1
|
Zhao H, Zhao H, Ji S. A Mesenchymal stem cell Aging Framework, from Mechanisms to Strategies. Stem Cell Rev Rep 2024; 20:1420-1440. [PMID: 38727878 DOI: 10.1007/s12015-024-10732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 08/13/2024]
Abstract
Mesenchymal stem cells (MSCs) are extensively researched for therapeutic applications in tissue engineering and show significant potential for clinical use. Intrinsic or extrinsic factors causing senescence may lead to reduced proliferation, aberrant differentiation, weakened immunoregulation, and increased inflammation, ultimately limiting the potential of MSCs. It is crucial to comprehend the molecular pathways and internal processes responsible for the decline in MSC function due to senescence in order to devise innovative approaches for rejuvenating senescent MSCs and enhancing MSC treatment. We investigate the main molecular processes involved in senescence, aiming to provide a thorough understanding of senescence-related issues in MSCs. Additionally, we analyze the most recent advancements in cutting-edge approaches to combat MSC senescence based on current research. We are curious whether the aging process of stem cells results in a permanent "memory" and if cellular reprogramming may potentially revert the aging epigenome to a more youthful state.
Collapse
Affiliation(s)
- Hongqing Zhao
- Nanbu County People's Hospital, Nanchong City, 637300, Sichuan Province, China
- Jinzhou Medical University, No.82 Songpo Road, Guta District, Jinzhou, 121001, Liaoning Province, China
| | - Houming Zhao
- Graduate School of PLA Medical College, Chinese PLA General Hospital, Beijing, 100083, China
| | - Shuaifei Ji
- Graduate School of PLA Medical College, Chinese PLA General Hospital, Beijing, 100083, China.
| |
Collapse
|
2
|
Bennett S, Tiollier E, Owens DJ, Brocherie F, Louis JB. Implications of Heat Stress-induced Metabolic Alterations for Endurance Training. Int J Sports Med 2024; 45:422-435. [PMID: 38401534 DOI: 10.1055/a-2251-3170] [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: 02/26/2024]
Abstract
Inducing a heat-acclimated phenotype via repeated heat stress improves exercise capacity and reduces athletes̓ risk of hyperthermia and heat illness. Given the increased number of international sporting events hosted in countries with warmer climates, heat acclimation strategies are increasingly popular among endurance athletes to optimize performance in hot environments. At the tissue level, completing endurance exercise under heat stress may augment endurance training adaptation, including mitochondrial and cardiovascular remodeling due to increased perturbations to cellular homeostasis as a consequence of metabolic and cardiovascular load, and this may improve endurance training adaptation and subsequent performance. This review provides an up-to-date overview of the metabolic impact of heat stress during endurance exercise, including proposed underlying mechanisms of altered substrate utilization. Against this metabolic backdrop, the current literature highlighting the role of heat stress in augmenting training adaptation and subsequent endurance performance will be presented with practical implications and opportunities for future research.
Collapse
Affiliation(s)
- Samuel Bennett
- Center for Biological Clocks Research, Texas A&M University, College Station, United States
| | - Eve Tiollier
- Laboratory Sport, Expertise and Performance, Research Department, Institut National du Sport de l'Expertise et de la Performance, Paris, France
| | - Daniel J Owens
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom of Great Britain and Northern Ireland
| | - Franck Brocherie
- Laboratory Sport, Expertise and Performance, Research Department, Institut National du Sport de l'Expertise et de la Performance, Paris, France
| | - Julien B Louis
- Laboratory Sport, Expertise and Performance, Research Department, Institut National du Sport de l'Expertise et de la Performance, Paris, France
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
3
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
4
|
Dong M, Zhang T, Liang X, Cheng X, Shi F, Yuan H, Zhang F, Jiang Q, Wang X. Sesamin alleviates lipid accumulation induced by oleic acid via PINK1/Parkin-mediated mitophagy in HepG2 cells. Biochem Biophys Res Commun 2024; 708:149815. [PMID: 38531220 DOI: 10.1016/j.bbrc.2024.149815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
Sesamin, a special compound present in sesame and sesame oil, has been reported a role in regulating lipid metabolism, while the underlying mechanisms remain unclear. Autophagy has been reported associated with lipid metabolism and regarded as a key modulator in liver steatosis. The present work aimed to investigate whether sesamin could exert its protective effects against lipid accumulation via modulating autophagy in HepG2 cells stimulated with oleic acid (OA). Cell viability was evaluated using the CCK-8 method, and triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein, cholesterol (LDL-C), alanine aminotransferase (ALT), along with aspartate aminotransferase (AST) were assessed by oil red O staining, transmission electron microscopy (TEM), and biochemical kits to investigate the lipid-lowering effects of sesamin. Differentially expressed genes were screened by RNA sequencing and validated using real-time quantitative PCR and Western blot. Autophagy and mitophagy related molecules were analyzed employing TEM, Western blot, and immunofluorescence. The data shows that in HepG2 cells stimulated by OA, sesamin reduces levels of TG, TC, LDL-C, ALT, and AST while elevating HDL-C, alleviates the lipid accumulation and improves fatty acid metabolism through modulating the levels of fat metabolism related genes including PCSK9, FABP1, CD36, and SOX4. Sesamin restores the suppressed autophagy in HepG2 cells caused by OA, which could be blocked by autophagy inhibitors. This indicates that sesamin improves fatty acid metabolism by enhancing autophagy levels, thereby mitigating the intracellular lipid accumulation. Furthermore, sesamin significantly enhances the mitophagy and improves mitochondrial homeostasis via activating the PINK/Parkin pathway. These data suggest that sesamin alleviates the excessive lipid accumulation in HepG2 caused by OA by restoring the impaired mitophagy via the PINK1/Parkin pathway, probably playing a preventive or therapeutic role in hepatic steatosis.
Collapse
Affiliation(s)
- Mengyun Dong
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Tianliang Zhang
- Experimental Center for Medical Research, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Xueli Liang
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Xinyi Cheng
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Fuyan Shi
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Hang Yuan
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Fengxiang Zhang
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qiqi Jiang
- Department of Gastroenterology, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, China.
| | - Xia Wang
- Department of Gastroenterology, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, 261000, China.
| |
Collapse
|
5
|
Zhou Z, Ma A, Moore TM, Wolf DM, Yang N, Tran P, Segawa M, Strumwasser AR, Ren W, Fu K, Wanagat J, van der Bliek AM, Crosbie-Watson R, Liesa M, Stiles L, Acin-Perez R, Mahata S, Shirihai O, Goodarzi MO, Handzlik M, Metallo CM, Walker DW, Hevener AL. Drp1 controls complex II assembly and skeletal muscle metabolism by Sdhaf2 action on mitochondria. SCIENCE ADVANCES 2024; 10:eadl0389. [PMID: 38569044 PMCID: PMC10990287 DOI: 10.1126/sciadv.adl0389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
The dynamin-related guanosine triphosphatase, Drp1 (encoded by Dnm1l), plays a central role in mitochondrial fission and is requisite for numerous cellular processes; however, its role in muscle metabolism remains unclear. Here, we show that, among human tissues, the highest number of gene correlations with DNM1L is in skeletal muscle. Knockdown of Drp1 (Drp1-KD) promoted mitochondrial hyperfusion in the muscle of male mice. Reduced fatty acid oxidation and impaired insulin action along with increased muscle succinate was observed in Drp1-KD muscle. Muscle Drp1-KD reduced complex II assembly and activity as a consequence of diminished mitochondrial translocation of succinate dehydrogenase assembly factor 2 (Sdhaf2). Restoration of Sdhaf2 normalized complex II activity, lipid oxidation, and insulin action in Drp1-KD myocytes. Drp1 is critical in maintaining mitochondrial complex II assembly, lipid oxidation, and insulin sensitivity, suggesting a mechanistic link between mitochondrial morphology and skeletal muscle metabolism, which is clinically relevant in combatting metabolic-related diseases.
Collapse
Affiliation(s)
- Zhenqi Zhou
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alice Ma
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy M. Moore
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Dane M. Wolf
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicole Yang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Tran
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mayuko Segawa
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Alexander R. Strumwasser
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wenjuan Ren
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kai Fu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jonathan Wanagat
- Division of Geriatrics, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | | | - Rachelle Crosbie-Watson
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marc Liesa
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linsey Stiles
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rebecca Acin-Perez
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sushil Mahata
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Orian Shirihai
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90095, USA
| | - Michal Handzlik
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christian M. Metallo
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - David W. Walker
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andrea L. Hevener
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Iris Cantor UCLA Women’s Health Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine and VA Greater Los Angeles Healthcare System GRECC, Los Angeles, CA 90073, USA
| |
Collapse
|
6
|
Noh SG, Ahn A, Davi SM, Lepley LK, Kwon OS. Quadriceps muscle atrophy after non-invasive anterior cruciate ligament injury: evidence linking to autophagy and mitophagy. Front Physiol 2024; 15:1341723. [PMID: 38496299 PMCID: PMC10940348 DOI: 10.3389/fphys.2024.1341723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction: Anterior cruciate ligament (ACL) injury is frequently accompanied by quadriceps muscle atrophy, a process closely linked to mitochondrial health and mitochondria-specific autophagy. However, the temporal progression of key quadricep atrophy-mediating events following ACL injury remains poorly understood. To advance our understanding, we conducted a longitudinal study to elucidate key parameters in quadriceps autophagy and mitophagy. Methods: Long-Evans rats were euthanized at 7, 14, 28, and 56 days after non-invasive ACL injury that was induced via tibial compression overload; controls were not injured. Vastus lateralis muscle was extracted, and subsequent immunoblotting analysis was conducted using primary antibodies targeting key proteins involved in autophagy and mitophagy cellular processes. Results: Our findings demonstrated dynamic changes in autophagy and mitophagy markers in the quadriceps muscle during the recovery period after ACL injury. The early response to the injury was characterized by the induction of autophagy at 14 days (Beclin1), indicating an initial cellular response to the injury. Subsequently, at 14 days we observed increase in the elongation of autophagosomes (Atg4B), suggesting a potential remodeling process. The autophagosome flux was also augmented between 14- and 28 days (LC3-II/LC3-I ratio and p62). Notably, at 56 days, markers associated with the elimination of damaged mitochondria were elevated (PINK1, Parkin, and VDAC1), indicating a possible ongoing cellular repair and restoration process. Conclusion: These data highlight the complexity of muscle recovery after ACL injury and underscore the overlooked but crucial role of autophagy and mitophagy in promoting the recovery process.
Collapse
Affiliation(s)
- Sung Gi Noh
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
| | - Ahram Ahn
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
| | - Steven M. Davi
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
- Cooperative Studies Program Coordinating Center (CSPCC), VA Connecticut Healthcare System, West Haven, CT, United States
| | - Lindsey K. Lepley
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
- Department of Orthopaedic Surgery and Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
7
|
Li X, Han Y, Meng Y, Yin L. Small RNA-big impact: exosomal miRNAs in mitochondrial dysfunction in various diseases. RNA Biol 2024; 21:1-20. [PMID: 38174992 PMCID: PMC10773649 DOI: 10.1080/15476286.2023.2293343] [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] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Mitochondria are multitasking organelles involved in maintaining the cell homoeostasis. Beyond its well-established role in cellular bioenergetics, mitochondria also function as signal organelles to propagate various cellular outcomes. However, mitochondria have a self-destructive arsenal of factors driving the development of diseases caused by mitochondrial dysfunction. Extracellular vesicles (EVs), a heterogeneous group of membranous nano-sized vesicles, are present in a variety of bodily fluids. EVs serve as mediators for intercellular interaction. Exosomes are a class of small EVs (30-100 nm) released by most cells. Exosomes carry various cargo including microRNAs (miRNAs), a class of short noncoding RNAs. Recent studies have closely associated exosomal miRNAs with various human diseases, including diseases caused by mitochondrial dysfunction, which are a group of complex multifactorial diseases and have not been comprehensively described. In this review, we first briefly introduce the characteristics of EVs. Then, we focus on possible mechanisms regarding exosome-mitochondria interaction through integrating signalling networks. Moreover, we summarize recent advances in the knowledge of the role of exosomal miRNAs in various diseases, describing how mitochondria are changed in disease status. Finally, we propose future research directions to provide a novel therapeutic strategy that could slow the disease progress mediated by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Xiaqing Li
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University Guangzhou, Guangdong, China
- Central laboratory, The Fifth Hospital Affiliated to Jinan University, Heyuan, China
| | - Yi Han
- Traditional Chinese Medicine Department, People’s Hospital of Yanjiang District, Ziyang, Sichuan, China
| | - Yu Meng
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University Guangzhou, Guangdong, China
- Central laboratory, The Fifth Hospital Affiliated to Jinan University, Heyuan, China
| | - Lianghong Yin
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Wang T, Wang X, Fu T, Ma Y, Wang Q, Zhang S, Zhang X, Zhou H, Chang X, Tong Y. Roles of mitochondrial dynamics and mitophagy in diabetic myocardial microvascular injury. Cell Stress Chaperones 2023; 28:675-688. [PMID: 37755621 PMCID: PMC10746668 DOI: 10.1007/s12192-023-01384-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Myocardial microvessels are composed of a monolayer of endothelial cells, which play a crucial role in maintaining vascular barrier function, luminal latency, vascular tone, and myocardial perfusion. Endothelial dysfunction is a key factor in the development of cardiac microvascular injury and diabetic cardiomyopathy. In addition to their role in glucose oxidation and energy metabolism, mitochondria also participate in non-metabolic processes such as apoptosis, intracellular ion handling, and redox balancing. Mitochondrial dynamics and mitophagy are responsible for regulating the quality and quantity of mitochondria in response to hyperglycemia. However, these endogenous homeostatic mechanisms can both preserve and/or disrupt non-metabolic mitochondrial functions during diabetic endothelial damage and cardiac microvascular injury. This review provides an overview of the molecular features and regulatory mechanisms of mitochondrial dynamics and mitophagy. Furthermore, we summarize findings from various investigations that suggest abnormal mitochondrial dynamics and defective mitophagy contribute to the development of diabetic endothelial dysfunction and myocardial microvascular injury. Finally, we discuss different therapeutic strategies aimed at improving endothelial homeostasis and cardiac microvascular function through the enhancement of mitochondrial dynamics and mitophagy.
Collapse
Affiliation(s)
- Tong Wang
- Heilongjiang Academy of Chinese Medicine, Harbin, 150000, China
| | - Xinwei Wang
- Heilongjiang Academy of Chinese Medicine, Harbin, 150000, China
| | - Tong Fu
- Brandeis University, Waltham, MA, 02453, USA
| | - Yanchun Ma
- Heilongjiang Academy of Chinese Medicine, Harbin, 150000, China
| | - Qi Wang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Shuxiang Zhang
- Heilongjiang Academy of Chinese Medicine, Harbin, 150000, China
| | - Xiao Zhang
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, 100048, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, 100048, China
| | - Xing Chang
- Cardiovascular Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Ying Tong
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
9
|
Esmaeilzadeh A, Mohammadi V, Elahi R, Rezakhani N. The role of heat shock proteins (HSPs) in type 2 diabetes mellitus pathophysiology. J Diabetes Complications 2023; 37:108564. [PMID: 37852076 DOI: 10.1016/j.jdiacomp.2023.108564] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 10/20/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by sustained hyperglycemia caused by impaired insulin signaling and secretion. Metabolic stress, caused by an inappropriate diet, is one of the major hallmarks provoking inflammation, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction. Heat shock proteins (HSPs) are a group of highly conserved proteins that have a crucial role in chaperoning damaged and misfolded proteins to avoid disruption of cellular homeostasis under stress conditions. To do this, HSPs interact with diverse intra-and extracellular pathways among which are the insulin signaling, insulin secretion, and apoptosis pathways. Therefore, HSP dysfunction, e.g. HSP70, may lead to disruption of the pathways responsible for insulin secretion and uptake. Consistently, the altered expression of other HSPs and genetic polymorphisms in HSP-producing genes in diabetic subjects has made HSPs hot research in T2DM. This paper provides a comprehensive overview of the role of different HSPs in T2DM pathogenesis, affected cellular pathways, and the potential therapeutic strategies targeting HSPs in T2DM.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Vahid Mohammadi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Negin Rezakhani
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
10
|
Sklifasovskaya AP, Blagonravov M, Ryabinina A, Goryachev V, Syatkin S, Chibisov S, Akhmetova K, Prokofiev D, Agostinelli E. The role of heat shock proteins in the pathogenesis of heart failure (Review). Int J Mol Med 2023; 52:106. [PMID: 37772383 PMCID: PMC10558216 DOI: 10.3892/ijmm.2023.5309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
The influence of heat shock proteins (HSPs) on protein quality control systems in cardiomyocytes is currently under investigation. The effect of HSPs on the regulated cell death of cardiomyocytes (CMCs) is of great importance, since they play a major role in the implementation of compensatory and adaptive mechanisms in the event of cardiac damage. HSPs mediate a number of mechanisms that activate the apoptotic cascade, playing both pro‑ and anti‑apoptotic roles depending on their location in the cell. Another type of cell death, autophagy, can in some cases lead to cell death, while in other situations it acts as a cell survival mechanism. The present review considered the characteristics of the expression of HSPs of different molecular weights in CMCs in myocardial damage caused by heart failure, as well as their role in the realization of certain types of regulated cell death.
Collapse
Affiliation(s)
| | | | - Anna Ryabinina
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | | | - Sergey Syatkin
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Sergey Chibisov
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Karina Akhmetova
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Daniil Prokofiev
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Faculty of Medicine and Dentistry, Sapienza University of Rome, University Hospital Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation, ETS-ONLUS, I-00159 Rome, Italy
| |
Collapse
|
11
|
Titus AS, Sung EA, Zablocki D, Sadoshima J. Mitophagy for cardioprotection. Basic Res Cardiol 2023; 118:42. [PMID: 37798455 PMCID: PMC10556134 DOI: 10.1007/s00395-023-01009-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Mitochondrial function is maintained by several strictly coordinated mechanisms, collectively termed mitochondrial quality control mechanisms, including fusion and fission, degradation, and biogenesis. As the primary source of energy in cardiomyocytes, mitochondria are the central organelle for maintaining cardiac function. Since adult cardiomyocytes in humans rarely divide, the number of dysfunctional mitochondria cannot easily be diluted through cell division. Thus, efficient degradation of dysfunctional mitochondria is crucial to maintaining cellular function. Mitophagy, a mitochondria specific form of autophagy, is a major mechanism by which damaged or unnecessary mitochondria are targeted and eliminated. Mitophagy is active in cardiomyocytes at baseline and in response to stress, and plays an essential role in maintaining the quality of mitochondria in cardiomyocytes. Mitophagy is mediated through multiple mechanisms in the heart, and each of these mechanisms can partially compensate for the loss of another mechanism. However, insufficient levels of mitophagy eventually lead to mitochondrial dysfunction and the development of heart failure. In this review, we discuss the molecular mechanisms of mitophagy in the heart and the role of mitophagy in cardiac pathophysiology, with the focus on recent findings in the field.
Collapse
Affiliation(s)
- Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA.
| |
Collapse
|
12
|
Nejat S, Menikdiwela KR, Efotte A, Scoggin S, Vandanmagsar B, Thornalley PJ, Dehbi M, Moustaid-Moussa N. Genetic Deletion of DNAJB3 Using CRISPR-Cas9, Produced Discordant Phenotypes. Genes (Basel) 2023; 14:1857. [PMID: 37895206 PMCID: PMC10606339 DOI: 10.3390/genes14101857] [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: 08/18/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Several pathways and/or genes have been shown to be dysregulated in obesity-induced insulin resistance (IR) and type 2 diabetes (T2D). We previously showed, for the first time, impaired expression of DNAJB3 mRNA and protein in subjects with obesity, which was concomitant with increased metabolic stress. Restoring the normal expression of DNAJB3 attenuated metabolic stress and improved insulin signaling both in vivo and in vitro, suggesting a protective role of DNAJB3 against obesity and T2D. The precise underlying mechanisms remained, however, unclear. This study was designed to confirm the human studies in a mouse model of dietary obesity-induced insulin resistance, and, if validated, to understand the underlying mechanisms. We hypothesized that mice lacking DNAJB3 would be more prone to high-fat (HF)-diet-induced increase in body weight and body fat, inflammation, glucose intolerance and insulin resistance as compared with wild-type (WT) littermates. Three DNAJB3 knockout (KO) lines were generated (KO 30, 44 and 47), using CRISPR-Cas9. Male and female KO and WT mice were fed a HF diet (45% kcal fat) for 16 weeks. Body weight was measured biweekly, and a glucose tolerance test (GTT) and insulin tolerance test (ITT) were conducted at week 13 and 14, respectively. Body composition was determined monthly by nuclear magnetic resonance (NMR). Following euthanasia, white adipose tissue (WAT) and skeletal muscle were harvested for further analyses. Compared with WT mice, male and female KO 47 mice demonstrated higher body weight and fat mass. Similarly, KO 47 mice also showed a slower rate of glucose clearance in GTT that was consistent with decreased mRNA expression of the GLUT4 gene in WAT but not in the muscle. Both male and female KO 47 mice exhibited higher mRNA levels of the pro-inflammatory marker TNF-a in WAT only, whereas increased mRNA levels of MCP1 chemokine and the ER stress marker BiP/Grp78 were observed in male but not in female KO 47 mice. However, we did not observe the same changes in the other KO lines. Taken together, the phenotype of the DNAJB3 KO 47 mice was consistent with the metabolic changes and low levels of DNAJB3 reported in human subjects. These findings suggest that DNAJB3 may play an important role in metabolic functions and glucose homeostasis, which warrants further phenotyping and intervention studies in other KO 47 and other KO mice, as well as investigating this protein as a potential therapeutic target for obesity and T2D.
Collapse
Affiliation(s)
- Shadi Nejat
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (S.N.); (K.R.M.); (A.E.); (S.S.)
| | - Kalhara R. Menikdiwela
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (S.N.); (K.R.M.); (A.E.); (S.S.)
| | - Aliyah Efotte
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (S.N.); (K.R.M.); (A.E.); (S.S.)
| | - Shane Scoggin
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (S.N.); (K.R.M.); (A.E.); (S.S.)
| | | | - Paul J. Thornalley
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar; (P.J.T.); (M.D.)
| | - Mohammed Dehbi
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar; (P.J.T.); (M.D.)
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (S.N.); (K.R.M.); (A.E.); (S.S.)
| |
Collapse
|
13
|
Jiang X, Yang M, Fang Y, Yang Z, Dai X, Gu P, Feng W, Chen Y. A Photo-Activated Thermoelectric Catalyst for Ferroptosis-/Pyroptosis-Boosted Tumor Nanotherapy. Adv Healthc Mater 2023; 12:e2300699. [PMID: 37086391 DOI: 10.1002/adhm.202300699] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/03/2023] [Indexed: 04/23/2023]
Abstract
Phototherapy including photothermal therapy (PTT) and photodynamic therapy (PDT) has gradually come into the limelight for oncological treatment due to its noninvasiveness, high specificity, and low side effects. However, upregulated heat-shock proteins (HSPs) and reactive oxygen species (ROS)-defensing system such as glutathione (GSH) or MutT homolog 1 (MTH1) protein in tumor microenvironment counteract the efficiency of single-modality therapy either PTT or PDT. Herein, the well-defined bismuth telluride nanoplates (Bi2 Te3 NPs) are engineered with a high-performance photo-thermo-electro-catalytic effect for tumor-synergistic treatment. Upon near-infrared light illumination, Bi2 Te3 NPs induce a significant temperature elevation for PTT, which effectively inhibits MTH1 expression. Especially, heating and cooling alteration caused temperature variations result in electron-hole separation for ROS generation, which not only damages HSPs to reduce the thermotolerance for enhance PTT, but also arouses tumor cell pyroptosis. Additionally, Bi2 Te3 NPs conspicuously reduce GSH, further improving ROS level and leading to decrease glutathione peroxidase 4 (GPX4) activity, which triggers tumor cell ferroptosis. Due to the photo-thermo-electro-catalytic synergistic therapy, Bi2 Te3 NPs are gifted with impressive tumor suppression on both ectopic and orthotopic ocular tumor models. This work highlights a high-performance multifunctional energy-conversion nanoplatform for reshaping tumor microenvironment to boost the tumor-therapeutic efficacy of phototherapy.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Muyue Yang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Ying Fang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Zhenyu Yang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
14
|
F AR, Quadrilatero J. Emerging role of mitophagy in myoblast differentiation and skeletal muscle remodeling. Semin Cell Dev Biol 2023; 143:54-65. [PMID: 34924331 DOI: 10.1016/j.semcdb.2021.11.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022]
Abstract
Mitochondrial turnover in the form of mitophagy is emerging as a central process in maintaining cellular function. The degradation of damaged mitochondria through mitophagy is particularly important in cells/tissues that exhibit high energy demands. Skeletal muscle is one such tissue that requires precise turnover of mitochondria in several conditions in order to optimize energy production and prevent bioenergetic crisis. For instance, the formation of skeletal muscle (i.e., myogenesis) is accompanied by robust turnover of low-functioning mitochondria to eventually allow the formation of high-functioning mitochondria. In mature skeletal muscle, alterations in mitophagy-related signaling occur during exercise, aging, and various disease states. Nonetheless, several questions regarding the direct role of mitophagy in various skeletal muscle conditions remain unknown. Furthermore, given the heterogenous nature of skeletal muscle with respect to various cellular and molecular properties, and the plasticity in these properties in various conditions, the involvement and characterization of mitophagy requires more careful consideration in this tissue. Therefore, this review will highlight the known mechanisms of mitophagy in skeletal muscle, and discuss their involvement during myogenesis and various skeletal muscle conditions. This review also provides important considerations for the accurate measurement of mitophagy and interpretation of data in skeletal muscle.
Collapse
Affiliation(s)
- Ahmad Rahman F
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Joe Quadrilatero
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
15
|
Undamatla R, Fagunloye OG, Chen J, Edmunds LR, Murali A, Mills A, Xie B, Pangburn MM, Sipula I, Gibson G, St Croix C, Jurczak MJ. Reduced mitophagy is an early feature of NAFLD and liver-specific PARKIN knockout hastens the onset of steatosis, inflammation and fibrosis. Sci Rep 2023; 13:7575. [PMID: 37165006 PMCID: PMC10172344 DOI: 10.1038/s41598-023-34710-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of pathologies that includes steatosis, steatohepatitis (NASH) and fibrosis and is strongly associated with insulin resistance and type 2 diabetes. Changes in mitochondrial function are implicated in the pathogenesis of NAFLD, particularly in the transition from steatosis to NASH. Mitophagy is a mitochondrial quality control mechanism that allows for the selective removal of damaged mitochondria from the cell via the autophagy pathway. While past work demonstrated a negative association between liver fat content and rates of mitophagy, when changes in mitophagy occur during the pathogenesis of NAFLD and whether such changes contribute to the primary endpoints associated with the disease are currently poorly defined. We therefore undertook the studies described here to establish when alterations in mitophagy occur during the pathogenesis of NAFLD, as well as to determine the effects of genetic inhibition of mitophagy via conditional deletion of a key mitophagy regulator, PARKIN, on the development of steatosis, insulin resistance, inflammation and fibrosis. We find that loss of mitophagy occurs early in the pathogenesis of NAFLD and that loss of PARKIN accelerates the onset of key NAFLD disease features. These observations suggest that loss of mitochondrial quality control in response to nutritional stress may contribute to mitochondrial dysfunction and the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- R Undamatla
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - O G Fagunloye
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - J Chen
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - L R Edmunds
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - A Murali
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - A Mills
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - B Xie
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - M M Pangburn
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - I Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA
| | - G Gibson
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - C St Croix
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - M J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, University of Pittsburgh, 200 Lothrop Street, BST W1060, Pittsburgh, PA, 15213, USA.
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Chatzinikita E, Maridaki M, Palikaras K, Koutsilieris M, Philippou A. The Role of Mitophagy in Skeletal Muscle Damage and Regeneration. Cells 2023; 12:716. [PMID: 36899852 PMCID: PMC10000750 DOI: 10.3390/cells12050716] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Mitochondria are cellular organelles that play an essential role in generating the chemical energy needed for the biochemical reactions in cells. Mitochondrial biogenesis, i.e., de novo mitochondria formation, results in enhanced cellular respiration, metabolic processes, and ATP generation, while autophagic clearance of mitochondria (mitophagy) is required to remove damaged or useless mitochondria. The balance between the opposing processes of mitochondrial biogenesis and mitophagy is highly regulated and crucial for the maintenance of the number and function of mitochondria as well as for the cellular homeostasis and adaptations to metabolic demands and extracellular stimuli. In skeletal muscle, mitochondria are essential for maintaining energy homeostasis, and the mitochondrial network exhibits complex behaviors and undergoes dynamic remodeling in response to various conditions and pathologies characterized by changes in muscle cell structure and metabolism, such as exercise, muscle damage, and myopathies. In particular, the involvement of mitochondrial remodeling in mediating skeletal muscle regeneration following damage has received increased attention, as modifications in mitophagy-related signals arise from exercise, while variations in mitochondrial restructuring pathways can lead to partial regeneration and impaired muscle function. Muscle regeneration (through myogenesis) following exercise-induced damage is characterized by a highly regulated, rapid turnover of poor-functioning mitochondria, permitting the synthesis of better-functioning mitochondria to occur. Nevertheless, essential aspects of mitochondrial remodeling during muscle regeneration remain poorly understood and warrant further characterization. In this review, we focus on the critical role of mitophagy for proper muscle cell regeneration following damage, highlighting the molecular mechanisms of the mitophagy-associated mitochondrial dynamics and network reformation.
Collapse
Affiliation(s)
- Eirini Chatzinikita
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Maria Maridaki
- Faculty of Physical Education and Sport Science, National and Kapodistrian University of Athens, 172 37 Athens, Greece
| | - Konstantinos Palikaras
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| |
Collapse
|
17
|
Niinuma SA, Lubbad L, Lubbad W, Moin ASM, Butler AE. The Role of Heat Shock Proteins in the Pathogenesis of Polycystic Ovarian Syndrome: A Review of the Literature. Int J Mol Sci 2023; 24:ijms24031838. [PMID: 36768170 PMCID: PMC9915177 DOI: 10.3390/ijms24031838] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine disorder in women of reproductive age and post-menopausal women. PCOS is a multifactorial heterogeneous disorder associated with a variety of etiologies, outcomes, and clinical manifestations. However, the pathophysiology of PCOS is still unclear. Heat shock proteins (HSPs) have recently been investigated for their role in the pathogenesis of PCOS. HSPs are a class of proteins that act as molecular chaperones and maintain cellular proteostasis. More recently, their actions beyond that of molecular chaperones have highlighted their pathogenic role in several diseases. In PCOS, different HSP family members show abnormal expression that affects the proliferation and apoptotic rates of ovarian cells as well as immunological processes. HSP dysregulation in the ovaries of PCOS subjects leads to a proliferation/apoptosis imbalance that mechanistically impacts follicle stage development, resulting in polycystic ovaries. Moreover, HSPs may play a role in the pathogenesis of PCOS-associated conditions. Recent studies on HSP activity during therapeutic interventions for PCOS suggest that modulating HSP activity may lead to novel treatment strategies. In this review, we summarize what is currently known regarding the role of HSPs in the pathogenesis of PCOS and their potential role in the treatment of PCOS, and we outline areas for future research.
Collapse
Affiliation(s)
- Sara Anjum Niinuma
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Laila Lubbad
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Walaa Lubbad
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Abu Saleh Md Moin
- Research Department, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
- Correspondence: or ; Tel.: +973-66760313
| |
Collapse
|
18
|
Undamatla R, Fagunloye OG, Chen J, Edmunds LR, Murali A, Mills A, Xie B, Pangburn MM, Sipula I, Gibson G, Croix CS, Jurczak MJ. Reduced hepatocyte mitophagy is an early feature of NAFLD pathogenesis and hastens the onset of steatosis, inflammation and fibrosis. RESEARCH SQUARE 2023:rs.3.rs-2469234. [PMID: 36711642 PMCID: PMC9882688 DOI: 10.21203/rs.3.rs-2469234/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of pathologies that includes steatosis, steatohepatitis (NASH) and fibrosis and is strongly associated with insulin resistance and type 2 diabetes. Changes in mitochondrial function are implicated in the pathogenesis of NAFLD, particularly in the transition from steatosis to NASH. Mitophagy is a mitochondrial quality control mechanism that allows for the selective removal of damaged mitochondria from the cell via the autophagy pathway. While past work demonstrated a negative association between liver fat content and rates of mitophagy, when changes in mitophagy occur during the pathogenesis of NAFLD and whether such changes contribute to the primary endpoints associated with the disease are currently poorly defined. We therefore undertook the studies described here to establish when alterations in mitophagy occur during the pathogenesis of NAFLD, as well as to determine the effects of genetic inhibition of mitophagy via conditional deletion of a key mitophagy regulator, PARKIN, on the development of steatosis, insulin resistance, inflammation and fibrosis. We find that loss of mitophagy occurs early in the pathogenesis of NAFLD and that loss of PARKIN hastens the onset but not severity of key NAFLD disease features. These observations suggest that loss of mitochondrial quality control in response to nutritional stress may contribute to mitochondrial dysfunction and the pathogenesis of NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ian Sipula
- University of Pittsburgh School of Medicine
| | | | | | | |
Collapse
|
19
|
Kuppuswami J, Senthilkumar GP. Nutri-stress, mitochondrial dysfunction, and insulin resistance-role of heat shock proteins. Cell Stress Chaperones 2023; 28:35-48. [PMID: 36441381 PMCID: PMC9877269 DOI: 10.1007/s12192-022-01314-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 10/05/2022] [Accepted: 11/17/2022] [Indexed: 11/29/2022] Open
Abstract
Excess nutrient flux into the cellular energy system results in a scenario of cellular metabolic stress in diseases involving insulin resistance, such as type 2 diabetes, referred to as nutri-stress and results in cellular bioenergetic imbalance, which leads to insulin resistance and disease. Under nutri-stress, the heat shock response system is compromised due to metabolic abnormalities that disturb energy homeostasis. Heat shock proteins (HSPs) are the chief protectors of intracellular homeostasis during stress. Heat shock response (HSR) impairment contributes to several metabolic pathways that aggravate chronic hyperglycaemia and insulin resistance, highlighting a central role in disease pathogenesis. This article discusses the role of nutri-stress-related molecular events in causing insulin resistance and the nature of the roles played by heat shock proteins in some of the crucial checkpoints of the molecular networks involved in insulin resistance. Ample evidence suggests that the heat shock machinery regulates critical pathways in mitochondrial function and energy metabolism and that cellular energy status highly influences it. Weakening of HSPs, therefore, leads to loss of their vital cytoprotective functions, propagating nutri-stress in the system. Further research into the mechanistic roles of HSPs in metabolic homeostasis will help widen our understanding of lifestyle diseases, their onset, and complications. These inducible proteins may be crucial to attenuating lifestyle risk factors and disease management.
Collapse
Affiliation(s)
- Jayashree Kuppuswami
- Department of Biochemistry, Jawaharlal Institute of Post-Graduate Medical Education and Research (JIPMER), Puducherry, 605006 India
| | | |
Collapse
|
20
|
Johnson CN, McCoin CS, Kueck PJ, Hawley AG, John CS, Thyfault JP, Swerdlow RH, Geiger PC, Morris JK. Relationship of Muscle Apolipoprotein E Expression with Markers of Cellular Stress, Metabolism, and Blood Biomarkers in Cognitively Healthy and Impaired Older Adults. J Alzheimers Dis 2023; 92:1027-1035. [PMID: 36847010 PMCID: PMC10116140 DOI: 10.3233/jad-221192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Individuals with mild cognitive impairment (MCI) have reduced lipid-stimulated mitochondrial respiration in skeletal muscle. A major risk factor for Alzheimer's disease (AD), the apolipoprotein E4 (APOE4) allele, is implicated in lipid metabolism and is associated with metabolic and oxidative stress that can result from dysfunctional mitochondria. Heat shock protein 72 (Hsp72) is protective against these stressors and is elevated in the AD brain. OBJECTIVE Our goal was to characterize skeletal muscle ApoE and Hsp72 protein expression in APOE4 carriers in relationship to cognitive status, muscle mitochondrial respiration and AD biomarkers. METHODS We analyzed previously collected skeletal muscle tissue from 24 APOE4 carriers (60y+) who were cognitively healthy (CH, n = 9) or MCI (n = 15). We measured ApoE and Hsp72 protein levels in muscle and phosphorylated tau181 (pTau181) levels in plasma, and leveraged previously collected data on APOE genotype, mitochondrial respiration during lipid oxidation, and VO2 max. RESULTS Muscle ApoE (p = 0.013) and plasma pTau181 levels (p < 0.001) were higher in MCI APOE4 carriers. Muscle ApoE positively correlated with plasma pTau181 in all APOE4 carriers (R2 = 0.338, p = 0.003). Hsp72 expression negatively correlated with ADP (R2 = 0.775, p = <0.001) and succinate-stimulated respiration (R2 = 0.405, p = 0.003) in skeletal muscle of MCI APOE4 carriers. Plasma pTau181 negatively tracked with VO2 max in all APOE4 carriers (R2 = 0.389, p = 0.003). Analyses were controlled for age. CONCLUSION This work supports a relationship between cellular stress in skeletal muscle and cognitive status in APOE4 carriers.
Collapse
Affiliation(s)
- Chelsea N. Johnson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Colin S. McCoin
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Diabetes Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paul J. Kueck
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Amelia G. Hawley
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Casey S. John
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - John P. Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Diabetes Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H. Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paige C. Geiger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Diabetes Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jill K. Morris
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas University Diabetes Institute, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
21
|
Li G, Yin W, Yang Y, Yang H, Chen Y, Liang Y, Zhang W, Xie T. Bibliometric Insights of Global Research Landscape in Mitophagy. Front Mol Biosci 2022; 9:851966. [PMID: 35923469 PMCID: PMC9340163 DOI: 10.3389/fmolb.2022.851966] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Autophagy is a highly regulated and evolutionarily conserved process in eukaryotes which is responsible for protein and organelle degradation. Although this process was described over 60 years ago, the selective autophagy of mitochondria (mitophagy) was recently coined in 2005. Research on the topic of mitophagy has made rapid progress in the past decade, which proposed to play critical roles in human health and disease. This study aimed to visualize the scientific outputs and research trends of mitophagy.Methods: Articles and reviews related to the topic of mitophagy were retrieved from the Web of Science Core Collection on 30 November 2021. Two kinds of software (CiteSpace and VOSviewer) were used to perform a visualized analysis of countries/regions, institutions, authors, journals, references, and keywords.Results: From 2005 to 2021, total 5844 publications on mitophagy were identified for final analysis. The annual number of publications grew yearly over the past 17 years. United States (N = 2025) and Chinese Academy of Sciences is the leading country and institute (N = 112) ranked by the number of publications, respectively. The most productive author was Jun Ren (N = 38) and Derek P. Narendra obtained the most co-cited times (2693 times). The journals with the highest output and the highest co-citation frequency were Autophagy (N = 208) and Journal of Biological Chemistry (co-citation: 17226), respectively. Analyses of references and keywords suggested that “mechanism of mitochondrial quality control”, “molecule and signaling pathway in mitophagy”, and “mitophagy related diseases” were research hotspots, and parkin-mediated mitophagy and its roles in skeletal muscle and inflammation-related diseases may be the frontiers of future research.Conclusion: Although mitophagy research has flourished and attracted attention from all over the world, the regional imbalance in the development of mitophagy research was observed. Our results provided a comprehensive global research landscape of mitophagy from 2005– 2021 from a perspective of bibliometrics, which may serve as a reference for future mitophagy studies.
Collapse
Affiliation(s)
- Guoli Li
- Department of Nephrology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- Changsha Clinical Research Center for Kidney Disease, Changsha, China
- Hunan Clinical Research Center for Chronic Kidney Disease, Changsha, China
| | - Wei Yin
- Department of Nephrology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- Changsha Clinical Research Center for Kidney Disease, Changsha, China
- Hunan Clinical Research Center for Chronic Kidney Disease, Changsha, China
| | - Yiya Yang
- Department of Nephrology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- Changsha Clinical Research Center for Kidney Disease, Changsha, China
- Hunan Clinical Research Center for Chronic Kidney Disease, Changsha, China
| | - Hongyu Yang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Yinyin Chen
- Department of Nephrology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- Changsha Clinical Research Center for Kidney Disease, Changsha, China
- Hunan Clinical Research Center for Chronic Kidney Disease, Changsha, China
| | - Yumei Liang
- Department of Nephrology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- Changsha Clinical Research Center for Kidney Disease, Changsha, China
- Hunan Clinical Research Center for Chronic Kidney Disease, Changsha, China
| | - Weiru Zhang
- Department of General Medicine, Xiangya Hospital, Central South University, Changsha, China
- International Collaborative Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
| | - Tingting Xie
- Department of General Medicine, Xiangya Hospital, Central South University, Changsha, China
- International Collaborative Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
- *Correspondence: Tingting Xie,
| |
Collapse
|
22
|
Heat shock proteins in adaptation to physical activity. UKRAINIAN BIOCHEMICAL JOURNAL 2022. [DOI: 10.15407/ubj94.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The review article presents the author’s model of one of the blocks of the integrated adaptation mechanism to physical activity and the accompanying moderate heat effects. The participation of heat shock proteins in the stabilization of the tertiary structure and in the restoration of the function of proteins damaged by temperature and physical stressors but performing catalytic, transport, reception or protective role and being involved in the processes of contraction- relaxation and muscle and bone tissue remodeling is discussed.
Collapse
|
23
|
Liang H, Zhang F, Wang W, Zhao W, Zhou J, Feng Y, Wu J, Li M, Bai X, Zeng Z, Niu J, Miao Y. Heat Shock Transcription Factor 2 Promotes Mitophagy of Intestinal Epithelial Cells Through PARL/PINK1/Parkin Pathway in Ulcerative Colitis. Front Pharmacol 2022; 13:893426. [PMID: 35860016 PMCID: PMC9289131 DOI: 10.3389/fphar.2022.893426] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/16/2022] [Indexed: 12/12/2022] Open
Abstract
The overactivation of NLRP3 inflammasome in intestinal epithelial cells (IECs) is among the important reasons for severe inflammation in ulcerative colitis (UC). We found that heat shock transcription factor 2 (HSF2), which is highly expressed in UC, could inhibit the activation of NLRP3 inflammasome and reduce IL-1β in IECs, but the mechanisms were still not clear. It has been reported that HSP72 regulated by HSF2 can enhance the mitophagy mediated by Parkin. The number of damaged mitochondria and the mitochondrial derived ROS (mtROS) can be reduced by mitophagy, which means the activity of NLRP3 inflammasome is inhibited. Therefore, we speculate that HSF2 might regulate the activation of NLRP3 inflammasome of IECs in UC through the mitophagy mediated by Parkin. This study proves that the number of damaged mitochondria in IECs, the level of mitophagy, and the level of ROS in intestinal mucosa are positively correlated with the severity of UC. In mice and cells, mitophagy was promoted by HSF2 through the PARL/PINK1/Parkin pathway. This study reveals the potential mechanisms of HSF2 decreasing mtROS of IECs in UC.
Collapse
Affiliation(s)
- Hao Liang
- Kunming Medical University, Kunming, China
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Fengrui Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Wen Wang
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Wei Zhao
- Kunming Medical University, Kunming, China
| | - Jiao Zhou
- Kunming Medical University, Kunming, China
| | - Yuran Feng
- Department of Ultrasound, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jing Wu
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Maojuan Li
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Xinyu Bai
- Kunming Medical University, Kunming, China
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Zhong Zeng
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Junkun Niu
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
- *Correspondence: Junkun Niu, ; Yinglei Miao,
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
- *Correspondence: Junkun Niu, ; Yinglei Miao,
| |
Collapse
|
24
|
Johnson CN, Jensen RS, Von Schulze AT, Geiger PC. Heat Therapy Can Improve Hepatic Mitochondrial Function and Glucose Control. Exerc Sport Sci Rev 2022; 50:162-170. [PMID: 35394967 DOI: 10.1249/jes.0000000000000296] [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: 11/21/2022]
Abstract
This review proposes the novel hypothesis that heat can be used as an alternative therapy to exercise to improve hepatic mitochondrial function and glucose regulation in patients with nonalcoholic fatty liver disease. Although exercise has proven benefits in treating nonalcoholic fatty liver disease, barriers to exercise in the majority of patients necessitate an alternative method of treatment.
Collapse
Affiliation(s)
- Chelsea N Johnson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Reilly S Jensen
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | | | - Paige C Geiger
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
25
|
Von Schulze AT, Geiger PC. Heat and Mitochondrial Bioenergetics. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
26
|
Wu R, Cao S, Li F, Feng S, Shu G, Wang L, Gao P, Zhu X, Zhu C, Wang S, Jiang Q. RNA-binding protein YBX1 promotes brown adipogenesis and thermogenesis via PINK1/PRKN-mediated mitophagy. FASEB J 2022; 36:e22219. [PMID: 35195911 DOI: 10.1096/fj.202101810rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/11/2022]
Abstract
Promoting the thermogenic function of brown adipose tissue (BAT) is a promising strategy to combat obesity and metabolic disorders. While much is known about the transcriptional regulation of BAT activation, however, the underlying mechanism of post-transcriptional control by RNA binding proteins remains largely unknown. Here, we found that RNA binding protein Y-box binding protein 1 (YBX1) expression was abundant in BAT and induced by cold exposure and a β-adrenergic agonist in mice. Loss-of-function experiments showed that YBX1 deficiency inhibited mouse primary brown adipocyte differentiation and thermogenic function. Further study showed that YBX1 positively regulates thermogenesis through enhancing mitophagy. Mechanistically, RNA immunoprecipitation identified that YBX1 directly targeted the transcripts of PTEN-induced kinase 1 (Pink1) and parkin RBR E3 ubiquitin-protein ligase (Prkn), two key regulators of mitophagy. RNA decay assay proved that loss of YBX1 decreased mRNA stability of Pink1 and Prkn, leading to reduced protein expression, thereby alleviating mitophagy and inhibiting thermogenic program. Importantly, in vivo experiments demonstrated that YBX1 overexpression in BAT promoted thermogenesis and mitophagy in mice. Collectively, our results reveal novel insight into the molecular mechanism of YBX1 in post-transcriptional regulation of PINK1/PRKN-mediated mitophagy and highlight the critical role of YBX1 in brown adipogenesis and thermogenesis.
Collapse
Affiliation(s)
- Ruifan Wu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shuting Cao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Fan Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shengchun Feng
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lina Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ping Gao
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaotong Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Canjun Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Songbo Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
27
|
Manfredi LH. Overheating or overcooling: heat transfer in the spot to fight against the pandemic obesity. Rev Endocr Metab Disord 2021; 22:665-680. [PMID: 33000381 DOI: 10.1007/s11154-020-09596-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 12/25/2022]
Abstract
The prevalence of obesity has nearly doubled worldwide over the past three and a half decades, reaching pandemic status. Obesity is associated with decreased life expectancy and with an increased risk of metabolic, cardiovascular, nervous system diseases. Hence, understanding the mechanisms involved in the onset and development of obesity is mandatory to promote planned health actions to revert this scenario. In this review, common aspects of cold exposure, a process of heat generation, and exercise, a process of heat dissipation, will be discussed as two opposite mechanisms of obesity, which can be oversimplified as caloric conservation. A common road between heat generation and dissipation is the mobilization of Free Faty Acids (FFA) and Carbohydrates (CHO). An increase in energy expenditure (immediate effect) and molecular/metabolic adaptations (chronic effect) are responses that depend on SNS activity in both conditions of heat transfer. This cycle of using and removing FFA and CHO from blood either for heat or force generation disrupt the key concept of obesity: energy accumulation. Despite efforts in making the anti-obesity pill, maybe it is time to consider that the world's population is living at thermoneutrality since temperature-controlled places and the lack of exercise are favoring caloric accumulation.
Collapse
Affiliation(s)
- Leandro Henrique Manfredi
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, Santa Catarina, Brazil.
| |
Collapse
|
28
|
Guerrero‐Castillo S, van Strien J, Brandt U, Arnold S. Ablation of mitochondrial DNA results in widespread remodeling of the mitochondrial complexome. EMBO J 2021; 40:e108648. [PMID: 34542926 PMCID: PMC8561636 DOI: 10.15252/embj.2021108648] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
So-called ρ0 cells lack mitochondrial DNA and are therefore incapable of aerobic ATP synthesis. How cells adapt to survive ablation of oxidative phosphorylation remains poorly understood. Complexome profiling analysis of ρ0 cells covered 1,002 mitochondrial proteins and revealed changes in abundance and organization of numerous multiprotein complexes including previously not described assemblies. Beyond multiple subassemblies of complexes that would normally contain components encoded by mitochondrial DNA, we observed widespread reorganization of the complexome. This included distinct changes in the expression pattern of adenine nucleotide carrier isoforms, other mitochondrial transporters, and components of the protein import machinery. Remarkably, ablation of mitochondrial DNA hardly affected the complexes organizing cristae junctions indicating that the altered cristae morphology in ρ0 mitochondria predominantly resulted from the loss of complex V dimers required to impose narrow curvatures to the inner membrane. Our data provide a comprehensive resource for in-depth analysis of remodeling of the mitochondrial complexome in response to respiratory deficiency.
Collapse
Affiliation(s)
- Sergio Guerrero‐Castillo
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
- University Children's Research@Kinder‐UKEUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Joeri van Strien
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Center for Molecular and Biomolecular InformaticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Ulrich Brandt
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| | - Susanne Arnold
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| |
Collapse
|
29
|
Rodriguez YA, Kaur S, Nolte E, Zheng Z, Blagg BSJ, Dobrowsky RT. Novologue Therapy Requires Heat Shock Protein 70 and Thioredoxin-Interacting Protein to Improve Mitochondrial Bioenergetics and Decrease Mitophagy in Diabetic Sensory Neurons. ACS Chem Neurosci 2021; 12:3049-3059. [PMID: 34340312 PMCID: PMC8456717 DOI: 10.1021/acschemneuro.1c00340] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a complication of diabetes whose pathophysiology is linked to altered mitochondrial bioenergetics (mtBE). KU-596 is a small molecule neurotherapeutic that reverses symptoms of DPN, improves sensory neuron mtBE, and decreases the pro-oxidant protein, thioredoxin-interacting protein (Txnip) in a heat shock protein 70 (Hsp70)-dependent manner. However, the mechanism by which KU-596 improves mtBE and the role of Txnip in drug efficacy remains unknown. Mitophagy is a quality-control mechanism that selectively targets damaged mitochondria for degradation. The goal of this study was to determine if KU-596 therapy improved DPN, mtBE, and mitophagy in an Hsp70- and Txnip-dependent manner. Mito-QC (MQC) mice express a mitochondrially targeted mCherry-GFP fusion protein that enables visualizing mitophagy. Diabetic MQC, MQC × Hsp70 knockout (KO), and MQC × Txnip KO mice developed sensory and nerve conduction dysfunctions consistent with the onset of DPN. KU-596 therapy improved these measures, and this was dependent on Hsp70 but not Txnip. In MQC mice, diabetes decreased mtBE and increased mitophagy and KU-596 treatment reversed these effects. In contrast, KU-596 was unable to improve mtBE and decrease mitophagy in MQC × Hsp70 and MQC × Txnip KO mice. These data suggest that Txnip is not necessary for the development of the sensory symptoms and mitochondrial dysfunction induced by diabetes. KU-596 therapy may improve mitochondrial tolerance to diabetic stress to decrease mitophagic clearance in an Hsp70- and Txnip-dependent manner.
Collapse
Affiliation(s)
- Yssa A Rodriguez
- Department of Pharmacology and Toxicology, University of Kansas, 5064 Malott Hall/1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Sukmanjit Kaur
- Department of Pharmacology and Toxicology, University of Kansas, 5064 Malott Hall/1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Erika Nolte
- Department of Pharmacology and Toxicology, University of Kansas, 5064 Malott Hall/1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Zhang Zheng
- Department of Chemistry and Biochemistry University of Notre Dame, 305 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry University of Notre Dame, 305 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Rick T Dobrowsky
- Department of Pharmacology and Toxicology, University of Kansas, 5064 Malott Hall/1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| |
Collapse
|
30
|
Zhu Y, Yang H, Deng J, Fan D. Ginsenoside Rg5 Improves Insulin Resistance and Mitochondrial Biogenesis of Liver via Regulation of the Sirt1/PGC-1α Signaling Pathway in db/db Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:8428-8439. [PMID: 34309383 DOI: 10.1021/acs.jafc.1c02476] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a common metabolic syndrome that decreases insulin sensitivity and mitochondrial biogenesis in the liver. Our previous study demonstrated that ginsenoside Rg5 (Rg5) could attenuate renal injury in diabetic mice but its underlying mechanism in mitochondrial biogenesis and insulin sensitivity remains poorly understood. In this study, we found that Rg5 intervention significantly inhibited blood glucose increases in db/db mice, improved liver function damage and hepatocyte apoptosis, and activated the IRS-1/phosphatidylinositol 3-kinase/AKT insulin metabolism signaling pathway. Rg5 treatment also increased the level of glycogen synthesis and activated sirtuin1 (Sirt1) to increase glucose uptake and insulin sensitivity in insulin-resistant HepG2 (IR-HepG2) cells. Rg5 intervention also effectively improved liver oxidative stress and inflammation in db/db mice and increased mitochondrial biogenesis caused by T2DM. Additionally, the Rg5 treatment increased the mitochondrial mass in IR-HepG2 cells and activated Sirt1 to regulate the Sirt1/PGC-1α/mitofusin-2 mitochondrial biosynthesis pathway. Our findings demonstrated that Rg5 enhanced liver mitochondrial biogenesis and insulin sensitivity in db/db mice by activating the Sirt1/PGC-1α signaling pathway, suggesting the potential of Rg5 as a natural product for T2DM interventions.
Collapse
Affiliation(s)
- Yanyan Zhu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotechnology & Biomedical Research Institute, Northwest University, 229 North Taibai Road, Xi'an 710069, China
| | - Haixia Yang
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Jianjun Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotechnology & Biomedical Research Institute, Northwest University, 229 North Taibai Road, Xi'an 710069, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotechnology & Biomedical Research Institute, Northwest University, 229 North Taibai Road, Xi'an 710069, China
| |
Collapse
|
31
|
Costa JSR, Fonseca GFAC, Ottone NCDS, Silva PA, Antonaccio RF, Silva G, Rocha MDSA, Coimbra CC, Esteves EA, Mang ZA, Amorim FT, Magalhães FDC. Strength training improves insulin resistance and differently affects mitochondria in skeletal muscle and visceral adipose tissue in high-fat fed mice. Life Sci 2021; 278:119639. [PMID: 34043987 DOI: 10.1016/j.lfs.2021.119639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
AIMS Strength training (ST) improves insulin resistance and glucose tolerance by yet unknown mechanisms. The aims of this study were to investigate the effects of ST on mitochondrial adaptation in skeletal muscle and adipose tissue, on heat shock protein 72 (Hsp72) in skeletal muscle, and on visceral adipocyte size in mice with high-fat diet (HFD)-induced insulin resistance. MATERIALS AND METHODS Male Balb/c mice were divided into sedentary control-chow (C-chow), strength trained-chow (ST-chow), sedentary control-HFD (C-HFD) and strength trained-HFD (ST-HFD). Diet was provided for 12 weeks, while ladder climbing ST was performed for the final six weeks of the study at a frequency of three days per week. KEY FINDINGS Strength training led to increased strength, muscular endurance, and skeletal muscle hypertrophy. Compared to the C-HFD group, mice in the ST-HFD group decreased their whole-body insulin resistance, improved their glucose tolerance, and had higher activation of the insulin pathway in skeletal muscle. ST increased citrate synthase (CS) activity in skeletal muscle, but this increase was blunted in ST-HFD. Conversely, HFD reduced adipose tissue CS activity regardless of training status. Hsp72 content was reduced in C-HFD, but returned to control levels in ST-HFD. Finally, reduced epididymal adipocyte size was observed in ST-HFD. SIGNIFICANCE These results suggest that the improvement in insulin resistance induced by ST is related to mitochondrial adaptation in skeletal muscle, but not in adipose tissue. Moreover, this improvement might be related to increased skeletal muscle Hsp72 and reduced epididymal adipocyte size.
Collapse
Affiliation(s)
- Juliana Sales Rodrigues Costa
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Graciene Fernandes Araújo Campos Fonseca
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Natielle Cecília Dos Santos Ottone
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Patrick Almeida Silva
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Romulo Fernandes Antonaccio
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Gabriela Silva
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Maíra da Silva Almeida Rocha
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Candido Celso Coimbra
- Endocrinology Laboratory, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Elizabethe Adriana Esteves
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Zachary A Mang
- Department of Health, Exercise, and Sport Science, University of New Mexico, Albuquerque, NM 87131, United States of America
| | - Fabiano Trigueiro Amorim
- Department of Health, Exercise, and Sport Science, University of New Mexico, Albuquerque, NM 87131, United States of America
| | - Flávio de Castro Magalhães
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Faculdade de Ciências Básicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil.
| |
Collapse
|
32
|
Diane A, Abunada H, Khattab N, Moin ASM, Butler AE, Dehbi M. Role of the DNAJ/HSP40 family in the pathogenesis of insulin resistance and type 2 diabetes. Ageing Res Rev 2021; 67:101313. [PMID: 33676026 DOI: 10.1016/j.arr.2021.101313] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/22/2021] [Accepted: 02/28/2021] [Indexed: 12/13/2022]
Abstract
Insulin resistance (IR) underpins a wide range of metabolic disorders including type 2 diabetes (T2D), metabolic syndrome and cardiovascular diseases. IR is characterized by a marked reduction in the magnitude and/or delayed onset of insulin to stimulate glucose disposal. This condition is due to defects in one or several intracellular intermediates of the insulin signaling cascade, ranging from insulin receptor substrate (IRS) inactivation to reduced glucose phosphorylation and oxidation. Genetic predisposition, as well as other precipitating factors such as aging, obesity, and sedentary lifestyles are among the risk factors underlying the pathogenesis of IR and its subsequent progression to T2D. One of the cardinal hallmarks of T2D is the impairment of the heat shock response (HSR). Human and animal studies provided compelling evidence of reduced expression of several components of the HSR (i.e. Heat shock proteins or HSPs) in diabetic samples in a manner that correlates with the degree of IR. Interventions that induce the HSR, irrespective of the means to achieve it, proved their effectiveness in enhancing insulin sensitivity and improving glycemic index. However, most of these studies have been focused on HSP70 family. In this review, we will focus on the novel role of DNAJ/HSP40 cochaperone family in metabolic diseases associated with IR.
Collapse
|
33
|
Triolo M, Hood DA. Manifestations of Age on Autophagy, Mitophagy and Lysosomes in Skeletal Muscle. Cells 2021; 10:cells10051054. [PMID: 33946883 PMCID: PMC8146406 DOI: 10.3390/cells10051054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/18/2023] Open
Abstract
Sarcopenia is the loss of both muscle mass and function with age. Although the molecular underpinnings of sarcopenia are not fully understood, numerous pathways are implicated, including autophagy, in which defective cargo is selectively identified and degraded at the lysosome. The specific tagging and degradation of mitochondria is termed mitophagy, a process important for the maintenance of an organelle pool that functions efficiently in energy production and with relatively low reactive oxygen species production. Emerging data, yet insufficient, have implicated various steps in this pathway as potential contributors to the aging muscle atrophy phenotype. Included in this is the lysosome, the end-stage organelle possessing a host of proteolytic and degradative enzymes, and a function devoted to the hydrolysis and breakdown of defective molecular complexes and organelles. This review provides a summary of our current understanding of how the autophagy-lysosome system is regulated in aging muscle, highlighting specific areas where knowledge gaps exist. Characterization of the autophagy pathway with a particular focus on the lysosome will undoubtedly pave the way for the development of novel therapeutic strategies to combat age-related muscle loss.
Collapse
Affiliation(s)
- Matthew Triolo
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| | - David A. Hood
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada;
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
- Correspondence: ; Tel.: +(416)-736-2100 (ext. 66640)
| |
Collapse
|
34
|
Zhu L, Luo X, Fu N, Chen L. Mitochondrial unfolded protein response: A novel pathway in metabolism and immunity. Pharmacol Res 2021; 168:105603. [PMID: 33838292 DOI: 10.1016/j.phrs.2021.105603] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/03/2021] [Accepted: 04/04/2021] [Indexed: 12/11/2022]
Abstract
Mitochondrial unfolded protein response (mitoUPR) is a mitochondria stress response to maintain mitochondrial proteostasis during stress. Increasing evidence suggests that mitoUPR participates in diverse physiological processes especially metabolism and immunity. Although mitoUPR regulates metabolism in many aspects, it is mainly reflected in the regulation of energy metabolism. During stress, mitoUPR alters energy metabolism via suppressing oxidative phosphorylation (OXPHOS) or increasing glycolysis. MitoUPR also alters energy metabolism and regulates diverse metabolic diseases such as diabetes, cancers, fatty liver and obesity. In addition, mitoUPR also participates in immune process during stress. MitoUPR can induce innate immune response during various infections and may regulate inflammatory response during diverse inflammations. Considering the pleiotropic actions of mitoUPR, mitoUPR may supply diverse therapeutic targets for metabolic diseases and immune diseases.
Collapse
Affiliation(s)
- Li Zhu
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Xuling Luo
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Nian Fu
- Department of Gastroenterology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
35
|
Von Schulze AT, Deng F, Fuller KNZ, Franczak E, Miller J, Allen J, McCoin CS, Shankar K, Ding WX, Thyfault JP, Geiger PC. Heat Treatment Improves Hepatic Mitochondrial Respiratory Efficiency via Mitochondrial Remodeling. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab001. [PMID: 33629069 PMCID: PMC7886620 DOI: 10.1093/function/zqab001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023]
Abstract
Nonacholic fatty liver disease, or hepatic steatosis, is the most common liver disorder affecting the western world and currently has no pharmacologic cure. Thus, many investigations have focused on alternative strategies to treat or prevent hepatic steatosis. Our laboratory has shown that chronic heat treatment (HT) mitigates glucose intolerance, insulin resistance, and hepatic steatosis in rodent models of obesity. Here, we investigate the direct bioenergetic mechanism(s) surrounding the metabolic effects of HT on hepatic mitochondria. Utilizing mitochondrial proteomics and respiratory function assays, we show that one bout of acute HT (42°C for 20 min) in male C57Bl/6J mice (n = 6/group) triggers a hepatic mitochondrial heat shock response resulting in acute reductions in respiratory capacity, degradation of key mitochondrial enzymes, and induction of mitophagy via mitochondrial ubiquitination. We also show that chronic bouts of HT and recurrent activation of the heat shock response enhances mitochondrial quality and respiratory function via compensatory adaptations in mitochondrial organization, gene expression, and transport even during 4 weeks of high-fat feeding (n = 6/group). Finally, utilizing a liver-specific heat shock protein 72 (HSP72) knockout model, we are the first to show that HSP72, a protein putatively driving the HT metabolic response, does not play a significant role in the hepatic mitochondrial adaptation to acute or chronic HT. However, HSP72 is required for the reductions in blood glucose observed with chronic HT. Our data are the first to suggest that chronic HT (1) improves hepatic mitochondrial respiratory efficiency via mitochondrial remodeling and (2) reduces blood glucose in a hepatic HSP72-dependent manner.
Collapse
Affiliation(s)
- Alex T Von Schulze
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fengyan Deng
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kelly N Z Fuller
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Edziu Franczak
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Josh Miller
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Julie Allen
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Colin S McCoin
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kartik Shankar
- Pediatrics, Section of Nutrition, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wen-Xing Ding
- Pharmacology, Toxicology & Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - John P Thyfault
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Paige C Geiger
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA,Address correspondence to P.C.G. (e-mail: )
| |
Collapse
|
36
|
Piñeros AR, Gao H, Wu W, Liu Y, Tersey SA, Mirmira RG. Single-Cell Transcriptional Profiling of Mouse Islets Following Short-Term Obesogenic Dietary Intervention. Metabolites 2020; 10:metabo10120513. [PMID: 33353164 PMCID: PMC7765825 DOI: 10.3390/metabo10120513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/22/2022] Open
Abstract
Obesity is closely associated with adipose tissue inflammation and insulin resistance. Dysglycemia and type 2 diabetes results when islet β cells fail to maintain appropriate insulin secretion in the face of insulin resistance. To clarify the early transcriptional events leading to β-cell failure in the setting of obesity, we fed male C57BL/6J mice an obesogenic, high-fat diet (60% kcal from fat) or a control diet (10% kcal from fat) for one week, and islets from these mice (from four high-fat- and three control-fed mice) were subjected to single-cell RNA sequencing (sc-RNAseq) analysis. Islet endocrine cell types (α cells, β cells, δ cells, PP cells) and other resident cell types (macrophages, T cells) were annotated by transcript profiles and visualized using Uniform Manifold Approximation and Projection for Dimension Reduction (UMAP) plots. UMAP analysis revealed distinct cell clusters (11 for β cells, 5 for α cells, 3 for δ cells, PP cells, ductal cells, endothelial cells), emphasizing the heterogeneity of cell populations in the islet. Collectively, the clusters containing the majority of β cells showed the fewest gene expression changes, whereas clusters harboring the minority of β cells showed the most changes. We identified that distinct β-cell clusters downregulate genes associated with the endoplasmic reticulum stress response and upregulate genes associated with insulin secretion, whereas others upregulate genes that impair insulin secretion, cell proliferation, and cell survival. Moreover, all β-cell clusters negatively regulate genes associated with immune response activation. Glucagon-producing α cells exhibited patterns similar to β cells but, again, in clusters containing the minority of α cells. Our data indicate that an early transcriptional response in islets to an obesogenic diet reflects an attempt by distinct populations of β cells to augment or impair cellular function and/or reduce inflammatory responses as possible harbingers of ensuing insulin resistance.
Collapse
Affiliation(s)
- Annie R. Piñeros
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.R.P.); (W.W.)
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.L.)
| | - Wenting Wu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.R.P.); (W.W.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.L.)
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.L.)
| | - Sarah A. Tersey
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Raghavendra G. Mirmira
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
- Correspondence:
| |
Collapse
|
37
|
Uda M, Yoshihara T, Ichinoseki-Sekine N, Baba T, Yoshioka T. Potential roles of neuronal nitric oxide synthase and the PTEN-induced kinase 1 (PINK1)/Parkin pathway for mitochondrial protein degradation in disuse-induced soleus muscle atrophy in adult rats. PLoS One 2020; 15:e0243660. [PMID: 33296434 PMCID: PMC7725317 DOI: 10.1371/journal.pone.0243660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Excessive nitric oxide (NO) production and mitochondrial dysfunction can activate protein degradation in disuse-induced skeletal muscle atrophy. However, the increase in NO production in atrophied muscles remains controversial. In addition, although several studies have investigated the PTEN-induced kinase 1 (PINK1)/Parkin pathway, a mitophagy pathway, in atrophied muscle, the involvement of this pathway in soleus muscle atrophy is unclear. In this study, we investigated the involvement of neuronal nitric oxide synthase (nNOS) and the PINK1/Parkin pathway in soleus muscle atrophy induced by 14 days of hindlimb unloading (HU) in adult rats. HU lowered the weight of the soleus muscles. nNOS expression showed an increase in atrophied soleus muscles. Although HU increased malondialdehyde as oxidative modification of the protein, it decreased 6-nitrotryptophan, a marker of protein nitration. Additionally, the nitrosocysteine content and S-nitrosylated Parkin were not altered, suggesting the absence of excessive nitrosative stress after HU. The expression of PINK1 and Parkin was also unchanged, whereas the expression of heat shock protein 70 (HSP70), which is required for Parkin activity, was reduced in atrophied soleus muscles. Moreover, we observed accumulation and reduced ubiquitination of high molecular weight mitofusin 2, which is a target of Parkin, in atrophied soleus muscles. These results indicate that excessive NO is not produced in atrophied soleus muscles despite nNOS accumulation, suggesting that excessive NO dose not mediate in soleus muscle atrophy at least after 14 days of HU. Furthermore, the PINK1/Parkin pathway may not play a role in mitophagy at this time point. In contrast, the activity of Parkin may be downregulated because of reduced HSP70 expression, which may contribute to attenuated degradation of target proteins in the atrophied soleus muscles after 14 days of HU. The present study provides new insights into the roles of nNOS and a protein degradation pathway in soleus muscle atrophy.
Collapse
Affiliation(s)
- Munehiro Uda
- School of Nursing, Hirosaki Gakuin University, Hirosaki, Aomori, Japan
- * E-mail: ,
| | - Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba, Japan
| | - Noriko Ichinoseki-Sekine
- Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba, Japan
- Faculty of Liberal Arts, The Open University of Japan, Chiba, Japan
| | - Takeshi Baba
- School of Medicine, Juntendo University, Inzai, Chiba, Japan
| | | |
Collapse
|
38
|
Moore TM, Zhou Z, Strumwasser AR, Cohn W, Lin AJ, Cory K, Whitney K, Ho T, Ho T, Lee JL, Rucker DH, Hoang AN, Widjaja K, Abrishami AD, Charugundla S, Stiles L, Whitelegge JP, Turcotte LP, Wanagat J, Hevener AL. Age-induced mitochondrial DNA point mutations are inadequate to alter metabolic homeostasis in response to nutrient challenge. Aging Cell 2020; 19:e13166. [PMID: 33049094 PMCID: PMC7681042 DOI: 10.1111/acel.13166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 04/10/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction is frequently associated with impairment in metabolic homeostasis and insulin action, and is thought to underlie cellular aging. However, it is unclear whether mitochondrial dysfunction is a cause or consequence of insulin resistance in humans. To determine the impact of intrinsic mitochondrial dysfunction on metabolism and insulin action, we performed comprehensive metabolic phenotyping of the polymerase gamma (PolG) D257A "mutator" mouse, a model known to accumulate supraphysiological mitochondrial DNA (mtDNA) point mutations. We utilized the heterozygous PolG mutator mouse (PolG+/mut ) because it accumulates mtDNA point mutations ~ 500-fold > wild-type mice (WT), but fails to develop an overt progeria phenotype, unlike PolGmut/mut animals. To determine whether mtDNA point mutations induce metabolic dysfunction, we examined male PolG+/mut mice at 6 and 12 months of age during normal chow feeding, after 24-hr starvation, and following high-fat diet (HFD) feeding. No marked differences were observed in glucose homeostasis, adiposity, protein/gene markers of metabolism, or oxygen consumption in muscle between WT and PolG+/mut mice during any of the conditions or ages studied. However, proteomic analyses performed on isolated mitochondria from 12-month-old PolG+/mut mouse muscle revealed alterations in the expression of mitochondrial ribosomal proteins, electron transport chain components, and oxidative stress-related factors compared with WT. These findings suggest that mtDNA point mutations at levels observed in mammalian aging are insufficient to disrupt metabolic homeostasis and insulin action in male mice.
Collapse
Affiliation(s)
- Timothy M. Moore
- Department of Biological SciencesDana & David Dornsife College of Letters, Arts, and SciencesUniversity of Southern CaliforniaLos AngelesCAUSA
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Zhenqi Zhou
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Alexander R. Strumwasser
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Whitaker Cohn
- Department of Psychiatry and Biobehavioral Sciences & The Semel Institute for Neuroscience and Human BehaviorUniversity of CaliforniaLos AngelesCAUSA
| | - Amanda J. Lin
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Kevin Cory
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Kate Whitney
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Theodore Ho
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Timothy Ho
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Joseph L. Lee
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Daniel H. Rucker
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Austin N. Hoang
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Kevin Widjaja
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Aaron D. Abrishami
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Sarada Charugundla
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Linsey Stiles
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Julian P. Whitelegge
- Department of Psychiatry and Biobehavioral Sciences & The Semel Institute for Neuroscience and Human BehaviorUniversity of CaliforniaLos AngelesCAUSA
| | - Lorraine P. Turcotte
- Department of Biological SciencesDana & David Dornsife College of Letters, Arts, and SciencesUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Jonathan Wanagat
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Andrea L. Hevener
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Iris Cantor‐UCLA Women's Health CenterUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
39
|
Von Schulze AT, Deng F, Morris JK, Geiger PC. Heat therapy: possible benefits for cognitive function and the aging brain. J Appl Physiol (1985) 2020; 129:1468-1476. [PMID: 32969779 DOI: 10.1152/japplphysiol.00168.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, yet there are no disease-modifying treatments available and there is no cure. It is becoming apparent that metabolic and vascular conditions such as type 2 diabetes (T2D) and hypertension promote the development and accumulation of Alzheimer's disease-related dementia pathologies. To this end, aerobic exercise, which is a common lifestyle intervention for both metabolic disease and hypertension, is shown to improve brain health during both healthy aging and dementia. However, noncompliance or other barriers to exercise response are common in exercise treatment paradigms. In addition, reduced intracellular proteostasis and mitochondrial function could contribute to the etiology of AD. Specifically, compromised chaperone systems [i.e., heat shock protein (HSP) systems] can contribute to protein aggregates (i.e., β-amyloid plaques and neurofibrillary tangles) and reduced mitochondrial quality control (i.e., mitophagy). Therefore, novel therapies that target whole body metabolism, the vasculature, and chaperone systems (like HSPs) are needed to effectively treat AD. This review focuses on the role of heat therapy in the treatment and prevention of AD. Heat therapy has been independently shown to reduce whole body insulin resistance, improve vascular function, activate interorgan cross talk via endocytic vesicles, and activate HSPs to improve mitochondrial function and proteostasis in a variety of tissues. Thus, heat therapy could offer immense clinical benefit to patients suffering from AD. Importantly, future studies in patients are needed to determine the safety and efficacy of heat therapy in preventing AD.
Collapse
Affiliation(s)
- Alex T Von Schulze
- Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Fengyan Deng
- Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Jill K Morris
- Department of Neurology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Paige C Geiger
- Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
40
|
Wang Y, Liu Y, Chen E, Pan Z. The role of mitochondrial dysfunction in mesenchymal stem cell senescence. Cell Tissue Res 2020; 382:457-462. [DOI: 10.1007/s00441-020-03272-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
|
41
|
Pallubinsky H, Phielix E, Dautzenberg B, Schaart G, Connell NJ, Wit‐Verheggen V, Havekes B, Baak MA, Schrauwen P, Marken Lichtenbelt WD. Passive exposure to heat improves glucose metabolism in overweight humans. Acta Physiol (Oxf) 2020; 229:e13488. [PMID: 32359193 PMCID: PMC7379279 DOI: 10.1111/apha.13488] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
AIM Heat exposure has been indicated to positively affect glucose metabolism. An involvement of heat shock protein 72 (HSP72) in the enhancement of insulin sensitivity upon heat exposure has been previously suggested. Here, we performed an intervention study exploring the effect of passive heat acclimation (PHA) on glucose metabolism and intracellular (a) HSP72 concentrations in overweight humans. METHODS Eleven non-diabetic overweight (BMI 27-35 kg/m2 ) participants underwent 10 consecutive days of PHA (4-6 h/day, 34.4 ± 0.2°C, 22.8 ± 2.7%RH). Before and after PHA, whole-body insulin sensitivity was assessed using a one-step hyperinsulinaemic-euglycaemic clamp, skeletal muscle biopsies were taken to measure intracellular iHSP72, energy expenditure and substrate oxidation were measured using indirect calorimetry and blood samples were drawn to assess markers of metabolic health. Thermophysiological adaptations were measured during a temperature ramp protocol before and after PHA. RESULTS Despite a lack of change in iHSP72, 10 days of PHA reduced basal (9.7 ± 1.4 pre- vs 8.4 ± 2.1 μmol · kg-1 · min-1 post-PHA, P = .038) and insulin-stimulated (2.1 ± 0.9 pre- vs 1.5 ± 0.8 μmol · kg-1 · min-1 post-PHA, P = .005) endogenous glucose production (EGP) and increased insulin suppression of EGP (78.5 ± 9.7% pre- vs 83.0 ± 7.9% post-PHA, P = .028). Consistently, fasting plasma glucose (6.0 ± 0.5 pre- vs 5.8 ± 0.4 mmol/L post-PHA, P = .013) and insulin concentrations (97 ± 55 pre- vs 84 ± 49 pmol/L post-PHA, P = .026) decreased significantly. Moreover, fat oxidation increased, and free fatty acids as well as cholesterol concentrations and mean arterial pressure decreased after PHA. CONCLUSION Our results show that PHA for 10 days improves glucose metabolism and enhances fat metabolism, without changes in iHSP72. Further exploration of the therapeutic role of heat in cardio-metabolic disorders should be considered.
Collapse
Affiliation(s)
- Hannah Pallubinsky
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Bas Dautzenberg
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Gert Schaart
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Niels J. Connell
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Vera Wit‐Verheggen
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Bas Havekes
- Department of Internal Medicine Division of Endocrinology Maastricht University Medical Centre+ Maastricht the Netherlands
| | - Marleen A. Baak
- Department of Human Biology NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| | - Wouter D. Marken Lichtenbelt
- Department of Nutrition and Movement Sciences NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Maastricht the Netherlands
| |
Collapse
|
42
|
Thakur SS, Swiderski K, Chhen VL, James JL, Cranna NJ, Islam AMT, Ryall JG, Lynch GS. HSP70 drives myoblast fusion during C2C12 myogenic differentiation. Biol Open 2020; 9:bio053918. [PMID: 32605905 PMCID: PMC7390621 DOI: 10.1242/bio.053918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/17/2020] [Indexed: 11/30/2022] Open
Abstract
In response to injury, skeletal muscle stem cells (MuSCs) undergo myogenesis where they become activated, proliferate rapidly, differentiate and undergo fusion to form multinucleated myotubes. Dramatic changes in cell size, shape, metabolism and motility occur during myogenesis, which cause cellular stress and alter proteostasis. The molecular chaperone heat shock protein 70 (HSP70) maintains proteostasis by regulating protein biosynthesis and folding, facilitating transport of polypeptides across intracellular membranes and preventing stress-induced protein unfolding/aggregation. Although HSP70 overexpression can exert beneficial effects in skeletal muscle diseases and enhance skeletal muscle repair after injury, its effect on myogenesis has not been investigated. Plasmid-mediated overexpression of HSP70 did not affect the rate of C2C12 proliferation or differentiation, but the median number of myonuclei per myotube and median myotube width in differentiated C2C12 myotubes were increased with HSP70 overexpression. These findings reveal that increased HSP70 expression can promote myoblast fusion, identifying a mechanism for its therapeutic potential to enhance muscle repair after injury.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Savant S Thakur
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| | - Kristy Swiderski
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| | - Victoria L Chhen
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| | - Janine L James
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| | - Nicki J Cranna
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| | - A M Taufiqual Islam
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| | - James G Ryall
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Physiology, University of Melbourne, Victoria, Australia 3010
| |
Collapse
|
43
|
Edmunds LR, Xie B, Mills AM, Huckestein BR, Undamatla R, Murali A, Pangburn MM, Martin J, Sipula I, Kaufman BA, Scott I, Jurczak MJ. Liver-specific Prkn knockout mice are more susceptible to diet-induced hepatic steatosis and insulin resistance. Mol Metab 2020; 41:101051. [PMID: 32653576 PMCID: PMC7399260 DOI: 10.1016/j.molmet.2020.101051] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 12/20/2022] Open
Abstract
Objective PARKIN is an E3 ubiquitin ligase that regulates mitochondrial quality control through a process called mitophagy. Recent human and rodent studies suggest that loss of hepatic mitophagy may occur during the pathogenesis of obesity-associated fatty liver and contribute to changes in mitochondrial metabolism associated with this disease. Whole-body Prkn knockout mice are paradoxically protected against diet-induced hepatic steatosis; however, liver-specific effects of Prkn deficiency cannot be discerned in this model due to pleotropic effects of germline Prkn deletion on energy balance and subsequent protection against diet-induced obesity. We therefore generated the first liver-specific Prkn knockout mouse strain (LKO) to directly address the role of hepatic Prkn. Methods Littermate control (WT) and LKO mice were fed regular chow (RC) or high-fat diet (HFD) and changes in body weight and composition were measured over time. Liver mitochondrial content was assessed using multiple, complementary techniques, and mitochondrial respiratory capacity was assessed using Oroboros O2K platform. Liver fat was measured biochemically and assessed histologically, while global changes in hepatic gene expression were measured by RNA-seq. Whole-body and tissue-specific insulin resistance were assessed by hyperinsulinemic-euglycemic clamp with isotopic tracers. Results Liver-specific deletion of Prkn had no effect on body weight or adiposity during RC or HFD feeding; however, hepatic steatosis was increased by 45% in HFD-fed LKO compared with WT mice (P < 0.05). While there were no differences in mitochondrial content between genotypes on either diet, mitochondrial respiratory capacity and efficiency in the liver were significantly reduced in LKO mice. Gene enrichment analyses from liver RNA-seq results suggested significant changes in pathways related to lipid metabolism and fibrosis in HFD-fed Prkn knockout mice. Finally, whole-body insulin sensitivity was reduced by 35% in HFD-fed LKO mice (P < 0.05), which was primarily due to increased hepatic insulin resistance (60% of whole-body effect; P = 0.11). Conclusions These data demonstrate that PARKIN contributes to mitochondrial homeostasis in the liver and plays a protective role against the pathogenesis of hepatic steatosis and insulin resistance. Mitochondrial respiratory capacity is reduced in liver-specific Prkn knockout mice. Liver-specific Prkn knockout mice develop more severe steatosis during high-fat diet feeding. Pathogenesis of NAFLD, including insulin resistance and markers of fibrosis, is enhanced in liver-specific Prkn knockout mice.
Collapse
Affiliation(s)
- Lia R Edmunds
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bingxian Xie
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amanda M Mills
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brydie R Huckestein
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ramya Undamatla
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anjana Murali
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martha M Pangburn
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Martin
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ian Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brett A Kaufman
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Iain Scott
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Moore TM, Lin AJ, Strumwasser AR, Cory K, Whitney K, Ho T, Ho T, Lee JL, Rucker DH, Nguyen CQ, Yackly A, Mahata SK, Wanagat J, Stiles L, Turcotte LP, Crosbie RH, Zhou Z. Mitochondrial Dysfunction Is an Early Consequence of Partial or Complete Dystrophin Loss in mdx Mice. Front Physiol 2020; 11:690. [PMID: 32636760 PMCID: PMC7317021 DOI: 10.3389/fphys.2020.00690] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by rapid wasting of skeletal muscle. Mitochondrial dysfunction is a well-known pathological feature of DMD. However, whether mitochondrial dysfunction occurs before muscle fiber damage in DMD pathology is not well known. Furthermore, the impact upon heterozygous female mdx carriers (mdx/+), who display dystrophin mosaicism, has received little attention. We hypothesized that dystrophin deletion leads to mitochondrial dysfunction, and that this may occur before myofiber necrosis. As a secondary complication to mitochondrial dysfunction, we also hypothesized metabolic abnormalities prior to the onset of muscle damage. In this study, we detected aberrant mitochondrial morphology, reduced cristae number, and large mitochondrial vacuoles from both male and female mdx mice prior to the onset of muscle damage. Furthermore, we systematically characterized mitochondria during disease progression starting before the onset of muscle damage, noting additional changes in mitochondrial DNA copy number and regulators of mitochondrial size. We further detected mild metabolic and mitochondrial impairments in female mdx carrier mice that were exacerbated with high-fat diet feeding. Lastly, inhibition of the strong autophagic program observed in adolescent mdx male mice via administration of the autophagy inhibitor leupeptin did not improve skeletal muscle pathology. These results are in line with previous data and suggest that before the onset of myofiber necrosis, mitochondrial and metabolic abnormalities are present within the mdx mouse.
Collapse
Affiliation(s)
- Timothy M. Moore
- Department of Biological Sciences, Dana & David Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Amanda J. Lin
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alexander R. Strumwasser
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Kevin Cory
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Kate Whitney
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Theodore Ho
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Timothy Ho
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Joseph L. Lee
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel H. Rucker
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christina Q. Nguyen
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aidan Yackly
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sushil K. Mahata
- VA San Diego Healthcare System, San Diego, CA, United States
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jonathan Wanagat
- Division of Geriatrics, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Linsey Stiles
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lorraine P. Turcotte
- Department of Biological Sciences, Dana & David Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Rachelle H. Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Zhenqi Zhou
- Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
45
|
Lakshmi PK, Kumar S, Pawar S, Kuriakose BB, Sudheesh MS, Pawar RS. Targeting metabolic syndrome with phytochemicals: Focus on the role of molecular chaperones and hormesis in drug discovery. Pharmacol Res 2020; 159:104925. [PMID: 32492491 DOI: 10.1016/j.phrs.2020.104925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 05/10/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022]
Abstract
Adaptive cellular stress response confers stress tolerance against inflammatory and metabolic disorders. In response to metabolic stress, the key mediator of cellular adaptation and tolerance is a class of molecules called the molecular chaperones (MCs). MCs are highly conserved molecules that play critical role in maintaining protein stability and functionality. Hormesis in this context is a unique adaptation mechanism where a low dose of a stressor (which is toxic at high dose) confers a stress-resistant adaptive cellular phenotype. Hormesis can be observed at different level of biological organization at various measurable endpoints. The MCs are believed to play a key role in adaptation during hormesis. Several phytochemicals are known for their hormetic response and are called phytochemical hormetins. The role of phytochemical-mediated hormesis on the adaptive cellular processes is proposed as a potential therapeutic approach to target inflammation associated with metabolic syndrome. However, the screening of phytochemical hormetins would require a paradigm shift in the methods currently used in drug discovery.
Collapse
Affiliation(s)
- P K Lakshmi
- Pharmacognosy and Phytochemistry Laboratory, Faculty of Pharmacy, VNS Group of Institutions, VNS Campus, Vidya Vihar, Neelbad-462044, Bhopal, MP, India
| | - Shweta Kumar
- Pharmacognosy and Phytochemistry Laboratory, Faculty of Pharmacy, VNS Group of Institutions, VNS Campus, Vidya Vihar, Neelbad-462044, Bhopal, MP, India
| | - Sulakshhna Pawar
- Ravi Shankar College of Pharmacy, Bypass Road, Bhanpur Square, Bhopal, MP 462010, India
| | - Beena Briget Kuriakose
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Khamis, Mushayt, Saudi Arabia
| | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Rajesh Singh Pawar
- Truba Institute of Pharmacy, Karond-Gandhi Nagar, By Pass Road, Bhopal, 462038, India.
| |
Collapse
|
46
|
Mulyani WRW, Sanjiwani MID, Sandra, Prabawa IPY, Lestari AAW, Wihandani DM, Suastika K, Saraswati MR, Bhargah A, Manuaba IBAP. Chaperone-Based Therapeutic Target Innovation: Heat Shock Protein 70 (HSP70) for Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2020; 13:559-568. [PMID: 32161482 PMCID: PMC7051252 DOI: 10.2147/dmso.s232133] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/06/2019] [Indexed: 12/24/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is still a global health problem. Current T2DM treatments are limited to curing the symptoms and have not been able to restore insulin sensitivity in insulin-sensitive tissues that have become resistant. In the past decade, some studies have shown the significant role of a chaperone family, heat shock protein 70 (HSP70), in insulin resistance pathogenesis that leads to T2DM. HSP70 is a cytoprotective molecular chaperone that functions in protein folding and degradation. In general, studies have shown that decreased concentration of HSP70 is able to induce inflammation process through JNK activation, inhibit fatty acid oxidation by mitochondria through mitophagy decrease and mitochondrial biogenesis, as well as activate SREBP-1c, one of the lipogenic gene transcription factors in ER stress. The overall molecular pathways are potentially leading to insulin resistance and T2DM. Increased expression of HSP70 in brain tissues is able to improve insulin sensitivity and glycemic control specifically. HSP70 modulation-targeting strategies (including long-term physical exercise, hot tub therapy (HTT), and administration of alfalfa-derived HSP70 (aHSP70)) in subjects with insulin resistance are proven to have therapeutic and preventive potency that are promising in T2DM management.
Collapse
Affiliation(s)
| | | | - Sandra
- Faculty of Medicine, Universitas Udayana, Bali, Indonesia
| | - I Putu Yuda Prabawa
- Department of Clinical Pathology, Faculty of Medicine, Universitas Udayana, Sanglah General Hospital, Bali, Indonesia
| | - Anak Agung Wiradewi Lestari
- Department of Clinical Pathology, Faculty of Medicine, Universitas Udayana, Sanglah General Hospital, Bali, Indonesia
| | - Desak Made Wihandani
- Department of Biochemistry, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
| | - Ketut Suastika
- Department of Internal Medicine, Faculty of Medicine, Universitas Udayana, Sanglah General Hospital, Bali, Indonesia
| | - Made Ratna Saraswati
- Department of Internal Medicine, Faculty of Medicine, Universitas Udayana, Sanglah General Hospital, Bali, Indonesia
| | - Agha Bhargah
- Faculty of Medicine, Universitas Udayana, Bali, Indonesia.,Cardiology Department, Faculty of Medicine, Universitas Udayana-Sanglah General Hospital, Bali, Indonesia
| | - Ida Bagus Amertha Putra Manuaba
- International Ph.D Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Medical and Health Education Unit, Faculty of Medicine, Universitas Udayana, Bali, Indonesia
| |
Collapse
|
47
|
Calcium Fluxes in Work-Related Muscle Disorder: Implications from a Rat Model. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5040818. [PMID: 31662979 PMCID: PMC6791278 DOI: 10.1155/2019/5040818] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/13/2019] [Accepted: 08/20/2019] [Indexed: 01/16/2023]
Abstract
Introduction Ca2+ regulatory excitation-contraction coupling properties are key topics of interest in the development of work-related muscle myalgia and may constitute an underlying cause of muscle pain and loss of force generating capacity. Method A well-established rat model of high repetition high force (HRHF) work was used to investigate if such exposure leads to an increase in cytosolic Ca2+ concentration ([Ca2+]i) and changes in sarcoplasmic reticulum (SR) vesicle Ca2+ uptake and release rates. Result Six weeks exposure of rats to HRHF increased indicators of fatigue, pain behaviors, and [Ca2+]i, the latter implied by around 50-100% increases in pCam, as well as in the Ca2+ handling proteins RyR1 and Casq1 accompanied by an ∼10% increased SR Ca2+ uptake rate in extensor and flexor muscles compared to those of control rats. This demonstrated a work-related altered myocellular Ca2+ regulation, SR Ca2+ handling, and SR protein expression. Discussion These disturbances may mirror intracellular changes in early stages of human work-related myalgic muscle. Increased uptake of Ca2+ into the SR may reflect an early adaptation to avoid a sustained detrimental increase in [Ca2+]i similar to the previous findings of deteriorated Ca2+ regulation and impaired function in fatigued human muscle.
Collapse
|
48
|
Wu H, Wang Y, Li W, Chen H, Du L, Liu D, Wang X, Xu T, Liu L, Chen Q. Deficiency of mitophagy receptor FUNDC1 impairs mitochondrial quality and aggravates dietary-induced obesity and metabolic syndrome. Autophagy 2019; 15:1882-1898. [PMID: 30898010 DOI: 10.1080/15548627.2019.1596482] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
There is overwhelming evidence for an association between impaired mitochondrial function and metabolic syndrome. Mitophagy, a process that selectively removes damaged mitochondria via a specialized form of autophagy, is essential for mitochondrial quality control (mitochondrial QC) and metabolic homeostasis. We thus addressed the potential role of defective mitophagy in the pathogenesis of metabolic disorders. Mice lacking Fundc1, a newly characterized mitophagy receptor, develop more severe obesity and insulin resistance when fed a high-fat diet (HFD). Ablation of Fundc1 results in defective mitophagy and impaired mitochondrial QC in vitro and in white adipose tissue (WAT). In addition, there is more pronounced WAT remodeling with more adipose tissue-associated macrophages infiltration, more M1 macrophage polarization and thus an elevated inflammatory response. Mechanistically, hyperactivation of MAPK/JNK leads to insulin insensitivity, which can be inhibited by knocking out Mapk8/Jnk1 in fundc1 KO mice. Our results demonstrate that dysregulated mitochondrial QC due to defective mitophagy receptor FUNDC1 links with metabolic disorders via MAPK signaling and inflammatory responses. Abbreviations: ATMs: adipose tissue macrophages; BAT: brown adipose tissue; BMDMs: bone marrow-derived macrophages; GOT1/AST: glutamic-oxaloacetic transaminase 1, soluble; GPT/ALT: glutamic pyruvic transaminase, soluble; H&E staining: hematoxylin and eosin staining; HFD: high-fat diet; LIR: LC3-interacting region; mitochondrial QC: mitochondrial quality control; mito-ROS: mitochondrial ROS; mtDNA: mitochondrial DNA; RT-PCR: real-time-PCR; T2D: type 2 diabetes; WAT: white adipose tissue.
Collapse
Affiliation(s)
- Hao Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - You Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing , China
| | - Wenhui Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,College of Life Sciences, University of Chinese Academy of Sciences , Beijing , China
| | - Hui Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Lei Du
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Dong Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,College of Life Sciences, University of Chinese Academy of Sciences , Beijing , China
| | - Xiaohui Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Tao Xu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing , China
| | - Lei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Quan Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,College of Life Sciences, University of Chinese Academy of Sciences , Beijing , China.,Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University , Tianjin , China
| |
Collapse
|
49
|
Moore TM, Zhou Z, Cohn W, Norheim F, Lin AJ, Kalajian N, Strumwasser AR, Cory K, Whitney K, Ho T, Ho T, Lee JL, Rucker DH, Shirihai O, van der Bliek AM, Whitelegge JP, Seldin MM, Lusis AJ, Lee S, Drevon CA, Mahata SK, Turcotte LP, Hevener AL. The impact of exercise on mitochondrial dynamics and the role of Drp1 in exercise performance and training adaptations in skeletal muscle. Mol Metab 2019; 21:51-67. [PMID: 30591411 PMCID: PMC6407367 DOI: 10.1016/j.molmet.2018.11.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Mitochondria are organelles primarily responsible for energy production, and recent evidence indicates that alterations in size, shape, location, and quantity occur in response to fluctuations in energy supply and demand. We tested the impact of acute and chronic exercise on mitochondrial dynamics signaling and determined the impact of the mitochondrial fission regulator Dynamin related protein (Drp)1 on exercise performance and muscle adaptations to training. METHODS Wildtype and muscle-specific Drp1 heterozygote (mDrp1+/-) mice, as well as dysglycemic (DG) and healthy normoglycemic men (control) performed acute and chronic exercise. The Hybrid Mouse Diversity Panel, including 100 murine strains of recombinant inbred mice, was used to identify muscle Dnm1L (encodes Drp1)-gene relationships. RESULTS Endurance exercise impacted all aspects of the mitochondrial life cycle, i.e. fission-fusion, biogenesis, and mitophagy. Dnm1L gene expression and Drp1Ser616 phosphorylation were markedly increased by acute exercise and declined to baseline during post-exercise recovery. Dnm1L expression was strongly associated with transcripts known to regulate mitochondrial metabolism and adaptations to exercise. Exercise increased the expression of DNM1L in skeletal muscle of healthy control and DG subjects, despite a 15% ↓(P = 0.01) in muscle DNM1L expression in DG at baseline. To interrogate the role of Dnm1L further, we exercise trained male mDrp1+/- mice and found that Drp1 deficiency reduced muscle endurance and running performance, and altered muscle adaptations in response to exercise training. CONCLUSION Our findings highlight the importance of mitochondrial dynamics, specifically Drp1 signaling, in the regulation of exercise performance and adaptations to endurance exercise training.
Collapse
Affiliation(s)
- Timothy M Moore
- Department of Biological Sciences, Dana & David Dornsife College of Letters, Arts, and Sciences, University of Southern California, CA 90089-0372, USA; David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Zhenqi Zhou
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Whitaker Cohn
- David Geffen School of Medicine, Department of Psychiatry and Biobehavioral Sciences, The Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Frode Norheim
- David Geffen School of Medicine, Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Amanda J Lin
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Nareg Kalajian
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Alexander R Strumwasser
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Kevin Cory
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Kate Whitney
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Theodore Ho
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Timothy Ho
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Joseph L Lee
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Daniel H Rucker
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Orian Shirihai
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Alexander M van der Bliek
- David Geffen School of Medicine, Department of Biological Chemistry, University of California, Los Angeles, CA 90095, USA
| | - Julian P Whitelegge
- David Geffen School of Medicine, Department of Psychiatry and Biobehavioral Sciences, The Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Marcus M Seldin
- David Geffen School of Medicine, Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Aldons J Lusis
- David Geffen School of Medicine, Human Genetics, University of California, Los Angeles, CA 90095, USA; David Geffen School of Medicine, Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Sindre Lee
- University Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christian A Drevon
- University Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sushil K Mahata
- VA San Diego Healthcare System, San Diego, CA 92161, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lorraine P Turcotte
- Department of Biological Sciences, Dana & David Dornsife College of Letters, Arts, and Sciences, University of Southern California, CA 90089-0372, USA
| | - Andrea L Hevener
- David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA; Iris Cantor-UCLA Women's Health Research Center, Los Angeles, CA 90095, USA.
| |
Collapse
|
50
|
Parker BL, Calkin AC, Seldin MM, Keating MF, Tarling EJ, Yang P, Moody SC, Liu Y, Zerenturk EJ, Needham EJ, Miller ML, Clifford BL, Morand P, Watt MJ, Meex RCR, Peng KY, Lee R, Jayawardana K, Pan C, Mellett NA, Weir JM, Lazarus R, Lusis AJ, Meikle PJ, James DE, de Aguiar Vallim TQ, Drew BG. An integrative systems genetic analysis of mammalian lipid metabolism. Nature 2019; 567:187-193. [PMID: 30814737 PMCID: PMC6656374 DOI: 10.1038/s41586-019-0984-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 01/23/2019] [Indexed: 12/16/2022]
Abstract
Dysregulation of lipid homeostasis is a precipitating event in the pathogenesis and progression of hepatosteatosis and metabolic syndrome. These conditions are highly prevalent in developed societies and currently have limited options for diagnostic and therapeutic intervention. Here, using a proteomic and lipidomic-wide systems genetic approach, we interrogated lipid regulatory networks in 107 genetically distinct mouse strains to reveal key insights into the control and network structure of mammalian lipid metabolism. These include the identification of plasma lipid signatures that predict pathological lipid abundance in the liver of mice and humans, defining subcellular localization and functionality of lipid-related proteins, and revealing functional protein and genetic variants that are predicted to modulate lipid abundance. Trans-omic analyses using these datasets facilitated the identification and validation of PSMD9 as a previously unknown lipid regulatory protein. Collectively, our study serves as a rich resource for probing mammalian lipid metabolism and provides opportunities for the discovery of therapeutic agents and biomarkers in the setting of hepatic lipotoxicity.
Collapse
Affiliation(s)
- Benjamin L Parker
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Anna C Calkin
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia.
- Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia.
| | - Marcus M Seldin
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Michael F Keating
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Elizabeth J Tarling
- Department of Medicine, Division of Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Pengyi Yang
- Charles Perkins Centre, School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia
| | - Sarah C Moody
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Yingying Liu
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Eser J Zerenturk
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Elise J Needham
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Matthew L Miller
- Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Bethan L Clifford
- Department of Medicine, Division of Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Pauline Morand
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Matthew J Watt
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Ruth C R Meex
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kang-Yu Peng
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Kaushala Jayawardana
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Calvin Pan
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Ross Lazarus
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Aldons J Lusis
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - David E James
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Brian G Drew
- Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia.
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia.
| |
Collapse
|