1
|
Cheng L, Zheng Q, Qiu K, Elmer Ker DF, Chen X, Yin Z. Mitochondrial destabilization in tendinopathy and potential therapeutic strategies. J Orthop Translat 2024; 49:49-61. [PMID: 39430132 PMCID: PMC11488423 DOI: 10.1016/j.jot.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 10/22/2024] Open
Abstract
Tendinopathy is a prevalent aging-related disorder characterized by pain, swelling, and impaired function, often resulting from micro-scarring and degeneration caused by overuse or trauma. Current interventions for tendinopathy have limited efficacy, highlighting the need for innovative therapies. Mitochondria play an underappreciated and yet crucial role in tenocytes function, including energy production, redox homeostasis, autophagy, and calcium regulation. Abnormalities in mitochondrial function may lead to cellular senescence. Within this context, this review provides an overview of the physiological functions of mitochondria in tendons and presents current insights into mitochondrial dysfunction in tendinopathy. It also proposes potential therapeutic strategies that focus on targeting mitochondrial health in tenocytes. These strategies include: (1) utilizing reactive oxygen species (ROS) scavengers to mitigate the detrimental effects of aberrant mitochondria, (2) employing mitochondria-protecting agents to reduce the production of dysfunctional mitochondria, and (3) supplementing with exogenous normal mitochondria. In conclusion, mitochondria-targeted therapies hold great promise for restoring mitochondrial function and improving outcomes in patients with tendinopathy. The translational potential of this article: Tendinopathy is challenging to treat effectively due to its poorly understood pathogenesis. This review thoroughly analyzes the role of mitochondria in tenocytes and proposes potential strategies for the mitochondrial treatment of tendinopathy. These findings establish a theoretical basis for future research and the clinical translation of mitochondrial therapy for tendinopathy.
Collapse
Affiliation(s)
- Linxiang Cheng
- Department of Orthopedic Surgery of Sir Run Run Shaw Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Qiangqiang Zheng
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, And Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Kaijie Qiu
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, And Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Xiao Chen
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, And Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Zi Yin
- Department of Orthopedic Surgery of Sir Run Run Shaw Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| |
Collapse
|
2
|
Qi LFR, Liu Y, Liu S, Xiang L, Liu Z, Liu Q, Zhao JQ, Xu X. Phillyrin promotes autophagosome formation in A53T-αSyn-induced Parkinson's disease model via modulation of REEP1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155952. [PMID: 39178680 DOI: 10.1016/j.phymed.2024.155952] [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: 04/15/2024] [Revised: 06/30/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND The preservation of autophagosome formation presents a promising strategy for tackling neurological disorders, such as Parkinson's disease (PD). Mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) serve not only as a focal point linked to various neurological disorders but also play a crucial role in supporting the biogenesis of autophagosomes. PURPOSE This investigation aimed to elucidate the neuroprotective properties of phillyrin against PD and its underlying mechanisms in promoting autophagosome formation. METHODS ER and mitochondria co-localization was assessed via fluorescent staining. Annexin V-fluorescein isothiocyanate (FITC) fluorescence was employed to quantify accessible cardiolipin (CL) on mitochondrial surfaces. The levels of CL within the MAM fraction of SH-SY5Y cells were evaluated using a CL probe assay kit. Monodansylcadaverine staining was utilized to detect autophagosome formation in SH-SY5Y cells. In an A53T-alpha-synuclein (αSyn)-induced PD mouse model, the anti-PD properties of phillyrin were assessed using open field, pole climbing, and rotarod tests, as well as immunohistochemistry staining of TH+ neurons in the brain sections. RESULTS In A53T-αSyn-treated SH-SY5Y cells, phillyrin facilitated autophagosome formation by suppressing CL externalization and restoring MAM integrity. Phillyrin enhanced the localization of receptor expression-enhancing protein 1 (REEP1) within MAM and mitochondria, bolstering MAM formation. Increased REEP1 levels in mitochondria, attributed to phillyrin, enhanced the interaction between REEP1 and NDPK-D, thereby reducing CL externalization. Furthermore, phillyrin exhibited a dose-dependent enhancement of motor function in mice, accompanied by an increase in the abundance of dopaminergic neurons within the substantia nigra. CONCLUSIONS These findings illuminate phillyrin's ability to enhance MAM formation through upregulation of REEP1 expression within MAM, while concurrently attenuating CL externalization via the REEP1-NDPK-D interaction. These mechanisms bolster autophagosome biogenesis, offering resilience against A53T-αSyn-induced PD. Thus, our study advances the understanding of phillyrin's complex mechanisms and underscores its potential as a therapeutic approach for PD, opening new avenues in natural product pharmacology.
Collapse
Affiliation(s)
- Li-Feng-Rong Qi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| | - Yuci Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shuai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lin Xiang
- Department of Translational Medicine Research Institute, Jiangsu Yifengrong Biotechnology Co., Ltd., Nanjing, Jiangsu, China
| | - Zhiyuan Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qingling Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Jin-Quan Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Pharmacy, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China; Center for Innovative Traditional Chinese Medicine Target and New Drug Research, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China.
| |
Collapse
|
3
|
Hoenger Ramazanova RD, Roumeliotis TI, Wright JC, Choudhary JS. PhoXplex: Combining Phospho-enrichable Cross-Linking with Isobaric Labeling for Quantitative Proteome-Wide Mapping of Protein Interfaces. J Proteome Res 2024; 23:5209-5220. [PMID: 39422127 PMCID: PMC11537259 DOI: 10.1021/acs.jproteome.4c00567] [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: 07/04/2024] [Revised: 09/07/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Integrating cross-linking mass spectrometry (XL-MS) into structural biology workflows provides valuable information about the spatial arrangement of amino acid stretches, which can guide elucidation of protein assembly architecture. Additionally, the combination of XL-MS with peptide quantitation techniques is a powerful approach to delineate protein interface dynamics across diverse conditions. While XL-MS is increasingly effective with isolated proteins or small complexes, its application to whole-cell samples poses technical challenges related to analysis depth and throughput. The use of enrichable cross-linkers has greatly improved the detectability of protein interfaces in a proteome-wide scale, facilitating global protein-protein interaction mapping. Therefore, bringing together enrichable cross-linking and multiplexed peptide quantification is an appealing approach to enable comparative characterization of structural attributes of proteins and protein interactions. Here, we combined phospho-enrichable cross-linking with TMT labeling to develop a streamline workflow (PhoXplex) for the detection of differential structural features across a panel of cell lines in a global scale. We achieved deep coverage with quantification of over 9000 cross-links and long loop-links in total including potentially novel interactions. Overlaying AlphaFold predictions and disorder protein annotations enables exploration of the quantitative cross-linking data set, to reveal possible associations between mutations and protein structures. Lastly, we discuss current shortcomings and perspectives for deep whole-cell profiling of protein interfaces at large-scale.
Collapse
Affiliation(s)
- Runa D. Hoenger Ramazanova
- Functional
Proteomics team, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, United Kingdom
| | - Theodoros I. Roumeliotis
- Functional
Proteomics team, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, United Kingdom
| | - James C. Wright
- Functional
Proteomics team, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, United Kingdom
| | - Jyoti S. Choudhary
- Functional
Proteomics team, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, United Kingdom
| |
Collapse
|
4
|
Zhu S, Liu X, Lu X, Liao Q, Luo H, Tian Y, Cheng X, Jiang Y, Liu G, Chen J. Biomaterials and tissue engineering in traumatic brain injury: novel perspectives on promoting neural regeneration. Neural Regen Res 2024; 19:2157-2174. [PMID: 38488550 PMCID: PMC11034597 DOI: 10.4103/1673-5374.391179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/13/2023] [Accepted: 11/20/2023] [Indexed: 04/24/2024] Open
Abstract
Traumatic brain injury is a serious medical condition that can be attributed to falls, motor vehicle accidents, sports injuries and acts of violence, causing a series of neural injuries and neuropsychiatric symptoms. However, limited accessibility to the injury sites, complicated histological and anatomical structure, intricate cellular and extracellular milieu, lack of regenerative capacity in the native cells, vast variety of damage routes, and the insufficient time available for treatment have restricted the widespread application of several therapeutic methods in cases of central nervous system injury. Tissue engineering and regenerative medicine have emerged as innovative approaches in the field of nerve regeneration. By combining biomaterials, stem cells, and growth factors, these approaches have provided a platform for developing effective treatments for neural injuries, which can offer the potential to restore neural function, improve patient outcomes, and reduce the need for drugs and invasive surgical procedures. Biomaterials have shown advantages in promoting neural development, inhibiting glial scar formation, and providing a suitable biomimetic neural microenvironment, which makes their application promising in the field of neural regeneration. For instance, bioactive scaffolds loaded with stem cells can provide a biocompatible and biodegradable milieu. Furthermore, stem cells-derived exosomes combine the advantages of stem cells, avoid the risk of immune rejection, cooperate with biomaterials to enhance their biological functions, and exert stable functions, thereby inducing angiogenesis and neural regeneration in patients with traumatic brain injury and promoting the recovery of brain function. Unfortunately, biomaterials have shown positive effects in the laboratory, but when similar materials are used in clinical studies of human central nervous system regeneration, their efficacy is unsatisfactory. Here, we review the characteristics and properties of various bioactive materials, followed by the introduction of applications based on biochemistry and cell molecules, and discuss the emerging role of biomaterials in promoting neural regeneration. Further, we summarize the adaptive biomaterials infused with exosomes produced from stem cells and stem cells themselves for the treatment of traumatic brain injury. Finally, we present the main limitations of biomaterials for the treatment of traumatic brain injury and offer insights into their future potential.
Collapse
Affiliation(s)
- Shihong Zhu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoyin Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiyue Lu
- Department of Anesthesiology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiang Liao
- Department of Pharmacy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Huiyang Luo
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- Department of Anesthesiology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Tian
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xu Cheng
- Department of Anesthesiology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Yaxin Jiang
- Out-patient Department, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Guangdi Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Jing Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
5
|
Qi L, Liu S, Fang Q, Qian C, Peng C, Liu Y, Yang P, Wu P, Shan L, Cui Q, Hua Q, Yang S, Ye C, Yang W, Li P, Xu X. Ginsenoside Rg3 Restores Mitochondrial Cardiolipin Homeostasis via GRB2 to Prevent Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403058. [PMID: 39159293 PMCID: PMC11497058 DOI: 10.1002/advs.202403058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/20/2024] [Indexed: 08/21/2024]
Abstract
Regulating cardiolipin to maintain mitochondrial homeostasis is a promising strategy for addressing Parkinson's disease (PD). Through a comprehensive screening and validation process involving multiple models, ginsenoside Rg3 (Rg3) as a compound capable of enhancing cardiolipin levels is identified. This augmentation in cardiolipin levels fosters mitochondrial homeostasis by bolstering mitochondrial unfolded protein response, promoting mitophagy, and enhancing mitochondrial oxidative phosphorylation. Consequently, this cascade enhances the survival of tyrosine hydroxylase positive (TH+) dopaminergic neurons, leading to an amelioration in motor performance within PD mouse models. Using limited proteolysis-small-molecule mapping combined with molecular docking analysis, it has confirmed Growth Factor Receptor-Bound Protein 2 (GRB2) as a molecular target for Rg3. Furthermore, these investigations reveal that Rg3 facilitates the interaction between GRB2 and TRKA (Neurotrophic Tyrosine Kinase, Receptor, Type 1), thus promotes EVI1 (Ecotropic Virus Integration Site 1 Protein Homolog) phosphorylation by ERK, subsequently increases CRLS1 (Cardiolipin Synthase 1) gene expression and boosts cardiolipin synthesis. The absence of GRB2 or CRLS1 significantly attenuates the beneficial effects of Rg3 on PD symptoms. Finally, Tenofovir Disoproxil Fumarate (TDF) that also promotes the binding between GRB2 and TRKA is further identified. The identified compounds, Rg3 and TDF, exhibit promising potential for the prevention of PD by bolstering cardiolipin expression and reinstating mitochondrial homeostasis.
Collapse
Affiliation(s)
- Li‐Feng‐Rong Qi
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Shuai Liu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| | - Qiuyuan Fang
- Department of Biophysics and Department of Neurosurgery of the First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Cheng Qian
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Chao Peng
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai Advanced Research InstituteChinese Academy of ScienceShanghai201210China
- Shanghai Science Research CenterChinese Academy of SciencesShanghai201204China
| | - Yuci Liu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Peng Yang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Ping Wu
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai Advanced Research InstituteChinese Academy of ScienceShanghai201210China
- Shanghai Science Research CenterChinese Academy of SciencesShanghai201204China
| | - Ling Shan
- Dept. Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesMeibergdreef 47Amsterdam1105BAthe Netherlands
| | - Qinghua Cui
- Department of Biomedical InformaticsSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Sciences of the Ministry of EducationCenter for Non‐Coding RNA MedicinePeking University Health Science Center BeijingBeijing100191China
| | - Qian Hua
- School of Life SciencesBeijing University of Chinese MedicineBeijing100029China
| | - Sen Yang
- Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Cunqi Ye
- Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Wei Yang
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| | - Ping Li
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Xiaojun Xu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| |
Collapse
|
6
|
Guo Z, Tian Y, Liu N, Chen Y, Chen X, Yuan G, Chang A, Chang X, Wu J, Zhou H. Mitochondrial Stress as a Central Player in the Pathogenesis of Hypoxia-Related Myocardial Dysfunction: New Insights. Int J Med Sci 2024; 21:2502-2509. [PMID: 39439461 PMCID: PMC11492880 DOI: 10.7150/ijms.99359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Hypoxic injury is a critical pathological factor in the development of various cardiovascular diseases, such as congenital heart disease, myocardial infarction, and heart failure. Mitochondrial quality control is essential for protecting cardiomyocytes from hypoxic damage. Under hypoxic conditions, disruptions in mitochondrial homeostasis result in excessive reactive oxygen species (ROS) production, imbalances in mitochondrial dynamics, and initiate pathological processes including oxidative stress, inflammatory responses, and apoptosis. Targeted interventions to enhance mitochondrial quality control, such as coenzyme Q10 and statins, have shown promise in mitigating hypoxia-induced mitochondrial dysfunction. These treatments offer potential therapeutic strategies for hypoxia-related cardiovascular diseases by regulating mitochondrial fission and fusion, restoring mitochondrial biogenesis, reducing ROS production, and promoting mitophagy.
Collapse
Affiliation(s)
- Zhijiang Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yingjie Tian
- Beijing University of Chinese Medicine, Beijing, 100028, China
| | - Nanyang Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ye Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaohan Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Guoxing Yuan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - An Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jie Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
7
|
Decker ST, Funai K. Mitochondrial membrane lipids in the regulation of bioenergetic flux. Cell Metab 2024; 36:1963-1978. [PMID: 39178855 PMCID: PMC11374467 DOI: 10.1016/j.cmet.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/12/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Oxidative phosphorylation (OXPHOS) occurs through and across the inner mitochondrial membrane (IMM). Mitochondrial membranes contain a distinct lipid composition, aided by lipid biosynthetic machinery localized in the IMM and class-specific lipid transporters that limit lipid traffic in and out of mitochondria. This unique lipid composition appears to be essential for functions of mitochondria, particularly OXPHOS, by its effects on direct lipid-to-protein interactions, membrane properties, and cristae ultrastructure. This review highlights the biological significance of mitochondrial lipids, with a particular spotlight on the role of lipids in mitochondrial bioenergetics. We describe pathways for the biosynthesis of mitochondrial lipids and provide evidence for their roles in physiology, their implications in human disease, and the mechanisms by which they regulate mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Stephen Thomas Decker
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
8
|
Hong WL, Huang H, Zeng X, Duan CY. Targeting mitochondrial quality control: new therapeutic strategies for major diseases. Mil Med Res 2024; 11:59. [PMID: 39164792 PMCID: PMC11337860 DOI: 10.1186/s40779-024-00556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 07/13/2024] [Indexed: 08/22/2024] Open
Abstract
Mitochondria play a crucial role in maintaining the normal physiological state of cells. Hence, ensuring mitochondrial quality control is imperative for the prevention and treatment of numerous diseases. Previous reviews on this topic have however been inconsistencies and lack of systematic organization. Therefore, this review aims to provide a comprehensive and systematic overview of mitochondrial quality control and explore the possibility of targeting the same for the treatment of major diseases. This review systematically summarizes three fundamental characteristics of mitochondrial quality control, including mitochondrial morphology and dynamics, function and metabolism, and protein expression and regulation. It also extensively examines how imbalances in mitochondrial quality are linked to major diseases, such as ischemia-hypoxia, inflammatory disorders, viral infections, metabolic dysregulations, degenerative conditions, and tumors. Additionally, the review explores innovative approaches to target mitochondrial quality control, including using small molecule drugs that regulate critical steps in maintaining mitochondrial quality, nanomolecular materials designed for precise targeting of mitochondria, and novel cellular therapies, such as vesicle therapy and mitochondrial transplantation. This review offers a novel perspective on comprehending the shared mechanisms underlying the occurrence and progression of major diseases and provides theoretical support and practical guidance for the clinical implementation of innovative therapeutic strategies that target mitochondrial quality control for treating major diseases.
Collapse
Affiliation(s)
- Wei-Long Hong
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - He Huang
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xue Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Chen-Yang Duan
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
9
|
Liu X, Wang FY, Chi S, Liu T, Yang HL, Zhong RJ, Li XY, Gao J. Mitochondria-targeting peptide SS-31 attenuates ferroptosis via inhibition of the p38 MAPK signaling pathway in the hippocampus of epileptic rats. Brain Res 2024; 1836:148882. [PMID: 38521160 DOI: 10.1016/j.brainres.2024.148882] [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: 01/01/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Ferroptosis is a newly identified form of non-apoptotic regulated cell death (RCD) andplaysanimportantrole in epileptogenesis. The p38 mitogen-activated protein kinase (p38 MAPK) pathway has been confirmed to be involved in ferroptosis. The mitochondria-targeting antioxidant Elamipretide (SS-31) can reduce the generation of lipid peroxidation and the buildup of reactive oxygen species (ROS). Collectively, our present study was to decipher whether SS-31 inhibits ferroptosis via the p38 MAPK signaling pathway in the rat epilepsy model induced by pilocarpine (PILO).Adult male Wistar rats were randomly divided into four groups: control group (CON group), epilepsy group (EP group), SS-31 treatment group (SS group), and p38 MAPK inhibitor (SB203580) treatment group (SB group). Our results demonstrated that the rat hippocampal neurons after epilepsy were followed by accumulated iron and malondialdehyde (MDA) content, upregulated phosphorylated p38 MAPK protein (P-p38) and nuclear factor erythroid 2-related factor 2 (Nrf2) levels, reduced glutathione peroxidase 4 (Gpx4) content, and depleted glutathione (GSH) activity. Morphologically, mitochondrial ultrastructural damage under electron microscopy was manifested by a partial increase in outer membrane density, disappearance of mitochondrial cristae, and mitochondrial shrinkage. SS-31 and SB203580 treatment blocked the initiation and progression of ferroptosis in the hippocampus of epileptic rats via reducing the severity of epileptic seizures, reversing the expression of Gpx4, P-p38 , decreasing the levels of iron and MDA, as well as increasing the activity of GSH and Nrf2. To summarize, our findings proved that ferroptosis was coupled with the pathology of epilepsy, and SS-31 can inhibit PILO-induced seizures by preventing ferroptosis, which may be connected to the inhibition of p38 MAPK phosphorylation, highlighting the potential therapeutic value for targeting ferroptosis process in individuals with seizure-related diseases.
Collapse
Affiliation(s)
- Xue Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Fei-Yu Wang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Song Chi
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Tao Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Hai-Lin Yang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Ru-Jie Zhong
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xiao-Yu Li
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
10
|
van Hameren G, Aboghazleh R, Parker E, Dreier JP, Kaufer D, Friedman A. From spreading depolarization to blood-brain barrier dysfunction: navigating traumatic brain injury for novel diagnosis and therapy. Nat Rev Neurol 2024; 20:408-425. [PMID: 38886512 DOI: 10.1038/s41582-024-00973-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Considerable strides in medical interventions during the acute phase of traumatic brain injury (TBI) have brought improved overall survival rates. However, following TBI, people often face ongoing, persistent and debilitating long-term complications. Here, we review the recent literature to propose possible mechanisms that lead from TBI to long-term complications, focusing particularly on the involvement of a compromised blood-brain barrier (BBB). We discuss evidence for the role of spreading depolarization as a key pathological mechanism associated with microvascular dysfunction and the transformation of astrocytes to an inflammatory phenotype. Finally, we summarize new predictive and diagnostic biomarkers and explore potential therapeutic targets for treating long-term complications of TBI.
Collapse
Affiliation(s)
- Gerben van Hameren
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Refat Aboghazleh
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ellen Parker
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Jens P Dreier
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada.
- Department of Cell Biology, Cognitive and Brain Sciences, Zelman Inter-Disciplinary Center of Brain Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
11
|
Zheng CM, Hou YC, Liao MT, Tsai KW, Hu WC, Yeh CC, Lu KC. Potential role of molecular hydrogen therapy on oxidative stress and redox signaling in chronic kidney disease. Biomed Pharmacother 2024; 176:116802. [PMID: 38795643 DOI: 10.1016/j.biopha.2024.116802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Oxidative stress plays a key role in chronic kidney disease (CKD) development and progression, inducing kidney cell damage, inflammation, and fibrosis. However, effective therapeutic interventions to slow down CKD advancement are currently lacking. The multifaceted pharmacological effects of molecular hydrogen (H2) have made it a promising therapeutic avenue. H2 is capable of capturing harmful •OH and ONOO- while maintaining the crucial reactive oxygen species (ROS) involved in cellular signaling. The NRF2-KEAP1 system, which manages cell redox balance, could be used to treat CKD. H2 activates this pathway, fortifying antioxidant defenses and scavenging ROS to counteract oxidative stress. H2 can improve NRF2 signaling by using the Wnt/β-catenin pathway and indirectly activate NRF2-KEAP1 in mitochondria. Additionally, H2 modulates NF-κB activity by regulating cellular redox status, inhibiting MAPK pathways, and maintaining Trx levels. Treatment with H2 also attenuates HIF signaling by neutralizing ROS while indirectly bolstering HIF-1α function. Furthermore, H2 affects FOXO factors and enhances the activity of antioxidant enzymes. Despite the encouraging results of bench studies, clinical trials are still limited and require further investigation. The focus of this review is on hydrogen's role in treating renal diseases, with a specific focus on oxidative stress and redox signaling regulation, and it discusses its potential clinical applications.
Collapse
Affiliation(s)
- Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, School of Medicine, College of Medicine, Taipei Medical University, New Taipei City 11031, Taiwan; TMU Research Centre of Urology and Kidney, Taipei Medical University, New Taipei City 11031, Taiwan
| | - Yi-Chou Hou
- Division of Nephrology, Department of Internal Medicine, Cardinal-Tien Hospital, School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan; Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Wang Tsai
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Medical Tzu Chi Foundation, New Taipei City 23142, Taiwan
| | - Chien-Chih Yeh
- Division of colon and Rectal Surgery, Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan; National Defense Medical Center, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 24352, Taiwan.
| |
Collapse
|
12
|
Silvaroli JA, Bisunke B, Kim JY, Stayton A, Jayne LA, Martinez SA, Nguyen C, Patel PS, Vanichapol T, Verma V, Akhter J, Bolisetty S, Madhavan SM, Kuscu C, Coss CC, Zepeda-Orozco D, Parikh SV, Satoskar AA, Davidson AJ, Eason JD, Szeto HH, Pabla NS, Bajwa A. Genome-Wide CRISPR Screen Identifies Phospholipid Scramblase 3 as the Biological Target of Mitoprotective Drug SS-31. J Am Soc Nephrol 2024; 35:681-695. [PMID: 38530359 PMCID: PMC11164119 DOI: 10.1681/asn.0000000000000338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
Key Points Szeto–Schiller-31–mediated mitoprotection is phospholipid scramblase 3–dependent. Phospholipid scramblase 3 is required for recovery after AKI. Background The synthetic tetrapeptide Szeto–Schiller (SS)-31 shows promise in alleviating mitochondrial dysfunction associated with common diseases. However, the precise pharmacological basis of its mitoprotective effects remains unknown. Methods To uncover the biological targets of SS-31, we performed a genome-scale clustered regularly interspaced short palindromic repeats screen in human kidney-2, a cell culture model where SS-31 mitigates cisplatin-associated cell death and mitochondrial dysfunction. The identified hit candidate gene was functionally validated using knockout cell lines, small interfering RNA-mediated downregulation, and tubular epithelial–specific conditional knockout mice. Biochemical interaction studies were also performed to examine the interaction of SS-31 with the identified target protein. Results Our primary screen and validation studies in hexokinase 2 and primary murine tubular epithelial cells showed that phospholipid scramblase 3 (PLSCR3), an understudied inner mitochondrial membrane protein, was essential for the protective effects of SS-31. For in vivo validation, we generated tubular epithelial–specific knockout mice and found that Plscr3 gene ablation did not influence kidney function under normal conditions or affect the severity of cisplatin and rhabdomyolysis-associated AKI. However, Plscr3 gene deletion completely abrogated the protective effects of SS-31 during cisplatin and rhabdomyolysis-associated AKI. Biochemical studies showed that SS-31 directly binds to a previously uncharacterized N -terminal domain and stimulates PLSCR3 scramblase activity. Finally, PLSCR3 protein expression was found to be increased in the kidneys of patients with AKI. Conclusions PLSCR3 was identified as the essential biological target that facilitated the mitoprotective effects of SS-31 in vitro and in vivo .
Collapse
Affiliation(s)
- Josie A. Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Bijay Bisunke
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Amanda Stayton
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Laura A. Jayne
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Shirely A. Martinez
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Christopher Nguyen
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Prisha S. Patel
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Thitinee Vanichapol
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Vivek Verma
- Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Juheb Akhter
- Department of Medicine, University of Alabama, Birmingham, Alabama
| | | | - Sethu M. Madhavan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Cem Kuscu
- Department of Surgery, College of Medicine, Transplant Research Institute, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Diana Zepeda-Orozco
- Department of Pediatrics, The Ohio State University College of Medicine and Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Samir V. Parikh
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Anjali A. Satoskar
- Division of Renal and Transplant Pathology, Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - James D. Eason
- Department of Surgery, College of Medicine, Transplant Research Institute, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hazel H. Szeto
- Social Profit Network Research Lab, Menlo Park, California
| | - Navjot S. Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Amandeep Bajwa
- Department of Genetics, Genomics, and Informatics; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Surgery, College of Medicine, Transplant Research Institute, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Microbiology, Immunology, and Biochemistry; College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
13
|
Cui P, Li X, Huang C, Lin D. Metabolomics-driven discovery of therapeutic targets for cancer cachexia. J Cachexia Sarcopenia Muscle 2024; 15:781-793. [PMID: 38644205 PMCID: PMC11154780 DOI: 10.1002/jcsm.13465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/07/2023] [Accepted: 01/09/2024] [Indexed: 04/23/2024] Open
Abstract
Cancer cachexia (CC) is a devastating metabolic syndrome characterized by skeletal muscle wasting and body weight loss, posing a significant burden on the health and survival of cancer patients. Despite ongoing efforts, effective treatments for CC are still lacking. Metabolomics, an advanced omics technique, offers a comprehensive analysis of small-molecule metabolites involved in cellular metabolism. In CC research, metabolomics has emerged as a valuable tool for identifying diagnostic biomarkers, unravelling molecular mechanisms and discovering potential therapeutic targets. A comprehensive search strategy was implemented to retrieve relevant articles from primary databases, including Web of Science, Google Scholar, Scopus and PubMed, for CC and metabolomics. Recent advancements in metabolomics have deepened our understanding of CC by uncovering key metabolic signatures and elucidating underlying mechanisms. By targeting crucial metabolic pathways including glucose metabolism, amino acid metabolism, fatty acid metabolism, bile acid metabolism, ketone body metabolism, steroid metabolism and mitochondrial energy metabolism, it becomes possible to restore metabolic balance and alleviate CC symptoms. This review provides a comprehensive summary of metabolomics studies in CC, focusing on the discovery of potential therapeutic targets and the evaluation of modulating specific metabolic pathways for CC treatment. By harnessing the insights derived from metabolomics, novel interventions for CC can be developed, leading to improved patient outcomes and enhanced quality of life.
Collapse
Affiliation(s)
- Pengfei Cui
- College of Food and PharmacyXuchang UniversityXuchangChina
| | - Xiaoyi Li
- Xuchang Central HospitalXuchangChina
| | - Caihua Huang
- Research and Communication Center of Exercise and HealthXiamen University of TechnologyXiamenChina
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
| |
Collapse
|
14
|
Saleh TA, Whitson JA, Keiser P, Prasad P, Jenkins BC, Sodeinde T, Mann C, Rabinovitch PS, McReynolds MR, Sweetwyne MT. Metabolite accumulation from oral NMN supplementation drives aging-specific kidney inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588624. [PMID: 38645109 PMCID: PMC11030441 DOI: 10.1101/2024.04.09.588624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The mitochondrial-rich renal tubule cells are key regulators of blood homeostasis via excretion and reabsorption of metabolic waste. With age, tubules are subject to increasing mitochondrial dysfunction and declining nicotinamide adenine dinucleotide (NAD+) levels, both hampering ATP production efficiency. We tested two mitochondrial interventions in young (6-mo) and aged (26-mo) adult male mice: elamipretide (ELAM), a tetrapeptide in clinical trials that improves mitochondrial structure and function, and nicotinamide mononucleotide (NMN), an NAD+ intermediate and commercially available oral supplement. Kidneys were analyzed from young and aged mice after eight weeks of treatment with ELAM (3 mg/kg/day), NMN (300 mg/kg/day), or from aged mice treated with the two interventions combined (ELAM+NMN). We hypothesized that combining pharmacologic treatments to ameliorate mitochondrial dysfunction and boost NAD+ levels, would more effectively reduce kidney aging than either intervention alone. Unexpectedly, in aged kidneys, NMN increased expression of genetic markers of inflammation (IL-1-beta; and Ccl2) and tubule injury (Kim-1). Metabolomics of endpoint sera showed that NMN-treated aged mice had higher circulating levels of uremic toxins than either aged controls or young NMN-treated mice. ELAM+NMN-treated aged mice accumulated uremic toxins like NMN-only aged mice, but reduced IL-1-beta; and Ccl2 kidney mRNA. This suggests that pre-existing mitochondrial dysfunction in aged kidney underlies susceptibility to inflammatory signaling with NMN supplementation in aged, but not young, mice. These findings demonstrate age and tissue dependent effects on downstream metabolic accumulation from NMN and highlight the need for targeted analysis of aged kidneys to assess the safety of anti-aging supplements in older populations.
Collapse
|
15
|
Gao L, Zuo XL, Dong LL, Zhou SF, Wang ZJ, Duan YS, Chen MY, Zhu QX, Zhang JX. Hepatocyte mitochondrial DNA mediates macrophage immune response in liver injury induced by trichloroethylene. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116317. [PMID: 38615641 DOI: 10.1016/j.ecoenv.2024.116317] [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: 09/19/2023] [Revised: 03/26/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
We have previously shown that excessive activation of macrophage proinflammatory activity plays a key role in TCE-induced immune liver injury, but the mechanism of polarization is unclear. Recent studies have shown that TLR9 activation plays an important regulatory role in macrophage polarization. In the present study, we demonstrated that elevated levels of oxidative stress in hepatocytes mediate the release of mtDNA into the bloodstream, leading to the activation of TLR9 in macrophages to regulate macrophage polarization. In vivo experiments revealed that pretreatment with SS-31, a mitochondria-targeting antioxidant peptide, reduced the level of oxidative stress in hepatocytes, leading to a decrease in mtDNA release. Importantly, SS-31 pretreatment inhibited TLR9 activation in macrophages, suggesting that hepatocyte mtDNA may activate TLR9 in macrophages. Further studies revealed that pharmacological inhibition of TLR9 by ODN2088 partially blocked macrophage activation, suggesting that the level of macrophage activation is dependent on TLR9 activation. In vitro experiments involving the extraction of mtDNA from TCE-sensitized mice treated with RAW264.7 cells further confirmed that hepatocyte mtDNA can activate TLR9 in mouse peritoneal macrophages, leading to macrophage polarization. In summary, our study comprehensively confirmed that TLR9 activation in macrophages is dependent on mtDNA released by elevated levels of oxidative stress in hepatocytes and that TLR9 activation in macrophages plays a key role in regulating macrophage polarization. These findings reveal the mechanism of macrophage activation in TCE-induced immune liver injury and provide new perspectives and therapeutic targets for the treatment of OMDT-induced immune liver injury.
Collapse
Affiliation(s)
- Lei Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xu-Lei Zuo
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Luo-Lun Dong
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Si-Fan Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Zhou-Jian Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Yuan-Sheng Duan
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Mu-Yue Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Qi-Xing Zhu
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.
| | - Jia-Xiang Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.
| |
Collapse
|
16
|
Yao X, Lu Q, Wu Y, Liu J, Liu N, Huang X, Xu C. Effect of Elamipretide on the Vitrification of Mouse Ovarian Tissue by Freezing. Biopreserv Biobank 2024. [PMID: 38648553 DOI: 10.1089/bio.2023.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
The importance of ovarian cortical cryopreservation in fertility preservation is receiving increasing attention from reproductive specialists, and mitochondrial dysfunction is an important cause of reduced ovarian tissue cryopreservation. Elamipretide (SS-31) is a novel mitochondria-targeted antioxidant. However, whether it has a protective effect on mouse ovarian tissue cryopreservation remains to be studied. In this study, we examined follicular morphology and viability, mitochondrial function and oxidative stress levels, apoptosis, and culture in vitro after vitrification cryoresuscitation operation by treating ovarian tissues with SS-31 in cryoprotectant resuscitation solution. At the end of the experiment, the addition of 100 μmol/L SS-31 significantly improved follicle quality and oocyte maturation rate in vitro (p < 0.05) and significantly reduced apoptosis (p < 0.05) and oxidative stress levels (superoxide dismutase, catalase, malondialdehyde, p < 0.05). Meanwhile, mitochondrial respiratory chain complex enzyme activity, mtDNA copy number (p < 0.05), and adenosine triphosphate (p < 0.05) content were significantly increased in the 100 μmol/L SS-31-treated group. In addition, the mRNA expression levels of mitochondrial energy metabolism- and biosynthesis-related genes (STRT1, PGC-1a, PPAR-a, TFAM, p < 0.05) were markedly upregulated (p < 0.05) in the 100 μmol/L SS-31 group. In conclusion, SS-31 improved the cryopreservation of ovarian tissues, and 100 μmol/L SS-31 was found to be the most effective.
Collapse
Affiliation(s)
- Xingfeng Yao
- Medical College, Guangxi University, Nanning, China
| | - Qingfang Lu
- Medical College, Guangxi University, Nanning, China
| | - Yuyin Wu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Juan Liu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Niang Liu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xiling Huang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Changlong Xu
- Reproductive Medical Center of Nanning Second People's Hospital, Nanning, China
| |
Collapse
|
17
|
Fuentes JM, Morcillo P. The Role of Cardiolipin in Mitochondrial Function and Neurodegenerative Diseases. Cells 2024; 13:609. [PMID: 38607048 PMCID: PMC11012098 DOI: 10.3390/cells13070609] [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: 02/18/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Cardiolipin (CL) is a mitochondria-exclusive phospholipid synthesized in the inner mitochondrial membrane. CL plays a key role in mitochondrial membranes, impacting a plethora of functions this organelle performs. Consequently, it is conceivable that abnormalities in the CL content, composition, and level of oxidation may negatively impact mitochondrial function and dynamics, with important implications in a variety of diseases. This review concentrates on papers published in recent years, combined with basic and underexplored research in CL. We capture new findings on its biological functions in the mitochondria, as well as its association with neurodegenerative diseases such as Alzheimer's disease or Parkinson's disease. Lastly, we explore the potential applications of CL as a biomarker and pharmacological target to mitigate mitochondrial dysfunction.
Collapse
Affiliation(s)
- José M. Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
| | - Patricia Morcillo
- Departmentof Neurology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
18
|
Li Y, Yang J, Chen X, Hu H, Lan N, Zhao J, Zheng L. Mitochondrial-targeting and NIR-responsive Mn 3O 4@PDA@Pd-SS31 nanozymes reduce oxidative stress and reverse mitochondrial dysfunction to alleviate osteoarthritis. Biomaterials 2024; 305:122449. [PMID: 38194734 DOI: 10.1016/j.biomaterials.2023.122449] [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: 09/04/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
Mitochondrial reactive oxygen species (mROS) play a crucial role in the process of osteoarthritis (OA), which may be a promising target for therapy of OA. In this study, novel mitochondrial-targeting and SOD-mimic Mn3O4@PDA@Pd-SS31 nanozymes with near-infrared (NIR) responsiveness and synergistic cascade to scavenge mROS were designed for the therapy of OA. Results showed that the nanozymes accelerated the release of Pd and Mn3O4 under NIR irradiation, exhibiting enhanced activities of SOD and CAT mimic enzymes with reversed mitochondrial dysfunction and promoted mitophagy to effectively scavenge mROS from chondrocytes, modulate the microenvironment of oxidative stress, and eventually inhibit the inflammatory response. Nanozymes were excreted in vivo through intestinal metabolic pathway and had good biocompatibility, effectively reducing the inflammatory response and relieving articular cartilage degeneration in OA joints, with a reduction of 93.7 % and 93.8 % in OARSCI scores for 4 and 8 weeks respectively. Thus, this study demonstrated that the mitochondria targeting and NIR responsive Mn3O4@PDA@Pd-SS31 nanozymes could efficiently scavenge mROS, repair damaged mitochondrial function and promote cartilage regeneration, which are promising for the treatment of OA in clinical applications.
Collapse
Affiliation(s)
- Yuquan Li
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530005, China
| | - Junxu Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xiaoming Chen
- Department of Spine Osteopathia, The First Affifiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hao Hu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441100, China
| | - Nihan Lan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application, Guangxi Key Laboratory of Regenerative Medicine,The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
19
|
Du X, Zeng Q, Luo Y, He L, Zhao Y, Li N, Han C, Zhang G, Liu W. Application research of novel peptide mitochondrial-targeted antioxidant SS-31 in mitigating mitochondrial dysfunction. Mitochondrion 2024; 75:101846. [PMID: 38237649 DOI: 10.1016/j.mito.2024.101846] [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: 09/20/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Due to the pivotal role of mitochondria in the generation of adenosine triphosphate (ATP) and the regulation of cellular homeostasis, mitochondrial dysfunction may exert a profound impact on various physiological systems, potentially precipitating a spectrum of distinct diseases. Consequently, research pertaining to mitochondrial therapeutics has assumed increasing significance, warranting heightened scrutiny. In recent years, the field of mitochondrial therapy has witnessed noteworthy advancements, with active exploration into diverse pharmacological agents aimed at ameliorating mitochondrial function. Elamipretide (SS-31), a novel synthetic mitochondrial-targeted antioxidant, has emerged as a promising candidate with extensive therapeutic potential. Its notable attributes encompass the mitigation of oxidative stress, the suppression of inflammatory processes, the maintenance of mitochondrial dynamics, and the prevention of cellular apoptosis. As such, SS-31 may emerge as a viable choice for the treatment of mitochondrial dysfunction-related ailments in the foreseeable future. This article extensively expounds upon the superiority of SS-31 over natural antioxidants and traditional mitochondrial-targeted antioxidants, delves into its mechanisms of modulating mitochondrial function, and comprehensively summarizes its applications in alleviating mitochondrial dysfunction-associated disorders. Furthermore, we offer a comprehensive outlook on the expansive prospects of SS-31's future development and application.
Collapse
Affiliation(s)
- Xinrong Du
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 611137, China; Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| | - Qin Zeng
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China; Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Yunchang Luo
- Biology Major, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Libing He
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| | - Yuhong Zhao
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China; School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu 610083, China.
| | - Ninjing Li
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 611137, China; Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| | - Changli Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 611137, China; Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| | - Guohui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| | - Weixin Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 611137, China; Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| |
Collapse
|
20
|
Liu SZ, Chiao YA, Rabinovitch PS, Marcinek DJ. Mitochondrial Targeted Interventions for Aging. Cold Spring Harb Perspect Med 2024; 14:a041199. [PMID: 37788882 PMCID: PMC10910403 DOI: 10.1101/cshperspect.a041199] [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: 10/05/2023]
Abstract
Changes in mitochondrial function play a critical role in the basic biology of aging and age-related disease. Mitochondria are typically thought of in the context of ATP production and oxidant production. However, it is clear that the mitochondria sit at a nexus of cell signaling where they affect metabolite, redox, and energy status, which influence many factors that contribute to the biology of aging, including stress responses, proteostasis, epigenetics, and inflammation. This has led to growing interest in identifying mitochondrial targeted interventions to delay or reverse age-related decline in function and promote healthy aging. In this review, we discuss the diverse roles of mitochondria in the cell. We then highlight some of the most promising strategies and compounds to target aging mitochondria in preclinical testing. Finally, we review the strategies and compounds that have advanced to clinical trials to test their ability to improve health in older adults.
Collapse
Affiliation(s)
- Sophia Z Liu
- Department of Radiology, University of Washington, Seattle, Washington 98195, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Peter S Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington 98195, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
21
|
Yap C, Wanga S, Wüst RCI, van Os BW, Pijls MME, Keijzer S, van Zanten E, Koolbergen DR, Driessen AHG, Balm R, Yeung KK, de Vries CJM, Houtkooper RH, Lindeman JHN, de Waard V. Doxycycline induces mitochondrial dysfunction in aortic smooth muscle cells. Vascul Pharmacol 2024; 154:107279. [PMID: 38272196 DOI: 10.1016/j.vph.2024.107279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/29/2023] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
The antibiotic doxycycline is known to inhibit inflammation and was therefore considered as a therapeutic to prevent abdominal aortic aneurysm (AAA) growth. Yet mitochondrial dysfunction is a key-characteristic of clinical AAA disease. We hypothesize that doxycycline impairs mitochondrial function in the aorta and aortic smooth muscle cells (SMCs). Doxycycline induced mitonuclear imbalance, reduced proliferation and diminished expression of typical contractile smooth muscle cell (SMC) proteins. To understand the underlying mechanism, we studied krüppel-like factor 4 (KLF4). The expression of this transcription factor was enhanced in SMCs after doxycycline treatment. Knockdown of KLF4, however, did not affect the doxycycline-induced SMC phenotypic changes. Then we used the bioenergetics drug elamipretide (SS-31). Doxycycline-induced loss of SMC contractility markers was not rescued, but mitochondrial genes and mitochondrial connectivity improved upon elamipretide. Thus while doxycycline is anti-inflammatory, it also induces mitochondrial dysfunction in aortic SMCs and causes SMC phenotypic switching, potentially contributing to aortic aneurysm pathology. The drug elamipretide helps mitigate the harmful effects of doxycycline on mitochondrial function in aortic SMC, and may be of interest for treatment of aneurysm diseases with pre-existing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Carmen Yap
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Shaynah Wanga
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Cardiology, Meibergdreef 9, Amsterdam, the Netherlands
| | - Rob C I Wüst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Behavioural and Movement Sciences, Myology, Boelelaan 1117, Amsterdam, the Netherlands
| | - Bram W van Os
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands
| | - Maud M E Pijls
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands
| | - Sofie Keijzer
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands
| | - Eva van Zanten
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands
| | - David R Koolbergen
- Amsterdam UMC location University of Amsterdam, Cardiothoracic Surgery, Meibergdreef 9, Amsterdam, the Netherlands
| | - Antoine H G Driessen
- Amsterdam UMC location University of Amsterdam, Cardiothoracic Surgery, Meibergdreef 9, Amsterdam, the Netherlands
| | - Ron Balm
- Amsterdam UMC location University of Amsterdam, Vascular Surgery, Meibergdreef 9, Amsterdam, the Netherlands
| | - Kak Khee Yeung
- Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Vascular Surgery, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands
| | - Carlie J M de Vries
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Riekelt H Houtkooper
- Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Laboratory Genetic Metabolic Diseases, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology, and Metabolism, Amsterdam, the Netherlands
| | - Jan H N Lindeman
- Leiden University Medical Center, Vascular Surgery, Leiden, the Netherlands
| | - Vivian de Waard
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Xing D, Jin Y, Jin B. A narrative review on inflammaging and late-onset hypogonadism. Front Endocrinol (Lausanne) 2024; 15:1291389. [PMID: 38298378 PMCID: PMC10827931 DOI: 10.3389/fendo.2024.1291389] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
The increasing life expectancy observed in recent years has resulted in a higher prevalence of late-onset hypogonadism (LOH) in older men. LOH is characterized by the decline in testosterone levels and can have significant impacts on physical and mental health. While the underlying causes of LOH are not fully understood, there is a growing interest in exploring the role of inflammaging in its development. Inflammaging is a concept that describes the chronic, low-grade, systemic inflammation that occurs as a result of aging. This inflammatory state has been implicated in the development of various age-related diseases. Several cellular and molecular mechanisms have been identified as contributors to inflammaging, including immune senescence, cellular senescence, autophagy defects, and mitochondrial dysfunction. Despite the extensive research on inflammaging, its relationship with LOH has not yet been thoroughly reviewed in the literature. To address this gap, we aim to review the latest findings related to inflammaging and its impact on the development of LOH. Additionally, we will explore interventions that target inflammaging as potential treatments for LOH.
Collapse
Affiliation(s)
- Dong Xing
- Medical College of Southeast University, Nanjing, Jiangsu, China
| | - Yihan Jin
- Reproductive Medicine Center, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Baofang Jin
- Andrology Department of Integrative Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
23
|
Hou Y, Tan E, Shi H, Ren X, Wan X, Wu W, Chen Y, Niu H, Zhu G, Li J, Li Y, Wang L. Mitochondrial oxidative damage reprograms lipid metabolism of renal tubular epithelial cells in the diabetic kidney. Cell Mol Life Sci 2024; 81:23. [PMID: 38200266 PMCID: PMC10781825 DOI: 10.1007/s00018-023-05078-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/12/2024]
Abstract
The functional and structural changes in the proximal tubule play an important role in the occurrence and development of diabetic kidney disease (DKD). Diabetes-induced metabolic changes, including lipid metabolism reprogramming, are reported to lead to changes in the state of tubular epithelial cells (TECs), and among all the disturbances in metabolism, mitochondria serve as central regulators. Mitochondrial dysfunction, accompanied by increased production of mitochondrial reactive oxygen species (mtROS), is considered one of the primary factors causing diabetic tubular injury. Most studies have discussed how altered metabolic flux drives mitochondrial oxidative stress during DKD. In the present study, we focused on targeting mitochondrial damage as an upstream factor in metabolic abnormalities under diabetic conditions in TECs. Using SS31, a tetrapeptide that protects the mitochondrial cristae structure, we demonstrated that mitochondrial oxidative damage contributes to TEC injury and lipid peroxidation caused by lipid accumulation. Mitochondria protected using SS31 significantly reversed the decreased expression of key enzymes and regulators of fatty acid oxidation (FAO), but had no obvious effect on major glucose metabolic rate-limiting enzymes. Mitochondrial oxidative stress facilitated renal Sphingosine-1-phosphate (S1P) deposition and SS31 limited the elevated Acer1, S1pr1 and SPHK1 activity, and the decreased Spns2 expression. These data suggest a role of mitochondrial oxidative damage in unbalanced lipid metabolism, including lipid droplet (LD) formulation, lipid peroxidation, and impaired FAO and sphingolipid homeostasis in DKD. An in vitro study demonstrated that high glucose drove elevated expression of cytosolic phospholipase A2 (cPLA2), which, in turn, was responsible for the altered lipid metabolism, including LD generation and S1P accumulation, in HK-2 cells. A mitochondria-targeted antioxidant inhibited the activation of cPLA2f isoforms. Taken together, these findings identify mechanistic links between mitochondrial oxidative metabolism and reprogrammed lipid metabolism in diabetic TECs, and provide further evidence for the nephroprotective effects of SS31 via influencing metabolic pathways.
Collapse
Affiliation(s)
- Yanjuan Hou
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Enxue Tan
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Honghong Shi
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Xiayu Ren
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Xing Wan
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Wenjie Wu
- Department of Orthopaedics, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Yiliang Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Hiumin Niu
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
- Department of Nephrology, Heping Hospital, Changzhi Medical College, Changzhi, China
| | - Guozhen Zhu
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Jing Li
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Province People's Hospital, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China
| | - Lihua Wang
- Department of Nephrology, Second Hospital, Shanxi Medical University, No.382, Wuyi Road, Taiyuan, Shanxi, 030000, China.
| |
Collapse
|
24
|
Li Y, Berliocchi L, Li Z, Rasmussen LJ. Interactions between mitochondrial dysfunction and other hallmarks of aging: Paving a path toward interventions that promote healthy old age. Aging Cell 2024; 23:e13942. [PMID: 37497653 PMCID: PMC10776122 DOI: 10.1111/acel.13942] [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: 03/21/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/28/2023] Open
Abstract
Current research on human aging has largely been guided by the milestone paper "hallmarks of aging," which were first proposed in the seminal 2013 paper by Lopez-Otin et al. Most studies have focused on one aging hallmark at a time, asking whether the underlying molecular perturbations are sufficient to drive the aging process and its associated phenotypes. More recently, researchers have begun to investigate whether aging phenotypes are driven by concurrent perturbations in molecular pathways linked to not one but to multiple hallmarks of aging and whether they present different patterns in organs and systems over time. Indeed, preliminary results suggest that more complex interactions between aging hallmarks must be considered and addressed, if we are to develop interventions that successfully promote healthy aging and/or delay aging-associated dysfunction and diseases. Here, we summarize some of the latest work and views on the interplay between hallmarks of aging, with a specific focus on mitochondrial dysfunction. Indeed, this represents a significant example of the complex crosstalk between hallmarks of aging and of the effects that an intervention targeted to a specific hallmark may have on the others. A better knowledge of these interconnections, of their cause-effect relationships, of their spatial and temporal sequence, will be very beneficial for the whole aging research field and for the identification of effective interventions in promoting healthy old age.
Collapse
Affiliation(s)
- Yuan Li
- Department of Cellular and Molecular Medicine, Center for Healthy AgingUniversity of CopenhagenCopenhagenDenmark
| | - Laura Berliocchi
- Department of Cellular and Molecular Medicine, Center for Healthy AgingUniversity of CopenhagenCopenhagenDenmark
- Department of Health SciencesUniversity Magna Græcia of CatanzaroCatanzaroItaly
| | - Zhiquan Li
- Department of Cellular and Molecular Medicine, Center for Healthy AgingUniversity of CopenhagenCopenhagenDenmark
| | - Lene Juel Rasmussen
- Department of Cellular and Molecular Medicine, Center for Healthy AgingUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
25
|
Tamucci JD, Alder NN, May ER. Peptide Power: Mechanistic Insights into the Effect of Mitochondria-Targeted Tetrapeptides on Membrane Electrostatics from Molecular Simulations. Mol Pharm 2023; 20:6114-6129. [PMID: 37904323 PMCID: PMC10841697 DOI: 10.1021/acs.molpharmaceut.3c00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Mitochondrial dysfunction is implicated in nine of the ten leading causes of death in the US, yet there are no FDA-approved therapeutics to treat it. Synthetic mitochondria-targeted peptides (MTPs), including the lead compound SS-31, offer promise, as they have been shown to restore healthy mitochondrial function and treat a variety of common diseases. At the cellular level, research has shown that MTPs accumulate strongly at the inner mitochondrial membrane (IMM), slow energy sinks (e.g., proton leaks), and improve ATP production. Modulation of electrostatic fields around the IMM has been implicated as a key aspect in the mechanism of action (MoA) of these peptides; however, molecular and mechanistic details have remained elusive. In this study, we employed all-atom molecular dynamics simulations (MD) to investigate the interactions of four MTPs with lipid bilayers and calculate their effect on structural and electrostatic properties. In agreement with previous experimental findings, we observed the modulation of the membrane surface and dipole potentials by MTPs. The simulations reveal that the MTPs achieve a reduction in the dipole potential by acting to disorder both lipid head groups and water layers proximal to the bilayer surface. We also find that MTPs decrease the bilayer thickness and increase the membrane's capacitance. These changes suggest that MTPs may enhance how much potential energy can be stored across the IMM at a given transmembrane potential difference. The MTPs also displace cations away from the bilayer surface, modulating the surface potential and offering an alternative mechanism for how these MTPs reduce mitochondrial energy sinks like proton leaks and mitigate Ca2+ accumulation stress. In conclusion, this study highlights the therapeutic potential of MTPs and underlines how interactions of MTPs with lipid bilayers serve as a fundamental component of their MoA.
Collapse
Affiliation(s)
- Jeffrey D Tamucci
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Nathan N Alder
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Eric R May
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
26
|
Pharaoh G, Kamat V, Kannan S, Stuppard RS, Whitson J, Martín-Pérez M, Qian WJ, MacCoss MJ, Villén J, Rabinovitch P, Campbell MD, Sweet IR, Marcinek DJ. The mitochondrially targeted peptide elamipretide (SS-31) improves ADP sensitivity in aged mitochondria by increasing uptake through the adenine nucleotide translocator (ANT). GeroScience 2023; 45:3529-3548. [PMID: 37462785 PMCID: PMC10643647 DOI: 10.1007/s11357-023-00861-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/23/2023] [Indexed: 07/28/2023] Open
Abstract
Aging muscle experiences functional decline in part mediated by impaired mitochondrial ADP sensitivity. Elamipretide (ELAM) rapidly improves physiological and mitochondrial function in aging and binds directly to the mitochondrial ADP transporter ANT. We hypothesized that ELAM improves ADP sensitivity in aging leading to rescued physiological function. We measured the response to ADP stimulation in young and old muscle mitochondria with ELAM treatment, in vivo heart and muscle function, and compared protein abundance, phosphorylation, and S-glutathionylation of ADP/ATP pathway proteins. ELAM treatment increased ADP sensitivity in old muscle mitochondria by increasing uptake of ADP through the ANT and rescued muscle force and heart systolic function. Protein abundance in the ADP/ATP transport and synthesis pathway was unchanged, but ELAM treatment decreased protein s-glutathionylation incuding of ANT. Mitochondrial ADP sensitivity is rapidly modifiable. This research supports the hypothesis that ELAM improves ANT function in aging and links mitochondrial ADP sensitivity to physiological function. ELAM binds directly to ANT and ATP synthase and ELAM treatment improves ADP sensitivity, increases ATP production, and improves physiological function in old muscles. ADP (adenosine diphosphate), ATP (adenosine triphosphate), VDAC (voltage-dependent anion channel), ANT (adenine nucleotide translocator), H+ (proton), ROS (reactive oxygen species), NADH (nicotinamide adenine dinucleotide), FADH2 (flavin adenine dinucleotide), O2 (oxygen), ELAM (elamipretide), -SH (free thiol), -SSG (glutathionylated protein).
Collapse
Affiliation(s)
- Gavin Pharaoh
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Varun Kamat
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Sricharan Kannan
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Rudolph S Stuppard
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Jeremy Whitson
- Department of Biology, High Point University, High Point, NC, 27268, USA
| | - Miguel Martín-Pérez
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08028, Barcelona, Spain
| | - Wei-Jun Qian
- Integrative Omics Group, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Judit Villén
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Peter Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Matthew D Campbell
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Ian R Sweet
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
27
|
Nie Y, Li J, Zhai X, Wang Z, Wang J, Wu Y, Zhao P, Yan G. Elamipretide(SS-31) Attenuates Idiopathic Pulmonary Fibrosis by Inhibiting the Nrf2-Dependent NLRP3 Inflammasome in Macrophages. Antioxidants (Basel) 2023; 12:2022. [PMID: 38136142 PMCID: PMC10740969 DOI: 10.3390/antiox12122022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/05/2023] [Accepted: 11/11/2023] [Indexed: 12/24/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fatal lung disease with a limited therapeutic strategy. Mitochondrial oxidative stress in macrophages is directly linked to IPF. Elamipretide(SS-31) is a mitochondrion-targeted peptide that has been shown to be safe and beneficial for multiple diseases. However, whether SS-31 alleviates IPF is unclear. In the present study, we used a bleomycin (BLM)-induced mouse model followed by SS-31 injection every other day to investigate its role in IPF and explore the possible mechanism. Our results showed that SS-31 treatment significantly suppressed BLM-induced pulmonary fibrosis and inflammation, with improved histological change, and decreased extracellular matrix deposition and inflammatory cytokines release. Impressively, the expression percentage of IL-1β and IL-18 was downregulated to lower than half with SS-31 treatment. Mechanistically, SS-31 inhibited IL-33- or lipopolysaccharide(LPS)/IL-4-induced production of IL-1β and IL-18 in macrophages by suppressing NOD-like receptor thermal protein domain associated protein 3(NLRP3) inflammasome activation. Nuclear factor erythroid 2-related factor 2(Nrf2) was dramatically upregulated along with improved mitochondrial function after SS-31 treatment in activated macrophages and BLM-induced mice. Conversely, there was no significant change after SS-31 treatment in Nrf2-/- mice and macrophages. These findings indicated that SS-31 protected against pulmonary fibrosis and inflammation by inhibiting the Nrf2-mediated NLRP3 inflammasome in macrophages. Our data provide initial evidence for the therapeutic efficacy of SS-31 in IPF.
Collapse
Affiliation(s)
- Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.N.); (J.L.); (Z.W.); (J.W.); (Y.W.)
| | - Jiao Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.N.); (J.L.); (Z.W.); (J.W.); (Y.W.)
| | - Xiaorun Zhai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.N.); (J.L.); (Z.W.); (J.W.); (Y.W.)
| | - Zhixu Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.N.); (J.L.); (Z.W.); (J.W.); (Y.W.)
| | - Junpeng Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.N.); (J.L.); (Z.W.); (J.W.); (Y.W.)
| | - Yaxian Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.N.); (J.L.); (Z.W.); (J.W.); (Y.W.)
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.N.); (J.L.); (Z.W.); (J.W.); (Y.W.)
| | - Gen Yan
- Department of Radiology, The Second Affiliated Hospital of Xiamen Medical College, Xiamen 361000, China
| |
Collapse
|
28
|
Kutschka I, Bertero E, Wasmus C, Xiao K, Yang L, Chen X, Oshima Y, Fischer M, Erk M, Arslan B, Alhasan L, Grosser D, Ermer KJ, Nickel A, Kohlhaas M, Eberl H, Rebs S, Streckfuss-Bömeke K, Schmitz W, Rehling P, Thum T, Higuchi T, Rabinowitz J, Maack C, Dudek J. Activation of the integrated stress response rewires cardiac metabolism in Barth syndrome. Basic Res Cardiol 2023; 118:47. [PMID: 37930434 PMCID: PMC10628049 DOI: 10.1007/s00395-023-01017-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 09/29/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023]
Abstract
Barth Syndrome (BTHS) is an inherited cardiomyopathy caused by defects in the mitochondrial transacylase TAFAZZIN (Taz), required for the synthesis of the phospholipid cardiolipin. BTHS is characterized by heart failure, increased propensity for arrhythmias and a blunted inotropic reserve. Defects in Ca2+-induced Krebs cycle activation contribute to these functional defects, but despite oxidation of pyridine nucleotides, no oxidative stress developed in the heart. Here, we investigated how retrograde signaling pathways orchestrate metabolic rewiring to compensate for mitochondrial defects. In mice with an inducible knockdown (KD) of TAFAZZIN, and in induced pluripotent stem cell-derived cardiac myocytes, mitochondrial uptake and oxidation of fatty acids was strongly decreased, while glucose uptake was increased. Unbiased transcriptomic analyses revealed that the activation of the eIF2α/ATF4 axis of the integrated stress response upregulates one-carbon metabolism, which diverts glycolytic intermediates towards the biosynthesis of serine and fuels the biosynthesis of glutathione. In addition, strong upregulation of the glutamate/cystine antiporter xCT increases cardiac cystine import required for glutathione synthesis. Increased glutamate uptake facilitates anaplerotic replenishment of the Krebs cycle, sustaining energy production and antioxidative pathways. These data indicate that ATF4-driven rewiring of metabolism compensates for defects in mitochondrial uptake of fatty acids to sustain energy production and antioxidation.
Collapse
Affiliation(s)
- Ilona Kutschka
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Edoardo Bertero
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino - Italian IRCCS Cardiology Network, Genoa, Italy
| | - Christina Wasmus
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Lifeng Yang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Rd, Shanghai, 200031, China
| | - Xinyu Chen
- Department of Nuclear Medicine, University Clinic Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Yasuhiro Oshima
- Department of Nuclear Medicine, University Clinic Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Marcus Fischer
- Division of Pediatric Cardiology and Intensive Care, University Hospital LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Manuela Erk
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Berkan Arslan
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Lin Alhasan
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Daria Grosser
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Katharina J Ermer
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Alexander Nickel
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Michael Kohlhaas
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Hanna Eberl
- Department for Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany
| | - Sabine Rebs
- Department for Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany
| | - Katrin Streckfuss-Bömeke
- Department for Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany
- Clinic for Cardiology and Pneumology, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen, Germany
| | - Werner Schmitz
- Department of Biochemistry and Molecular Biology, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Peter Rehling
- University Göttingen, Institute of Biochemistry and Molecular Cell Biology, Humboldtallee 23, 37072, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
- Rebirth Center for Translational Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Clinic Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Joshua Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
- Medical Clinic I, University Clinic Würzburg, Würzburg, Germany
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany.
| |
Collapse
|
29
|
Reed AL, Mitchell W, Alexandrescu AT, Alder NN. Interactions of amyloidogenic proteins with mitochondrial protein import machinery in aging-related neurodegenerative diseases. Front Physiol 2023; 14:1263420. [PMID: 38028797 PMCID: PMC10652799 DOI: 10.3389/fphys.2023.1263420] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Most mitochondrial proteins are targeted to the organelle by N-terminal mitochondrial targeting sequences (MTSs, or "presequences") that are recognized by the import machinery and subsequently cleaved to yield the mature protein. MTSs do not have conserved amino acid compositions, but share common physicochemical properties, including the ability to form amphipathic α-helical structures enriched with basic and hydrophobic residues on alternating faces. The lack of strict sequence conservation implies that some polypeptides can be mistargeted to mitochondria, especially under cellular stress. The pathogenic accumulation of proteins within mitochondria is implicated in many aging-related neurodegenerative diseases, including Alzheimer's, Parkinson's, and Huntington's diseases. Mechanistically, these diseases may originate in part from mitochondrial interactions with amyloid-β precursor protein (APP) or its cleavage product amyloid-β (Aβ), α-synuclein (α-syn), and mutant forms of huntingtin (mHtt), respectively, that are mediated in part through their associations with the mitochondrial protein import machinery. Emerging evidence suggests that these amyloidogenic proteins may present cryptic targeting signals that act as MTS mimetics and can be recognized by mitochondrial import receptors and transported into different mitochondrial compartments. Accumulation of these mistargeted proteins could overwhelm the import machinery and its associated quality control mechanisms, thereby contributing to neurological disease progression. Alternatively, the uptake of amyloidogenic proteins into mitochondria may be part of a protein quality control mechanism for clearance of cytotoxic proteins. Here we review the pathomechanisms of these diseases as they relate to mitochondrial protein import and effects on mitochondrial function, what features of APP/Aβ, α-syn and mHtt make them suitable substrates for the import machinery, and how this information can be leveraged for the development of therapeutic interventions.
Collapse
Affiliation(s)
- Ashley L. Reed
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| | - Wayne Mitchell
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Andrei T. Alexandrescu
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| | - Nathan N. Alder
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
30
|
Hevler JF, Heck AJR. Higher-Order Structural Organization of the Mitochondrial Proteome Charted by In Situ Cross-Linking Mass Spectrometry. Mol Cell Proteomics 2023; 22:100657. [PMID: 37805037 PMCID: PMC10651688 DOI: 10.1016/j.mcpro.2023.100657] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/14/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023] Open
Abstract
Mitochondria are densely packed with proteins, of which most are involved physically or more transiently in protein-protein interactions (PPIs). Mitochondria host among others all enzymes of the Krebs cycle and the oxidative phosphorylation pathway and are foremost associated with cellular bioenergetics. However, mitochondria are also important contributors to apoptotic cell death and contain their own genome indicating that they play additionally an eminent role in processes beyond bioenergetics. Despite intense efforts in identifying and characterizing mitochondrial protein complexes by structural biology and proteomics techniques, many PPIs have remained elusive. Several of these (membrane embedded) PPIs are less stable in vitro hampering their characterization by most contemporary methods in structural biology. Particularly in these cases, cross-linking mass spectrometry (XL-MS) has proven valuable for the in-depth characterization of mitochondrial protein complexes in situ. Here, we highlight experimental strategies for the analysis of proteome-wide PPIs in mitochondria using XL-MS. We showcase the ability of in situ XL-MS as a tool to map suborganelle interactions and topologies and aid in refining structural models of protein complexes. We describe some of the most recent technological advances in XL-MS that may benefit the in situ characterization of PPIs even further, especially when combined with electron microscopy and structural modeling.
Collapse
Affiliation(s)
- Johannes F Hevler
- Division of Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Albert J R Heck
- Division of Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Center, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Ruwolt M, Piazza I, Liu F. The potential of cross-linking mass spectrometry in the development of protein-protein interaction modulators. Curr Opin Struct Biol 2023; 82:102648. [PMID: 37423038 DOI: 10.1016/j.sbi.2023.102648] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023]
Abstract
Cross-linking mass spectrometry (XL-MS) can provide a wealth of information on endogenous protein-protein interaction (PPI) networks and protein binding interfaces. These features make XL-MS an attractive tool to support the development of PPI-targeting drugs. Though not yet widely used, applications of XL-MS to drug characterization are beginning to emerge. Here, we compare XL-MS to established structural proteomics methods in drug research, discuss the current state and remaining challenges of XL-MS technology, and provide a perspective on the future role XL-MS can play in drug development, with a particular emphasis on PPI modulators.
Collapse
Affiliation(s)
- Max Ruwolt
- Department of Structural Biology, Leibniz, Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Roessle-Str. 10, 13125 Berlin, Germany. https://twitter.com/@MRuwolt
| | - Ilaria Piazza
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC Berlin), Berlin, Germany.
| | - Fan Liu
- Department of Structural Biology, Leibniz, Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Roessle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Charitépl. 1, 10117 Berlin, Germany.
| |
Collapse
|
32
|
Yu C, Huang L. New advances in cross-linking mass spectrometry toward structural systems biology. Curr Opin Chem Biol 2023; 76:102357. [PMID: 37406423 PMCID: PMC11091472 DOI: 10.1016/j.cbpa.2023.102357] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 07/07/2023]
Abstract
Elucidating protein-protein interaction (PPI) networks and their structural features within cells is central to understanding fundamental biology and associations of cell phenotypes with human pathologies. Owing to technological advancements during the last decade, cross-linking mass spectrometry (XL-MS) has become an enabling technology for delineating interaction landscapes of proteomes as they exist in living systems. XL-MS is unique due to its capability to simultaneously capture PPIs from native environments and uncover interaction contacts though identification of cross-linked peptides, thereby permitting the determination of both identity and connectivity of PPIs in cells. In combination with high resolution structural tools such as cryo-electron microscopy and AI-assisted prediction, XL-MS has contributed significantly to elucidating architectures of large protein assemblies. This review highlights the latest developments in XL-MS technologies and their applications in proteome-wide analysis to advance structural systems biology.
Collapse
Affiliation(s)
- Clinton Yu
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Lan Huang
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
33
|
Campbell MD, Djukovic D, Raftery D, Marcinek DJ. Age-related changes of skeletal muscle metabolic response to contraction are also sex-dependent. J Physiol 2023:10.1113/JP285124. [PMID: 37742081 PMCID: PMC10959763 DOI: 10.1113/jp285124] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023] Open
Abstract
Mitochondria adapt to increased energy demands during muscle contraction by acutely altering metabolite fluxes and substrate oxidation. With age, an impaired mitochondrial metabolic response may contribute to reduced exercise tolerance and decreased skeletal muscle mass, specific force, increased overall fatty depositions in the skeletal muscle, frailty and depressed energy maintenance. We hypothesized that elevated energy stress in mitochondria with age alters the capacity of mitochondria to utilize different substrates following muscle contraction. To test this hypothesis, we used in vivo electrical stimulation to simulate high-intensity intervals (HII) or low intensity steady-state (LISS) exercise in young (5-7 months) and aged (27-29 months) male and female mice to characterize effects of age and sex on mitochondrial substrate utilization in skeletal muscle following contraction. Mitochondrial respiration using glutamate decreased in aged males following HII and glutamate oxidation was inhibited following HII in both the contracted and non-stimulated muscle of aged female muscle. Analyses of the muscle metabolome of female mice indicated that changes in metabolic pathways induced by HII and LISS contractions in young muscle are absent in aged muscle. To test improved mitochondrial function on substrate utilization following HII, we treated aged females with elamipretide (ELAM), a mitochondrially-targeted peptide shown to improve mitochondrial bioenergetics and restore redox status in aged muscle. ELAM removed inhibition of glutamate oxidation and showed increased metabolic pathway changes following HII, suggesting rescuing redox status and improving bioenergetic function in mitochondria from aged muscle increases glutamate utilization and enhances the metabolic response to muscle contraction in aged muscle. KEY POINTS: Acute local contraction of gastrocnemius can systemically alter mitochondrial respiration in non-stimulated muscle. Age-related changes in mitochondrial respiration using glutamate or palmitoyl carnitine following contraction are sex-dependent. Respiration using glutamate after high-intensity contraction is inhibited in aged female muscle. Metabolite level and pathway changes following muscle contraction decrease with age in female mice. Treatment with the mitochondrially-targeted peptide elamipretide can partially rescue metabolite response to muscle contraction.
Collapse
Affiliation(s)
| | - Danijel Djukovic
- Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
| | - Daniel Raftery
- Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
| | | |
Collapse
|
34
|
Deng QS, Gao Y, Rui BY, Li XR, Liu PL, Han ZY, Wei ZY, Zhang CR, Wang F, Dawes H, Zhu TH, Tao SC, Guo SC. Double-network hydrogel enhanced by SS31-loaded mesoporous polydopamine nanoparticles: Symphonic collaboration of near-infrared photothermal antibacterial effect and mitochondrial maintenance for full-thickness wound healing in diabetes mellitus. Bioact Mater 2023; 27:409-428. [PMID: 37152712 PMCID: PMC10160601 DOI: 10.1016/j.bioactmat.2023.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/24/2023] [Accepted: 04/02/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetic wound healing has become a serious healthcare challenge. The high-glucose environment leads to persistent bacterial infection and mitochondrial dysfunction, resulting in chronic inflammation, abnormal vascular function, and tissue necrosis. To solve these issues, we developed a double-network hydrogel, constructed with pluronic F127 diacrylate (F127DA) and hyaluronic acid methacrylate (HAMA), and enhanced by SS31-loaded mesoporous polydopamine nanoparticles (MPDA NPs). As components, SS31, a mitochondria-targeted peptide, maintains mitochondrial function, reduces mitochondrial reactive oxygen species (ROS) and thus regulates macrophage polarization, as well as promoting cell proliferation and migration, while MPDA NPs not only scavenge ROS and exert an anti-bacterial effect by photothermal treatment under near-infrared light irradiation, but also control release of SS31 in response to ROS. This F127DA/HAMA-MPDA@SS31 (FH-M@S) hydrogel has characteristics of adhesion, superior biocompatibility and mechanical properties which can adapt to irregular wounds at different body sites and provide sustained release of MPDA@SS31 (M@S) NPs. In addition, in a diabetic rat full thickness skin defect model, the FH-M@S hydrogel promoted macrophage M2 polarization, collagen deposition, neovascularization and wound healing. Therefore, the FH-M@S hydrogel exhibits promising therapeutic potential for skin regeneration.
Collapse
Affiliation(s)
- Qing-Song Deng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yuan Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Bi-Yu Rui
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
| | - Xu-Ran Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Po-Lin Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zi-Yin Han
- Department of Rheumatology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, No.29, Xinglongxiang, Tianning District, Changzhou, 213000, China
| | - Zhan-Ying Wei
- Shanghai Clinical Research Centre of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chang-Ru Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Fei Wang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Helen Dawes
- Faculty of Health and Life Science, Oxford Brookes University, Headington Road, Oxford, OX3 0BP, UK
- NIHR Oxford Health Biomedical Research Centre, Oxford, OX3 7JX, UK
- College of Medicine and Health, St Lukes Campus, University of Exeter, Heavitree Road, Exeter, EX1 2LU, UK
| | - Tong-He Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-Coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
| | - Shi-Cong Tao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Corresponding author. Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Shang-Chun Guo
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- Corresponding author. Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
35
|
Muñoz JP, Basei FL, Rojas ML, Galvis D, Zorzano A. Mechanisms of Modulation of Mitochondrial Architecture. Biomolecules 2023; 13:1225. [PMID: 37627290 PMCID: PMC10452872 DOI: 10.3390/biom13081225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrial network architecture plays a critical role in cellular physiology. Indeed, alterations in the shape of mitochondria upon exposure to cellular stress can cause the dysfunction of these organelles. In this scenario, mitochondrial dynamics proteins and the phospholipid composition of the mitochondrial membrane are key for fine-tuning the modulation of mitochondrial architecture. In addition, several factors including post-translational modifications such as the phosphorylation, acetylation, SUMOylation, and o-GlcNAcylation of mitochondrial dynamics proteins contribute to shaping the plasticity of this architecture. In this regard, several studies have evidenced that, upon metabolic stress, mitochondrial dynamics proteins are post-translationally modified, leading to the alteration of mitochondrial architecture. Interestingly, several proteins that sustain the mitochondrial lipid composition also modulate mitochondrial morphology and organelle communication. In this context, pharmacological studies have revealed that the modulation of mitochondrial shape and function emerges as a potential therapeutic strategy for metabolic diseases. Here, we review the factors that modulate mitochondrial architecture.
Collapse
Affiliation(s)
- Juan Pablo Muñoz
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Fernanda Luisa Basei
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas, 13083-871 Campinas, SP, Brazil
| | - María Laura Rojas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - David Galvis
- Programa de Química Farmacéutica, Universidad CES, Medellín 050031, Colombia
| | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
36
|
Qi X, Rusch NJ, Fan J, Mora CJ, Xie L, Mu S, Rabinovitch PS, Zhang H. Mitochondrial proton leak in cardiac aging. GeroScience 2023; 45:2135-2143. [PMID: 36856945 PMCID: PMC10651624 DOI: 10.1007/s11357-023-00757-x] [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: 01/10/2023] [Accepted: 02/16/2023] [Indexed: 03/02/2023] Open
Abstract
Age-associated diseases are becoming progressively more prevalent, reflecting the increased lifespan of the world's population. However, the fundamental mechanisms of physiologic aging are poorly understood, and in particular, the molecular pathways that mediate cardiac aging and its associated dysfunction are unclear. Here, we focus on certain ion flux abnormalities of the mitochondria that may contribute to cardiac aging and age-related heart failure. Using oxidative phosphorylation, mitochondria pump protons from the matrix to the intermembrane space to generate a proton gradient across the inner membrane. The protons are returned to the matrix by the ATPase complex within the membrane to generate ATP. However, a portion of protons leak back to the matrix and do not drive ATP production, and this event is called proton leak or uncoupling. Accumulating evidence suggests that mitochondrial proton leak is increased in the cardiac myocytes of aged hearts. In this mini-review, we discuss the measurement methods and major sites of mitochondrial proton leak with an emphasis on the adenine nucleotide transporter 1 (ANT1), and explore the possibility of inhibiting augmented mitochondrial proton leak as a therapeutic intervention to mitigate cardiac aging.
Collapse
Affiliation(s)
- Xingyun Qi
- Department of Biology, Rutgers University, Camden, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Jiaojiao Fan
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Christoph J Mora
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Lixin Xie
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Shengyu Mu
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Peter S Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA.
| | - Huiliang Zhang
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, USA.
| |
Collapse
|
37
|
Zhao T, He F, Zhao K, Yuxia L, Li H, Liu X, Cen J, Duan S. A Triple-Targeted Rutin-Based Self-Assembled Delivery Vector for Treating Ischemic Stroke by Vascular Normalization and Anti-Inflammation via ACE2/Ang1-7 Signaling. ACS CENTRAL SCIENCE 2023; 9:1180-1199. [PMID: 37396868 PMCID: PMC10311651 DOI: 10.1021/acscentsci.3c00377] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Indexed: 07/04/2023]
Abstract
Changes in the cerebral microenvironment caused by acute ischemic stroke-reperfusion are the main obstacle to the recovery of neurological function and an important cause of stroke recurrence after thrombolytic therapy. The intracerebral microenvironment after ischemia-reperfusion reduces the neuroplasticity of the penumbra and ultimately leads to permanent neurological damage. To overcome this challenge, we developed a triple-targeted self-assembled nanodelivery system, which combines the neuroprotective drug rutin with hyaluronic acid through esterification to form a conjugate, and then connected SS-31, a small peptide that can penetrate the blood brain barrier and target mitochondria. Brain targeting, CD44-mediated endocytosis, hyaluronidase 1-mediated degradation, and the acidic environment synergistically promoted the enrichment of nanoparticles and drug release in the injured area. Results demonstrate that rutin has a high affinity for ACE2 receptors on the cell membrane and can directly activate ACE2/Ang1-7 signaling, maintain neuroinflammation, and promote penumbra angiogenesis and normal neovascularization. Importantly, this delivery system enhanced the overall plasticity of the injured area and significantly reduced neurological damage after stroke. The relevant mechanism was expounded from the aspects of behavior, histology, and molecular cytology. All results suggest that our delivery system may be an effective and safe strategy for the treatment of acute ischemic stroke-reperfusion injury.
Collapse
Affiliation(s)
- Tingkui Zhao
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Fujin He
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Keqing Zhao
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Lin Yuxia
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Huanyu Li
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xingru Liu
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Juan Cen
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Shaofeng Duan
- Key
Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University, Kaifeng 475004, China
- Institute
for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng 475004, China
- Henan
International Joint Laboratory of Chinese Medicine Efficacy, Henan University, Kaifeng 475004, China
| |
Collapse
|
38
|
Dai S, Liu S, Zhou C, Yu F, Zhu G, Zhang W, Deng H, Burlingame A, Yu W, Wang T, Li N. Capturing the hierarchically assorted modules of protein-protein interactions in the organized nucleome. MOLECULAR PLANT 2023; 16:930-961. [PMID: 36960533 DOI: 10.1016/j.molp.2023.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/16/2023] [Accepted: 03/21/2023] [Indexed: 05/04/2023]
Abstract
Nuclear proteins are major constituents and key regulators of nucleome topological organization and manipulators of nuclear events. To decipher the global connectivity of nuclear proteins and the hierarchically organized modules of their interactions, we conducted two rounds of cross-linking mass spectrometry (XL-MS) analysis, one of which followed a quantitative double chemical cross-linking mass spectrometry (in vivoqXL-MS) workflow, and identified 24,140 unique crosslinks in total from the nuclei of soybean seedlings. This in vivo quantitative interactomics enabled the identification of 5340 crosslinks that can be converted into 1297 nuclear protein-protein interactions (PPIs), 1220 (94%) of which were non-confirmative (or novel) nuclear PPIs compared with those in repositories. There were 250 and 26 novel interactors of histones and the nucleolar box C/D small nucleolar ribonucleoprotein complex, respectively. Modulomic analysis of orthologous Arabidopsis PPIs produced 27 and 24 master nuclear PPI modules (NPIMs) that contain the condensate-forming protein(s) and the intrinsically disordered region-containing proteins, respectively. These NPIMs successfully captured previously reported nuclear protein complexes and nuclear bodies in the nucleus. Surprisingly, these NPIMs were hierarchically assorted into four higher-order communities in a nucleomic graph, including genome and nucleolus communities. This combinatorial pipeline of 4C quantitative interactomics and PPI network modularization revealed 17 ethylene-specific module variants that participate in a broad range of nuclear events. The pipeline was able to capture both nuclear protein complexes and nuclear bodies, construct the topological architectures of PPI modules and module variants in the nucleome, and probably map the protein compositions of biomolecular condensates.
Collapse
Affiliation(s)
- Shuaijian Dai
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Shichang Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Chen Zhou
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Fengchao Yu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Guang Zhu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Wenhao Zhang
- Tsinghua-Peking Joint Centre for Life Sciences, Centre for Structural Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Haiteng Deng
- Tsinghua-Peking Joint Centre for Life Sciences, Centre for Structural Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Al Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Weichuan Yu
- The HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, Guangdong 518057, China; Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| | - Tingliang Wang
- Tsinghua-Peking Joint Centre for Life Sciences, Centre for Structural Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Ning Li
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China; Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China; The HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, Guangdong 518057, China.
| |
Collapse
|
39
|
Chen B, Daneshgar N, Lee HC, Song LS, Dai DF. Mitochondrial Oxidative Stress Mediates Bradyarrhythmia in Leigh Syndrome Mitochondrial Disease Mice. Antioxidants (Basel) 2023; 12:antiox12051001. [PMID: 37237867 PMCID: PMC10215409 DOI: 10.3390/antiox12051001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondrial oxidative stress has been implicated in aging and several cardiovascular diseases, including heart failure and cardiomyopathy, ventricular tachycardia, and atrial fibrillation. The role of mitochondrial oxidative stress in bradyarrhythmia is less clear. Mice with a germline deletion of Ndufs4 subunit respiratory complex I develop severe mitochondrial encephalomyopathy resembling Leigh Syndrome (LS). Several types of cardiac bradyarrhythmia are present in LS mice, including a frequent sinus node dysfunction and episodic atrioventricular (AV) block. Treatment with the mitochondrial antioxidant Mitotempo or mitochondrial protective peptide SS31 significantly ameliorated the bradyarrhythmia and extended the lifespan of LS mice. Using an ex vivo Langendorff perfused heart with live confocal imaging of mitochondrial and total cellular reactive oxygen species (ROS), we showed increased ROS in the LS heart, which was potentiated by ischemia-reperfusion. A simultaneous ECG recording showed a sinus node dysfunction and AV block concurrent with the severity of the oxidative stress. Treatment with Mitotempo abolished ROS and restored the sinus rhythm. Our study reveals robust evidence of the direct mechanistic roles of mitochondrial and total ROS in bradyarrhythmia in the setting of LS mitochondrial cardiomyopathy. Our study also supports the potential clinical application of mitochondrial-targeted antioxidants or SS31 for the treatment of LS patients.
Collapse
Affiliation(s)
- Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Nastaran Daneshgar
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Hsiang-Chun Lee
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Dao-Fu Dai
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
40
|
Fairley LH, Das S, Dharwal V, Amorim N, Hegarty KJ, Wadhwa R, Mounika G, Hansbro PM. Mitochondria-Targeted Antioxidants as a Therapeutic Strategy for Chronic Obstructive Pulmonary Disease. Antioxidants (Basel) 2023; 12:973. [PMID: 37107348 PMCID: PMC10135688 DOI: 10.3390/antiox12040973] [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: 02/28/2023] [Revised: 03/29/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Oxidative stress is a major hallmark of COPD, contributing to inflammatory signaling, corticosteroid resistance, DNA damage, and accelerated lung aging and cellular senescence. Evidence suggests that oxidative damage is not solely due to exogenous exposure to inhaled irritants, but also endogenous sources of oxidants in the form of reactive oxygen species (ROS). Mitochondria, the major producers of ROS, exhibit impaired structure and function in COPD, resulting in reduced oxidative capacity and excessive ROS production. Antioxidants have been shown to protect against ROS-induced oxidative damage in COPD, by reducing ROS levels, reducing inflammation, and protecting against the development of emphysema. However, currently available antioxidants are not routinely used in the management of COPD, suggesting the need for more effective antioxidant agents. In recent years, a number of mitochondria-targeted antioxidant (MTA) compounds have been developed that are capable of crossing the mitochondria lipid bilayer, offering a more targeted approach to reducing ROS at its source. In particular, MTAs have been shown to illicit greater protective effects compared to non-targeted, cellular antioxidants by further reducing apoptosis and offering greater protection against mtDNA damage, suggesting they are promising therapeutic agents for the treatment of COPD. Here, we review evidence for the therapeutic potential of MTAs as a treatment for chronic lung disease and discuss current challenges and future directions.
Collapse
Affiliation(s)
- Lauren H. Fairley
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| | - Shatarupa Das
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| | - Vivek Dharwal
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| | - Nadia Amorim
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| | - Karl J. Hegarty
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| | - Ridhima Wadhwa
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
- Discipline of Pharmacy, Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Guntipally Mounika
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| | - Philip M. Hansbro
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2050, Australia
| |
Collapse
|
41
|
Mitochondrial connexin43 and mitochondrial K ATP channels modulate triggered arrhythmias in mouse ventricular muscle. Pflugers Arch 2023; 475:477-488. [PMID: 36707457 DOI: 10.1007/s00424-023-02789-w] [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: 09/07/2022] [Revised: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 01/29/2023]
Abstract
Connexin43 (Cx43) exits as hemichannels in the inner mitochondrial membrane. We examined how mitochondrial Cx43 and mitochondrial KATP channels affect the occurrence of triggered arrhythmias. To generate cardiac-specific Cx43-deficient (cCx43-/-) mice, Cx43flox/flox mice were crossed with α-MHC (Myh6)-cre+/- mice. The resulting offspring, Cx43flox/flox/Myh6-cre+/- mice (cCx43-/- mice) and their littermates (cCx43+/+ mice), were used. Trabeculae were dissected from the right ventricles of mouse hearts. Cardiomyocytes were enzymatically isolated from the ventricles of mouse hearts. Force was measured with a strain gauge in trabeculae (22°C). To assess arrhythmia susceptibility, the minimal extracellular Ca2+ concentration ([Ca2+]o,min), at which arrhythmias were induced by electrical stimulation, was determined in trabeculae. ROS production was estimated with 2',7'-dichlorofluorescein (DCF), mitochondrial membrane potential with tetramethylrhodamine methyl ester (TMRM), and Ca2+ spark frequency with fluo-4 and confocal microscopy in cardiomyocytes. ROS production within the mitochondria was estimated with MitoSoxRed and mitochondrial Ca2+ with rhod-2 in trabeculae. Diazoxide was used to activate mitochondrial KATP. Most of cCx43-/- mice died suddenly within 8 weeks. Cx43 was present in the inner mitochondrial membrane in cCx43+/+ mice but not in cCx43-/- mice. In cCx43-/- mice, the [Ca2+]o,min was lower, and Ca2+ spark frequency, the slope of DCF fluorescence intensity, MitoSoxRed fluorescence, and rhod-2 fluorescence were higher. TMRM fluorescence was more decreased in cCx43-/- mice. Most of these changes were suppressed by diazoxide. In addition, in cCx43-/- mice, antioxidant peptide SS-31 and N-acetyl-L-cysteine increased the [Ca2+]o,min. These results suggest that Cx43 deficiency activates Ca2+ leak from the SR, probably due to depolarization of mitochondrial membrane potential, an increase in mitochondrial Ca2+, and an increase in ROS production, thereby causing triggered arrhythmias, and that Cx43 hemichannel deficiency may be compensated by activation of mitochondrial KATP channels in mouse hearts.
Collapse
|
42
|
Berry BJ, Pharaoh GA, Marcinek DJ. From mitochondria to cells to humans: Targeting bioenergetics in aging and disease. Int J Biochem Cell Biol 2023; 157:106391. [PMID: 36806357 PMCID: PMC10033341 DOI: 10.1016/j.biocel.2023.106391] [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: 02/04/2023] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
In vivo control over metabolism is at the cutting edge of biomedical research. The particulars of mitochondrial function are especially important to understand in vivo to progress metabolic therapies that will be relevant for diseases of aging. Understanding the differences between how mitochondria function in vitro versus in vivo will be a necessary challenge to overcome to achieve mitochondrial medicine. In this article we outline how discoveries in invertebrate models will be informative for understanding the basic biology of mitochondria to streamline translation to mammals and eventually to humans. Further, we highlight examples of how what is known about mitochondria in vitro is translatable to in vivo models and, in some cases, to human diseases.
Collapse
Affiliation(s)
- Brandon J Berry
- University of Washington Medical Center, Department of Laboratory Medicine and Pathology, 1959 NE Pacific St., Seattle, WA 98195, USA.
| | - Gavin A Pharaoh
- University of Washington, Department of Radiology, and Institute for Stem Cell and Regenerative Medicine, South Lake Union Campus, 850 Republican St., Brotman D142, Box 358050, Seattle, WA 98109, USA.
| | - David J Marcinek
- University of Washington Medical Center, Department of Laboratory Medicine and Pathology, 1959 NE Pacific St., Seattle, WA 98195, USA; University of Washington, Department of Radiology, and Institute for Stem Cell and Regenerative Medicine, South Lake Union Campus, 850 Republican St., Brotman D142, Box 358050, Seattle, WA 98109, USA.
| |
Collapse
|
43
|
Nanodrugs alleviate acute kidney injury: Manipulate RONS at kidney. Bioact Mater 2023; 22:141-167. [PMID: 36203963 PMCID: PMC9526023 DOI: 10.1016/j.bioactmat.2022.09.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/12/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023] Open
Abstract
Currently, there are no clinical drugs available to treat acute kidney injury (AKI). Given the high prevalence and high mortality rate of AKI, the development of drugs to effectively treat AKI is a huge unmet medical need and a research hotspot. Although existing evidence fully demonstrates that reactive oxygen and nitrogen species (RONS) burst at the AKI site is a major contributor to AKI progression, the heterogeneity, complexity, and unique physiological structure of the kidney make most antioxidant and anti-inflammatory small molecule drugs ineffective because of the lack of kidney targeting and side effects. Recently, nanodrugs with intrinsic kidney targeting through the control of size, shape, and surface properties have opened exciting prospects for the treatment of AKI. Many antioxidant nanodrugs have emerged to address the limitations of current AKI treatments. In this review, we systematically summarized for the first time about the emerging nanodrugs that exploit the pathological and physiological features of the kidney to overcome the limitations of traditional small-molecule drugs to achieve high AKI efficacy. First, we analyzed the pathological structural characteristics of AKI and the main pathological mechanism of AKI: hypoxia, harmful substance accumulation-induced RONS burst at the renal site despite the multifactorial initiation and heterogeneity of AKI. Subsequently, we introduced the strategies used to improve renal targeting and reviewed advances of nanodrugs for AKI: nano-RONS-sacrificial agents, antioxidant nanozymes, and nanocarriers for antioxidants and anti-inflammatory drugs. These nanodrugs have demonstrated excellent therapeutic effects, such as greatly reducing oxidative stress damage, restoring renal function, and low side effects. Finally, we discussed the challenges and future directions for translating nanodrugs into clinical AKI treatment. AKI is a common clinical acute syndrome with high morbidity and mortality but without effective clinical drug available. Hypoxia and accumulation of toxic substances are key pathological features of various heterogeneous AKI. Excessive RONS is the core of the pathological mechanism of AKI. The development of nanodrugs is expected to achieve successful treatment in AKI.
Collapse
|
44
|
Hu H, Chen X, Zhao K, Zheng W, Gao C. Recent Advances in Biomaterials-Based Therapies for Alleviation and Regeneration of Traumatic Brain Injury. Macromol Biosci 2023; 23:e2200577. [PMID: 36758541 DOI: 10.1002/mabi.202200577] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Indexed: 02/11/2023]
Abstract
Traumatic brain injury (TBI), a major public health problem accompanied with numerous complications, usually leads to serve disability and huge financial burden. The adverse and unfavorable pathological environment triggers a series of secondary injuries, resulting in serious loss of nerve function and huge obstacle of endogenous nerve regeneration. With the advances in adaptive tissue regeneration biomaterials, regulation of detrimental microenvironment to reduce the secondary injury and to promote the neurogenesis becomes possible. The adaptive biomaterials could respond and regulate biochemical, cellular, and physiological events in the secondary injury, including excitotoxicity, oxidative stress, and neuroinflammation, to rebuild circumstances suitable for regeneration. In this review, the development of pathology after TBI is discussed, followed by the introduction of adaptive biomaterials based on various pathological characteristics. The adaptive biomaterials carried with neurotrophic factors and stem cells for TBI treatment are then summarized. Finally, the current drawbacks and future perspective of biomaterials for TBI treatment are suggested.
Collapse
Affiliation(s)
- Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiping Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Kefei Zhao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.,Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| |
Collapse
|
45
|
Pharaoh G, Kamat V, Kannan S, Stuppard RS, Whitson J, Martin-Perez M, Qian WJ, MacCoss MJ, Villen J, Rabinovitch P, Campbell MD, Sweet IR, Marcinek DJ. Elamipretide Improves ADP Sensitivity in Aged Mitochondria by Increasing Uptake through the Adenine Nucleotide Translocator (ANT). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.525989. [PMID: 36778398 PMCID: PMC9915686 DOI: 10.1101/2023.02.01.525989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aging muscle experiences functional decline in part mediated by impaired mitochondrial ADP sensitivity. Elamipretide (ELAM) rapidly improves physiological and mitochondrial function in aging and binds directly to the mitochondrial ADP transporter ANT. We hypothesized that ELAM improves ADP sensitivity in aging leading to rescued physiological function. We measured the response to ADP stimulation in young and old muscle mitochondria with ELAM treatment, in vivo heart and muscle function, and compared protein abundance, phosphorylation, and S-glutathionylation of ADP/ATP pathway proteins. ELAM treatment increased ADP sensitivity in old muscle mitochondria by increasing uptake of ADP through the ANT and rescued muscle force and heart systolic function. Protein abundance in the ADP/ATP transport and synthesis pathway was unchanged, but ELAM treatment decreased protein s-glutathionylation incuding of ANT. Mitochondrial ADP sensitivity is rapidly modifiable. This research supports the hypothesis that ELAM improves ANT function in aging and links mitochondrial ADP sensitivity to physiological function. Abstract Figure
Collapse
Affiliation(s)
- Gavin Pharaoh
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Varun Kamat
- Department of Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - Sricharan Kannan
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Rudolph S. Stuppard
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Jeremy Whitson
- Department of Biology, High Point University, High Point, NC, 27268, USA
| | - Miguel Martin-Perez
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, 08028, Spain
| | - Wei-Jun Qian
- Integrative Omics Group, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Michael J. MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Judit Villen
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Peter Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Matthew D. Campbell
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Ian R. Sweet
- Department of Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - David J. Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
46
|
Targeting mitochondrial impairment for the treatment of cardiovascular diseases: From hypertension to ischemia-reperfusion injury, searching for new pharmacological targets. Biochem Pharmacol 2023; 208:115405. [PMID: 36603686 DOI: 10.1016/j.bcp.2022.115405] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Mitochondria and mitochondrial proteins represent a group of promising pharmacological target candidates in the search of new molecular targets and drugs to counteract the onset of hypertension and more in general cardiovascular diseases (CVDs). Indeed, several mitochondrial pathways result impaired in CVDs, showing ATP depletion and ROS production as common traits of cardiac tissue degeneration. Thus, targeting mitochondrial dysfunction in cardiomyocytes can represent a successful strategy to prevent heart failure. In this context, the identification of new pharmacological targets among mitochondrial proteins paves the way for the design of new selective drugs. Thanks to the advances in omics approaches, to a greater availability of mitochondrial crystallized protein structures and to the development of new computational approaches for protein 3D-modelling and drug design, it is now possible to investigate in detail impaired mitochondrial pathways in CVDs. Furthermore, it is possible to design new powerful drugs able to hit the selected pharmacological targets in a highly selective way to rescue mitochondrial dysfunction and prevent cardiac tissue degeneration. The role of mitochondrial dysfunction in the onset of CVDs appears increasingly evident, as reflected by the impairment of proteins involved in lipid peroxidation, mitochondrial dynamics, respiratory chain complexes, and membrane polarization maintenance in CVD patients. Conversely, little is known about proteins responsible for the cross-talk between mitochondria and cytoplasm in cardiomyocytes. Mitochondrial transporters of the SLC25A family, in particular, are responsible for the translocation of nucleotides (e.g., ATP), amino acids (e.g., aspartate, glutamate, ornithine), organic acids (e.g. malate and 2-oxoglutarate), and other cofactors (e.g., inorganic phosphate, NAD+, FAD, carnitine, CoA derivatives) between the mitochondrial and cytosolic compartments. Thus, mitochondrial transporters play a key role in the mitochondria-cytosol cross-talk by leading metabolic pathways such as the malate/aspartate shuttle, the carnitine shuttle, the ATP export from mitochondria, and the regulation of permeability transition pore opening. Since all these pathways are crucial for maintaining healthy cardiomyocytes, mitochondrial carriers emerge as an interesting class of new possible pharmacological targets for CVD treatments.
Collapse
|
47
|
Ishkaeva RA, Salakhieva DV, Garifullin R, Alshadidi R, Laikov AV, Yergeshov AA, Kamalov MI, Abdullin TI. A new triphenylphosphonium-conjugated amphipathic cationic peptide with improved cell-penetrating and ROS-targeting properties. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 4:100148. [PMID: 36593927 PMCID: PMC9804109 DOI: 10.1016/j.crphar.2022.100148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
We study for the first time whether triphenylphosphonium (TPP) moiety can improve cellular delivery and redox properties of amphipathic cationic peptides based on YRFK/YrFK cell-penetrating and cytoprotective motif. TPP moiety was found to increase reducing activity of both stereoisomeric peptides in solution and on electrode surface in association with TPP-mediated intramolecular interactions. Among TPP-conjugated peptides, newly synthesized TPP3-YrFK featured both increased antioxidant efficacy and proteolytic resistance. TPP-conjugated peptides preferably mitigated endogenic ROS in mitochondria and cytoplasm of model glioblastoma cells with increased oxidative status. This anti-ROS effect was accompanied by mild reversible decrease of reduced glutathione level in the cells with relatively weak change in glutathione redox forms ratio. Such low interference with cell redox status is in accordance with non-cytotoxic nature of the compounds. Intracellular concentrations of label-free peptides were analyzed by LC-MS/MS, which showed substantial TPP-promoted penetration of YrFK motif across cell plasma membrane. However, according to ΔΨm analysis, TPP moiety did not profoundly enhance peptide interaction with mitochondrial inner membrane. Our study clarifies the role of TPP moiety in cellular delivery of amphipathic cationic oligopeptides. The results suggest TPP moiety as a multi-functional modifier for the oligopeptides which is capable of improving cellular pharmacokinetics and antioxidant activity as well as targeting increased ROS levels. The results encourage further investigation of TPP3-YrFK as a peptide antioxidant with multiple benefits.
Collapse
Key Words
- ABTS, 2,2′-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid)
- Amphipathic cationic peptides
- CCCP, carbonyl cyanide 3-chlorophenylhydrazone
- CD, circular dichroism
- Cellular pharmacokinetics
- DCFDA, 2′,7′-dichlorofluorescin diacetate
- GSH, reduced glutathione
- HBSS, Hank's balanced salt solution
- Intramolecular interaction
- LC–MS/MS, liquid chromatography tandem mass-spectrometry
- MCB, monochlorobimane
- MRM, multiple reaction monitoring
- ROS targeting
- ROS, reactive oxygen species
- Redox activity
- SPPS, solid-phase peptide synthesis
- TPP, triphenylphosphonium
- Triphenylphosphonium cation
- aa, amino acid
Collapse
Affiliation(s)
- Rezeda A. Ishkaeva
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia
| | - Diana V. Salakhieva
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia
| | - Ruslan Garifullin
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia,Department of Aeronautical Engineering, University of Turkish Aeronautical Association, Türkkuşu Kampüsü, 06790, Ankara, Turkey
| | - Raghad Alshadidi
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia
| | - Alexander V. Laikov
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia
| | - Abdulla A. Yergeshov
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia
| | - Marat I. Kamalov
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia
| | - Timur I. Abdullin
- Department of Biochemistry, Biotechnology, Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Volga Region Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia,Corresponding author. Department of Biochemistry, Biotechnology and Pharmacology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., 420008, Kazan, Russia.
| |
Collapse
|
48
|
Eggelbusch M, Shi A, Broeksma BC, Vázquez-Cruz M, Soares MN, de Wit GMJ, Everts B, Jaspers RT, Wüst RCI. The NLRP3 inflammasome contributes to inflammation-induced morphological and metabolic alterations in skeletal muscle. J Cachexia Sarcopenia Muscle 2022; 13:3048-3061. [PMID: 35978267 PMCID: PMC9745466 DOI: 10.1002/jcsm.13062] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/09/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Systemic inflammation is associated with skeletal muscle atrophy and metabolic dysfunction. Although the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome contributes to cytokine production in immune cells, its role in skeletal muscle is poorly understood. Here, we studied the link between inflammation, NLRP3, muscle morphology, and metabolism in in vitro cultured C2C12 myotubes, independent of immune cell involvement. METHODS Differentiated C2C12 myotubes were treated with lipopolysaccharide (LPS; 0, 10, and 100-200 ng/mL) to induce activation of the NLRP3 inflammasome with and without MCC950, a pharmacological inhibitor of NLRP3-induced IL-1β production. We assessed markers of the NLRP3 inflammasome, cell diameter, reactive oxygen species, and mitochondrial function. RESULTS NLRP3 gene expression and protein concentrations increased in a time-dependent and dose-dependent manner. Intracellular IL-1β concentration significantly increased (P < 0.0001), but significantly less with MCC950 (P = 0.03), suggestive of moderate activation of the NLRP3 inflammasome in cultured myotubes upon LPS stimulation. LPS suppressed myotube growth after 24 h (P = 0.03), and myotubes remained smaller up to 72 h (P = 0.0009). Exposure of myotubes to IL-1β caused similar alterations in cell morphology, and MCC950 mitigated these LPS-induced differences in cell diameter. NLRP3 appeared to co-localize with mitochondria, more so upon exposure to LPS. Mitochondrial reactive oxygen species were higher after LPS (P = 0.03), but not after addition of MCC950. Myotubes had higher glycolytic rates, and mitochondria were more fragmented upon LPS exposure, which was not altered by MCC950 supplementation. CONCLUSIONS LPS-induced activation of the NLRP3 inflammasome in cultured myotubes contributes to morphological and metabolic alterations, likely due to its mitochondrial association.
Collapse
Affiliation(s)
- Moritz Eggelbusch
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Nutrition and Dietetics, Amsterdam UMC location VUmc, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Faculty of Sports and Nutrition, Center of Expertise Urban Vitality, Amsterdam University of Applied Sciences, Amsterdam, The Netherlands
| | - Andi Shi
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Bonnie C Broeksma
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Mariana Vázquez-Cruz
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Madu N Soares
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Gerard M J de Wit
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, University of Leiden, Leiden, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Rob C I Wüst
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
49
|
Campbell MD, Martín-Pérez M, Egertson JD, Gaffrey MJ, Wang L, Bammler T, Rabinovitch PS, MacCoss M, Qian WJ, Villen J, Marcinek D. Elamipretide effects on the skeletal muscle phosphoproteome in aged female mice. GeroScience 2022; 44:2913-2924. [PMID: 36322234 PMCID: PMC9768078 DOI: 10.1007/s11357-022-00679-0] [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: 09/19/2022] [Accepted: 10/20/2022] [Indexed: 12/24/2022] Open
Abstract
The age-related decline in skeletal muscle mass and function is known as sarcopenia. Sarcopenia progresses based on complex processes involving protein dynamics, cell signaling, oxidative stress, and repair. We have previously found that 8-week treatment with elamipretide improves skeletal muscle function, reverses redox stress, and restores protein S-glutathionylation changes in aged female mice. This study tested whether 8-week treatment with elamipretide also affects global phosphorylation in skeletal muscle consistent with functional improvements and S-glutathionylation. Using female 6-7-month-old mice and 28-29-month-old mice, we found that phosphorylation changes did not relate to S-glutathionylation modifications, but that treatment with elamipretide did partially reverse age-related changes in protein phosphorylation in mouse skeletal muscle.
Collapse
Affiliation(s)
- Matthew D Campbell
- Department of Radiology, University of Washington, South Lake Union Campus, 850 Republican St., Brotman D142, Box 358050, Seattle, WA, 98109, USA
| | | | - Jarrett D Egertson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Matthew J Gaffrey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Peter S Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Judit Villen
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - David Marcinek
- Department of Radiology, University of Washington, South Lake Union Campus, 850 Republican St., Brotman D142, Box 358050, Seattle, WA, 98109, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
50
|
Valencia AP, Whitson JA, Wang S, Nguyen L, den Hartigh LJ, Rabinovitch PS, Marcinek DJ. Aging Increases Susceptibility to Develop Cardiac Hypertrophy following High Sugar Consumption. Nutrients 2022; 14:4645. [PMID: 36364920 PMCID: PMC9655368 DOI: 10.3390/nu14214645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 02/15/2024] Open
Abstract
Aging and poor diet are independent risk factors for heart disease, but the impact of high-sucrose (HS) consumption in the aging heart is understudied. Aging leads to impairments in mitochondrial function that result in muscle dysfunction (e.g., cardiac remodeling and sarcopenia). We tested whether HS diet (60%kcal sucrose) would accelerate muscle dysfunction in 24-month-old male CB6F1 mice. By week 1 on HS diet, mice developed significant cardiac hypertrophy compared to age-matched chow-fed controls. The increased weight of the heart persisted throughout the 4-week treatment, while body weight and strength declined more rapidly than controls. We then tested whether HS diet could worsen cardiac dysfunction in old mice and if the mitochondrial-targeted drug, elamipretide (ELAM), could prevent the diet-induced effect. Old and young mice were treated with either ELAM or saline as a control for 2 weeks, and provided with HS diet or chow on the last week. As demonstrated in the previous experiment, old mice had age-related cardiac hypertrophy that worsened after one week on HS and was prevented by ELAM treatment, while the HS diet had no detectable effect on hypertrophy in the young mice. As expected, mitochondrial respiration and reactive oxygen species (ROS) production were altered by age, but were not significantly affected by HS diet or ELAM. Our findings highlight the vulnerability of the aged heart to HS diet that can be prevented by systemic targeting of the mitochondria with ELAM.
Collapse
Affiliation(s)
- Ana P. Valencia
- Department of Radiology, University of Washington, Seattle, WA 98109, USA
| | - Jeremy A. Whitson
- Department of Biology, High Point University, High Point, NC 27268, USA
| | - Shari Wang
- Department of Medicine, Metabolism, University of Washington, Seattle, WA 98109, USA
| | - Leon Nguyen
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Laura J. den Hartigh
- Department of Medicine, Metabolism, University of Washington, Seattle, WA 98109, USA
| | | | - David J. Marcinek
- Department of Radiology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|