1
|
Mori K, Togo A, Yamashita K, Sakuragi S, Bannai H, Umezawa T, Ohta K, Asahi T, Nozaki C, Kataoka K. Mitochondrial damage and ER stress in CB1 receptor antagonist-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neuropharmacology 2025; 273:110440. [PMID: 40185361 DOI: 10.1016/j.neuropharm.2025.110440] [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: 01/03/2025] [Revised: 03/13/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Cannabinoid receptor type 1 (CB1R) is the key modulator of neuronal viability. CB1R antagonists provide neuroprotective effects on neurotoxicity caused by e.g. neuronal injury. However, the underlying mechanisms and potential limitations of CB1R antagonism remain unclear. Here we investigated the impact of environmental conditions on CB1R antagonist effects. We have found that cell-permeable CB1R antagonists, rimonabant and AM251, induced cell death in human neuroblastoma SH-SY5Y cells under serum-free conditions. Mitochondrial morphological analysis revealed mitochondrial swelling characterized by their network fragmentation and cristae reduction. Phosphoproteomics analysis showed the ER stress signaling pathway PERK/eIF2α/ATF4/CHOP, leading to caspase-dependent apoptosis. These results suggest that CB1R antagonists promote apoptosis via mitochondrial damage and ER stress under serum-free conditions in SH-SY5Y cells. Our findings indicate that while CB1R antagonists may be neuroprotective in certain conditions, they may also pose a neurotoxic risk in environments characterized by cellular stress or nutrient deprivation.
Collapse
Affiliation(s)
- Kazuaki Mori
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Kota Yamashita
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Shigeo Sakuragi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Hiroko Bannai
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Taishi Umezawa
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Keisuke Ohta
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Toru Asahi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan
| | - Chihiro Nozaki
- Global Center for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Kosuke Kataoka
- Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan; Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan.
| |
Collapse
|
2
|
Proust A, Wilkinson KA, Wilkinson RJ. Effects of M. tuberculosis and HIV-1 infection on in vitro blood-brain barrier function. J Neuroinflammation 2025; 22:141. [PMID: 40420159 PMCID: PMC12107840 DOI: 10.1186/s12974-025-03467-7] [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: 01/13/2025] [Accepted: 05/11/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Tuberculous meningitis is the most severe form of tuberculosis and HIV-1 co-infection worsens the already poor prognosis. However, how Mycobacterium tuberculosis crosses the blood-brain barrier and how HIV-1 influences tuberculous meningitis pathogenesis remains unclear. METHODS Using human pericytes, astrocytes, endothelial cells, and microglia alone and combined in an in vitro blood-brain barrier model, we investigated the effect of Mycobacterium tuberculosis +/- HIV-1 co-infection on central nervous system cell entry and function. Cells and the blood-brain barrier model were infected with Mycobacterium tuberculosis and/or HIV-1 and we evaluated the effects of both infection on (i) cells susceptibility to Mycobacterium tuberculosis and its growth in cells by flow cytometry; (ii) modulation of blood-brain barrier permeability and Mycobacterium tuberculosis passage through it; (iii) viral and bacterial cytopathogenicity using the xCELLigence system; (iv) cell metabolic activity and ROS release using colorimetric assays; (v) extracellular glutamate concentration by fluorometric assay; (vi) the inflammatory response by Luminex; and (vii) endoplasmic reticulum stress by quantitative PCR. RESULTS We demonstrated that Mycobacterium tuberculosis infects and multiplies in all cell types with HIV-1 increasing entry to astrocytes and pericytes, and growth in HIV-1 positive pericytes and endothelial cells. Mycobacterium tuberculosis also induces an increase of the blood-brain barrier permeability resulting in translocation of bacilli across it. Cytopathic effects include (i) increased markers of cellular stress (mitochondrial metabolic activity, unfolded protein response); (ii) ROS release; (iii) the induction of neurotoxic astrocytes; (iv) and the secretion of the excitotoxic neurotransmitter glutamate. Lastly, we observed distinct cell-type specific production of inflammatory and effector mediators. CONCLUSION These results indicate that Mycobacterium tuberculosis can translocate the blood-brain barrier directly to initiate meningitis.
Collapse
Affiliation(s)
- Alizé Proust
- The Francis Crick Institute, Midland Road, London, NW1 1AT, UK.
| | - Katalin A Wilkinson
- The Francis Crick Institute, Midland Road, London, NW1 1AT, UK
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, Department of Medicine, University of Cape Town, Observatory, 7925, Republic of South Africa
| | - Robert J Wilkinson
- The Francis Crick Institute, Midland Road, London, NW1 1AT, UK
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, Department of Medicine, University of Cape Town, Observatory, 7925, Republic of South Africa
- Department of Infectious Diseases, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
3
|
Marotta G, Osti D, Zaccheroni E, Costanza B, Faletti S, Marinaro A, Richichi C, Mesa D, Rodighiero S, Soriani C, Migliaccio E, Ruscitto F, Priami C, Sigismund S, Manetti F, Polli D, Beznusenko GV, Rusu MC, Favero F, Corà D, Silvestris DA, Gallo A, Gambino V, Alfieri F, Gandini S, Schmitt MJ, Gargiulo G, Noberini R, Bonaldi T, Pelicci G. Metabolic traits shape responses to LSD1-directed therapy in glioblastoma tumor-initiating cells. SCIENCE ADVANCES 2025; 11:eadt2724. [PMID: 40408499 DOI: 10.1126/sciadv.adt2724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/17/2025] [Indexed: 05/25/2025]
Abstract
Lysine-specific histone demethylase 1A (LSD1) is an epigenetic regulator involved in various biological processes, including metabolic pathways. We demonstrated the therapeutic potential of its pharmacological inhibition in glioblastoma using DDP_38003 (LSD1i), which selectively targets tumor-initiating cells (TICs) by hampering their adaptability to stress. Through biological, metabolic, and omic approaches, we now show that LSD1i acts as an endoplasmic reticulum (ER) stressor, activating the integrated stress response and altering mitochondrial structure and function. These effects impair TICs' oxidative metabolism and generate reactive oxygen species, further amplifying cellular stress. LSD1i also impairs TICs' glycolytic activity, causing their metabolic decline. TICs with enhanced glycolysis benefit from LSD1-directed therapy. Conversely, metabolically silent TICs mantain ER and mitochondrial homeostasis, adapting to stress conditions, including LSD1i treatment. A dropout short hairpin RNA screening identifies postglycosylphosphatidylinositol attachment to proteins inositol deacylase 1 (PGAP1) as a mediator of resistance to LSD1i. Disruptions in ER and mitochondrial balance holds promise for improving LSD1-targeted therapy efficacy and overcoming treatment resistance.
Collapse
Affiliation(s)
- Giulia Marotta
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Daniela Osti
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Elena Zaccheroni
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Brunella Costanza
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Stefania Faletti
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Human Technopole, Viale Rita Levi-Montalcini, 1, Milan 20157, Italy
| | - Adriana Marinaro
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Cristina Richichi
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Deborah Mesa
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Simona Rodighiero
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Chiara Soriani
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Enrica Migliaccio
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Federica Ruscitto
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Chiara Priami
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Sara Sigismund
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan Italy
| | | | - Dario Polli
- Department of Physics, Politecnico di Milano, Milan, Italy
- CNR Institute for Photonics and Nanotechnology (CNR-IFN), Milan, Italy
| | | | - Mara-Camelia Rusu
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Francesco Favero
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara 28100, Italy
| | - Davide Corà
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara 28100, Italy
| | - Domenico A Silvestris
- Unit of Genetics and Epigenetic of Pediatric Cancer, Oncohaematology Department, IRCCS Ospedale Pediatrico Bambino Gesù, Viale di San Paolo 15, Rome 00146, Italy
| | - Angela Gallo
- Unit of Genetics and Epigenetic of Pediatric Cancer, Oncohaematology Department, IRCCS Ospedale Pediatrico Bambino Gesù, Viale di San Paolo 15, Rome 00146, Italy
| | - Valentina Gambino
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Fabio Alfieri
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Matthias J Schmitt
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Gaetano Gargiulo
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Roberta Noberini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Tiziana Bonaldi
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy
| |
Collapse
|
4
|
Geng J, Yuan Q, Ni C, Zhang Y, Liu X, Zhu X, Hao X, Liang G, Wang D, Fan H. Ca 2+ transfer via enhancing ER-Mito coupling contributed to BDE-47- induced hippocampal neuronal necroptosis and cognitive dysfunction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118396. [PMID: 40412245 DOI: 10.1016/j.ecoenv.2025.118396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/11/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
2,2,4,4-Tetrabromodiphenyl ether (BDE-47), a ubiquitous environmental pollutant, has gained increasing attention due to its high level in biological samples and potential neurotoxicity. Recent studies have indicated that the receptor interacting protein kinase 1 (RIPK1)-mediated necroptosis is implicated in BDE-47 cytotoxicity. However, little is known about the underlying mechanism and whether the necroptosis participates in BDE-47-induced neuronal injury and cognitive impairment. Our results indicated that exposure to BDE-47 triggered RIPK1-dependent neuronal necroptosis in mice hippocampi and HT-22 mouse hippocampal neurons. Necrostain-1 (Nec-1), a specific RIPK1 inhibitor, suppressed the RIPK1/RIPK3/mixed lineage kinase-like domain protein (MLKL) signaling and rescued neuronal survival in BDE-47-treated HT-22 neurons. Mechanically, increased mitochondrial Ca2+ influx precipitated the opening of the mitochondrial permeability transition pore (mPTP), leading to occurrence of hippocampal neuronal necroptosis under BDE-47 stress. BDE-47 exposure induced excessive mitochondria-associated endoplasmic reticulum membranes (MAMs) formation and promoted ER-to-mitochondria Ca2+ transfer, while diminishing ER-mitochondrial contacts by Glucose-regulated protein 75 (Grp75)-deficiency remarkably prevented mitochondria Ca2+ overload and opening of mPTP as well as neuronal necroptosis. Notably, Nec-1 pre-treatment could substantially mitigate neuronal/synaptic damage and cognitive impairment in BDE-47-exposed mice. Collectively, these data suggest that BDE-47 exposure intensified endoplasmic reticulum (ER)-mitochondrial (Mito) contact and facilitated Ca2+ transfer from ER towards mitochondria, resulting in mPTP opening-mediated hippocampal neuronal necroptosis and subsequent cognitive dysfunction. Our study shed new light on the mechanisms underlying BDE-47 neurotoxicity and provided a novel therapeutic strategy through targeting RIPK1 kinase activity.
Collapse
Affiliation(s)
- Junhong Geng
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Quan Yuan
- Henan Province Rongkang Hospital, Luoyang, China
| | - Chaofang Ni
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Yarong Zhang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoli Liu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoying Zhu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xueqin Hao
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Gaofeng Liang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Dongmei Wang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| | - Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
5
|
Dong L, Zhang G, Shen Z, Hong X, Xing Y, Wu Y, Yang W, Zhang B, Shi Z. Degradation of WE43 Magnesium Alloy in Vivo and Its Degradation Products on Macrophages. ACS OMEGA 2025; 10:17280-17295. [PMID: 40352546 PMCID: PMC12059945 DOI: 10.1021/acsomega.4c09349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/14/2025]
Abstract
Due to their biocompatibility, biodegradability, and suitable mechanical properties, magnesium-based biodegradable implants are emerging as a promising alternative to traditional metal implants. The Mg-4Y-3RE (WE43) biodegradable alloy is among the most extensively studied and widely utilized magnesium alloys in clinical applications. As an absorbable and degradable metallic material, magnesium alloys undergo gradual degradation, wear, and fracture within the body. These alloys reduce the long-term risks associated with permanent implants but generate insoluble byproducts that accumulate in surrounding tissues. Following the implantation of magnesium alloys, granulation tissue and fibrous encapsulation typically form around the material. However, limited research has addressed the interaction between insoluble byproducts of magnesium alloys and macrophages. This study focused on the biological effects of macrophages during the second stage of the host inflammatory response in the degradation process of magnesium alloy. Using subcutaneous implantation of WE43 magnesium alloy sheets, observations were made regarding the degradation components, morphological changes in surrounding tissues, and the biological effects of macrophages upon phagocytosis of insoluble byproducts. The primary degradation products of WE43 in vivo were identified as Ca3 (PO4)2, Mg3(PO4)2, Na3PO4, NaCa (PO4), MgSO4, MgCO3, NaCl, Mg24Y5, and Mg12YNd. Postimplantation, levels of IL-1β and IL-18 in adjacent tissues significantly increased (p < 0.05). By 8 weeks, compared to nitinol alloy, significant thickening of the fibrous capsule (p < 0.05) was observed, accompanied by substantial inflammatory cell infiltration, vascularization, and the presence of macrophages and multinucleated giant cells. Macrophages were observed extending pseudopodia to enclose and phagocytose particles, forming phagosomes and creating a relatively isolated microenvironment around the engulfed substances, where further particle degradation occurred. Following the phagocytosis of degradation products, macrophages exhibited increased lysosome numbers, mitochondrial swelling and damage, phagolysosome formation, and autophagosome development. Furthermore, the degradation products were observed to induce elevated reactive oxygen species (ROS) production in macrophages, activation of P2X7 receptors, enhanced IL-6 secretion, endoplasmic reticulum stress, autophagy, and activation of the NLRP3 inflammasome pathway. This study provides novel insights and contributes a theoretical foundation for a more comprehensive understanding of magnesium alloy degradation in vivo.
Collapse
Affiliation(s)
- Li Dong
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Guangde Zhang
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Zhiyuan Shen
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Xiaojian Hong
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Yongli Xing
- Department
of Medical Imaging, Second Hospital of Harbin, Harbin 150056, China
| | - Yue Wu
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Wei Yang
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Binmei Zhang
- Department
of Cardiology, The Fourth Affiliated Hospital
of Harbin Medical University, Harbin 150001, China
| | - Zhiyu Shi
- Department
of Cardiology, The First Affiliated Hospital
of Harbin Medical University, Harbin 150007, China
| |
Collapse
|
6
|
Diokmetzidou A, Scorrano L. Mitochondria-membranous organelle contacts at a glance. J Cell Sci 2025; 138:jcs263895. [PMID: 40357586 DOI: 10.1242/jcs.263895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Mitochondrial contact sites are specialized interfaces where mitochondria physically interact with other organelles. Stabilized by molecular tethers and defined by unique proteomic and lipidomic profiles, these sites enable direct interorganellar communication and functional coordination, playing crucial roles in cellular physiology and homeostasis. Recent advances have expanded our knowledge of contact site-resident proteins, illuminated the dynamic and adaptive nature of these interfaces, and clarified their contribution to pathophysiology. In this Cell Science at a Glance article and the accompanying poster, we summarize the mitochondrial contacts that have been characterized in mammals, the molecular mechanisms underlying their formation, and their principal functions.
Collapse
Affiliation(s)
- Antigoni Diokmetzidou
- Department of Biology, University of Padova, 35121 Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Luca Scorrano
- Department of Biology, University of Padova, 35121 Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| |
Collapse
|
7
|
Chen B, Lyssiotis CA, Shah YM. Mitochondria-organelle crosstalk in establishing compartmentalized metabolic homeostasis. Mol Cell 2025; 85:1487-1508. [PMID: 40250411 DOI: 10.1016/j.molcel.2025.03.003] [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: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 04/20/2025]
Abstract
Mitochondria serve as central hubs in cellular metabolism by sensing, integrating, and responding to metabolic demands. This integrative function is achieved through inter-organellar communication, involving the exchange of metabolites, lipids, and signaling molecules. The functional diversity of metabolite exchange and pathway interactions is enabled by compartmentalization within organelle membranes. Membrane contact sites (MCSs) are critical for facilitating mitochondria-organelle communication, creating specialized microdomains that enhance the efficiency of metabolite and lipid exchange. MCS dynamics, regulated by tethering proteins, adapt to changing cellular conditions. Dysregulation of mitochondrial-organelle interactions at MCSs is increasingly recognized as a contributing factor in the pathogenesis of multiple diseases. Emerging technologies, such as advanced microscopy, biosensors, chemical-biology tools, and functional genomics, are revolutionizing our understanding of inter-organellar communication. These approaches provide novel insights into the role of these interactions in both normal cellular physiology and disease states. This review will highlight the roles of metabolite transporters, lipid-transfer proteins, and mitochondria-organelle interfaces in the coordination of metabolism and transport.
Collapse
Affiliation(s)
- Brandon Chen
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| | - Yatrik M Shah
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
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
|
9
|
Mikołajczyk-Korona A, Dziedzic R, Wójcik K, Olchawa M, Sarna T, Pięta J, Jakiela B, Zaręba L, Bazan JG, Potaczek DP, Kosałka-Węgiel J, Socha M, Kuszmiersz P, Padjas A, Bazan-Socha S. Enhanced systemic oxidative stress response in patients with idiopathic inflammatory myopathies. Arthritis Res Ther 2025; 27:50. [PMID: 40055731 PMCID: PMC11887258 DOI: 10.1186/s13075-025-03511-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/18/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Idiopathic inflammatory myopathies (IIM) are characterized by chronic inflammation, endothelial dysfunction, and muscle tissue mitochondrial defect, leading to the local oxidative stress response. However, data on its systemic intensity and correlation with IIM clinical and laboratory characteristics remains scarce. METHODS In clinically stable dermatomyositis (n = 18) and myositis (n = 38) patients and matched controls (n = 50), we measured global oxidative stress response in peripheral blood using a novel coumarin boronic acid (CBA) assay enabling real-time detection of protein hydroperoxides (HP) formed in serum. RESULTS We documented 36% faster kinetics (p < 0.001) and a 68% increase in the cumulative (p = 0.003) fluorescent product generation in the IIM group compared to the control, which indicates higher HP formation associated with systemic oxidative stress. The dynamics of fluorescent product growth were similar in the dermatomyositis and myositis groups. Interestingly, myositis patients with a marked increase in HP formation were characterized by lower serum myoglobin levels (p = 0.038). There was also an inverse correlation between serum myoglobin and the kinetics of HP formation (e.g., for cumulative in-time generation r = -0.35, p = 0.03). The systemic oxidative stress response measures were not related to clinical characteristics of the disease and treatment, internal medicine comorbidities, smoking status, or autoantibody profile. CONCLUSIONS IIM are characterized by a global pro-oxidant imbalance reflected by enhanced HP generation in serum. Furthermore, muscle weakening without active signs of muscle damage may be related to the increased local and systemic oxidative stress response, suggesting non-inflammatory pathomechanism of the disease that our technically undemanding assay may evaluate.
Collapse
Affiliation(s)
- Anna Mikołajczyk-Korona
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-668, Poland
| | - Radosław Dziedzic
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, św. Łazarza 16, Kraków, 31-530, Poland
| | - Krzysztof Wójcik
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-668, Poland
| | - Magdalena Olchawa
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland
| | - Tadeusz Sarna
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Kraków, 30-387, Poland
| | - Jakub Pięta
- Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, Łódź, 90-924, Poland
| | - Bogdan Jakiela
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-668, Poland
| | - Lech Zaręba
- Institute of Computer Science, University of Rzeszów, Pigonia 1, Rzeszów, 35-310, Poland
| | - Jan G Bazan
- Institute of Computer Science, University of Rzeszów, Pigonia 1, Rzeszów, 35-310, Poland
| | - Daniel P Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Philipps-University Marburg, Marburg, 35043, Germany
- Bioscientia MVZ Labor Mittelhessen GmbH, Gießen, 35394, Germany
| | - Joanna Kosałka-Węgiel
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Mateusz Socha
- Students' Scientific Group of Immune Diseases and Hypercoagulation, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-668, Poland
| | - Piotr Kuszmiersz
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Agnieszka Padjas
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-668, Poland
| | - Stanisława Bazan-Socha
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-668, Poland.
| |
Collapse
|
10
|
Sánchez-Mendoza LM, González-Reyes JA, Rodríguez-López S, Calvo-Rubio M, Calero-Rodríguez P, de Cabo R, Burón MI, Villalba JM. Sex-dependent adaptations in heart mitochondria from transgenic mice overexpressing cytochrome b 5 reductase-3. Mitochondrion 2025; 81:102004. [PMID: 39793940 PMCID: PMC11875916 DOI: 10.1016/j.mito.2025.102004] [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: 09/25/2024] [Revised: 12/16/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
Cytochrome b5 reductase 3 (CYB5R3) overexpression upregulates mitochondrial biogenesis, function, and abundance in skeletal muscle and kidneys, and mimics some of the salutary effects of calorie restriction, with the most striking effects being observed in females. We aimed to investigate the mitochondrial adaptations prompted by CYB5R3 overexpression in the heart, an organ surprisingly overlooked in studies focused on this long-lived transgenic model despite the critical role played by CYB5R3 in supporting cardiomyocytes mitochondrial respiration. Given that CYB5R3 effects have been found to be sex-dependent, we focused our research on both males and females. CYB5R3 was efficiently overexpressed in cardiac tissue from transgenic mice, without any difference between sexes. The abundance of electron transport chain complexes markers and cytochrome c was higher in males than in females. CYB5R3 overexpression downregulated the levels of complexes markers in males but not females, without decreasing oxygen consumption capacity. CYB5R3 increased the size and abundance of cardiomyocytes mitochondria, and reduced thickness and preserved the length of mitochondria-endoplasmic reticulum contact sites in heart from males but not females. Metabolic changes were also highlighted in transgenic mice, with an upregulation of fatty acid oxidation markers, particularly in males. Our results support that CYB5R3 overexpression upregulates markers consistent with enhanced mitochondrial function in the heart, producing most of these actions in males, with illustrates the complexity of the CYB5R3-overexpressing transgenic model.
Collapse
Affiliation(s)
- Luz Marina Sánchez-Mendoza
- Departamento de Biología Celular Fisiología e Inmunología Universidad de Córdoba Campus de Excelencia Internacional Agroalimentario ceiA3 Córdoba Spain.
| | - José A González-Reyes
- Departamento de Biología Celular Fisiología e Inmunología Universidad de Córdoba Campus de Excelencia Internacional Agroalimentario ceiA3 Córdoba Spain.
| | - Sandra Rodríguez-López
- Departamento de Biología Celular Fisiología e Inmunología Universidad de Córdoba Campus de Excelencia Internacional Agroalimentario ceiA3 Córdoba Spain.
| | - Miguel Calvo-Rubio
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Pilar Calero-Rodríguez
- Departamento de Biología Celular Fisiología e Inmunología Universidad de Córdoba Campus de Excelencia Internacional Agroalimentario ceiA3 Córdoba Spain.
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - M Isabel Burón
- Departamento de Biología Celular Fisiología e Inmunología Universidad de Córdoba Campus de Excelencia Internacional Agroalimentario ceiA3 Córdoba Spain.
| | - José M Villalba
- Departamento de Biología Celular Fisiología e Inmunología Universidad de Córdoba Campus de Excelencia Internacional Agroalimentario ceiA3 Córdoba Spain.
| |
Collapse
|
11
|
Zellmer JC, Tarantino MB, Kim M, Lomoio S, Maesako M, Hajnóczky G, Bhattacharyya R. Stabilization of mitochondria-associated endoplasmic reticulum membranes regulates Aβ generation in a three-dimensional neural model of Alzheimer's disease. Alzheimers Dement 2025; 21:e14417. [PMID: 39713841 PMCID: PMC11848173 DOI: 10.1002/alz.14417] [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: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/21/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION We previously demonstrated that regulating mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) affects axonal Aβ generation in a well-characterized three-dimensional (3D) neural Alzheimer's disease (AD) model. MAMs vary in thickness and length, impacting their functions. Here, we examined the effect of MAM thickness on Aβ in our 3D neural model of AD. METHODS We employed fluorescence resonance energy transfer (FRET) or fluorescence-based MAM stabilizers, electron microscopy, Aβ enzyme-linked immunosorbent assay (ELISA), and live-cell imaging with kymography to assess how stabilizing MAMs of different gap widths influence Aβ production and MAM axonal mobility. RESULTS Stabilizing tight MAMs (∼6 nm gap width) significantly increased Aβ levels, whereas basal (∼25 nm) and loose MAMs (∼40 nm) maintained or reduced Aβ levels, respectively. Tight MAMs reduced mitochondrial axonal velocity compared to basal MAMs, while loose MAMs showed severely reduced axonal distribution. DISCUSSION Our findings suggest that stabilizing MAMs of specific gap widths, particularly in axons, without complete destabilization could be an effective therapeutic strategy for AD. HIGHLIGHTS The stabilization of MAMs exacerbates or ameliorates Aβ generation from AD neurons in a MAM gap width-dependent manner. A specific stabilization threshold within the MAM gap width spectrum shifts the amyloidogenic process to non-amyloidogenic. Tight MAMs slow down mitochondrial axonal transport compared to lose MAMs offering a quantitative method for measuring MAM stabilization.
Collapse
Affiliation(s)
- Jacob C. Zellmer
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Marina B. Tarantino
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Michelle Kim
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Selene Lomoio
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Masato Maesako
- Alzheimer's Disease Research UnitMassGeneral Institute for Neurodegenerative DiseaseMassachusetts General Hospital/Harvard Medical SchoolCharlestownMassachusettsUSA
| | - György Hajnóczky
- MitoCare CenterDepartment of PathologyAnatomy & Cell BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Raja Bhattacharyya
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| |
Collapse
|
12
|
Le HT, Yu J, Ahn HS, Kim MJ, Chae IG, Cho HN, Kim J, Park HK, Kwon HN, Chae HJ, Kang BH, Seo JK, Kim K, Back SH. eIF2α phosphorylation-ATF4 axis-mediated transcriptional reprogramming mitigates mitochondrial impairment during ER stress. Mol Cells 2025; 48:100176. [PMID: 39756584 PMCID: PMC11786836 DOI: 10.1016/j.mocell.2024.100176] [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: 10/03/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025] Open
Abstract
Eukaryotic translation initiation factor 2α (eIF2α) phosphorylation, which regulates all 3 unfolded protein response pathways, helps maintain cellular homeostasis and overcome endoplasmic reticulum (ER) stress through transcriptional and translational reprogramming. However, transcriptional regulation of mitochondrial homeostasis by eIF2α phosphorylation during ER stress is not fully understood. Here, we report that the eIF2α phosphorylation-activating transcription factor 4 (ATF4) axis is required for the expression of multiple transcription factors, including nuclear factor erythroid 2-related factor 2 and its target genes responsible for mitochondrial homeostasis during ER stress. eIF2α phosphorylation-deficient (A/A) cells displayed dysregulated mitochondrial dynamics and mitochondrial DNA replication, decreased expression of oxidative phosphorylation complex proteins, and impaired mitochondrial functions during ER stress. ATF4 overexpression suppressed impairment of mitochondrial homeostasis in A/A cells during ER stress by promoting the expression of downstream transcription factors and their target genes. Our findings underscore the importance of the eIF2α phosphorylation-ATF4 axis for maintaining mitochondrial homeostasis through transcriptional reprogramming during ER stress.
Collapse
Affiliation(s)
- Hien Thi Le
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Jiyoung Yu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Hee Sung Ahn
- AMC Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Mi-Jeong Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - In Gyeong Chae
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Hyun-Nam Cho
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Juhee Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Hye-Kyung Park
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Hyuk Nam Kwon
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Han-Jung Chae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Korea
| | - Byoung Heon Kang
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Jeong Kon Seo
- Central Research Facilities (UCRF), Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea.
| | - Kyunggon Kim
- Department of Digital Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
| | - Sung Hoon Back
- Basic-Clinical Convergence Research Center, School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea.
| |
Collapse
|
13
|
Parker HN, Haberman KL, Ojo T, Watkins J, Nambiar A, Morales K, Zechmann B, Taube JH. Twist-Induced Epithelial-to-Mesenchymal Transition Confers Specific Metabolic and Mitochondrial Alterations. Cells 2025; 14:80. [PMID: 39851508 PMCID: PMC11763985 DOI: 10.3390/cells14020080] [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: 08/22/2024] [Revised: 11/19/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Cells undergo significant epigenetic and phenotypic change during the epithelial-to-mesenchymal transition (EMT), a process observed in development, wound healing, and cancer metastasis. EMT confers several advantageous characteristics, including enhanced migration and invasion, resistance to cell death, and altered metabolism. In disease, these adaptations could be leveraged as therapeutic targets. Here, we analyze Twist-induced EMT in non-transformed HMLE cells as well as a breast cancer cell line with (MDA-MB-231) and without (MCF7) EMT features to compare differences in metabolic pathways and mitochondrial morphology. Analysis of oxidative and glycolytic metabolism reveals a general EMT-associated glycolytic metabolic phenotype accompanied by increased ATP production. Furthermore, a decrease in mitochondrial size was also associated with EMT-positive cells. However, mitochondrial elongation and spatial dynamics were not consistently altered, as HMLE Twist cells exhibit more rounded and dispersed mitochondria compared to control, while MDA-MB-231 cells exhibit more elongated and clustered mitochondria compared to MCF7 cells. These results provide further insight as to the contextual nature of EMT conferred properties.
Collapse
Affiliation(s)
- Haleigh N. Parker
- Department of Biology, Baylor University, Waco, TX 76798, USA; (H.N.P.)
| | - Kayla L. Haberman
- Department of Biology, Baylor University, Waco, TX 76798, USA; (H.N.P.)
| | - Tolulope Ojo
- Department of Biology, Baylor University, Waco, TX 76798, USA; (H.N.P.)
| | - Juli Watkins
- Department of Biology, Baylor University, Waco, TX 76798, USA; (H.N.P.)
| | - Adhwaitha Nambiar
- Department of Biology, Baylor University, Waco, TX 76798, USA; (H.N.P.)
| | - Kayla Morales
- Department of Biology, Baylor University, Waco, TX 76798, USA; (H.N.P.)
| | - Bernd Zechmann
- Center for Microscopy and Imaging, Baylor University, Waco, TX 76798, USA
| | - Joseph H. Taube
- Department of Biology, Baylor University, Waco, TX 76798, USA; (H.N.P.)
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
14
|
Ho WT, Chang JS, Lei CJ, Chen TC, Wang JK, Chang SW, Yang MH, Jou TS, Wang IJ. ROCK Inhibitor Enhances Resilience Against Metabolic Stress Through Increasing Bioenergetic Capacity in Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2025; 66:51. [PMID: 39847368 PMCID: PMC11759620 DOI: 10.1167/iovs.66.1.51] [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: 07/09/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose To investigate the effect of Rho-associated protein kinase (ROCK) inhibitor Y27632 on bioenergetic capacity and resilience of corneal endothelial cells (CECs) under metabolic stress. Methods Bovine CECs (BCECs) were treated with Y27632 and subjected to bioenergetic profiling using the Seahorse XFp Analyzer. The effects on adenosine triphosphate (ATP) production through oxidative phosphorylation and glycolysis were measured. BCECs were also challenged with monensin to induce metabolic stress. Cell viability, apoptosis, intracellular sodium levels, and hexokinase localization were assessed using calcein AM assay, flow cytometry, fluorescence imaging, and immunostaining, respectively. Results Y27632 increased maximal ATP production rates via both oxidative phosphorylation and glycolysis, thereby expanding the overall bioenergetic capacity in BCECs. Under monensin-induced metabolic stress, ROCK inhibitor pretreatment significantly enhanced glycolytic ATP production and reduced apoptosis compared with untreated cells. Y27632 also facilitated sodium export by increasing Na/K-ATPase activity, as evidenced by lower intracellular sodium levels. Additionally, Y27632 promoted the translocation of hexokinase 2 to mitochondria under stress conditions, thereby enhancing glycolytic capacity. The effect of Y27632 on cell viability and sodium export was abrogated when cells were forced to rely on oxidative phosphorylation in galactose media, indicating that the protective effects of Y27632 are dependent on glycolytic ATP production under monensin stress. Conclusions ROCK inhibitor Y27632 enhances the bioenergetic capacity of BCECs, allowing the cells to better withstand metabolic stress by rapidly generating ATP to meet increased energy demands, maintaining ion homeostasis and reducing apoptosis.
Collapse
Affiliation(s)
- Wei-Ting Ho
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jung-Shen Chang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Jen Lei
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsan-Chi Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Jia-Kang Wang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Electrical Engineering, Yuan Ze University, Taoyuan, Taiwan
| | - Shu-Wen Chang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzuu-Shuh Jou
- College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taiwan
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
15
|
Kaniuka O, Deregowska A, Bandura Y, Sabadashka M, Chala D, Kulachkovskyi O, Kubis H, Adamczyk-Grochala J, Sybirna N. Upregulation of GRP78 is accompanied by decreased antioxidant response and mitophagy promotion in streptozotocin-induced type 1 diabetes in rats. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167531. [PMID: 39353543 DOI: 10.1016/j.bbadis.2024.167531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Endoplasmic reticulum stress, oxidative stress, and mitochondrial dysfunction are interconnected processes involved in the pathogenesis of diabetes mellitus (DM). In the present study, we demonstrate a distinct unfolded protein response (UPR) signaling pathways in two mammalian models of DM: β-TC-6 cell line and streptozotocin-induced type 1 diabetes model in rats. However, a feature common to both systems was the upregulation of the GRP78 protein. Moreover, in vivo studies showed the disruption of the antioxidant system and an escalation of mitophagy against the background of a depletion of the level of ATP in pancreatic cells. In conclusion, we suggest that glucotoxic conditions induced GRP78 upregulation, and next cause depletion of the antioxidant pool and disruption of the functioning of antioxidant defense enzymes and in consequence promote mitophagy in pancreatic cells. Therefore, GRP78 may be considered as a potential therapeutic factor in patients with diabetes.
Collapse
Affiliation(s)
- O Kaniuka
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - A Deregowska
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - Yu Bandura
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - M Sabadashka
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - D Chala
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - O Kulachkovskyi
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - H Kubis
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - J Adamczyk-Grochala
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - N Sybirna
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| |
Collapse
|
16
|
Makio T, Chen J, Simmen T. ER stress as a sentinel mechanism for ER Ca 2+ homeostasis. Cell Calcium 2024; 124:102961. [PMID: 39471738 DOI: 10.1016/j.ceca.2024.102961] [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: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Endoplasmic reticulum (ER) stress is triggered upon the interference with oxidative protein folding that aims to produce fully folded, disulfide-bonded and glycosylated proteins, which are then competent to exit the ER. Many of the enzymes catalyzing this process require the binding of Ca2+ ions, including the chaperones BiP/GRP78, calnexin and calreticulin. The induction of ER stress with a variety of drugs interferes with chaperone Ca2+ binding, increases cytosolic Ca2+through the opening of ER Ca2+ channels, and activates store-operated Ca2+ entry (SOCE). Posttranslational modifications (PTMs) of the ER Ca2+ handling proteins through ER stress-dependent phosphorylation or oxidation control these mechanisms, as demonstrated in the case of the sarco/endoplasmic reticulum ATPase (SERCA), inositol 1,4,5 trisphosphate receptors (IP3Rs) or stromal interaction molecule 1 (STIM1). Their aim is to restore ER Ca2+ homeostasis but also to increase Ca2+ transfer from the ER to mitochondria during ER stress. This latter function boosts ER bioenergetics, but also triggers apoptosis if ER Ca2+ signaling persists. ER Ca2+ toolkit oxidative modifications upon ER stress can occur within the ER lumen or in the adjacent cytosol. Enzymes involved in this redox control include ER oxidoreductin 1 (ERO1) or the thioredoxin-family protein disulfide isomerases (PDI) and ERp57. A tight, but adaptive connection between ER Ca2+ content, ER stress and mitochondrial readouts allows for the proper functioning of many tissues, including skeletal muscle, the liver, and the pancreas, where ER stress either maintains or compromises their function, depending on its extent and context. Upon mutation of key regulators of ER Ca2+ signaling, diseases such as muscular defects (e.g., from mutated selenoprotein N, SEPN1/SELENON), or diabetes (e.g., from mutated PERK) are the result.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada.
| |
Collapse
|
17
|
Jia Z, Li H, Xu K, Li R, Yang S, Chen L, Zhang Q, Li S, Sun X. MAM-mediated mitophagy and endoplasmic reticulum stress: the hidden regulators of ischemic stroke. Front Cell Neurosci 2024; 18:1470144. [PMID: 39640236 PMCID: PMC11617170 DOI: 10.3389/fncel.2024.1470144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemic stroke (IS) is the predominant subtype of stroke and a leading contributor to global mortality. The mitochondrial-associated endoplasmic reticulum membrane (MAM) is a specialized region that facilitates communication between the endoplasmic reticulum and mitochondria, and has been extensively investigated in the context of neurodegenerative diseases. Nevertheless, its precise involvement in IS remains elusive. This literature review elucidates the intricate involvement of MAM in mitophagy and endoplasmic reticulum stress during IS. PINK1, FUNDC1, Beclin1, and Mfn2 are highly concentrated in the MAM and play a crucial role in regulating mitochondrial autophagy. GRP78, IRE1, PERK, and Sig-1R participate in the unfolded protein response (UPR) within the MAM, regulating endoplasmic reticulum stress during IS. Hence, the diverse molecules on MAM operate independently and interact with each other, collectively contributing to the pathogenesis of IS as the covert orchestrator.
Collapse
Affiliation(s)
- Ziyi Jia
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongtao Li
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ke Xu
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ruobing Li
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyu Yang
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Long Chen
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qianwen Zhang
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shulin Li
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaowei Sun
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
18
|
Chen X, Yang Y, Zhou Z, Yu H, Zhang S, Huang S, Wei Z, Ren K, Jin Y. Unraveling the complex interplay between Mitochondria-Associated Membranes (MAMs) and cardiovascular Inflammation: Molecular mechanisms and therapeutic implications. Int Immunopharmacol 2024; 141:112930. [PMID: 39146786 DOI: 10.1016/j.intimp.2024.112930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant public health concern because of their associations with inflammation, oxidative stress, and abnormal remodeling of the heart and blood vessels. In this review, we discuss the intricate interplay between mitochondria-associated membranes (MAMs) and cardiovascular inflammation, highlighting their role in key cellular processes such as calcium homeostasis, lipid metabolism, oxidative stress management, and ERS. We explored how these functions impact the pathogenesis and progression of various CVDs, including myocardial ischemia-reperfusion injury, atherosclerosis, diabetic cardiomyopathy, cardiovascular aging, heart failure, and pulmonary hypertension. Additionally, we examined current therapeutic strategies targeting MAM-related pathways and proteins, emphasizing the potential of MAMs as therapeutic targets. Our review aims to provide new insights into the mechanisms of cardiovascular inflammation and propose novel therapeutic approaches to improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Xing Chen
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zheng Zhou
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haihan Yu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shuwei Zhang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siyuan Huang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Ziqing Wei
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
19
|
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
|
20
|
Chen B, Stark DC, Jadhav PV, Lynn-Nguyen TM, Halligan BS, Rossiter NJ, Sindoni N, Shin M, Paulo JA, Chang M, Koo I, Koshkin S, Eyunni S, Ronchi P, Paulsen MT, Morlacchi P, Hanna DA, Lin J, Guerra RM, Pagliarini DJ, Banerjee R, Parolia A, Ljungman ME, Patterson AD, Mancias JD, Mosalaganti S, Sexton JZ, Calì T, Lyssiotis CA, Shah YM. BRD4-mediated epigenetic regulation of endoplasmic reticulum-mitochondria contact sites is governed by the mitochondrial complex III. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578646. [PMID: 38352460 PMCID: PMC10862858 DOI: 10.1101/2024.02.02.578646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2024]
Abstract
Inter-organellar communication is critical for cellular metabolic homeostasis. One of the most abundant inter-organellar interactions are those at the endoplasmic reticulum and mitochondria contact sites (ERMCS). However, a detailed understanding of the mechanisms governing ERMCS regulation and their roles in cellular metabolism are limited by a lack of tools that permit temporal induction and reversal. Through unbiased screening approaches, we identified fedratinib, an FDA-approved drug, that dramatically increases ERMCS abundance by inhibiting the epigenetic modifier BRD4. Fedratinib rapidly and reversibly modulates mitochondrial and ER morphology and alters metabolic homeostasis. Moreover, ERMCS modulation depends on mitochondria electron transport chain complex III function. Comparison of fedratinib activity to other reported inducers of ERMCS revealed common mechanisms of induction and function, providing clarity and union to a growing body of experimental observations. In total, our results uncovered a novel epigenetic signaling pathway and an endogenous metabolic regulator that connects ERMCS and cellular metabolism.
Collapse
|
21
|
Knedlik T, Giacomello M. Temporal dynamics of membrane contact sites. Nat Cell Biol 2024; 26:1822-1824. [PMID: 39482355 DOI: 10.1038/s41556-024-01539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Affiliation(s)
- Tomas Knedlik
- Department of Biology, University of Padua, Padua, Italy
| | | |
Collapse
|
22
|
Chen Q, Ren Z, Dang L, Liu Z, Wang S, Chen X, Qiu G, Sun C. Hoxa5 alleviates adipose tissue metabolic distortions in high-fat diet mice associated with a reduction in MERC. BMC Biol 2024; 22:247. [PMID: 39468535 PMCID: PMC11520472 DOI: 10.1186/s12915-024-02047-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/16/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Mitochondria-endoplasmic reticulum membrane contact (MERC) is an important mode of intercellular organelle communication and plays a crucial role in adipose tissue metabolism. Functionality of Hoxa5 is an important transcription factor involved in adipose tissue fate determination and metabolic regulation, but the relationship between Hoxa5 and MERC is not well understood. RESULTS In our study, we established an obesity model mouse by high-fat diet (HFD), induced the alteration of Hoxa5 expression by adenoviral transfection, and explored the effect of Hoxa5 on MERC dysfunction and metabolic distortions of adipose tissue with the help of transmission electron microscopy, calcium ion probe staining, and other detection means. The results showed Hoxa5 was able to reduce MERC production, alleviate endoplasmic reticulum stress (ERS) and calcium over-transport, and affect cGAS-STING-mediated innate immune response affecting adipose tissue energy metabolism, as well as affect the AKT-IP3R pathway to alleviate insulin resistance and ameliorate metabolic distortions in adipose tissue of mice. CONCLUSIONS Our results suggest that Hoxa5 can ameliorate high-fat diet-induced MERC overproduction and related functional abnormalities, in which finding is expected to provide new ideas for the improvement of obesity-related metabolic distortions.
Collapse
Affiliation(s)
- Qi Chen
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China
| | - Zeyu Ren
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China
| | - Liping Dang
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China
| | - Zunhai Liu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China
| | - Simeng Wang
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China
| | - Xinhao Chen
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China
| | - Guiping Qiu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xi Nong Roud, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
23
|
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
|
24
|
Dafinca R, Tosat-Bitrian C, Carroll E, Vahsen BF, Gilbert-Jaramillo J, Scaber J, Feneberg E, Johnson E, Talbot K. Dynactin-1 mediates rescue of impaired axonal transport due to reduced mitochondrial bioenergetics in amyotrophic lateral sclerosis motor neurons. Brain Commun 2024; 6:fcae350. [PMID: 39440303 PMCID: PMC11495216 DOI: 10.1093/braincomms/fcae350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/07/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of the motor system with complex determinants, including genetic and non-genetic factors. A key pathological signature of ALS is the cytoplasmic mislocalization and aggregation of TDP-43 in affected motor neurons, which is found in 97% of cases. Recent reports have shown that mitochondrial dysfunction plays a significant role in motor neuron degeneration in ALS, and TDP-43 modulates several mitochondrial transcripts. In this study, we used induced pluripotent stem cell-derived motor neurons from ALS patients with TDP-43 mutations and a transgenic TDP-43M337V mouse model to determine how TDP-43 mutations alter mitochondrial function and axonal transport. We detected significantly reduced mitochondrial respiration and ATP production in patient induced pluripotent stem cell-derived motor neurons, linked to an interaction between TDP-43M337V with ATPB and COX5A. A downstream reduction in speed of retrograde axonal transport in patient induced pluripotent stem cell-derived motor neurons was detected, which correlated with downregulation of the motor protein complex, DCTN1/dynein. Overexpression of DCTN1 in patient induced pluripotent stem cell-derived motor neurons significantly increased the percentage of retrograde travelling mitochondria and reduced the percentage of stationary mitochondria. This study shows that ALS induced pluripotent stem cell-derived motor neurons with mutations in TDP-43 have deficiencies in essential mitochondrial functions with downstream effects on retrograde axonal transport, which can be partially rescued by DCTN1 overexpression.
Collapse
Affiliation(s)
- Ruxandra Dafinca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, Oxford OX1 3QU, UK
| | - Carlota Tosat-Bitrian
- Margarita Salas Center for Biological Research, University of Madrid, Madrid 28040, Spain
| | - Emily Carroll
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, Oxford OX1 3QU, UK
| | - Björn F Vahsen
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, Oxford OX1 3QU, UK
| | - Javier Gilbert-Jaramillo
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, Oxford OX1 3QU, UK
| | - Jakub Scaber
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, Oxford OX1 3QU, UK
| | - Emily Feneberg
- Department of Neurology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, Oxford OX1 3QU, UK
| |
Collapse
|
25
|
Repas J, Frlic T, Snedec T, Kopitar AN, Sourij H, Janež A, Pavlin M. Physiologically Achievable Concentration of 2-Deoxy-D-Glucose Stimulates IFN-γ Secretion in Activated T Cells In Vitro. Int J Mol Sci 2024; 25:10384. [PMID: 39408714 PMCID: PMC11476708 DOI: 10.3390/ijms251910384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
2-deoxy-D-glucose (2DG) is a glycolysis and protein N-glycosylation inhibitor with promising anti-tumor and immunomodulatory effects. However, 2DG can also suppress T cell function, including IFN-γ secretion. Few human T cell studies have studied low-dose 2DG, which can increase IFN-γ in a Jurkat clone. We therefore investigated 2DG's effect on IFN-γ in activated human T cells from PBMCs, with 2DG treatment commenced either concurrently with activation or 48 h after activation. Concurrent 2DG treatment decreased IFN-γ secretion in a dose-dependent manner. However, 2DG treatment of pre-activated T cells had a hormetic effect on IFN-γ, with 0.15-0.6 mM 2DG (achievable in vivo) increasing and >2.4 mM 2DG reducing its secretion. In contrast, IL-2 levels declined monotonously with increasing 2DG concentration. Lower 2DG concentrations reduced PD-1 and increased CD69 expression regardless of treatment timing. The absence of increased T-bet or Eomes expression or IFNG transcription suggests another downstream mechanism. 2DG dose-dependently induced the unfolded protein response, suggesting a possible role in increased IFN-γ secretion, possibly by increasing the ER folding capacity for IFN-γ via increased chaperone expression. Overall, low-dose, short-term 2DG exposure could potentially improve the T cell anti-tumor response.
Collapse
Affiliation(s)
- Jernej Repas
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Tjaša Frlic
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Tadeja Snedec
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Andreja Nataša Kopitar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Medical University Graz, 8010 Graz, Austria;
| | - Andrej Janež
- Clinical Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
| | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
- Group for Nano- and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
26
|
Ulfig A, Jakob U. Cellular oxidants and the proteostasis network: balance between activation and destruction. Trends Biochem Sci 2024; 49:761-774. [PMID: 39168791 PMCID: PMC11731897 DOI: 10.1016/j.tibs.2024.07.001] [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: 04/05/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 08/23/2024]
Abstract
Loss of protein homeostasis (proteostasis) is a common hallmark of aging and age-associated diseases. Considered as the guardian of proteostasis, the proteostasis network (PN) acts to preserve the functionality of proteins during their lifetime. However, its activity declines with age, leading to disease manifestation. While reactive oxygen species (ROS) were traditionally considered culprits in this process, recent research challenges this view. While harmful at high concentrations, moderate ROS levels protect the cell against age-mediated onset of proteotoxicity by activating molecular chaperones, stress response pathways, and autophagy. This review explores the nuanced roles of ROS in proteostasis and discusses the most recent findings regarding the redox regulation of the PN and its potential in extending healthspan and delaying age-related pathologies.
Collapse
Affiliation(s)
- Agnes Ulfig
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ursula Jakob
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA; Biological Chemistry Department, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Guo Q, Deng T, Du Y, Yao W, Tian W, Liao H, Wang Y, Li J, Yan W, Li Y. Impact of DEHP on mitochondria-associated endoplasmic reticulum membranes and reproductive toxicity in ovary. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116679. [PMID: 38981393 DOI: 10.1016/j.ecoenv.2024.116679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/12/2024] [Accepted: 06/30/2024] [Indexed: 07/11/2024]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a widely recognized environmental endocrine disruptor that potentially impacts female reproductive function, although the specific mechanisms leading to such impairment remain unclear. A growing body of research has revealed that the endoplasmic reticulum and mitochondrial function significantly influence oocyte quality. The structure of mitochondria-associated endoplasmic reticulum membranes (MAMs) is crucial for facilitating the exchange of Ca2+, lipids, and metabolites. This study aimed to investigate the alterations in the composition and function of MAMs after DEHP exposure and to elucidate the underlying mechanisms of ovarian toxicity. The female mice were exposed to DEHP at doses of 5 and 500 mg/kg/day for one month. The results revealed that DEHP exposure led to reduced serum anti-Müllerian hormone levels and increased atretic follicles in mice. DEHP induced endoplasmic reticulum stress and disrupted calcium homeostasis in oocytes. Furthermore, DEHP impaired the mitochondrial function of oocytes and reduced their membrane potential, and promoting apoptosis. Similar results were observed in human granulosa cells after exposure to mono-(2-ethylhexyl) phthalate (MEHP, metabolites of DEHP) in vitro. Proteomic analysis and transmission electron microscopy revealed modifications in the functional proteins and structure of the MAMs, and the suppression of oxidative phosphorylation pathways. The findings of this investigation provide a new perspective on the mechanism underlying the reproductive toxicity of DEHP in females.
Collapse
Affiliation(s)
- Qingchun Guo
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Taoran Deng
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yaoyao Du
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Wen Yao
- General Hospital of Central Theater Command, Wuhan, Hubei, PR China
| | - Wenqu Tian
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Hongmei Liao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yi Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Juan Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Wei Yan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Yufeng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
28
|
Liao Q, Yang Y, Li Y, Zhang J, Fan K, Guo Y, Chen J, Chen Y, Zhu P, Huang L, Liu Z. Targeting TANK-binding kinase 1 attenuates painful diabetic neuropathy via inhibiting microglia pyroptosis. Cell Commun Signal 2024; 22:368. [PMID: 39030571 PMCID: PMC11264750 DOI: 10.1186/s12964-024-01723-6] [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: 02/23/2024] [Accepted: 06/22/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Painful diabetic neuropathy (PDN) is closely linked to inflammation, which has been demonstrated to be associated with pyroptosis. Emerging evidence has implicated TANK-binding kinase 1 (TBK1) in various inflammatory diseases. However, it remains unknown whether activated TBK1 causes hyperalgesia via pyroptosis. METHODS PDN mice model of type 1 or type 2 diabetic was induced by C57BL/6J or BKS-DB mice with Lepr gene mutation. For type 2 diabetes PDN model, TBK1-siRNA, Caspase-1 inhibitor Ac-YVAD-cmk or TBK1 inhibitor amlexanox (AMX) were delivered by intrathecal injection or intragastric administration. The pain threshold and plantar skin blood perfusion were evaluated through animal experiments. The assessments of spinal cord, dorsal root ganglion, sciatic nerve, plantar skin and serum included western blotting, immunofluorescence, ELISA, and transmission electron microscopy. RESULTS In the PDN mouse model, we found that TBK1 was significantly activated in the spinal dorsal horn (SDH) and mainly located in microglia, and intrathecal injection of chemically modified TBK1-siRNA could improve hyperalgesia. Herein, we described the mechanism that TBK1 could activate the noncanonical nuclear factor κB (NF-κB) pathway, mediate the activation of NLRP3 inflammasome, trigger microglia pyroptosis, and ultimately induce PDN, which could be reversed following TBK1-siRNA injection. We also found that systemic administration of AMX, a TBK1 inhibitor, could effectively improve peripheral nerve injury. These results revealed the key role of TBK1 in PDN and that TBK1 inhibitor AMX could be a potential strategy for treating PDN. CONCLUSIONS Our findings revealed a novel causal role of TBK1 in pathogenesis of PDN, which raises the possibility of applying amlexanox to selectively target TBK1 as a potential therapeutic strategy for PDN.
Collapse
Affiliation(s)
- Qinming Liao
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Yimei Yang
- Department of Neurosurgery, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510030, Guangdong, China
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Yilu Li
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Jun Zhang
- Department of Neurosurgery, Dalang Hospital, Dongguan, 523775, Guangdong, China
| | - Keke Fan
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518000, Guangdong, China
| | - Yihao Guo
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510220, Guangdong, China
| | - Jun Chen
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Yinhao Chen
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Pian Zhu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510220, Guangdong, China
- Department of Anesthesiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032, China
| | - Lijin Huang
- Department of Neurosurgery, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510030, Guangdong, China.
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China.
| | - Zhongjie Liu
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518000, Guangdong, China.
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510220, Guangdong, China.
| |
Collapse
|
29
|
Lee CT, Bell M, Bonilla-Quintana M, Rangamani P. Biophysical Modeling of Synaptic Plasticity. Annu Rev Biophys 2024; 53:397-426. [PMID: 38382115 DOI: 10.1146/annurev-biophys-072123-124954] [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: 02/23/2024]
Abstract
Dendritic spines are small, bulbous compartments that function as postsynaptic sites and undergo intense biochemical and biophysical activity. The role of the myriad signaling pathways that are implicated in synaptic plasticity is well studied. A recent abundance of quantitative experimental data has made the events associated with synaptic plasticity amenable to quantitative biophysical modeling. Spines are also fascinating biophysical computational units because spine geometry, signal transduction, and mechanics work in a complex feedback loop to tune synaptic plasticity. In this sense, ideas from modeling cell motility can inspire us to develop multiscale approaches for predictive modeling of synaptic plasticity. In this article, we review the key steps in postsynaptic plasticity with a specific focus on the impact of spine geometry on signaling, cytoskeleton rearrangement, and membrane mechanics. We summarize the main experimental observations and highlight how theory and computation can aid our understanding of these complex processes.
Collapse
Affiliation(s)
- Christopher T Lee
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Miriam Bell
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Mayte Bonilla-Quintana
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA;
| |
Collapse
|
30
|
Chen C, Dong X, Zhang W, Chang X, Gao W. Dialogue between mitochondria and endoplasmic reticulum-potential therapeutic targets for age-related cardiovascular diseases. Front Pharmacol 2024; 15:1389202. [PMID: 38939842 PMCID: PMC11208709 DOI: 10.3389/fphar.2024.1389202] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/30/2024] [Indexed: 06/29/2024] Open
Abstract
Mitochondria-associated endoplasmic reticulum membranes (MAMs) act as physical membrane contact sites facilitating material exchange and signal transmission between mitochondria and endoplasmic reticulum (ER), thereby regulating processes such as Ca2+/lipid transport, mitochondrial dynamics, autophagy, ER stress, inflammation, and apoptosis, among other pathological mechanisms. Emerging evidence underscores the pivotal role of MAMs in cardiovascular diseases (CVDs), particularly in aging-related pathologies. Aging significantly influences the structure and function of the heart and the arterial system, possibly due to the accumulation of reactive oxygen species (ROS) resulting from reduced antioxidant capacity and the age-related decline in organelle function, including mitochondria. Therefore, this paper begins by describing the composition, structure, and function of MAMs, followed by an exploration of the degenerative changes in MAMs and the cardiovascular system during aging. Subsequently, it discusses the regulatory pathways and approaches targeting MAMs in aging-related CVDs, to provide novel treatment strategies for managing CVDs in aging populations.
Collapse
Affiliation(s)
- Chen Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xueyan Dong
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wang Zhang
- Shandong Provincial Mental Health Center, Jinan, China
| | - Xing Chang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wulin Gao
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
31
|
Licini C, Morroni G, Lucarini G, Vitto VAM, Orlando F, Missiroli S, D'Achille G, Perrone M, Spadoni T, Graciotti L, Bigossi G, Provinciali M, Offidani A, Mattioli-Belmonte M, Cirioni O, Pinton P, Simonetti O, Marchi S. ER-mitochondria association negatively affects wound healing by regulating NLRP3 activation. Cell Death Dis 2024; 15:407. [PMID: 38862500 PMCID: PMC11167056 DOI: 10.1038/s41419-024-06765-9] [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: 02/01/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is the most common causative agent of acute bacterial skin and skin-structure infections (ABSSSI), one of the major challenges to the health system worldwide. Although the use of antibiotics as the first line of intervention for MRSA-infected wounds is recommended, important side effects could occur, including cytotoxicity or immune dysregulation, thus affecting the repair process. Here, we show that the oxazolidinone antibiotic linezolid (LZD) impairs wound healing by aberrantly increasing interleukin 1 β (IL-1β) production in keratinocytes. Mechanistically, LZD triggers a reactive oxygen species (ROS)-independent mitochondrial damage that culminates in increased tethering between the endoplasmic reticulum (ER) and mitochondria, which in turn activates the NLR family pyrin domain-containing 3 (NLRP3) inflammasome complex by promoting its assembly to the mitochondrial surface. Downregulation of ER-mitochondria contact formation is sufficient to inhibit the LZD-driven NLRP3 inflammasome activation and IL-1β production, restoring wound closure. These results identify the ER-mitochondria association as a key factor for NLRP3 activation and reveal a new mechanism in the regulation of the wound healing process that might be clinically relevant.
Collapse
Affiliation(s)
- Caterina Licini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Gianluca Morroni
- Microbiology Unit, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Guendalina Lucarini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Veronica Angela Maria Vitto
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Sonia Missiroli
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Gloria D'Achille
- Microbiology Unit, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Mariasole Perrone
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Tatiana Spadoni
- Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Laura Graciotti
- Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Giorgia Bigossi
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121, Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121, Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121, Ancona, Italy
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy.
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy.
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121, Ancona, Italy.
| |
Collapse
|
32
|
Huang X, Jiang F, Ma Y, Zhu K, Wang Z, Hua Z, Yu J, Zhang L. A bibliometric analysis of endoplasmic reticulum stress and atherosclerosis. Front Physiol 2024; 15:1392454. [PMID: 38938744 PMCID: PMC11210825 DOI: 10.3389/fphys.2024.1392454] [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: 03/25/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
The mechanisms underlying the occurrence and development of atherosclerosis (AS) are diverse, among which endoplasmic reticulum stress (ERS) is an important mechanism that should not be overlooked. However, up to now, there has been no bibliometric study on the relationship between ERS and AS. To understand the research progress in ERS and AS, this paper conducted a statistical analysis of publications in this field using bibliometrics. A total of 1,035 records were retrieved from the Web of Science Core Collection. CiteSpace, VOSviewer, and the R package "bibliometric" were used to analyze the spatiotemporal distribution, countries, authors, institutions, journals, references, and keywords of the literature, and to present the basic information of this field through visualized maps, as well as determine the collaboration relationships among researchers in this field. This field has gradually developed and stabilized over the past 20 years. The current research hotspots in this field mainly include the relationship between ERS and AS-related cells, the mechanisms by which ERS promotes AS, related diseases, and associated cytokines, etc. Vascular calcification, endothelial dysfunction, NLRP3 inflammasome, and heart failure represent the frontier research in this field and are becoming new research hotspots. It is hoped that this study will provide new insights for research and clinical work in the field of ERS and AS.
Collapse
Affiliation(s)
- Xinyu Huang
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Feng Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Yongbo Ma
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Kunpeng Zhu
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Zhenyuan Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Zhen Hua
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Jie Yu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Lei Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| |
Collapse
|
33
|
Casas-Martinez JC, Samali A, McDonagh B. Redox regulation of UPR signalling and mitochondrial ER contact sites. Cell Mol Life Sci 2024; 81:250. [PMID: 38847861 PMCID: PMC11335286 DOI: 10.1007/s00018-024-05286-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/11/2024] [Accepted: 05/18/2024] [Indexed: 06/13/2024]
Abstract
Mitochondria and the endoplasmic reticulum (ER) have a synergistic relationship and are key regulatory hubs in maintaining cell homeostasis. Communication between these organelles is mediated by mitochondria ER contact sites (MERCS), allowing the exchange of material and information, modulating calcium homeostasis, redox signalling, lipid transfer and the regulation of mitochondrial dynamics. MERCS are dynamic structures that allow cells to respond to changes in the intracellular environment under normal homeostatic conditions, while their assembly/disassembly are affected by pathophysiological conditions such as ageing and disease. Disruption of protein folding in the ER lumen can activate the Unfolded Protein Response (UPR), promoting the remodelling of ER membranes and MERCS formation. The UPR stress receptor kinases PERK and IRE1, are located at or close to MERCS. UPR signalling can be adaptive or maladaptive, depending on whether the disruption in protein folding or ER stress is transient or sustained. Adaptive UPR signalling via MERCS can increase mitochondrial calcium import, metabolism and dynamics, while maladaptive UPR signalling can result in excessive calcium import and activation of apoptotic pathways. Targeting UPR signalling and the assembly of MERCS is an attractive therapeutic approach for a range of age-related conditions such as neurodegeneration and sarcopenia. This review highlights the emerging evidence related to the role of redox mediated UPR activation in orchestrating inter-organelle communication between the ER and mitochondria, and ultimately the determination of cell function and fate.
Collapse
Affiliation(s)
- Jose C Casas-Martinez
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
- Apoptosis Research Centre, University of Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, University of Galway, Galway, Ireland
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland.
- Apoptosis Research Centre, University of Galway, Galway, Ireland.
| |
Collapse
|
34
|
Balhara M, Neikirk K, Marshall A, Hinton A, Kirabo A. Endoplasmic Reticulum Stress in Hypertension and Salt Sensitivity of Blood Pressure. Curr Hypertens Rep 2024; 26:273-290. [PMID: 38602583 PMCID: PMC11166838 DOI: 10.1007/s11906-024-01300-9] [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] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Hypertension is a principal risk factor for cardiovascular morbidity and mortality, with its severity exacerbated by high sodium intake, particularly in individuals with salt-sensitive blood pressure. However, the mechanisms underlying hypertension and salt sensitivity are only partly understood. Herein, we review potential interactions in hypertension pathophysiology involving the immune system, endoplasmic reticulum (ER) stress, the unfolded protein response (UPR), and proteostasis pathways; identify knowledge gaps; and discuss future directions. RECENT FINDINGS Recent advancements by our research group and others reveal interactions within and between adaptive and innate immune responses in hypertension pathophysiology. The salt-immune-hypertension axis is further supported by the discovery of the role of dendritic cells in hypertension, marked by isolevuglandin (IsoLG) formation. Alongside these broadened understandings of immune-mediated salt sensitivity, the contributions of T cells to hypertension have been recently challenged by groups whose findings did not support increased resistance of Rag-1-deficient mice to Ang II infusion. Hypertension has also been linked to ER stress and the UPR. Notably, a holistic approach is needed because the UPR engages in crosstalk with autophagy, the ubiquitin proteasome, and other proteostasis pathways, that may all involve hypertension. There is a critical need for studies to establish cause and effect relationships between ER stress and the UPR in hypertension pathophysiology in humans and to determine whether the immune system and ER stress function mainly to exacerbate or initiate hypertension and target organ injury. This review of recent studies proposes new avenues for future research for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maria Balhara
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA.
- Vanderbilt Center for Immunobiology, Nashville, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, USA.
- Vanderbilt Institute for Global Health, Nashville, USA.
| |
Collapse
|
35
|
Kim HS, Lee D, Shen S. Endoplasmic reticular stress as an emerging therapeutic target for chronic pain: a narrative review. Br J Anaesth 2024; 132:707-724. [PMID: 38378384 PMCID: PMC10925894 DOI: 10.1016/j.bja.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 02/22/2024] Open
Abstract
Chronic pain is a severely debilitating condition with enormous socioeconomic costs. Current treatment regimens with nonsteroidal anti-inflammatory drugs (NSAIDs), steroids, or opioids have been largely unsatisfactory with uncertain benefits or severe long-term side effects. This is mainly because chronic pain has a multifactorial aetiology. Although conventional pain medications can alleviate pain by keeping several dysfunctional pathways under control, they can mask other underlying pathological causes, ultimately worsening nerve pathologies and pain outcome. Recent preclinical studies have shown that endoplasmic reticulum (ER) stress could be a central hub for triggering multiple molecular cascades involved in the development of chronic pain. Several ER stress inhibitors and unfolded protein response modulators, which have been tested in randomised clinical trials or apprpoved by the US Food and Drug Administration for other chronic diseases, significantly alleviated hyperalgesia in multiple preclinical pain models. Although the role of ER stress in neurodegenerative disorders, metabolic disorders, and cancer has been well established, research on ER stress and chronic pain is still in its infancy. Here, we critically analyse preclinical studies and explore how ER stress can mechanistically act as a central node to drive development and progression of chronic pain. We also discuss therapeutic prospects, benefits, and pitfalls of using ER stress inhibitors and unfolded protein response modulators for managing intractable chronic pain. In the future, targeting ER stress to impact multiple molecular networks might be an attractive therapeutic strategy against chronic pain refractory to steroids, NSAIDs, or opioids. This novel therapeutic strategy could provide solutions for the opioid crisis and public health challenge.
Collapse
Affiliation(s)
- Harper S Kim
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donghwan Lee
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shiqian Shen
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Hinton A, Katti P, Mungai M, Hall DD, Koval O, Shao J, Vue Z, Lopez EG, Rostami R, Neikirk K, Ponce J, Streeter J, Schickling B, Bacevac S, Grueter C, Marshall A, Beasley HK, Do Koo Y, Bodine SC, Nava NGR, Quintana AM, Song LS, Grumbach I, Pereira RO, Glancy B, Abel ED. ATF4-dependent increase in mitochondrial-endoplasmic reticulum tethering following OPA1 deletion in skeletal muscle. J Cell Physiol 2024; 239:e31204. [PMID: 38419397 PMCID: PMC11144302 DOI: 10.1002/jcp.31204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024]
Abstract
Mitochondria and endoplasmic reticulum (ER) contact sites (MERCs) are protein- and lipid-enriched hubs that mediate interorganellar communication by contributing to the dynamic transfer of Ca2+, lipid, and other metabolites between these organelles. Defective MERCs are associated with cellular oxidative stress, neurodegenerative disease, and cardiac and skeletal muscle pathology via mechanisms that are poorly understood. We previously demonstrated that skeletal muscle-specific knockdown (KD) of the mitochondrial fusion mediator optic atrophy 1 (OPA1) induced ER stress and correlated with an induction of Mitofusin-2, a known MERC protein. In the present study, we tested the hypothesis that Opa1 downregulation in skeletal muscle cells alters MERC formation by evaluating multiple myocyte systems, including from mice and Drosophila, and in primary myotubes. Our results revealed that OPA1 deficiency induced tighter and more frequent MERCs in concert with a greater abundance of MERC proteins involved in calcium exchange. Additionally, loss of OPA1 increased the expression of activating transcription factor 4 (ATF4), an integrated stress response (ISR) pathway effector. Reducing Atf4 expression prevented the OPA1-loss-induced tightening of MERC structures. OPA1 reduction was associated with decreased mitochondrial and sarcoplasmic reticulum, a specialized form of ER, calcium, which was reversed following ATF4 repression. These data suggest that mitochondrial stress, induced by OPA1 deficiency, regulates skeletal muscle MERC formation in an ATF4-dependent manner.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Prasanna Katti
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA, 20892
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Duane D. Hall
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
| | - Olha Koval
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Jianqiang Shao
- Central Microscopy Research Facility, Iowa City, IA USA 52242
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Edgar Garza Lopez
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
| | - Rahmati Rostami
- Department of Genetic Medicine, Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA, 10065
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Jessica Ponce
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Jennifer Streeter
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Brandon Schickling
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Department of Medicine, Duke University, Durham, NC, USA 27708
| | - Serif Bacevac
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Chad Grueter
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Sue C. Bodine
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA 73104
| | - Nayeli G. Reyes Nava
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA 79968
| | - Anita M. Quintana
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA 79968
| | - Long-Sheng Song
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Isabella Grumbach
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Renata O. Pereira
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA, 20892
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA 20892
| | - E. Dale Abel
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
- Department of Medicine, UCLA School of Medicine, Los Angeles, CA, USA 90095
| |
Collapse
|
37
|
Germani S, Van Ho AT, Cherubini A, Varone E, Chernorudskiy A, Renna GM, Fumagalli S, Gobbi M, Lucchetti J, Bolis M, Guarrera L, Craparotta I, Rastelli G, Piccoli G, de Napoli C, Nogara L, Poggio E, Brini M, Cattaneo A, Bachi A, Simmen T, Calì T, Quijano-Roy S, Boncompagni S, Blaauw B, Ferreiro A, Zito E. SEPN1-related myopathy depends on the oxidoreductase ERO1A and is druggable with the chemical chaperone TUDCA. Cell Rep Med 2024; 5:101439. [PMID: 38402623 PMCID: PMC10982971 DOI: 10.1016/j.xcrm.2024.101439] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/06/2023] [Accepted: 01/31/2024] [Indexed: 02/27/2024]
Abstract
Selenoprotein N (SEPN1) is a protein of the endoplasmic reticulum (ER) whose inherited defects originate SEPN1-related myopathy (SEPN1-RM). Here, we identify an interaction between SEPN1 and the ER-stress-induced oxidoreductase ERO1A. SEPN1 and ERO1A, both enriched in mitochondria-associated membranes (MAMs), are involved in the redox regulation of proteins. ERO1A depletion in SEPN1 knockout cells restores ER redox, re-equilibrates short-range MAMs, and rescues mitochondrial bioenergetics. ERO1A knockout in a mouse background of SEPN1 loss blunts ER stress and improves multiple MAM functions, including Ca2+ levels and bioenergetics, thus reversing diaphragmatic weakness. The treatment of SEPN1 knockout mice with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) mirrors the results of ERO1A loss. Importantly, muscle biopsies from patients with SEPN1-RM exhibit ERO1A overexpression, and TUDCA-treated SEPN1-RM patient-derived primary myoblasts show improvement in bioenergetics. These findings point to ERO1A as a biomarker and a viable target for intervention and to TUDCA as a pharmacological treatment for SEPN1-RM.
Collapse
Affiliation(s)
- Serena Germani
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Andrew Tri Van Ho
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France
| | | | - Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | | | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jacopo Lucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Bioinformatics Core Unit, Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Giorgia Rastelli
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgia Piccoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cosimo de Napoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Department of Pharmaceutical Sciences, University of Padova, Padova, Italy
| | - Elena Poggio
- Department of Biology, University of Padova, Padova, Italy
| | - Marisa Brini
- Department of Pharmaceutical Sciences, University of Padova, Padova, Italy; Department of Biology, University of Padova, Padova, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy
| | | | - Angela Bachi
- IFOM-ETS AIRC Institute of Molecular Oncology, Milan, Italy
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy; Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Susana Quijano-Roy
- APHP-Université Paris-Saclay, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, FILNEMUS, ERN-Euro-NMD, Creteil, France; Pediatric Neurology and ICU Department, DMU Santé Enfant Adolescent (SEA), Raymond Poincaré University Hospital, Garches, France
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Venetian Institute of Molecular Medicine, Padova, Italy.
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France; APHP, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, Neuromyology Department, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
38
|
Zhai DS, Wang XS, Yang L, Jiang YL, Jin YC, Yan YX, Song DK, Zhang K, Han ZK, Liu MY, Wu YM, Ma X, Qi JY, Yang F, Tian F, Li XB, Liu SB. TOM40 mediates the effect of TSPO on postpartum depression partially through regulating calcium homeostasis in microglia. J Affect Disord 2024; 348:283-296. [PMID: 38159656 DOI: 10.1016/j.jad.2023.12.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/03/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
AIMS To assess the effect of the translocator protein 18 kDa (TSPO) on postpartum depression and explore its mechanism. METHODS Postpartum depression (PPD) mouse model was established, and flow cytometry, immunofluorescence, Western blot analysis, real-time quantitative PCR, adeno-associated virus (AAV), co-immunoprecipitation-mass spectrometry and immunofluorescence co-staining were used to detect the effect of TSPO ligand ZBD-2 on PPD mice. RESULTS ZBD-2 inhibits the overactivation of microglia in the hippocampus and amygdala of PPD model mice. ZBD-2 not only inhibited the inflammation but also repressed the burst of reactive oxygen species (ROS) and mitochondrial ROS (mtROS). Meanwhile, ZBD-2 protects mitochondria from LPS-induced damages through inhibiting the influx of calcium. ZBD-2 modulated the calcium influx by increasing the level of translocase of the outer mitochondrial membrane 40 (TOM40) and reducing the interaction of TSPO and TOM40. In addition, the effect of ZBD-2 was partially dependent on anti-oxidative process. Knockdown of TOM40 by adeno-associated virus (AAV) in the hippocampus or amygdala dramatically reduced the effect of ZBD-2 on PPD, indicating that TOM40 mediates the effect of ZBD-2 on PPD. CONCLUSIONS TOM40 is required for the effect of ZBD-2 on treating anxiety and depression in PPD mice. This study reveals the role of microglia TSPO in PPD development and provides the new therapeutic strategy for PPD.
Collapse
Affiliation(s)
- Dong-Sheng Zhai
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xin-Shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yong-Li Jiang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu-Chen Jin
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yu-Xuan Yan
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Da-Ke Song
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Zu-Kang Han
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ming-Ying Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xue Ma
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Jing-Yu Qi
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Fan Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Fei Tian
- Teaching Experimental Center, Fourth Military Medical University, Xi'an, China
| | - Xu-Bo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
39
|
Michalak M. Calreticulin: Endoplasmic reticulum Ca 2+ gatekeeper. J Cell Mol Med 2024; 28:e17839. [PMID: 37424156 PMCID: PMC10902585 DOI: 10.1111/jcmm.17839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Endoplasmic reticulum (ER) luminal Ca2+ is vital for the function of the ER and regulates many cellular processes. Calreticulin is a highly conserved, ER-resident Ca2+ binding protein and lectin-like chaperone. Over four decades of studying calreticulin demonstrate that this protein plays a crucial role in maintaining Ca2+ supply under different physiological conditions, in managing access to Ca2+ and how Ca2+ is used depending on the environmental events and in making sure that Ca2+ is not misused. Calreticulin plays a role of ER luminal Ca2+ sensor to manage Ca2+-dependent ER luminal events including maintaining interaction with its partners, Ca2+ handling molecules, substrates and stress sensors. The protein is strategically positioned in the lumen of the ER from where the protein manages access to and distribution of Ca2+ for many cellular Ca2+-signalling events. The importance of calreticulin Ca2+ pool extends beyond the ER and includes influence of cellular processes involved in many aspects of cellular pathophysiology. Abnormal handling of the ER Ca2+ contributes to many pathologies from heart failure to neurodegeneration and metabolic diseases.
Collapse
Affiliation(s)
- Marek Michalak
- Department of BiochemistryUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
40
|
Zhang R, Wang XX, Xie JF, Yao TT, Guo QW, Wang Q, Ding Z, Zhang JP, Zhang MR, Xu LC. Cypermethrin induces Sertoli cell apoptosis through endoplasmic reticulum-mitochondrial coupling involving IP3R1-GRP75-VDAC1. Reprod Toxicol 2024; 124:108552. [PMID: 38296003 DOI: 10.1016/j.reprotox.2024.108552] [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: 10/15/2023] [Revised: 01/06/2024] [Accepted: 01/25/2024] [Indexed: 02/18/2024]
Abstract
A widely used type II pyrethroid pesticide cypermethrin (CYP) is one of endocrine disrupting chemicals (EDCs) with anti-androgenic activity to induce male reproductive toxicology. However, the mechanisms have not been fully elucidated. This study was to explore the effects of CYP on apoptosis of mouse Sertoli cells (TM4) and the roles of endoplasmic reticulum (ER)-mitochondria coupling involving 1,4,5-trisphosphate receptor type1-glucose-regulated protein 75-voltage-dependent anion channel 1 (IP3R1-GRP75-VDAC1). TM4 were cultured with different concentrations of CYP. Flow cytometry, calcium (Ca2+) fluorescent probe, transmission electron microscopy and confocal microscopy, and western blot were to examine apoptosis of TM4, mitochondrial Ca2+, ER-mitochondria coupling, and expressions of related proteins. CYP was found to increase apoptotic rates of TM4 significantly. CYP was shown to significantly increase expressions of cleaved caspase-3, cleaved poly ADP-ribose polymerase (PARP). Concentration of mitochondrial Ca2+ was increased by CYP treatment significantly. CYP significantly enhanced ER-mitochondria coupling. CYP was shown to increase expressions of IP3R, Grp75 and VDAC1 significantly. We suggest that CYP induces apoptosis in TM4 cells by facilitating mitochondrial Ca2+ overload regulated by ER-mitochondria coupling involving IP3R1-GRP75-VDAC1. This study identifies a novel mechanism of CYP-induced apoptosis in Sertoli cells.
Collapse
Affiliation(s)
- Rui Zhang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xu-Xu Wang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jia-Fei Xie
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ting-Ting Yao
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Qian-Wen Guo
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Qi Wang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Zhen Ding
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jin-Peng Zhang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Mei-Rong Zhang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Li-Chun Xu
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, 209 Tong-Shan Road, Xuzhou 221004, Jiangsu, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
41
|
Giamogante F, Barazzuol L, Maiorca F, Poggio E, Esposito A, Masato A, Napolitano G, Vagnoni A, Calì T, Brini M. A SPLICS reporter reveals [Formula: see text]-synuclein regulation of lysosome-mitochondria contacts which affects TFEB nuclear translocation. Nat Commun 2024; 15:1516. [PMID: 38374070 PMCID: PMC10876553 DOI: 10.1038/s41467-024-46007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024] Open
Abstract
Mitochondrial and lysosomal activities are crucial to maintain cellular homeostasis: optimal coordination is achieved at their membrane contact sites where distinct protein machineries regulate organelle network dynamics, ions and metabolites exchange. Here we describe a genetically encoded SPLICS reporter for short- and long- juxtapositions between mitochondria and lysosomes. We report the existence of narrow and wide lysosome-mitochondria contacts differently modulated by mitophagy, autophagy and genetic manipulation of tethering factors. The overexpression of α-synuclein (α-syn) reduces the apposition of mitochondria/lysosomes membranes and affects their privileged Ca2+ transfer, impinging on TFEB nuclear translocation. We observe enhanced TFEB nuclear translocation in α-syn-overexpressing cells. We propose that α-syn, by interfering with mitochondria/lysosomes tethering impacts on local Ca2+ regulated pathways, among which TFEB mediated signaling, and in turn mitochondrial and lysosomal function. Defects in mitochondria and lysosome represent a common hallmark of neurodegenerative diseases: targeting their communication could open therapeutic avenues.
Collapse
Affiliation(s)
- Flavia Giamogante
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | - Lucia Barazzuol
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | | | - Elena Poggio
- Department of Biology (DIBIO), University of Padova, Padova, Italy
| | - Alessandra Esposito
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Anna Masato
- Department of Biology (DIBIO), University of Padova, Padova, Italy
- UK-Dementia Research Institute at UCL, University College London, London, UK
| | - Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Alessio Vagnoni
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Tito Calì
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy.
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.
- Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy.
| | - Marisa Brini
- Department of Biology (DIBIO), University of Padova, Padova, Italy.
- Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy.
- Department of Pharmaceutical and Pharmacological Sciences (DSF), University of Padova, Padova, Italy.
| |
Collapse
|
42
|
Lu B, Chen X, Ma Y, Gui M, Yao L, Li J, Wang M, Zhou X, Fu D. So close, yet so far away: the relationship between MAM and cardiac disease. Front Cardiovasc Med 2024; 11:1353533. [PMID: 38374992 PMCID: PMC10875081 DOI: 10.3389/fcvm.2024.1353533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Mitochondria-associated membrane (MAM) serve as crucial contact sites between mitochondria and the endoplasmic reticulum (ER). Recent research has highlighted the significance of MAM, which serve as a platform for various protein molecules, in processes such as calcium signaling, ATP production, mitochondrial structure and function, and autophagy. Cardiac diseases caused by any reason can lead to changes in myocardial structure and function, significantly impacting human health. Notably, MAM exhibits various regulatory effects to maintain cellular balance in several cardiac diseases conditions, such as obesity, diabetes mellitus, and cardiotoxicity. MAM proteins independently or interact with their counterparts, forming essential tethers between the ER and mitochondria in cardiomyocytes. This review provides an overview of key MAM regulators, detailing their structure and functions. Additionally, it explores the connection between MAM and various cardiac injuries, suggesting that precise genetic, pharmacological, and physical regulation of MAM may be a promising strategy for preventing and treating heart failure.
Collapse
Affiliation(s)
- Bo Lu
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Xiaozhe Chen
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yulong Ma
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingtai Gui
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Yao
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianhua Li
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingzhu Wang
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xunjie Zhou
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Deyu Fu
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
43
|
Obara CJ, Nixon-Abell J, Moore AS, Riccio F, Hoffman DP, Shtengel G, Xu CS, Schaefer K, Pasolli HA, Masson JB, Hess HF, Calderon CP, Blackstone C, Lippincott-Schwartz J. Motion of VAPB molecules reveals ER-mitochondria contact site subdomains. Nature 2024; 626:169-176. [PMID: 38267577 PMCID: PMC10830423 DOI: 10.1038/s41586-023-06956-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 12/08/2023] [Indexed: 01/26/2024]
Abstract
To coordinate cellular physiology, eukaryotic cells rely on the rapid exchange of molecules at specialized organelle-organelle contact sites1,2. Endoplasmic reticulum-mitochondrial contact sites (ERMCSs) are particularly vital communication hubs, playing key roles in the exchange of signalling molecules, lipids and metabolites3,4. ERMCSs are maintained by interactions between complementary tethering molecules on the surface of each organelle5,6. However, due to the extreme sensitivity of these membrane interfaces to experimental perturbation7,8, a clear understanding of their nanoscale organization and regulation is still lacking. Here we combine three-dimensional electron microscopy with high-speed molecular tracking of a model organelle tether, Vesicle-associated membrane protein (VAMP)-associated protein B (VAPB), to map the structure and diffusion landscape of ERMCSs. We uncovered dynamic subdomains within VAPB contact sites that correlate with ER membrane curvature and undergo rapid remodelling. We show that VAPB molecules enter and leave ERMCSs within seconds, despite the contact site itself remaining stable over much longer time scales. This metastability allows ERMCSs to remodel with changes in the physiological environment to accommodate metabolic needs of the cell. An amyotrophic lateral sclerosis-associated mutation in VAPB perturbs these subdomains, likely impairing their remodelling capacity and resulting in impaired interorganelle communication. These results establish high-speed single-molecule imaging as a new tool for mapping the structure of contact site interfaces and reveal that the diffusion landscape of VAPB at contact sites is a crucial component of ERMCS homeostasis.
Collapse
Affiliation(s)
| | - Jonathon Nixon-Abell
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
- Cambridge Institute for Medical Research (CIMR), Cambridge, UK
| | - Andrew S Moore
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Federica Riccio
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
- Centre for Gene Therapy & Regenerative Medicine, King's College London, London, UK
| | - David P Hoffman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- 10x Genomics, Pleasanton, CA, USA
| | - Gleb Shtengel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - C Shan Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kathy Schaefer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - H Amalia Pasolli
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jean-Baptiste Masson
- Decision and Bayesian Computation, Neuroscience, & Computational Biology Departments, CNRS UMR 3751, Institut Pasteur, Université de Paris, Paris, France
| | - Harald F Hess
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Christopher P Calderon
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
- Ursa Analytics, Inc., Denver, CO, USA
| | - Craig Blackstone
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
44
|
Ahlstedt BA, Ganji R, Mukkavalli S, Paulo JA, Gygi SP, Raman M. UBXN1 maintains ER proteostasis and represses UPR activation by modulating translation. EMBO Rep 2024; 25:672-703. [PMID: 38177917 PMCID: PMC10897191 DOI: 10.1038/s44319-023-00027-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
ER protein homeostasis (proteostasis) is essential for proper folding and maturation of proteins in the secretory pathway. Loss of ER proteostasis can lead to the accumulation of misfolded or aberrant proteins in the ER and triggers the unfolded protein response (UPR). In this study, we find that the p97 adaptor UBXN1 is an important negative regulator of the UPR. Loss of UBXN1 sensitizes cells to ER stress and activates the UPR. This leads to widespread upregulation of the ER stress transcriptional program. Using comparative, quantitative proteomics we show that deletion of UBXN1 results in a significant enrichment of proteins involved in ER-quality control processes including those involved in protein folding and import. Notably, we find that loss of UBXN1 does not perturb p97-dependent ER-associated degradation (ERAD). Our studies indicate that loss of UBXN1 increases translation in both resting and ER-stressed cells. Surprisingly, this process is independent of p97 function. Taken together, our studies have identified a new role for UBXN1 in repressing translation and maintaining ER proteostasis in a p97 independent manner.
Collapse
Affiliation(s)
- Brittany A Ahlstedt
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
- ALPCA diagnostics, Salem, NH, USA
| | - Rakesh Ganji
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Sirisha Mukkavalli
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
- Dana Farber Cancer Research Institute, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology Harvard Medical School, Boston, MA, USA
| | - Steve P Gygi
- Department of Cell Biology Harvard Medical School, Boston, MA, USA
| | - Malavika Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
45
|
Paredes F, Navarro-Marquez M, Quiroga C, Jiménez-Gallegos D, Yeligar SM, Parra V, Müller M, Chiong M, Quest AFG, San Martin A, Lavandero S. HERPUD1 governs tumor cell mitochondrial function via inositol 1,4,5-trisphosphate receptor-mediated calcium signaling. Free Radic Biol Med 2024; 211:24-34. [PMID: 38043868 DOI: 10.1016/j.freeradbiomed.2023.11.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
The intricate relationship between calcium (Ca2+) homeostasis and mitochondrial function is crucial for cellular metabolic adaptation in tumor cells. Ca2+-initiated signaling maintains mitochondrial respiratory capacity and ATP synthesis, influencing critical cellular processes in cancer development. Previous studies by our group have shown that the homocysteine-inducible ER Protein with Ubiquitin-Like Domain 1 (HERPUD1) regulates inositol 1,4,5-trisphosphate receptor (ITPR3) levels and intracellular Ca2+ signals in tumor cells. This study explores the role of HERPUD1 in regulating mitochondrial function and tumor cell migration by controlling ITPR3-dependent Ca2+ signals. We found HERPUD1 levels correlated with mitochondrial function in tumor cells, with HERPUD1 deficiency leading to enhanced mitochondrial activity. HERPUD1 knockdown increased intracellular Ca2+ release and mitochondrial Ca2+ influx, which was prevented using the ITPR3 antagonist xestospongin C or the Ca2+ chelator BAPTA-AM. Furthermore, HERPUD1 expression reduced tumor cell migration by controlling ITPR3-mediated Ca2+ signals. HERPUD1-deficient cells exhibited increased migratory capacity, which was attenuated by treatment with xestospongin C or BAPTA-AM. Additionally, HERPUD1 deficiency led to reactive oxygen species-dependent activation of paxillin and FAK proteins, which are associated with enhanced cell migration. Our findings highlight the pivotal role of HERPUD1 in regulating mitochondrial function and cell migration by controlling intracellular Ca2+ signals mediated by ITPR3. Understanding the interplay between HERPUD1 and mitochondrial Ca2+ regulation provides insights into potential therapeutic targets for cancer treatment and other pathologies involving altered energy metabolism.
Collapse
Affiliation(s)
- Felipe Paredes
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, USA
| | - Mario Navarro-Marquez
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Clara Quiroga
- Advanced Center for Chronic Diseases, Division de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Danica Jiménez-Gallegos
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Samantha M Yeligar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA; Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - Valentina Parra
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alejandra San Martin
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, USA; Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile; Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
46
|
Makio T, Simmen T. Not So Rare: Diseases Based on Mutant Proteins Controlling Endoplasmic Reticulum-Mitochondria Contact (MERC) Tethering. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241261228. [PMID: 39070058 PMCID: PMC11273598 DOI: 10.1177/25152564241261228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/30/2024]
Abstract
Mitochondria-endoplasmic reticulum contacts (MERCs), also called endoplasmic reticulum (ER)-mitochondria contact sites (ERMCS), are the membrane domains, where these two organelles exchange lipids, Ca2+ ions, and reactive oxygen species. This crosstalk is a major determinant of cell metabolism, since it allows the ER to control mitochondrial oxidative phosphorylation and the Krebs cycle, while conversely, it allows the mitochondria to provide sufficient ATP to control ER proteostasis. MERC metabolic signaling is under the control of tethers and a multitude of regulatory proteins. Many of these proteins have recently been discovered to give rise to rare diseases if their genes are mutated. Surprisingly, these diseases share important hallmarks and cause neurological defects, sometimes paired with, or replaced by skeletal muscle deficiency. Typical symptoms include developmental delay, intellectual disability, facial dysmorphism and ophthalmologic defects. Seizures, epilepsy, deafness, ataxia, or peripheral neuropathy can also occur upon mutation of a MERC protein. Given that most MERC tethers and regulatory proteins have secondary functions, some MERC protein-based diseases do not fit into this categorization. Typically, however, the proteins affected in those diseases have dominant functions unrelated to their roles in MERCs tethering or their regulation. We are discussing avenues to pharmacologically target genetic diseases leading to MERC defects, based on our novel insight that MERC defects lead to common characteristics in rare diseases. These shared characteristics of MERCs disorders raise the hope that they may allow for similar treatment options.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
47
|
Kohler A, Kohler V. Better Together: Interorganellar Communication in the Regulation of Proteostasis. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241272245. [PMID: 39385949 PMCID: PMC11462569 DOI: 10.1177/25152564241272245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 10/12/2024]
Abstract
An extensive network of chaperones and folding factors is responsible for maintaining a functional proteome, which is the basis for cellular life. The underlying proteostatic mechanisms are not isolated within organelles, rather they are connected over organellar borders via signalling processes or direct association via contact sites. This review aims to provide a conceptual understanding of proteostatic mechanisms across organelle borders, not focussing on individual organelles. This discussion highlights the precision of these finely tuned systems, emphasising the complicated balance between cellular protection and adaptation to stress. In this review, we discuss widely accepted aspects while shedding light on newly discovered perspectives.
Collapse
Affiliation(s)
- Andreas Kohler
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87 Umeå, Sweden
| | - Verena Kohler
- Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
48
|
Caron C, Bertolin G. Cristae shaping and dynamics in mitochondrial function. J Cell Sci 2024; 137:jcs260986. [PMID: 38197774 DOI: 10.1242/jcs.260986] [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: 01/11/2024] Open
Abstract
Mitochondria are multifunctional organelles of key importance for cell homeostasis. The outer mitochondrial membrane (OMM) envelops the organelle, and the inner mitochondrial membrane (IMM) is folded into invaginations called cristae. As cristae composition and functions depend on the cell type and stress conditions, they recently started to be considered as a dynamic compartment. A number of proteins are known to play a role in cristae architecture, such as OPA1, MIC60, LETM1, the prohibitin (PHB) complex and the F1FO ATP synthase. Furthermore, phospholipids are involved in the maintenance of cristae ultrastructure and dynamics. The use of new technologies, including super-resolution microscopy to visualize cristae dynamics with superior spatiotemporal resolution, as well as high-content techniques and datasets have not only allowed the identification of new cristae proteins but also helped to explore cristae plasticity. However, a number of open questions remain in the field, such as whether cristae-resident proteins are capable of changing localization within mitochondria, or whether mitochondrial proteins can exit mitochondria through export. In this Review, we present the current view on cristae morphology, stability and composition, and address important outstanding issues that might pave the way to future discoveries.
Collapse
Affiliation(s)
- Claire Caron
- Univ. Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France
| | - Giulia Bertolin
- Univ. Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France
| |
Collapse
|
49
|
Tong D, Zhou J, Zhou J, Wang X, Gao B, Rui X, Liu L, Chen Q, Huang C. LAMC2 mitigates ER stress by enhancing ER-mitochondria interaction via binding to MYH9 and MYH10. Cancer Gene Ther 2024; 31:43-57. [PMID: 37891404 PMCID: PMC10794146 DOI: 10.1038/s41417-023-00680-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/01/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023]
Abstract
Highly proliferative and metastatic tumors are constantly exposed to both intrinsic and extrinsic factors that induce adaptation to stressful conditions. Chronic adaptation to endoplasmic reticulum (ER) ER stress is common to many different types of cancers, and poses a major challenge for acquired drug resistance. Here we report that LAMC2, an extracellular matrix protein upregulated in many types of cancers, is localized in the ER of lung, breast, and liver cancer cells. Under tunicamycin-induced ER stress, protein level of LAMC2 is upregulated. Transfection of cancer cells with LAMC2 resulted in the attenuation of ER stress phenotype, accompanied by elevation in mitochondrial membrane potential as well as reduction in reactive oxygen species (ROS) levels and apoptosis. In addition, LAMC2 forms protein complexes with MYH9 and MYH10 to promote mitochondrial aggregation and increased ER-mitochondria interaction at the perinuclear region. Moreover, overexpression of LAMC2 counteracts the effects of ER stress and promotes tumor growth in vivo. Taken together, our results revealed that in complex with MYH9 and MYH10, LAMC2 is essential for promoting ER-mitochondria interaction to alleviate ER stress and allow cancer cells to adapt and proliferate under stressful conditions. This study provides new insights and highlights the promising potential of LAMC2 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Dongdong Tong
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Jun Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Jing Zhou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Xiaofei Wang
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Beibei Gao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Xiaoyi Rui
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Liying Liu
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - QiaoYi Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China.
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China.
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, 710061, Xi'an, China.
| |
Collapse
|
50
|
Zhang J, Zhang Z, Wang X, Liu Y, Yu Q, Wang K, Fang Y, Lenahan C, Chen M, Chen S. Connection between oxidative stress and subcellular organelle in subarachnoid hemorrhage: Novel mechanisms and therapeutic implications. CNS Neurosci Ther 2023; 29:3672-3683. [PMID: 37408392 PMCID: PMC10651993 DOI: 10.1111/cns.14348] [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: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/24/2023] [Indexed: 07/07/2023] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH) is one of the most devastating forms of stroke, with limited treatment modalities and poor patient outcomes. Previous studies have proposed multiple prognostic factors; however, relative research on treatment has not yet yielded favorable clinical outcomes. Moreover, recent studies have suggested that early brain injury (EBI) occurring within 72 h after SAH may contribute to its poor clinical outcomes. Oxidative stress is recognized as one of the main mechanisms of EBI, which causes damage to various subcellular organelles, including the mitochondria, nucleus, endoplasmic reticulum (ER), and lysosomes. This could lead to significant impairment of numerous cellular functions, such as energy supply, protein synthesis, and autophagy, which may directly contribute to the development of EBI and poor long-term prognostic outcomes. In this review, the mechanisms underlying the connection between oxidative stress and subcellular organelles after SAH are discussed, and promising therapeutic options based on these mechanisms are summarized.
Collapse
Affiliation(s)
- Jiahao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Zeyu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Qian Yu
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Kaikai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Cameron Lenahan
- Center for Neuroscience ResearchLoma Linda University School of MedicineLoma LindaCaliforniaUSA
| | - Maohua Chen
- Department of Neurosurgery, Wenzhou Central HospitalAffiliated Dingli Clinical Institute of Wenzhou Medical UniversityWenzhouChina
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| |
Collapse
|