, Jagiellonian University Medical College, Medyczna 7, 30-688 Kraków, Poland
| | - Leszek Konieczny
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kopernika 7, 31-034 Kraków, Poland
| |
Collapse
|
27
|
Labat-de-Hoz L, Jiménez MÁ, Correas I, Alonso MA. Regulation of formin INF2 and its alteration in INF2-linked inherited disorders. Cell Mol Life Sci 2024; 81:463. [PMID: 39586895 PMCID: PMC11589041 DOI: 10.1007/s00018-024-05499-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/21/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
Formins are proteins that catalyze the formation of linear filaments made of actin. INF2, a formin, is crucial for correct vesicular transport, microtubule stability and mitochondrial division. Its activity is regulated by a complex of cyclase-associated protein and lysine-acetylated G-actin (KAc-actin), which helps INF2 adopt an inactive conformation through the association of its N-terminal diaphanous inhibitory domain (DID) with its C-terminal diaphanous autoinhibitory domain. INF2 activation can occur through calmodulin binding, KAc-actin deacetylation, G-actin binding, or association with the Cdc42 GTPase. Mutations in the INF2 DID are linked to focal segmental glomerulosclerosis (FSGS), affecting podocytes, and Charcot-Marie-Tooth disease, which affects Schwann cells and leads to axonal loss. At least 80 pathogenic DID variants of INF2 have been identified, with potential for many more. These mutations disrupt INF2 regulation, leading to excessive actin polymerization. This in turn causes altered intracellular trafficking, abnormal mitochondrial dynamics, and profound transcriptional reprogramming via the MRTF/SRF complex, resulting in mitotic abnormalities and p53-mediated cell death. This sequence of events could be responsible for progressive podocyte loss during glomerular degeneration in FSGS patients. Pharmacological targeting of INF2 or actin polymerization could offer the therapeutic potential to halt the progression of FSGS and improve outcomes for patients with INF2-linked disease.
Collapse
Affiliation(s)
- Leticia Labat-de-Hoz
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049, Madrid, Spain
| | - M Ángeles Jiménez
- Instituto de Química Física (IQF) Blas Cabrera, Consejo Superior de Investigaciones Científicas, 28006, Madrid, Spain
| | - Isabel Correas
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049, Madrid, Spain
- Department of Molecular Biology, UAM, 28049, Madrid, Spain
| | - Miguel A Alonso
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049, Madrid, Spain.
| |
Collapse
|
28
|
Hofstadter WA, Park JW, Lum KK, Chen S, Cristea IM. HCMV strain- and cell type-specific alterations in membrane contact sites point to the convergent regulation of organelle remodeling. J Virol 2024; 98:e0109924. [PMID: 39480111 PMCID: PMC11575408 DOI: 10.1128/jvi.01099-24] [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: 06/22/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Viruses are ubiquitous entities that infect organisms across the kingdoms of life. While viruses can infect a range of cells, tissues, and organisms, this aspect is often not explored in cell culture analyses. There is limited information about which infection-induced changes are shared or distinct in different cellular environments. The prevalent pathogen human cytomegalovirus (HCMV) remodels the structure and function of subcellular organelles and their interconnected networks formed by membrane contact sites (MCSs). A large portion of this knowledge has been derived from fibroblasts infected with a lab-adapted HCMV strain. Here, we assess strain- and cell type-specific alterations in MCSs and organelle remodeling induced by HCMV. Integrating quantitative mass spectrometry, super-resolution microscopy, and molecular virology assays, we compare infections with lab-adapted and low-passage HCMV strains in fibroblast and epithelial cells. We determine that, despite baseline proteome disparities between uninfected fibroblast and epithelial cells, infection induces convergent changes and is remarkably similar. We show that hallmarks of HCMV infection in fibroblasts, mitochondria-endoplasmic reticulum (ER) encapsulations and peroxisome proliferation, are also conserved in infected epithelial and macrophage-like cells. Exploring cell type-specific differences, we demonstrate that fibroblasts rely on endosomal cholesterol transport while epithelial cells rely on cholesterol from the Golgi. Despite these mechanistic differences, infections in both cell types result in phenotypically similar cholesterol accumulation at the viral assembly complex. Our findings highlight the adaptability of HCMV, in that infections can be tailored to the initial cell state by inducing both shared and unique proteome alterations, ultimately promoting a unified pro-viral environment.IMPORTANCEHuman cytomegalovirus (HCMV) establishes infections in diverse cell types throughout the body and is connected to a litany of diseases associated with each of these tissues. However, it is still not fully understood how HCMV replication varies in distinct cell types. Here, we compare HCMV replication with lab-adapted and low-passage strains in two primary sites of infection, lung fibroblasts and retinal epithelial cells. We discover that, despite displaying disparate protein compositions prior to infection, these cell types undergo convergent alterations upon HCMV infection, reaching a more similar cellular state late in infection. We find that remodeling of the subcellular landscape is a pervasive feature of HCMV infection, through alterations to both organelle structure-function and the interconnected networks they form via membrane contact sites. Our findings show how HCMV infection in different cell types induces both shared and divergent changes to cellular processes, ultimately leading to a more unified state.
Collapse
Affiliation(s)
| | - Ji Woo Park
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Krystal K. Lum
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Sophia Chen
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
29
|
Kors S, Schlaitz AL. Dynamic remodelling of the endoplasmic reticulum for mitosis. J Cell Sci 2024; 137:jcs261444. [PMID: 39584405 DOI: 10.1242/jcs.261444] [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: 11/26/2024] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic and continuous membrane network with roles in many cellular processes. The importance and maintenance of ER structure and function have been extensively studied in interphase cells, yet recent findings also indicate crucial roles of the ER in mitosis. During mitosis, the ER is remodelled significantly with respect to composition and morphology but persists as a continuous network. The ER interacts with microtubules, actin and intermediate filaments, and concomitant with the mitotic restructuring of all cytoskeletal systems, ER dynamics and distribution change. The ER is a metabolic hub and several examples of altered ER functions during mitosis have been described. However, we lack an overall understanding of the ER metabolic pathways and functions that are active during mitosis. In this Review, we will discuss mitotic changes to the ER at different organizational levels to explore how the mitotic ER, with its distinct properties, might support cell division.
Collapse
Affiliation(s)
- Suzan Kors
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany
| | - Anne-Lore Schlaitz
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany
| |
Collapse
|
30
|
Subramanian B, Williams S, Karp S, Hennino MF, Jacas S, Lee M, Riella CV, Alper SL, Higgs HN, Pollak MR. INF2 mutations cause kidney disease through a gain-of-function mechanism. SCIENCE ADVANCES 2024; 10:eadr1017. [PMID: 39536114 PMCID: PMC11559609 DOI: 10.1126/sciadv.adr1017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Heterozygosity for inverted formin-2 (INF2) mutations causes focal segmental glomerulosclerosis (FSGS) with or without Charcot-Marie-Tooth disease. A key question is whether the disease is caused by gain-of-function effects on INF2 or loss of function (haploinsufficiency). Despite established roles in multiple cellular processes, neither INF2 knockout mice nor mice with a disease-associated point mutation display an evident kidney or neurologic phenotype. Here, we compared responses to puromycin aminonucleoside (PAN)-induced kidney injury between INF2 R218Q and INF2 knockout mice. R218Q INF2 mice are susceptible to glomerular disease, in contrast to INF2 knockout mice. Colocalization, coimmunoprecipitation analyses, and cellular actin measurements showed that INF2 R218Q confers a gain-of-function effect on the actin cytoskeleton. RNA expression analysis showed that adhesion and mitochondria-related pathways were enriched in the PAN-treated R218Q mice. Both podocytes from INF2 R218Q mice and human kidney organoids with an INF2 mutation (S186P) recapitulate adhesion and mitochondrial phenotypes. Thus, gain-of-function mechanisms drive INF2-related FSGS and explain this disease's autosomal dominant inheritance.
Collapse
Affiliation(s)
- Balajikarthick Subramanian
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Kidney Bioengineering Resource Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sarah Williams
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sophie Karp
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Marie-Flore Hennino
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sonako Jacas
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Miriam Lee
- Department of Biochemistry, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Cristian V. Riella
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Seth L. Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Henry N. Higgs
- Department of Biochemistry, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Martin R. Pollak
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Kidney Bioengineering Resource Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
31
|
García Casas P, Rossini M, Påvénius L, Saeed M, Arnst N, Sonda S, Fernandes T, D'Arsiè I, Bruzzone M, Berno V, Raimondi A, Sassano ML, Naia L, Barbieri E, Sigismund S, Agostinis P, Sturlese M, Niemeyer BA, Brismar H, Ankarcrona M, Gautier A, Pizzo P, Filadi R. Simultaneous detection of membrane contact dynamics and associated Ca 2+ signals by reversible chemogenetic reporters. Nat Commun 2024; 15:9775. [PMID: 39532847 PMCID: PMC11557831 DOI: 10.1038/s41467-024-52985-0] [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: 02/01/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
Membrane contact sites (MCSs) are hubs allowing various cell organelles to coordinate their activities. The dynamic nature of these sites and their small size hinder analysis by current imaging techniques. To overcome these limitations, we here design a series of reversible chemogenetic reporters incorporating improved, low-affinity variants of splitFAST, and study the dynamics of different MCSs at high spatiotemporal resolution, both in vitro and in vivo. We demonstrate that these versatile reporters suit different experimental setups well, allowing one to address challenging biological questions. Using these probes, we identify a pathway in which calcium (Ca2+) signalling dynamically regulates endoplasmic reticulum-mitochondria juxtaposition, characterizing the underlying mechanism. Finally, by integrating Ca2+-sensing capabilities into the splitFAST technology, we introduce PRINCESS (PRobe for INterorganelle Ca2+-Exchange Sites based on SplitFAST), a class of reporters to simultaneously detect MCSs and measure the associated Ca2+ dynamics using a single biosensor.
Collapse
Affiliation(s)
- Paloma García Casas
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Medicine, Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), University of Valladolid and CSIC, Valladolid, Spain
| | - Michela Rossini
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Institute of Neuroscience, National Research Council (CNR), Padua, Italy
| | - Linnea Påvénius
- Science for Life Laboratory,, Karolinska Institutet, Stockholm, Sweden
| | - Mezida Saeed
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Nikita Arnst
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Sonia Sonda
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Institute of Neuroscience, National Research Council (CNR), Padua, Italy
| | - Tânia Fernandes
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Irene D'Arsiè
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Institute of Neuroscience, National Research Council (CNR), Padua, Italy
| | - Matteo Bruzzone
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Valeria Berno
- ALEMBIC, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Raimondi
- ALEMBIC, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, Institute for Research in Biomedicine, CH-6500, Bellinzona, Switzerland
| | - Maria Livia Sassano
- Cell Death Research and Therapy lab, Department of Cellular and Molecular Medicine, and Center for Cancer Biology-VIB, KU Leuven, Leuven, Belgium
| | - Luana Naia
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | | | - Sara Sigismund
- IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Patrizia Agostinis
- Cell Death Research and Therapy lab, Department of Cellular and Molecular Medicine, and Center for Cancer Biology-VIB, KU Leuven, Leuven, Belgium
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Hjalmar Brismar
- Science for Life Laboratory,, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Maria Ankarcrona
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Arnaud Gautier
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Laboratoire des Biomolécules, LBM, 75005, Paris, France
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
- Institute of Neuroscience, National Research Council (CNR), Padua, Italy.
- Centro Studi per la Neurodegenerazione (CESNE), University of Padua, Padua, Italy.
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
- Institute of Neuroscience, National Research Council (CNR), Padua, Italy.
| |
Collapse
|
32
|
Chen L, Hao J, Zhang J, Wu J, Ren Z. Rosiglitazone-induced white adipocyte browning is regulated by actin and Myh9. Life Sci 2024; 359:123217. [PMID: 39510170 DOI: 10.1016/j.lfs.2024.123217] [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: 08/03/2024] [Revised: 10/22/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
AIMS This study investigates the role of actin polymerization and Myh9 in mediating lipid droplet (LD) fission during rosiglitazone-induced browning of white adipocytes. The aim is to understand how LD splitting might contribute to the beige conversion of white adipose tissue, providing insights into adipocyte plasticity and metabolic regulation. MATERIALS AND METHODS C3H10 T1/2-differentiated adipocytes were used as a classical model to study white adipocyte browning. Rosiglitazone was applied to induce browning, and the interactions between LDs and actin, as well as the distribution of Myh9, were assessed using immunofluorescence and Western blotting. In vivo, we employed a microfilament inhibitor to block actin polymerization in cold-stimulated mice and evaluated changes in LD morphology and browning. Furthermore, dynamic live-cell imaging using confocal microscopy was conducted to observe the real-time behavior of LDs during the browning process and to determine whether they undergo fission. MAIN FINDINGS Our results demonstrate that rosiglitazone significantly induces LD size reduction, a process correlated with the increased contact of LDs with microfilaments. Inhibition of actin polymerization prevented both the reduction in LD size and the browning of white adipocytes, indicating that actin plays a critical role. Myh9 was enriched at the LD fission sites, forming a structure resembling a contractile ring. Overexpression of Myh9 promoted the shrinkage of LD, suggesting that it may be involved in LD fission. SIGNIFICANCE This study identifies actin and Myh9 as key regulators of LD fission in rosiglitazone-induced browning of white adipocytes, offering new insights into the cellular mechanisms of adipocyte plasticity. The findings propose a novel pathway by which LD dynamics contribute to the beige conversion of white fat, with potential implications for metabolic disease therapies targeting adipocyte function and energy expenditure.
Collapse
Affiliation(s)
- Lupeng Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jingjie Hao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Junzhi Zhang
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China.
| |
Collapse
|
33
|
Zhu HW, Wang YP, Zhang QF, Wang KD, Huang Y, Xiang RL. F-actin/DRP1 axis-mediated mitochondrial fission promotes mitophagy in diabetic submandibular glands. Oral Dis 2024; 30:5429-5444. [PMID: 38735833 DOI: 10.1111/odi.14983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Diabetes is accompanied by a high prevalence of hyposalivation, causing severe damage to oral and systemic health. Mitochondrial dynamics play important roles in the pathogenesis of various diabetic complications; however, little is known about their roles in diabetic hyposalivation. MATERIALS AND METHODS A diabetic mouse model and a high glucose (HG)-induced diabetic submandibular gland (SMG) cell model were employed. RESULTS More mitochondria surrounded by autophagosomes and higher expression of mitophagy-related proteins were detected in the SMGs of diabetic mice and HG-treated SMG cells. In diabetic SMGs, dynamin-related protein 1 (DRP1) was upregulated, whereas mitofusin-2 was downregulated both in vivo and in vitro. Shortened mitochondria and impaired mitochondrial functions were observed in the HG group. A DRP1-specific inhibitor, mdivi-1, suppressed mitochondrial fission and mitophagy, as well as restored mitochondrial functions in the HG condition. Moreover, the interaction of F-actin and DRP1 was enhanced in the diabetic group. Inhibiting F-actin with cytochalasin D repaired the injured effects of HG on mitochondrial dynamics and functions. Conversely, the F-actin-polymerization-inducer jasplakinolide aggravated mitochondrial fission and dysfunction. CONCLUSIONS F-actin contributes to HG-evoked mitochondrial fission by interacting with DRP1, which induces mitophagy and impairs mitochondrial function in SMG cells, ultimately damaging the SMG.
Collapse
Affiliation(s)
- Hou-Wei Zhu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomotalogical Disease Diagnosis and Treatment, Xiamen, China
| | - Yi-Ping Wang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomotalogical Disease Diagnosis and Treatment, Xiamen, China
| | - Qiu-Fang Zhang
- Xiamen Key Laboratory of Stomotalogical Disease Diagnosis and Treatment, Stomatological Hospital of Xiamen Medical College, Xiamen, China
| | - Kai-Di Wang
- Xiamen Key Laboratory of Stomotalogical Disease Diagnosis and Treatment, Stomatological Hospital of Xiamen Medical College, Xiamen, China
| | - Yan Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomotalogical Disease Diagnosis and Treatment, Xiamen, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
34
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
35
|
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
|
36
|
Landoni JC, Kleele T, Winter J, Stepp W, Manley S. Mitochondrial Structure, Dynamics, and Physiology: Light Microscopy to Disentangle the Network. Annu Rev Cell Dev Biol 2024; 40:219-240. [PMID: 38976811 DOI: 10.1146/annurev-cellbio-111822-114733] [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] [Indexed: 07/10/2024]
Abstract
Mitochondria serve as energetic and signaling hubs of the cell: This function results from the complex interplay between their structure, function, dynamics, interactions, and molecular organization. The ability to observe and quantify these properties often represents the puzzle piece critical for deciphering the mechanisms behind mitochondrial function and dysfunction. Fluorescence microscopy addresses this critical need and has become increasingly powerful with the advent of superresolution methods and context-sensitive fluorescent probes. In this review, we delve into advanced light microscopy methods and analyses for studying mitochondrial ultrastructure, dynamics, and physiology, and highlight notable discoveries they enabled.
Collapse
Affiliation(s)
- Juan C Landoni
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Tatjana Kleele
- Institute of Biochemistry, Swiss Federal Institute of Technology Zürich (ETH), Zürich, Switzerland;
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Julius Winter
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Willi Stepp
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Suliana Manley
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| |
Collapse
|
37
|
Guo L. F-ATP synthase inhibitory factor 1 and mitochondria-organelle interactions: New insight and implications. Pharmacol Res 2024; 208:107393. [PMID: 39233058 DOI: 10.1016/j.phrs.2024.107393] [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: 05/25/2024] [Revised: 08/08/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitochondria are metabolic hub, and act as primary sites for reactive oxygen species (ROS) and metabolites generation. Mitochondrial Ca2+ uptake contributes to Ca2+ storage. Mitochondria-organelle interactions are important for cellular metabolic adaptation, biosynthesis, redox balance, cell fate. Organelle communications are mediated by Ca2+/ROS signals, vesicle transport and membrane contact sites. The permeability transition pore (PTP) is an unselective channel that provides a release pathway for Ca2+/ROS, mtDNA and metabolites. F-ATP synthase inhibitory factor 1 (IF1) participates in regulation of PTP opening and is required for the translocation of transcriptional factors c-Myc/PGC1α to mitochondria to stimulate metabolic switch. IF1, a mitochondrial specific protein, has been suggested to regulate other organelles including nucleus, endoplasmic reticulum and lysosomes. IF1 may be able to mediate mitochondria-organelle interactions and cellular physiology through regulation of PTP activity.
Collapse
Affiliation(s)
- Lishu Guo
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Anesthesiology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
38
|
Luo JS, Zhai WH, Ding LL, Zhang XJ, Han J, Ning JQ, Chen XM, Jiang WC, Yan RY, Chen MJ. MAMs and Mitochondrial Quality Control: Overview and Their Role in Alzheimer's Disease. Neurochem Res 2024; 49:2682-2698. [PMID: 39002091 DOI: 10.1007/s11064-024-04205-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) represents the most widespread neurodegenerative disorder, distinguished by a gradual onset and slow progression, presenting a substantial challenge to global public health. The mitochondrial-associated membrane (MAMs) functions as a crucial center for signal transduction and material transport between mitochondria and the endoplasmic reticulum, playing a pivotal role in various pathological mechanisms of AD. The dysregulation of mitochondrial quality control systems is considered a fundamental factor in the development of AD, leading to mitochondrial dysfunction and subsequent neurodegenerative events. Recent studies have emphasized the role of MAMs in regulating mitochondrial quality control. This review will delve into the molecular mechanisms underlying the imbalance in mitochondrial quality control in AD and provide a comprehensive overview of the role of MAMs in regulating mitochondrial quality control.
Collapse
Affiliation(s)
- Jian-Sheng Luo
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Wen-Hu Zhai
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Ling-Ling Ding
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Xian-Jie Zhang
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Jia Han
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Jia-Qi Ning
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Xue-Meng Chen
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Wen-Cai Jiang
- Department of Anesthesiology, Deyang People's Hospital, Deyang, 618000, China
| | - Ru-Yu Yan
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Meng-Jie Chen
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| |
Collapse
|
39
|
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
|
40
|
Tran QTH, Kondo N, Ueda H, Matsuo Y, Tsukaguchi H. Altered Endoplasmic Reticulum Integrity and Organelle Interactions in Living Cells Expressing INF2 Variants. Int J Mol Sci 2024; 25:9783. [PMID: 39337270 PMCID: PMC11431639 DOI: 10.3390/ijms25189783] [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: 07/26/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The cytoskeleton mediates fundamental cellular processes by organizing inter-organelle interactions. Pathogenic variants of inverted formin 2 (INF2) CAAX isoform, an actin assembly factor that is predominantly expressed in the endoplasmic reticulum (ER), are linked to focal segmental glomerulosclerosis (FSGS) and Charcot-Marie-Tooth (CMT) neuropathy. To investigate how pathogenic INF2 variants alter ER integrity, we used high-resolution live imaging of HeLa cells. Cells expressing wild-type (WT) INF2 showed a predominant tubular ER with perinuclear clustering. Cells expressing INF2 FSGS variants that cause mild and intermediate disease induced more sheet-like ER, a pattern similar to that seen for cells expressing WT-INF2 that were treated with actin and microtubule (MT) inhibitors. Dual CMT-FSGS INF2 variants led to more severe ER dysmorphism, with a diffuse, fragmented ER and coarse INF2 aggregates. Proper organization of both F-actin and MT was needed to modulate the tubule vs. sheet conformation balance, while MT arrays regulated spatial expansion of tubular ER in the cell periphery. Pathogenic INF2 variants also induced mitochondria fragmentation and dysregulated mitochondria distribution. Such mitochondrial abnormalities were more prominent for cells expressing CMT-FSGS compared to those with FSGS variants, indicating that the severity of the dysfunction is linked to the degree of cytoskeletal disorganization. Our observations suggest that pathogenic INF2 variants disrupt ER continuity by altering interactions between the ER and the cytoskeleton that in turn impairs inter-organelle communication, especially at ER-mitochondria contact sites. ER continuity defects may be a common disease mechanism involved in both peripheral neuropathy and glomerulopathy.
Collapse
Affiliation(s)
- Quynh Thuy Huong Tran
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
| | - Naoyuki Kondo
- Department of Molecular Genetics, Institute of Biochemical Science, Kansai Medical University, Hirakata 573-1010, Japan
| | - Hiroko Ueda
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
| | - Yoshiyuki Matsuo
- Central Research Center, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Japan
| | - Hiroyasu Tsukaguchi
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Hirakata 573-1010, Japan
- Clinical Genetics Center, Kansai Medical University Hospital, Hirakata 573-1191, Japan
| |
Collapse
|
41
|
Wong HTC, Lang AE, Stein C, Drerup CM. ALS-Linked VapB P56S Mutation Alters Neuronal Mitochondrial Turnover at the Synapse. J Neurosci 2024; 44:e0879242024. [PMID: 39054069 PMCID: PMC11358610 DOI: 10.1523/jneurosci.0879-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Mitochondrial population maintenance in neurons is essential for neuron function and survival. Contact sites between mitochondria and the endoplasmic reticulum (ER) are poised to regulate mitochondrial homeostasis in neurons. These contact sites can facilitate transfer of calcium and lipids between the organelles and have been shown to regulate aspects of mitochondrial dynamics. Vesicle-associated membrane protein-associated protein B (VapB) is an ER membrane protein present at a subset of ER-mitochondrial contact sites. A proline-to-serine mutation in VapB at amino acid 56 (P56S) correlates with susceptibility to amyotrophic lateral sclerosis (ALS) type 8. Given the relationship between failed mitochondrial health and neurodegenerative disease, we investigated the function of VapB in mitochondrial population maintenance. We demonstrated that transgenic expression of VapBP56S in zebrafish larvae (sex undetermined) increased mitochondrial biogenesis, causing increased mitochondrial population size in the axon terminal. Expression of wild-type VapB did not alter biogenesis but, instead, increased mitophagy in the axon terminal. Using genetic manipulations to independently increase mitochondrial biogenesis, we show that biogenesis is normally balanced by mitophagy to maintain a constant mitochondrial population size. VapBP56S transgenics fail to increase mitophagy to compensate for the increase in mitochondrial biogenesis, suggesting an impaired mitophagic response. Finally, using a synthetic ER-mitochondrial tether, we show that VapB's function in mitochondrial turnover is likely independent of ER-mitochondrial tethering by contact sites. Our findings demonstrate that VapB can control mitochondrial turnover in the axon terminal, and this function is altered by the P56S ALS-linked mutation.
Collapse
Affiliation(s)
- Hiu-Tung C Wong
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Angelica E Lang
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Chris Stein
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Catherine M Drerup
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
42
|
Hofstadter WA, Cook KC, Tsopurashvili E, Gebauer R, Pražák V, Machala EA, Park JW, Grünewald K, Quemin ERJ, Cristea IM. Infection-induced peripheral mitochondria fission drives ER encapsulations and inter-mitochondria contacts that rescue bioenergetics. Nat Commun 2024; 15:7352. [PMID: 39187492 PMCID: PMC11347691 DOI: 10.1038/s41467-024-51680-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
The dynamic regulation of mitochondria shape via fission and fusion is critical for cellular responses to stimuli. In homeostatic cells, two modes of mitochondrial fission, midzone and peripheral, provide a decision fork between either proliferation or clearance of mitochondria. However, the relationship between specific mitochondria shapes and functions remains unclear in many biological contexts. While commonly associated with decreased bioenergetics, fragmented mitochondria paradoxically exhibit elevated respiration in several disease states, including infection with the prevalent pathogen human cytomegalovirus (HCMV) and metastatic melanoma. Here, incorporating super-resolution microscopy with mass spectrometry and metabolic assays, we use HCMV infection to establish a molecular mechanism for maintaining respiration within a fragmented mitochondria population. We establish that HCMV induces fragmentation through peripheral mitochondrial fission coupled with suppression of mitochondria fusion. Unlike uninfected cells, the progeny of peripheral fission enter mitochondria-ER encapsulations (MENCs) where they are protected from degradation and bioenergetically stabilized during infection. MENCs also stabilize pro-viral inter-mitochondria contacts (IMCs), which electrochemically link mitochondria and promote respiration. Demonstrating a broader relevance, we show that the fragmented mitochondria within metastatic melanoma cells also form MENCs. Our findings establish a mechanism where mitochondria fragmentation can promote increased respiration, a feature relevant in the context of human diseases.
Collapse
Affiliation(s)
| | - Katelyn C Cook
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - Robert Gebauer
- Department of Chemistry, MIN Faculty, Universität Hamburg, Hamburg, Germany
- Department of Structural Cell Biology of Viruses, Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
| | - Vojtěch Pražák
- Department of Chemistry, MIN Faculty, Universität Hamburg, Hamburg, Germany
- Department of Structural Cell Biology of Viruses, Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
| | - Emily A Machala
- Department of Chemistry, MIN Faculty, Universität Hamburg, Hamburg, Germany
- Department of Structural Cell Biology of Viruses, Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
| | - Ji Woo Park
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Kay Grünewald
- Department of Chemistry, MIN Faculty, Universität Hamburg, Hamburg, Germany
- Department of Structural Cell Biology of Viruses, Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
| | - Emmanuelle R J Quemin
- Department of Chemistry, MIN Faculty, Universität Hamburg, Hamburg, Germany
- Department of Structural Cell Biology of Viruses, Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
- Department of Virology, Institute for Integrative Biology of the Cell, CNRS UMR9198, Gif-sur-Yvette, France
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
43
|
Pokharel MD, Fu P, Garcia-Flores A, Yegambaram M, Lu Q, Sun X, Unwalla H, Aggarwal S, Fineman JR, Wang T, Black SM. Inflammatory lung injury is associated with endothelial cell mitochondrial fission and requires the nitration of RhoA and cytoskeletal remodeling. Free Radic Biol Med 2024; 221:125-135. [PMID: 38734269 PMCID: PMC11179967 DOI: 10.1016/j.freeradbiomed.2024.05.019] [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: 02/20/2024] [Revised: 04/12/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024]
Abstract
Higher levels of extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a TLR4 agonist, are associated with poor clinical outcomes in sepsis-induced acute lung injury (ALI). Little is known regarding the mechanisms by which eNAMPT is involved in ALI. Our recent work has identified a crucial role for mitochondrial dysfunction in ALI. Thus, this study aimed to determine if eNAMPT-mediated inflammatory injury is associated with the loss of mitochondrial function. Our data show that eNAMPT disrupted mitochondrial bioenergetics. This was associated with cytoskeleton remodeling and the loss of endothelial barrier integrity. These changes were associated with enhanced mitochondrial fission and blocked when Rho-kinase (ROCK) was inhibited. The increases in mitochondrial fission were also associated with the nitration-mediated activation of the small GTPase activator of ROCK, RhoA. Blocking RhoA nitration decreased eNAMPT-mediated mitochondrial fission and endothelial barrier dysfunction. The increase in fission was linked to a RhoA-ROCK mediated increase in Drp1 (dynamin-related protein 1) at serine(S)616. Another TLR4 agonist, lipopolysaccharide (LPS), also increased mitochondrial fission in a Drp1 and RhoA-ROCK-dependent manner. To validate our findings in vivo, we challenged C57BL/6 mice with eNAMPT in the presence and absence of the Drp1 inhibitor, Mdivi-1. Mdivi-1 treatment protected against eNAMPT-induced lung inflammation, edema, and lung injury. These studies demonstrate that mitochondrial fission-dependent disruption of mitochondrial function is essential in TLR4-mediated inflammatory lung injury and identify a key role for RhoA-ROCK signaling. Reducing mitochondrial fission could be a potential therapeutic strategy to improve ARDS outcomes.
Collapse
Affiliation(s)
- Marissa D Pokharel
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA; Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | | | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Hoshang Unwalla
- Department of Immunology and Nano-Medicine, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Saurabh Aggarwal
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; Department of Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Stephen M Black
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA; Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA.
| |
Collapse
|
44
|
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
|
45
|
Liu D, Qin H, Gao Y, Sun M, Wang M. Cardiovascular disease: Mitochondrial dynamics and mitophagy crosstalk mechanisms with novel programmed cell death and macrophage polarisation. Pharmacol Res 2024; 206:107258. [PMID: 38909638 DOI: 10.1016/j.phrs.2024.107258] [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: 02/20/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024]
Abstract
Several cardiovascular illnesses are associated with aberrant activation of cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis, and macrophage polarisation as hallmarks contributing to vascular damage and abnormal cardiac function. Meanwhile, these three novel forms of cellular dysfunction are closely related to mitochondrial homeostasis. Mitochondria are the main organelles that supply energy and maintain cellular homeostasis. Mitochondrial stability is maintained through a series of regulatory pathways, such as mitochondrial fission, mitochondrial fusion and mitophagy. Studies have shown that mitochondrial dysfunction (e.g., impaired mitochondrial dynamics and mitophagy) promotes ROS production, leading to oxidative stress, which induces cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage M1 phenotypic polarisation. Therefore, an in-depth knowledge of the dynamic regulation of mitochondria during cellular pyroptosis, ferroptosis, necroptosis, cuproptosis, disulfidptosis and macrophage polarisation is necessary to understand cardiovascular disease development. This paper systematically summarises the impact of changes in mitochondrial dynamics and mitophagy on regulating novel cellular dysfunctions and macrophage polarisation to promote an in-depth understanding of the pathogenesis of cardiovascular diseases and provide corresponding theoretical references for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Dandan Liu
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Hewei Qin
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China; Department of Rehabilitation Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 450002, China.
| | - Yang Gao
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengyan Sun
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Mengnan Wang
- School of Rehabilitation Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
46
|
Read TA, Cisterna BA, Skruber K, Ahmadieh S, Liu TM, Vitriol JA, Shi Y, Black JB, Butler MT, Lindamood HL, Lefebvre AE, Cherezova A, Ilatovskaya DV, Bear JE, Weintraub NL, Vitriol EA. The actin binding protein profilin 1 localizes inside mitochondria and is critical for their function. EMBO Rep 2024; 25:3240-3262. [PMID: 39026010 PMCID: PMC11316047 DOI: 10.1038/s44319-024-00209-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 06/16/2024] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The monomer-binding protein profilin 1 (PFN1) plays a crucial role in actin polymerization. However, mutations in PFN1 are also linked to hereditary amyotrophic lateral sclerosis, resulting in a broad range of cellular pathologies which cannot be explained by its primary function as a cytosolic actin assembly factor. This implies that there are important, undiscovered roles for PFN1 in cellular physiology. Here we screened knockout cells for novel phenotypes associated with PFN1 loss of function and discovered that mitophagy was significantly upregulated. Indeed, despite successful autophagosome formation, fusion with the lysosome, and activation of additional mitochondrial quality control pathways, PFN1 knockout cells accumulate depolarized, dysmorphic mitochondria with altered metabolic properties. Surprisingly, we also discovered that PFN1 is present inside mitochondria and provide evidence that mitochondrial defects associated with PFN1 loss are not caused by reduced actin polymerization in the cytosol. These findings suggest a previously unrecognized role for PFN1 in maintaining mitochondrial integrity and highlight new pathogenic mechanisms that can result from PFN1 dysregulation.
Collapse
Affiliation(s)
- Tracy-Ann Read
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Bruno A Cisterna
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Kristen Skruber
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Samah Ahmadieh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Tatiana M Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Josefine A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Yang Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Population Health Sciences, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Joseph B Black
- Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Halli L Lindamood
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | - Alena Cherezova
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
47
|
Froehlich T, Jenner A, Cavarischia-Rega C, Fagbadebo FO, Lurz Y, Frecot DI, Kaiser PD, Nueske S, Scholz AM, Schäffer E, Garcia-Saez AJ, Macek B, Rothbauer U. Nanobodies as novel tools to monitor the mitochondrial fission factor Drp1. Life Sci Alliance 2024; 7:e202402608. [PMID: 38816213 PMCID: PMC11140114 DOI: 10.26508/lsa.202402608] [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: 01/22/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
In cells, mitochondria undergo constant fusion and fission. An essential factor for fission is the mammalian dynamin-related protein 1 (Drp1). Dysregulation of Drp1 is associated with neurodegenerative diseases including Parkinson's, cardiovascular diseases and cancer, making Drp1 a pivotal biomarker for monitoring mitochondrial status and potential pathophysiological conditions. Here, we developed nanobodies (Nbs) as versatile binding molecules for proteomics, advanced microscopy and live cell imaging of Drp1. To specifically enrich endogenous Drp1 with interacting proteins for proteomics, we functionalized high-affinity Nbs into advanced capture matrices. Furthermore, we detected Drp1 by bivalent Nbs combined with site-directed fluorophore labelling in super-resolution STORM microscopy. For real-time imaging of Drp1, we intracellularly expressed fluorescently labelled Nbs, so-called chromobodies (Cbs). To improve the signal-to-noise ratio, we further converted Cbs into a "turnover-accelerated" format. With these imaging probes, we visualized the dynamics of endogenous Drp1 upon compound-induced mitochondrial fission in living cells. Considering the wide range of research applications, the presented Nb toolset will open up new possibilities for advanced functional studies of Drp1 in disease-relevant models.
Collapse
Affiliation(s)
- Theresa Froehlich
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Jenner
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Claudia Cavarischia-Rega
- Quantitative Proteomics, Department of Biology, Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | - Yannic Lurz
- Center for Plant Molecular Biology (ZMBP), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Desiree I Frecot
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Philipp D Kaiser
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Stefan Nueske
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Armin M Scholz
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Erik Schäffer
- Center for Plant Molecular Biology (ZMBP), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ana J Garcia-Saez
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Boris Macek
- Quantitative Proteomics, Department of Biology, Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| |
Collapse
|
48
|
Elezaby A, Lin AJ, Vijayan V, Pokhrel S, Kraemer BR, Bechara LRG, Larus I, Sun J, Baena V, Syed ZA, Murphy E, Glancy B, Ostberg NP, Queliconi BB, Campos JC, Ferreira JCB, Haileselassie B, Mochly-Rosen D. Cardiac troponin I directly binds and inhibits mitochondrial ATP synthase with a noncanonical role in the post-ischemic heart. NATURE CARDIOVASCULAR RESEARCH 2024; 3:987-1002. [PMID: 39196031 PMCID: PMC11700703 DOI: 10.1038/s44161-024-00512-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/21/2024] [Indexed: 08/29/2024]
Abstract
Cardiac troponin I (cTnI) is a key regulator of cardiomyocyte contraction. However, its role in mitochondria is unknown. Here we show that cTnI localized to mitochondria in the heart, inhibited mitochondrial functions when stably expressed in noncardiac cells and increased the opening of the mitochondrial permeability transition pore under oxidative stress. Direct, specific and saturable binding of cTnI to F1FO-ATP synthase was demonstrated in vitro using immune-captured ATP synthase and in cells using proximity ligation assay. cTnI binding doubled ATPase activity, whereas skeletal troponin I and several human pathogenic cTnI variants associated with familial hypertrophic cardiomyopathy did not. A rationally designed peptide, P888, inhibited cTnI binding to ATP synthase, inhibited cTnI-induced increase in ATPase activity in vitro and reduced cardiac injury following transient ischemia in vivo. We suggest that cTnI-bound ATP synthase results in lower ATP levels, and releasing this interaction during cardiac ischemia-reperfusion may increase the reservoir of functional mitochondria to reduce cardiac injury.
Collapse
Affiliation(s)
- Aly Elezaby
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Amanda J Lin
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Vijith Vijayan
- Department of Pediatrics, Division of Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Suman Pokhrel
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin R Kraemer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Luiz R G Bechara
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Isabel Larus
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Junhui Sun
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Valentina Baena
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zulfeqhar A Syed
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth Murphy
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brian Glancy
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- National Institute of Arthritis, Musculoskeletal, and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicolai P Ostberg
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bruno B Queliconi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Juliane C Campos
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Julio C B Ferreira
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Bereketeab Haileselassie
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Division of Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
49
|
Palmer NJ, Barrie KR, Dominguez R. Mechanisms of actin filament severing and elongation by formins. Nature 2024; 632:437-442. [PMID: 38843827 PMCID: PMC11375965 DOI: 10.1038/s41586-024-07637-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/31/2024] [Indexed: 08/02/2024]
Abstract
Humans express 15 formins that play crucial roles in actin-based processes, including cytokinesis, cell motility and mechanotransduction1,2. However, the lack of structures bound to the actin filament (F-actin) has been a major impediment to understanding formin function. Whereas formins are known for their ability to nucleate and elongate F-actin3-7, some formins can additionally depolymerize, sever or bundle F-actin. Two mammalian formins, inverted formin 2 (INF2) and diaphanous 1 (DIA1, encoded by DIAPH1), exemplify this diversity. INF2 shows potent severing activity but elongates weakly8-11 whereas DIA1 has potent elongation activity but does not sever4,8. Using cryo-electron microscopy (cryo-EM) we show five structural states of INF2 and two of DIA1 bound to the middle and barbed end of F-actin. INF2 and DIA1 bind differently to these sites, consistent with their distinct activities. The formin-homology 2 and Wiskott-Aldrich syndrome protein-homology 2 (FH2 and WH2, respectively) domains of INF2 are positioned to sever F-actin, whereas DIA1 appears unsuited for severing. These structures also show how profilin-actin is delivered to the fast-growing barbed end, and how this is followed by a transition of the incoming monomer into the F-actin conformation and the release of profilin. Combined, the seven structures presented here provide step-by-step visualization of the mechanisms of F-actin severing and elongation by formins.
Collapse
Affiliation(s)
- Nicholas J Palmer
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle R Barrie
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Roberto Dominguez
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Pilic J, Gottschalk B, Bourgeois B, Habisch H, Koshenov Z, Oflaz FE, Erdogan YC, Miri SM, Yiğit EN, Aydın MŞ, Öztürk G, Eroglu E, Shoshan-Barmatz V, Madl T, Graier WF, Malli R. Hexokinase 1 forms rings that regulate mitochondrial fission during energy stress. Mol Cell 2024; 84:2732-2746.e5. [PMID: 38981483 DOI: 10.1016/j.molcel.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/30/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024]
Abstract
Metabolic enzymes can adapt during energy stress, but the consequences of these adaptations remain understudied. Here, we discovered that hexokinase 1 (HK1), a key glycolytic enzyme, forms rings around mitochondria during energy stress. These HK1-rings constrict mitochondria at contact sites with the endoplasmic reticulum (ER) and mitochondrial dynamics protein (MiD51). HK1-rings prevent mitochondrial fission by displacing the dynamin-related protein 1 (Drp1) from mitochondrial fission factor (Mff) and mitochondrial fission 1 protein (Fis1). The disassembly of HK1-rings during energy restoration correlated with mitochondrial fission. Mechanistically, we identified that the lack of ATP and glucose-6-phosphate (G6P) promotes the formation of HK1-rings. Mutations that affect the formation of HK1-rings showed that HK1-rings rewire cellular metabolism toward increased TCA cycle activity. Our findings highlight that HK1 is an energy stress sensor that regulates the shape, connectivity, and metabolic activity of mitochondria. Thus, the formation of HK1-rings may affect mitochondrial function in energy-stress-related pathologies.
Collapse
Affiliation(s)
- Johannes Pilic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Benjamin Bourgeois
- BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria; Otto Loewi Research Center, Medical Chemistry, Medical University of Graz, 8010 Graz, Austria
| | - Hansjörg Habisch
- Otto Loewi Research Center, Medical Chemistry, Medical University of Graz, 8010 Graz, Austria
| | - Zhanat Koshenov
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Furkan E Oflaz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Yusuf C Erdogan
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria
| | - Seyed M Miri
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye; Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, 34956 Istanbul, Türkiye
| | - Esra N Yiğit
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye; Department of Physiology, International School of Medicine, Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Mehmet Ş Aydın
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Emrah Eroglu
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Türkiye; Department of Physiology, International School of Medicine, Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Varda Shoshan-Barmatz
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Tobias Madl
- BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria; Otto Loewi Research Center, Medical Chemistry, Medical University of Graz, 8010 Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria; BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, 8010 Graz, Austria; BioTechMed Graz, Mozartgasse 12/2, 8010 Graz, Austria; Center for Medical Research, CF Bioimaging, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria.
| |
Collapse
|