1
|
Boshnakovska A, Pronto JR, Gall T, Aich A, Prochazka J, Nichtova Z, Sedlacek R, Sobitov I, Ainatzi S, Lenz C, Katschinski DM, Urlaub H, Voigt N, Rehling P, Kremer LS. SMIM20 promotes complex IV biogenesis and Ca 2+ signaling in mice heart. Cell Rep 2025; 44:115723. [PMID: 40402744 DOI: 10.1016/j.celrep.2025.115723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/18/2025] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
Mitochondria are key to cellular energetics, metabolism, and signaling. Their dysfunction is linked to devastating diseases, including mitochondrial disorders, diabetes, neurodegenerative diseases, cardiac disorders, and cancer. Here, we present a knockout mouse model lacking the complex IV assembly factor SMIM20/MITRAC7. SMIM20-/- mice display cardiac pathology with reduced heart weight and cardiac output. Heart mitochondria present with reduced levels of complex IV associated with increased complex I activity, have altered fatty acid oxidation, and display elevated levels of ROS production. Interestingly, mutant mouse ventricular myocytes show unphysiological Ca2+ handling, which can be attributed to the increase in mitochondrial ROS production. Our study presents an example of a tissue-specific phenotype in the context of OXPHOS dysfunction. Moreover, our data suggest a link between complex IV dysfunction and Ca2+ handling at the endoplasmic reticulum through ROS signaling.
Collapse
Affiliation(s)
- Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany; German Center for Child and Adolescent Health (DZKJ), 37075 Göttingen, Germany
| | - Julius Ryan Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, 37075 Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Tanja Gall
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Abhishek Aich
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the CAS, 142 20 Prague, Czech Republic
| | - Zuzana Nichtova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the CAS, 142 20 Prague, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the CAS, 142 20 Prague, Czech Republic
| | - Izzatullo Sobitov
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, 37075 Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Sofia Ainatzi
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Christof Lenz
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany; Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Dörthe M Katschinski
- Department of Cardiovascular Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany; Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, 37075 Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany; German Center for Child and Adolescent Health (DZKJ), 37075 Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany; Max Planck Institute for Multidisciplinary Science, 37077 Göttingen, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology, Translational Neuroinflammation and Automated Microscopy, 37075 Göttingen, Germany.
| | - Laura S Kremer
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany.
| |
Collapse
|
2
|
Carvalho G, Nguyen TVH, Repolês B, Forslund JME, Wijethunga WMRR, Ranjbarian F, Mendes IC, Gorospe CM, Chaudhari N, Falabella M, Doimo M, Wanrooij S, Pitceathly RDS, Hofer A, Wanrooij PH. Activating AMPK improves pathological phenotypes due to mtDNA depletion. FEBS J 2025; 292:2359-2380. [PMID: 39918244 PMCID: PMC12062783 DOI: 10.1111/febs.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 05/11/2025]
Abstract
AMP-activated protein kinase (AMPK) is a master regulator of cellular energy homeostasis that also plays a role in preserving mitochondrial function and integrity. Upon a disturbance in the cellular energy state that increases AMP levels, AMPK activity promotes a switch from anabolic to catabolic metabolism to restore energy homeostasis. However, the level of severity of mitochondrial dysfunction required to trigger AMPK activation is currently unclear, as is whether stimulation of AMPK using specific agonists can improve the cellular phenotype following mitochondrial dysfunction. Using a cellular model of mitochondrial disease characterized by progressive mitochondrial DNA (mtDNA) depletion and deteriorating mitochondrial metabolism, we show that mitochondria-associated AMPK becomes activated early in the course of the advancing mitochondrial dysfunction, before any quantifiable decrease in the ATP/(AMP + ADP) ratio or respiratory chain activity. Moreover, stimulation of AMPK activity using the specific small-molecule agonist A-769662 alleviated the mitochondrial phenotypes caused by the mtDNA depletion and restored normal mitochondrial membrane potential. Notably, the agonist treatment was able to partially restore mtDNA levels in cells with severe mtDNA depletion, while it had no impact on mtDNA levels of control cells. The beneficial impact of the agonist on mitochondrial membrane potential was also observed in cells from patients suffering from mtDNA depletion. These findings improve our understanding of the effects of specific small-molecule activators of AMPK on mitochondrial and cellular function and suggest a potential application for these compounds in disease states involving mtDNA depletion.
Collapse
Affiliation(s)
- Gustavo Carvalho
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Tran V. H. Nguyen
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Bruno Repolês
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | | | | | | | - Isabela C. Mendes
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | | | - Namrata Chaudhari
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Micol Falabella
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUK
| | - Mara Doimo
- Clinical Genetics Unit, Department of Women and Children's HealthPadua UniversityPaduaItaly
| | - Sjoerd Wanrooij
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Robert D. S. Pitceathly
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular DiseasesThe National Hospital for Neurology and NeurosurgeryLondonUK
| | - Anders Hofer
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | | |
Collapse
|
3
|
D’Egidio F, Qosja E, Ammannito F, Topi S, d’Angelo M, Cimini A, Castelli V. Antioxidant and Anti-Inflammatory Defenses in Huntington's Disease: Roles of NRF2 and PGC-1α, and Therapeutic Strategies. Life (Basel) 2025; 15:577. [PMID: 40283130 PMCID: PMC12028459 DOI: 10.3390/life15040577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/21/2025] [Accepted: 03/29/2025] [Indexed: 04/29/2025] Open
Abstract
Huntington's disease (HD) is a detrimental neurodegenerative disease caused by the expansion of a CAG triplet in the HTT gene. This mutation leads to the production of mutant Huntingtin (Htt) protein with toxic gain-of-function. The mHtt is responsible in several ways for the establishment of an intricate pathogenetic scenario in affected cells, particularly in HD neurons. Among the features of HD, oxidative stress plays a relevant role in the progression of the disease at the cellular level. Mitochondrial dysfunction, bioenergetic deficits, Reactive Oxygen Species (ROS) production, neuroinflammation, and general reduction of antioxidant levels are all involved in the promotion of a toxic oxidative environment, eventually causing cell death. Nonetheless, neuronal cells exert antioxidant molecules to build up defense mechanisms. Key components of these defensive mechanisms are the nuclear factor erythroid 2-related factor 2 (NRF2) and peroxisome proliferator-activated receptor gamma coactivator-1 α (PGC-1α). Thus, this review aims to describe the involvement of oxidative stress in HD by exploring the roles of NRF2 and PGC-1α, crucial actors in this play. Finally, antioxidant therapeutic strategies targeting such markers are discussed.
Collapse
Affiliation(s)
- Francesco D’Egidio
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (F.A.)
| | - Elvira Qosja
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania; (E.Q.); (S.T.)
| | - Fabrizio Ammannito
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (F.A.)
| | - Skender Topi
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania; (E.Q.); (S.T.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (F.A.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (F.A.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (F.A.)
| |
Collapse
|
4
|
Liang L, Kuang X, He Y, Zhu L, Lau P, Li X, Luo D, Gong L, Zhou W, Zhang F, Liang X, Li Z, Hu B, Liu D, Ding T, Li H, Zhao S, Su J, Hung MC, Liu J, Liu H, Chen X. Alterations in PD-L1 succinylation shape anti-tumor immune responses in melanoma. Nat Genet 2025; 57:680-693. [PMID: 40069506 PMCID: PMC11906371 DOI: 10.1038/s41588-025-02077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/06/2025] [Indexed: 03/15/2025]
Abstract
Tumors undergo metabolic reprogramming to meet the energetic, synthetic and redox demands essential for malignancy, often characterized by increased glycolysis and lactate production. However, the role of mitochondrial metabolism in tumor immunity remains unclear. The present study integrates spatial transcriptomics, bulk transcriptomics and proteomics, revealing a strong link between the metabolite succinyl-CoA and tumor immunity as well as the efficacy of anti-programmed cell death protein-1 (PD-1) therapy in patients with melanoma. Elevated succinyl-CoA levels, through α-ketoglutarate or succinate supplementation, enhanced T cell-mediated tumor elimination, both in vitro and in vivo. Mechanistically, succinylation of the ligand of PD-1 (PD-L1) at lysine 129 led to its degradation. Increased carnitine palmitoyltransferase 1A (CPT1A), identified as a succinyltransferase for PD-L1, boosted anti-tumor activity. Preclinically, bezafibrate, a hyperlipidemia drug, upregulated CPT1A and synergized with CTLA-4 monoclonal antibody to inhibit tumor growth. Clinically, higher PD-L1 and lower CPT1A levels in tumors correlated with better anti-PD-1 therapy responses, suggesting potential biomarkers for prediction of treatment efficacy.
Collapse
Affiliation(s)
- Long Liang
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Medical Genetics & School of Life Sciences, Central South University, Changsha, China
| | - Xinwei Kuang
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Yi He
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Lin Zhu
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Poyee Lau
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Xin Li
- Medical Genetics & School of Life Sciences, Central South University, Changsha, China
| | - Dingan Luo
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Lan Gong
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Wenbin Zhou
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Fanglin Zhang
- Medical Genetics & School of Life Sciences, Central South University, Changsha, China
| | - Xiaowei Liang
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Zhuofeng Li
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Bin Hu
- Medical Genetics & School of Life Sciences, Central South University, Changsha, China
| | - Dandan Liu
- Medical Genetics & School of Life Sciences, Central South University, Changsha, China
| | - Tao Ding
- Department of Statistical Science, University College London, London, UK
| | - Hui Li
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Jing Liu
- Medical Genetics & School of Life Sciences, Central South University, Changsha, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital & School of Life Sciences & Furong Laboratory, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.
| |
Collapse
|
5
|
Mohamed Yusoff AA, Mohd Khair SZN, Abd Radzak SM. Mitochondrial DNA copy number alterations: Key players in the complexity of glioblastoma (Review). Mol Med Rep 2025; 31:78. [PMID: 39886971 PMCID: PMC11795256 DOI: 10.3892/mmr.2025.13443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025] Open
Abstract
Renowned as a highly invasive and lethal tumor derived from neural stem cells in the central nervous system, glioblastoma (GBM) exhibits substantial histopathological variation and genomic complexity, which drive its rapid progression and therapeutic resistance. Alterations in mitochondrial DNA (mtDNA) copy number (CN) serve a crucial role in GBM development and progression, affecting various aspects of tumor biology, including energy production, oxidative stress regulation and cellular adaptability. Fluctuations in mtDNA levels, whether elevated or diminished, can impair mitochondrial function, potentially disrupting oxidative phosphorylation and amplifying reactive oxygen species generation, thereby fueling tumor growth and influencing treatment responses. Understanding the mechanisms of mtDNA‑CN variations, and their interplay with genetic and environmental elements in the tumor microenvironment, is essential for advancing diagnostic and therapeutic strategies. Targeting mtDNA alterations could strengthen treatment efficacy, mitigate resistance and ultimately enhance the prognosis of patients with this aggressive brain tumor. The present review summarizes the existing literature on mtDNA alterations, specifically emphasizing variations in mtDNA‑CN and their association with GBM by surveying articles published between 1996 and 2024, sourced from databases such as Scopus, PubMed and Google Scholar. In addition, the review provides a brief overview of mitochondrial genome architecture, knowledge regarding the regulation of mtDNA integrity and CN, and how mitochondria significantly impact GBM tumorigenesis. This review further presents information on therapeutic approaches for restoring mtDNA‑CN that contribute to optimized mitochondrial function and improved health outcomes.
Collapse
Affiliation(s)
- Abdul Aziz Mohamed Yusoff
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | | | - Siti Muslihah Abd Radzak
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| |
Collapse
|
6
|
Yang T, Lu Z, Song H, Chen Y, Jiang M, Zhan K, Zhao G. Knockout of hexokinase 2 regulates mitochondrial dysfunction and activates the NLRP3 signal pathway in the rumen epithelial cells of dairy cows. Int J Biol Macromol 2025; 289:138831. [PMID: 39701238 DOI: 10.1016/j.ijbiomac.2024.138831] [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: 07/26/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Hexokinase 2 (HK2) plays a vital role in mitochondrial homeostasis; however, the molecular mechanisms underlying its involvement in high-concentrate diet-induced damage in the ruminal epithelium of dairy cows are poorly understood. This study aimed to explore the regulatory role of HK2 in mitochondrial function and responses to inflammation in the rumen of dairy cows fed a high-concentrate diet. Our results showed that, compared with a low-concentrate (LC) diet, feeding a high-concentrate (HC) diet increased oxidative stress and reduced relative antioxidant gene expression levels and enzyme activities in the ruminal epithelium. Furthermore, the expression of genes related to mitochondrial biosynthesis and structure decreased in the HC group, concomitant with nuclear oligomerization domain (NOD)-like receptor 3 (NLRP3) signaling pathway activation, which compromised normal rumen epithelium function. Meanwhile, transcription results showed the same trend in HK2-knockout bovine rumen epithelial cells (HK2KO BRECs) related to wild-type (WT) BRECs. Notably, the knockout of HK2 aggravated mitochondrial dysfunction, resulting in the impairment of mitochondrial morphology and quality, a reduction in mitochondrial membrane potential (MMP), mitochondrial permeability transition pore (MPTP) opening, increased reactive oxygen species (ROS) generation, and decreased expression of antioxidant genes. These changes led to upregulating genes and proteins in the NLRP3 pathway and activating proinflammatory response. In addition, metabolomic results showed that knockout HK2 altered the glycerophospholipid metabolic pathway. This study provides new strategies for mitigating high-concentrate diet-induced injury in the ruminal epithelium of dairy cows.
Collapse
Affiliation(s)
- Tianyu Yang
- Laboratory Animal Center, Xuzhou Medical University, Xuzhou, China; Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhiqi Lu
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Ningxia Dairy Science and Innovation Center of Bright Farming Company Limited, Zhongwei, China
| | - Han Song
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yuhang Chen
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Maocheng Jiang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Kang Zhan
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Guoqi Zhao
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
7
|
Palzkill VR, Tan J, Moparthy D, Tice AL, Ferreira LF, Ryan TE. A 6-Minute Limb Function Assessment for Therapeutic Testing in Experimental Peripheral Artery Disease Models. JACC Basic Transl Sci 2025; 10:88-103. [PMID: 39906594 PMCID: PMC11788496 DOI: 10.1016/j.jacbts.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 02/06/2025]
Abstract
In this study, we present a novel 6-minute limb function test that allows for the congruent assessment of muscular performance and hemodynamics in preclinical models of peripheral artery disease. Using several experimental conditions, the results demonstrate the superior efficacy of the 6-minute limb function test to detect differences in the response to hindlimb ischemia across several interventions, including where traditional perfusion recovery, capillary density, and muscle strength measures were unable to detect interventional differences, thus allowing for more rigorous assessment of preclinical therapies before clinical translation.
Collapse
Affiliation(s)
- Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, Florida, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, Florida, USA
| | - Divyansha Moparthy
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, Florida, USA
| | - Abigail L. Tice
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, Florida, USA
| | - Leonardo F. Ferreira
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, Florida, USA
- Center for Exercise Science, The University of Florida, Gainesville, Florida, USA
- The Myology Institute, The University of Florida, Gainesville, Florida, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, Florida, USA
- Center for Exercise Science, The University of Florida, Gainesville, Florida, USA
- The Myology Institute, The University of Florida, Gainesville, Florida, USA
| |
Collapse
|
8
|
Jin H, Xue B, Chen X, Ma T, Ma Y, Zou H, Zhu J, Tong X, Song R, Meng W, Liu Z. Polystyrene microplastics induced spermatogenesis disorder via disrupting mitochondrial function through the regulation of the Sirt1-Pgc1α signaling pathway in male mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 364:125364. [PMID: 39577614 DOI: 10.1016/j.envpol.2024.125364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
Microplastics (MPs) have emerged as hazardous substances, eliciting widespread concern regarding their potential toxicity. Although our previous research has indicated that polystyrene MPs (PS-MPs) might cause male reproductive toxicity in mammals, their precise effects on sperm motility parameters and acrosomal development remain uncertain. Herein, the effects on sperm motility of PS-MPs at varied particle sizes (0.5 μm, 4 μm and 10 μm) and the underlying mechanisms were examined. The results revealed that PS-MPs caused a decrease in sperm motility, accompanied by abnormalities in the structure and function of the sperm acrosome. Meanwhile, PS-MPs triggered the elevation of intracellular reactive oxygen species levels and the abnormal expression of antioxidant enzymes (γH2AX, GPX4, Peroxiredoxin 5 and SDHB), indicating disruption of the sperm antioxidant system. Furthermore, we observed aberrant expression of key factors involved in mitochondrial fission/fusion (Drp1, Fis1, Mfn1, Mfn2) and biogenesis (Tfam, Nrf1, Pgc1α), potentially resulting in disrupted mitochondrial dynamics and biogenesis in mice testis and Sertoli cells exposed to PS-MPs. Additionally, PS-MPs induced mitochondrial dysfunction by regulating the Sirt1-Pgc1α signaling pathway. Our data provided novel insights into potential mechanisms underlying the spermatogenesis disorders triggered by PS-MPs.
Collapse
Affiliation(s)
- Haibo Jin
- School of Public Health, Yangzhou University, Yangzhou, Jiangsu, 225000, China; College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Bowen Xue
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Xuefang Chen
- School of Public Health, Yangzhou University, Yangzhou, Jiangsu, 225000, China; College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Disease, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Jiaqiao Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Xishuai Tong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Weiqian Meng
- College of Health Science, Rush University, Chicago, IL, 60612, USA.
| | - Zongping Liu
- School of Public Health, Yangzhou University, Yangzhou, Jiangsu, 225000, China; College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
9
|
Ye F, Wei C, Wu A. The potential mechanism of mitochondrial homeostasis in postoperative neurocognitive disorders: an in-depth review. Ann Med 2024; 56:2411012. [PMID: 39450938 PMCID: PMC11514427 DOI: 10.1080/07853890.2024.2411012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 10/26/2024] Open
Abstract
Postoperative neurocognitive disorders (PND) are the most common neurological disorders following surgery and anaesthesia before and within 12 months after surgery, with a high prevalence in the geriatric population. PND can severely deteriorate the quality of life of patients, especially among the elderly, mainly manifested as memory loss, attention, decline and language comprehension disorders, mostly in elderly patients, with an incidence as high as 31%. Previous studies have also raised the possibility of accelerated cognitive decline and underlying neuropathological processes associated with diseases that affect cognitive performance (e.g. Alzheimer's dementia) for reasons related to anaesthesia and surgery. Currently, most research on PND has focused on various molecular pathways, especially in the geriatric population. The various hypotheses that have been proposed regarding the mechanisms imply peripheral neuroinflammation, oxidative stress, mitochondrial homeostasis, synaptic function, autophagy disorder, blood-brain barrier dysfunction, the microbiota-gut-brain axis and lack of neurotrophic support. However, the underlying pathogenesis and molecular mechanisms of PND have not yet been uncovered. Recent research has focused on mitochondrial homeostasis. In this paper, we present a review of various studies to better understand and characterize the mechanisms of associated cognitive dysfunction. As the biochemical basis of PND becomes more clearly defined, future treatments based on mitochondrial homeostasis modulation can prove to be very promising.
Collapse
Affiliation(s)
- Fan Ye
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Ye D, Hao Z, Tang S, Velkov T, Dai C. Aflatoxin Exposure-Caused Male Reproductive Toxicity: Molecular Mechanisms, Detoxification, and Future Directions. Biomolecules 2024; 14:1460. [PMID: 39595635 PMCID: PMC11592228 DOI: 10.3390/biom14111460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Widespread endocrine disorders and infertility caused by environmental and food pollutants have drawn considerable global attention. Aflatoxins (AFTs), a prominent class of mycotoxins, are recognized as one of the key contributors to environmental and food contamination. Aflatoxin B1 (AFB1) is the most potent and toxic pollutant among them and is known to cause multiple toxic effects, including neuro-, nephro-, hepato-, immune-, and genotoxicity. Recently, concerns have been raised regarding AFB1-induced infertility in both animals and humans. Exposure to AFB1 can disrupt the structure and functionality of reproductive organs, leading to gametogenesis impairment in males, subsequently reducing fertility. The potential molecular mechanisms have been demonstrated to involve oxidative stress, cell cycle arrest, apoptosis, inflammatory responses, and autophagy. Furthermore, several signaling pathways, including nuclear factor erythroid 2-related factor 2; NOD-, LRR-, and pyrin domain-containing protein 3; nuclear factor kappa-B; p53; p21; phosphoinositide 3-kinase/protein kinase B; the mammalian target of rapamycin; adenosine 5'-monophosphate-activated protein kinase; and mitochondrial apoptotic pathways, are implicated in these processes. Various interventions, including the use of small molecules, Chinese herbal extracts, probiotic supplementation, and camel milk, have shown efficacy in ameliorating AFB1-induced male reproductive toxicity, by targeting these signaling pathways. This review provides a comprehensive summary of the harmful impacts of AFB1 exposure on male reproductive organs in mammals, highlighting the potential molecular mechanisms and protective agents.
Collapse
Affiliation(s)
- Dongyun Ye
- Department of Obstetrics and Gynecology, Ezhou Central Hospital, Hubei University of Science and Technology, Ezhou 436000, China
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Shusheng Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Tony Velkov
- Department of Pharmacology, Biodiscovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| |
Collapse
|
11
|
Zhang Z, Yu R, Shi Q, Wu ZJ, Li Q, Mu J, Chen B, Shi J, Ni R, Wu L, Li Q, Fu J, Li R, Sun X, Wang J, He L, Kuang Y, Sang Q, Wang L. COX15 deficiency causes oocyte ferroptosis. Proc Natl Acad Sci U S A 2024; 121:e2406174121. [PMID: 39471219 PMCID: PMC11551447 DOI: 10.1073/pnas.2406174121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024] Open
Abstract
Mitochondria play diverse roles in mammalian physiology. The architecture, activity, and physiological functions of mitochondria in oocytes are largely different from those in somatic cells, but the mitochondrial proteins related to oocyte quality and reproductive longevity remain largely unknown. Here, using whole-exome sequencing data from 1,024 women (characterized by oocyte maturation arrest and degenerated or morphologically abnormal oocytes) and 2,868 healthy controls, we performed a population and gene-based burden test for mitochondrial genes and identified a candidate gene, cytochrome c oxidase assembly protein 15 (COX15). We report that biallelic COX15 pathogenic variants cause human oocyte ferroptosis and female infertility in a recessive inheritance pattern. COX15 variants impaired mitochondrial respiration in Saccharomyces cerevisiae and led to reduced protein levels in HeLa cells. Oocyte-specific deletion of Cox15 led to impaired Fe2+ and reactive oxygen species homeostasis that caused mitochondrial dysfunction and ultimately sensitized oocytes to ferroptosis. In addition, ferrostatin-1 (an inhibitor of ferroptosis) could rescue the oocyte ferroptosis phenotype in vitro and ex vivo. Our findings not only provide a genetic diagnostic marker for oocyte development defects but also expand the spectrum of mitochondrial disorders to female infertility and contribute to unique insights into the role of ferroptosis in human oocyte defects.
Collapse
Affiliation(s)
- Zhihua Zhang
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Ran Yu
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Qiuwen Shi
- Reproductive Medicine Center, The Third Affiliated Hospital, Guangxi Medical University, Nanning530031, Guangxi, China
| | - Zhi-Jing Wu
- The State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou310024, China
| | - Qingchun Li
- Department of Reproductive Medicine, Binzhou Medical University Hospital, Binzhou256603, China
| | - Jian Mu
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Biaobang Chen
- Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), National Health Commission of the People’s Republic of China, Shanghai200032, China
| | - Juanzi Shi
- Reproductive Medicine Center, Northwest Women’s and Children’s Hospital, Xi’an710069, China
| | - Renmin Ni
- Department of Reproductive Medicine, Kunming Angel Women’s and Children’s Hospital, Kunming650031, Yunnan, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
| | - Qiaoli Li
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and In Vitro Fertilization Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai200011, China
| | - Rong Li
- Reproductive Medicine Center, The Third Affiliated Hospital, Guangxi Medical University, Nanning530031, Guangxi, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and In Vitro Fertilization Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai200011, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai200438, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai200030, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
| | - Qing Sang
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Lei Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| |
Collapse
|
12
|
Sharma Y, Gupta JK, Babu MA, Singh S, Sindhu RK. Signaling Pathways Concerning Mitochondrial Dysfunction: Implications in Neurodegeneration and Possible Molecular Targets. J Mol Neurosci 2024; 74:101. [PMID: 39466510 DOI: 10.1007/s12031-024-02269-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/16/2024] [Indexed: 10/30/2024]
Abstract
Mitochondrion is an important organelle present in our cells responsible for meeting energy requirements. All higher organisms rely on efficient mitochondrial bioenergetic machinery to sustain life. No other respiratory process can produce as much power as generated by mitochondria in the form of ATPs. This review is written in order to get an insight into the magnificent working of mitochondrion and its implications in cellular homeostasis, bioenergetics, redox, calcium signaling, and cell death. However, if this machinery gets faulty, it may lead to several disease states. Mitochondrial dysfunctioning is of growing concern today as it is seen in the pathogenesis of several diseases which includes neurodegenerative disorders, cardiovascular disorders, diabetes mellitus, skeletal muscle defects, liver diseases, and so on. To cover all these aspects is beyond the scope of this article; hence, our study is restricted to neurodegenerative disorders only. Moreover, faulty functioning of this organelle can be one of the causes of early ageing in individuals. This review emphasizes mutations in the mitochondrial DNA, defects in oxidative phosphorylation, generation of ROS, and apoptosis. Researchers have looked into new approaches that might be able to control mitochondrial failure and show a lot of promise as treatments.
Collapse
Affiliation(s)
- Yati Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Jeetendra Kumar Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Sumitra Singh
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Rakesh K Sindhu
- School of Pharmacy, Sharda University, Gautam Buddha Nagar, Greater Noida, Uttar Paresdh, 201310, India.
| |
Collapse
|
13
|
Adelizzi A, Giri A, Di Donfrancesco A, Boito S, Prigione A, Bottani E, Bollati V, Tiranti V, Persico N, Brunetti D. Fetal and obstetrics manifestations of mitochondrial diseases. J Transl Med 2024; 22:853. [PMID: 39313811 PMCID: PMC11421203 DOI: 10.1186/s12967-024-05633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
During embryonic and neonatal development, mitochondria have essential effects on metabolic and energetic regulation, shaping cell fate decisions and leading to significant short- and long-term effects on embryonic and offspring health. Therefore, perturbation on mitochondrial function can have a pathological effect on pregnancy. Several shreds of evidence collected in preclinical models revealed that severe mitochondrial dysfunction is incompatible with life or leads to critical developmental defects, highlighting the importance of correct mitochondrial function during embryo-fetal development. The mechanism impairing the correct development is unknown and may include a dysfunctional metabolic switch in differentiating cells due to decreased ATP production or altered apoptotic signalling. Given the central role of mitochondria in embryonic and fetal development, the mitochondrial dysfunction typical of Mitochondrial Diseases (MDs) should, in principle, be detectable during pregnancy. However, little is known about the clinical manifestations of MDs in embryonic and fetal development. In this manuscript, we review preclinical and clinical evidence suggesting that MDs may affect fetal development and highlight the fetal and maternal outcomes that may provide a wake-up call for targeted genetic diagnosis.
Collapse
Affiliation(s)
- Alessia Adelizzi
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Anastasia Giri
- Fetal Medicine and Surgery Service, Ospedale Maggiore Policlinico, Fondazione IRCCS Ca' Granda, Milan, Italy
| | - Alessia Di Donfrancesco
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Simona Boito
- Fetal Medicine and Surgery Service, Ospedale Maggiore Policlinico, Fondazione IRCCS Ca' Granda, Milan, Italy
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Emanuela Bottani
- Department of Diagnostics and Public Health, University of Verona, Verona, 37124, Italy
| | - Valentina Bollati
- Dipartimento di Scienze Cliniche e di Comunità, Dipartimento di Eccellenza, University of Milan, Milan, 2023-2027, Italy
| | - Valeria Tiranti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Nicola Persico
- Fetal Medicine and Surgery Service, Ospedale Maggiore Policlinico, Fondazione IRCCS Ca' Granda, Milan, Italy.
- Dipartimento di Scienze Cliniche e di Comunità, Dipartimento di Eccellenza, University of Milan, Milan, 2023-2027, Italy.
| | - Dario Brunetti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy.
- Dipartimento di Scienze Cliniche e di Comunità, Dipartimento di Eccellenza, University of Milan, Milan, 2023-2027, Italy.
| |
Collapse
|
14
|
Van Hove JLK, Friederich MW, Hock DH, Stroud DA, Caruana NJ, Christians U, Schniedewind B, Michel CR, Reisdorph R, Lopez Gonzalez EDJ, Brenner C, Donovan TE, Lee JC, Chatfield KC, Larson AA, Baker PR, McCandless SE, Moore Burk MF. ACAD9 treatment with bezafibrate and nicotinamide riboside temporarily stabilizes cardiomyopathy and lactic acidosis. Mitochondrion 2024; 78:101905. [PMID: 38797357 PMCID: PMC11390326 DOI: 10.1016/j.mito.2024.101905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Pathogenic ACAD9 variants cause complex I deficiency. Patients presenting in infancy unresponsive to riboflavin have high mortality. A six-month-old infant presented with riboflavin unresponsive lactic acidosis and life-threatening cardiomyopathy. Treatment with high dose bezafibrate and nicotinamide riboside resulted in marked clinical improvement including reduced lactate and NT-pro-brain type natriuretic peptide levels, with stabilized echocardiographic measures. After a long stable period, the child succumbed from cardiac failure with infection at 10.5 months. Therapy was well tolerated. Peak bezafibrate levels exceeded its EC50. The clinical improvement with this treatment illustrates its potential, but weak PPAR agonist activity of bezafibrate limited its efficacy.
Collapse
Affiliation(s)
- Johan L K Van Hove
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA; Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, Aurora, CO 80045, USA.
| | - Marisa W Friederich
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA; Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Daniella H Hock
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3052, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria 3052, Australia; Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - David A Stroud
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3052, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria 3052, Australia; Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Nikeisha J Caruana
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Uwe Christians
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Björn Schniedewind
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cole R Michel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Richard Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Edwin D J Lopez Gonzalez
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Tonia E Donovan
- Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Jessica C Lee
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Kathryn C Chatfield
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA; Department of Pediatrics, Section of Cardiology, University of Colorado, Aurora, CO, USA
| | - Austin A Larson
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Peter R Baker
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Shawn E McCandless
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Meghan F Moore Burk
- Department of Physical Medicine and Rehabilitation, Children's Hospital Colorado, 13121 East 16(th) Avenue, Aurora, CO, USA
| |
Collapse
|
15
|
de Luxán-Delgado B, Potes Y, Rubio-González A, Solano JJ, Boga JA, Antuña E, Cachán-Vega C, Bermejo-Millo JC, Menéndez-Coto N, García-González C, Pereira GC, Caballero B, Coto-Montes A, Vega-Naredo I. Melatonin Alleviates Liver Mitochondrial Dysfunction in Leptin-Deficient Mice. Int J Mol Sci 2024; 25:8677. [PMID: 39201365 PMCID: PMC11354344 DOI: 10.3390/ijms25168677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Despite efforts to elucidate the cellular adaptations induced by obesity, cellular bioenergetics is currently considered a crucial target. New strategies to delay the onset of the hazardous adaptations induced by obesity are needed. Therefore, we evaluated the effects of 4 weeks of melatonin treatment on mitochondrial function and lipid metabolism in the livers of leptin-deficient mice. Our results revealed that the absence of leptin increased lipid storage in the liver and induced significant mitochondrial alterations, which were ultimately responsible for defective ATP production and reactive oxygen species overproduction. Moreover, leptin deficiency promoted mitochondrial biogenesis, fusion, and outer membrane permeabilization. Melatonin treatment reduced the bioenergetic deficit found in ob/ob mice, alleviating some mitochondrial alterations in the electron transport chain machinery, biogenesis, dynamics, respiration, ATP production, and mitochondrial outer membrane permeabilization. Given the role of melatonin in maintaining mitochondrial homeostasis, it could be used as a therapeutic agent against adipogenic steatosis.
Collapse
Affiliation(s)
- Beatriz de Luxán-Delgado
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
| | - Yaiza Potes
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Adrian Rubio-González
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Juan José Solano
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Geriatrics Service, Monte Naranco Hospital, 33012 Oviedo, Spain
| | - José Antonio Boga
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Microbiology Department, Hospital Universitario Central de Asturias, Avenida de Roma s/n, 33011 Oviedo, Spain
| | - Eduardo Antuña
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Cristina Cachán-Vega
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Juan Carlos Bermejo-Millo
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Nerea Menéndez-Coto
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
| | - Claudia García-González
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
| | - Gonçalo C. Pereira
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK;
| | - Beatriz Caballero
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Ana Coto-Montes
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| | - Ignacio Vega-Naredo
- Department of Morphology and Cell Biology, University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain; (B.d.L.-D.); (Y.P.); (A.R.-G.); (E.A.); (J.C.B.-M.); (N.M.-C.); (B.C.); (A.C.-M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Av. Del Hospital Universitario, 33011 Oviedo, Spain; (J.J.S.); (J.A.B.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Julián Clavería s/n, 33006 Oviedo, Spain
| |
Collapse
|
16
|
Yang TH, Kang EYC, Lin PH, Yu BBC, Wang JHH, Chen V, Wang NK. Mitochondria in Retinal Ganglion Cells: Unraveling the Metabolic Nexus and Oxidative Stress. Int J Mol Sci 2024; 25:8626. [PMID: 39201313 PMCID: PMC11354650 DOI: 10.3390/ijms25168626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
This review explored the role of mitochondria in retinal ganglion cells (RGCs), which are essential for visual processing. Mitochondrial dysfunction is a key factor in the pathogenesis of various vision-related disorders, including glaucoma, hereditary optic neuropathy, and age-related macular degeneration. This review highlighted the critical role of mitochondria in RGCs, which provide metabolic support, regulate cellular health, and respond to cellular stress while also producing reactive oxygen species (ROS) that can damage cellular components. Maintaining mitochondrial function is essential for meeting RGCs' high metabolic demands and ensuring redox homeostasis, which is crucial for their proper function and visual health. Oxidative stress, exacerbated by factors like elevated intraocular pressure and environmental factors, contributes to diseases such as glaucoma and age-related vision loss by triggering cellular damage pathways. Strategies targeting mitochondrial function or bolstering antioxidant defenses include mitochondrial-based therapies, gene therapies, and mitochondrial transplantation. These advances can offer potential strategies for addressing mitochondrial dysfunction in the retina, with implications that extend beyond ocular diseases.
Collapse
Affiliation(s)
- Tsai-Hsuan Yang
- Department of Education, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Eugene Yu-Chuan Kang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
| | - Pei-Hsuan Lin
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- National Taiwan University Hospital, Yunlin 640203, Taiwan
| | - Benjamin Ben-Chi Yu
- Fu Foundation School of Engineering & Applied Science, Columbia University, New York, NY 10027, USA;
| | - Jason Hung-Hsuan Wang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- Columbian College of Arts and Sciences, George Washington University, Washington, DC 20052, USA
| | - Vincent Chen
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada
| | - Nan-Kai Wang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
17
|
Adami R, Pezzotta M, Cadile F, Cuniolo B, Rovati G, Canepari M, Bottai D. Physiological Features of the Neural Stem Cells Obtained from an Animal Model of Spinal Muscular Atrophy and Their Response to Antioxidant Curcumin. Int J Mol Sci 2024; 25:8364. [PMID: 39125934 PMCID: PMC11313061 DOI: 10.3390/ijms25158364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
The most prevalent rare genetic disease affecting young individuals is spinal muscular atrophy (SMA), which is caused by a loss-of-function mutation in the telomeric gene survival motor neuron (SMN) 1. The high heterogeneity of the SMA pathophysiology is determined by the number of copies of SMN2, a separate centromeric gene that can transcribe for the same protein, although it is expressed at a slower rate. SMA affects motor neurons. However, a variety of different tissues and organs may also be affected depending on the severity of the condition. Novel pharmacological treatments, such as Spinraza, Onasemnogene abeparvovec-xioi, and Evrysdi, are considered to be disease modifiers because their use can change the phenotypes of the patients. Since oxidative stress has been reported in SMA-affected cells, we studied the impact of antioxidant therapy on neural stem cells (NSCs) that have the potential to differentiate into motor neurons. Antioxidants can act through various pathways; for example, some of them exert their function through nuclear factor (erythroid-derived 2)-like 2 (NRF2). We found that curcumin is able to induce positive effects in healthy and SMA-affected NSCs by activating the nuclear translocation of NRF2, which may use a different mechanism than canonical redox regulation through the antioxidant-response elements and the production of antioxidant molecules.
Collapse
Affiliation(s)
- Raffaella Adami
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Matteo Pezzotta
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Francesca Cadile
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy; (F.C.); (M.C.)
| | - Beatrice Cuniolo
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Gianenrico Rovati
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| | - Monica Canepari
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy; (F.C.); (M.C.)
| | - Daniele Bottai
- Section of Pharmacology and Biosciences, Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy; (R.A.); (M.P.); (B.C.); (G.R.)
| |
Collapse
|
18
|
Agostini F, Pereyra L, Dale J, Yambire KF, Maglioni S, Schiavi A, Ventura N, Milosevic I, Raimundo N. Upregulation of cholesterol synthesis by lysosomal defects requires a functional mitochondrial respiratory chain. J Biol Chem 2024; 300:107403. [PMID: 38782205 PMCID: PMC11254723 DOI: 10.1016/j.jbc.2024.107403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Mitochondria and lysosomes are two organelles that carry out both signaling and metabolic roles in cells. Recent evidence has shown that mitochondria and lysosomes are dependent on one another, as primary defects in one cause secondary defects in the other. Although there are functional impairments in both cases, the signaling consequences of primary mitochondrial dysfunction and lysosomal defects are dissimilar. Here, we used RNA sequencing to obtain transcriptomes from cells with primary mitochondrial or lysosomal defects to identify the global cellular consequences associated with mitochondrial or lysosomal dysfunction. We used these data to determine the pathways affected by defects in both organelles, which revealed a prominent role for the cholesterol synthesis pathway. We observed a transcriptional upregulation of this pathway in cellular and murine models of lysosomal defects, while it is transcriptionally downregulated in cellular and murine models of mitochondrial defects. We identified a role for the posttranscriptional regulation of transcription factor SREBF1, a master regulator of cholesterol and lipid biosynthesis, in models of mitochondrial respiratory chain deficiency. Furthermore, we found that retention of Ca2+ in lysosomes of cells with mitochondrial respiratory chain defects contributes to the differential regulation of the cholesterol synthesis pathway in the mitochondrial and lysosomal defects tested. Finally, we verified in vivo, using a model of mitochondria-associated disease in Caenorhabditis elegans that normalization of lysosomal Ca2+ levels results in partial rescue of the developmental delay induced by the respiratory chain deficiency.
Collapse
Affiliation(s)
- Francesco Agostini
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Leonardo Pereyra
- Department of Cellular Biochemistry, University Medical Center, Goettingen, Germany
| | - Justin Dale
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - King Faisal Yambire
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, New York, USA
| | - Silvia Maglioni
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Alfonso Schiavi
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Natascia Ventura
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Ira Milosevic
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Multidisciplinary Institute for Ageing, University of Coimbra, Coimbra, Portugal
| | - Nuno Raimundo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
19
|
Khaghani F, Hemmati M, Ebrahimi M, Salmaninejad A. Emerging Multi-omic Approaches to the Molecular Diagnosis of Mitochondrial Disease and Available Strategies for Treatment and Prevention. Curr Genomics 2024; 25:358-379. [PMID: 39323625 PMCID: PMC11420563 DOI: 10.2174/0113892029308327240612110334] [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: 02/02/2024] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 09/27/2024] Open
Abstract
Mitochondria are semi-autonomous organelles present in several copies within most cells in the human body that are controlled by the precise collaboration of mitochondrial DNA (mtDNA) and nuclear DNA (nDNA) encoding mitochondrial proteins. They play important roles in numerous metabolic pathways, such as the synthesis of adenosine triphosphate (ATP), the predominant energy substrate of the cell generated through oxidative phosphorylation (OXPHOS), intracellular calcium homeostasis, metabolite biosynthesis, aging, cell cycles, and so forth. Previous studies revealed that dysfunction of these multi-functional organelles, which may arise due to mutations in either the nuclear or mitochondrial genome, leads to a diverse group of clinically and genetically heterogeneous disorders. These diseases include neurodegenerative and metabolic disorders as well as cardiac and skeletal myopathies in both adults and newborns. The plethora of phenotypes and defects displayed leads to challenges in the diagnosis and treatment of mitochondrial diseases. In this regard, the related literature proposed several diagnostic options, such as high throughput mitochondrial genomics and omics technologies, as well as numerous therapeutic options, such as pharmacological approaches, manipulating the mitochondrial genome, increasing the mitochondria content of the affected cells, and recently mitochondrial diseases transmission prevention. Therefore, the present article attempted to review the latest advances and challenges in diagnostic and therapeutic options for mitochondrial diseases.
Collapse
Affiliation(s)
- Faeze Khaghani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
- Medical Genetic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboobeh Hemmati
- Medical Genetic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Ebrahimi
- Department of Animal Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Arash Salmaninejad
- Medical Genetic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Regenerative Medicine, Organ Procurement and Transplantation Multi-Disciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
20
|
Choksey A, Carter RD, Thackray BD, Ball V, Kennedy BWC, Ha LHT, Sharma E, Broxholme J, Castro-Guarda M, Murphy MP, Heather LC, Tyler DJ, Timm KN. AICAR confers prophylactic cardioprotection in doxorubicin-induced heart failure in rats. J Mol Cell Cardiol 2024; 191:12-22. [PMID: 38643934 DOI: 10.1016/j.yjmcc.2024.04.011] [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: 11/30/2022] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Doxorubicin (DOX) is a widely used chemotherapeutic agent that can cause serious cardiotoxic side effects, leading to heart failure (HF). Impaired mitochondrial function is thought to be key factor driving progression into HF. We have previously shown in a rat model of DOX-HF that heart failure with reduced ejection fraction correlates with mitochondrial loss and dysfunction. Adenosine monophosphate-dependent kinase (AMPK) is a cellular energy sensor, regulating mitochondrial biogenesis and energy metabolism, including fatty acid oxidation. We hypothesised that AMPK activation could restore mitochondrial function and therefore be a novel cardioprotective strategy for the prevention of DOX-HF. Consequently, we set out to assess whether 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside (AICAR), an activator of AMPK, could prevent cardiac functional decline in this chronic intravenous rat model of DOX-HF. In line with our hypothesis, AICAR improved cardiac systolic function. AICAR furthermore improved cardiac mitochondrial fatty acid oxidation, independent of mitochondrial number, and in the absence of observable AMPK-activation. In addition, we found that AICAR prevented loss of myocardial mass. RNAseq analysis showed that this may be driven by normalisation of pathways associated with ribosome function and protein synthesis, which are impaired in DOX-treated rat hearts. AICAR furthermore prevented dyslipidemia and excessive body-weight loss in DOX-treated rats, which may contribute to preservation of myocardial mass. Though it is unclear whether AICAR exerted its cardioprotective effect through cardiac or extra-cardiac AMPK-activation or via an AMPK-independent effect, these results show promise for the use of AICAR as a cardioprotective agent in DOX-HF to both preserve cardiac function and mass.
Collapse
Affiliation(s)
- Anurag Choksey
- Department of Physiology Anatomy and Genetics, University of Oxford, UK
| | - Ryan D Carter
- Department of Physiology Anatomy and Genetics, University of Oxford, UK; Doctoral Training Centre, University of Oxford, Keble Road, Oxford, OX1 3NP, UK
| | | | - Vicky Ball
- Department of Physiology Anatomy and Genetics, University of Oxford, UK
| | - Brett W C Kennedy
- Department of Physiology Anatomy and Genetics, University of Oxford, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK
| | | | - Eshita Sharma
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7BN, UK
| | - John Broxholme
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7BN, UK
| | | | | | - Lisa C Heather
- Department of Physiology Anatomy and Genetics, University of Oxford, UK
| | - Damian J Tyler
- Department of Physiology Anatomy and Genetics, University of Oxford, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK
| | - Kerstin N Timm
- Department of Physiology Anatomy and Genetics, University of Oxford, UK; Department of Pharmacology, University of Oxford, UK.
| |
Collapse
|
21
|
Zong Y, Li H, Liao P, Chen L, Pan Y, Zheng Y, Zhang C, Liu D, Zheng M, Gao J. Mitochondrial dysfunction: mechanisms and advances in therapy. Signal Transduct Target Ther 2024; 9:124. [PMID: 38744846 PMCID: PMC11094169 DOI: 10.1038/s41392-024-01839-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 12/05/2023] [Accepted: 04/21/2024] [Indexed: 05/16/2024] Open
Abstract
Mitochondria, with their intricate networks of functions and information processing, are pivotal in both health regulation and disease progression. Particularly, mitochondrial dysfunctions are identified in many common pathologies, including cardiovascular diseases, neurodegeneration, metabolic syndrome, and cancer. However, the multifaceted nature and elusive phenotypic threshold of mitochondrial dysfunction complicate our understanding of their contributions to diseases. Nonetheless, these complexities do not prevent mitochondria from being among the most important therapeutic targets. In recent years, strategies targeting mitochondrial dysfunction have continuously emerged and transitioned to clinical trials. Advanced intervention such as using healthy mitochondria to replenish or replace damaged mitochondria, has shown promise in preclinical trials of various diseases. Mitochondrial components, including mtDNA, mitochondria-located microRNA, and associated proteins can be potential therapeutic agents to augment mitochondrial function in immunometabolic diseases and tissue injuries. Here, we review current knowledge of mitochondrial pathophysiology in concrete examples of common diseases. We also summarize current strategies to treat mitochondrial dysfunction from the perspective of dietary supplements and targeted therapies, as well as the clinical translational situation of related pharmacology agents. Finally, this review discusses the innovations and potential applications of mitochondrial transplantation as an advanced and promising treatment.
Collapse
Affiliation(s)
- Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Long Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yao Pan
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yongqiang Zheng
- Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Minghao Zheng
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
22
|
Song N, Mei S, Wang X, Hu G, Lu M. Focusing on mitochondria in the brain: from biology to therapeutics. Transl Neurodegener 2024; 13:23. [PMID: 38632601 PMCID: PMC11022390 DOI: 10.1186/s40035-024-00409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Mitochondria have multiple functions such as supplying energy, regulating the redox status, and producing proteins encoded by an independent genome. They are closely related to the physiology and pathology of many organs and tissues, among which the brain is particularly prominent. The brain demands 20% of the resting metabolic rate and holds highly active mitochondrial activities. Considerable research shows that mitochondria are closely related to brain function, while mitochondrial defects induce or exacerbate pathology in the brain. In this review, we provide comprehensive research advances of mitochondrial biology involved in brain functions, as well as the mitochondria-dependent cellular events in brain physiology and pathology. Furthermore, various perspectives are explored to better identify the mitochondrial roles in neurological diseases and the neurophenotypes of mitochondrial diseases. Finally, mitochondrial therapies are discussed. Mitochondrial-targeting therapeutics are showing great potentials in the treatment of brain diseases.
Collapse
Affiliation(s)
- Nanshan Song
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shuyuan Mei
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Xiangxu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
- Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
23
|
Palzkill VR, Tan J, Tice AL, Ferriera LF, Ryan TE. A 6-minute Limb Function Assessment for Therapeutic Testing in Experimental Peripheral Artery Disease Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586197. [PMID: 38585832 PMCID: PMC10996543 DOI: 10.1101/2024.03.21.586197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background The translation of promising therapies from pre-clinical models of hindlimb ischemia (HLI) to patients with peripheral artery disease (PAD) has been inadequate. While this failure is multifactorial, primary outcome measures in preclinical HLI models and clinical trials involving patients with PAD are not aligned well. For example, laser Doppler perfusion recovery measured under resting conditions is the most used outcome in HLI studies, whereas clinical trials involving patients with PAD primarily assess walking performance. Here, we sought to develop a 6-min limb function test for preclinical HLI models that assess muscular performance and hemodynamics congruently. Methods We developed an in situ 6-min limb function test that involves repeated isotonic (shortening) contractions performed against a submaximal load. Continuous measurement of muscle blood flow was performed using laser Doppler flowmetry. Quantification of muscle power, work, and perfusion are obtained across the test. To assess the efficacy of this test, we performed HLI via femoral artery ligation on several mouse strains: C57BL6J, BALBc/J, and MCK-PGC1α (muscle-specific overexpression of PGC1α). Additional experiments were performed using an exercise intervention (voluntary wheel running) following HLI. Results The 6-min limb function test was successful at detecting differences in limb function of C57BL6/J and BALBc/J mice subjected to HLI with effect sizes superior to laser Doppler perfusion recovery. C57BL6/J mice randomized to exercise therapy following HLI had smaller decline in muscle power, greater hyperemia, and performed more work across the 6-min limb function test compared to non-exercise controls with HLI. Mice with muscle-specific overexpression of PGC1α had no differences in perfusion recovery in resting conditions, but exhibited greater capillary density, increased muscle mass and absolute force levels, and performed more work across the 6-min limb function test compared to their wildtype littermates without the transgene. Conclusion These results demonstrate the efficacy of the 6-min limb function test to detect differences in the response to HLI across several interventions including where traditional perfusion recovery, capillary density, and muscle strength measures were unable to detect therapeutic differences.
Collapse
Affiliation(s)
- Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | | | - Leonardo F. Ferriera
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
| |
Collapse
|
24
|
Agostini F, Pereyra L, Dale J, Yambire KF, Maglioni S, Schiavi A, Ventura N, Milosevic I, Raimundo N. Up-regulation of cholesterol synthesis by lysosomal defects requires a functional mitochondrial respiratory chain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583589. [PMID: 38496624 PMCID: PMC10942416 DOI: 10.1101/2024.03.06.583589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Mitochondria and lysosomes are two organelles that carry out both signaling and metabolic roles in the cells. Recent evidence has shown that mitochondria and lysosomes are dependent on one another, as primary defects in one cause secondary defects in the other. Nevertheless, the signaling consequences of primary mitochondrial malfunction and of primary lysosomal defects are not similar, despite in both cases there are impairments of mitochondria and of lysosomes. Here, we used RNA sequencing to obtain transcriptomes from cells with primary mitochondrial or lysosomal defects, to identify what are the global cellular consequences that are associated with malfunction of mitochondria or lysosomes. We used these data to determine what are the pathways that are affected by defects in both organelles, which revealed a prominent role for the cholesterol synthesis pathway. This pathway is transcriptionally up-regulated in cellular and mouse models of lysosomal defects and is transcriptionally down-regulated in cellular and mouse models of mitochondrial defects. We identified a role for post-transcriptional regulation of the transcription factor SREBF1, a master regulator of cholesterol and lipid biosynthesis, in models of mitochondrial respiratory chain deficiency. Furthermore, the retention of Ca 2+ in the lysosomes of cells with mitochondrial respiratory chain defects contributes to the differential regulation of the cholesterol synthesis pathway in the mitochondrial and lysosomal defects tested. Finally, we verified in vivo , using models of mitochondria-associated diseases in C. elegans , that normalization of lysosomal Ca 2+ levels results in partial rescue of the developmental arrest induced by the respiratory chain deficiency.
Collapse
|
25
|
Manoli I, Sysol JR, Head PE, Epping MW, Gavrilova O, Crocker MK, Sloan JL, Koutsoukos SA, Wang C, Ktena YP, Mendelson S, Pass AR, Zerfas PM, Hoffmann V, Vernon HJ, Fletcher LA, Reynolds JC, Tsokos MG, Stratakis CA, Voss SD, Chen KY, Brown RJ, Hamosh A, Berry GT, Chen XS, Yanovski JA, Venditti CP. Lipodystrophy in methylmalonic acidemia associated with elevated FGF21 and abnormal methylmalonylation. JCI Insight 2024; 9:e174097. [PMID: 38271099 DOI: 10.1172/jci.insight.174097] [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: 07/24/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
A distinct adipose tissue distribution pattern was observed in patients with methylmalonyl-CoA mutase deficiency, an inborn error of branched-chain amino acid (BCAA) metabolism, characterized by centripetal obesity with proximal upper and lower extremity fat deposition and paucity of visceral fat, that resembles familial multiple lipomatosis syndrome. To explore brown and white fat physiology in methylmalonic acidemia (MMA), body composition, adipokines, and inflammatory markers were assessed in 46 patients with MMA and 99 matched controls. Fibroblast growth factor 21 levels were associated with acyl-CoA accretion, aberrant methylmalonylation in adipose tissue, and an attenuated inflammatory cytokine profile. In parallel, brown and white fat were examined in a liver-specific transgenic MMA mouse model (Mmut-/- TgINS-Alb-Mmut). The MMA mice exhibited abnormal nonshivering thermogenesis with whitened brown fat and had an ineffective transcriptional response to cold stress. Treatment of the MMA mice with bezafibrates led to clinical improvement with beiging of subcutaneous fat depots, which resembled the distribution seen in the patients. These studies defined what we believe to be a novel lipodystrophy phenotype in patients with defects in the terminal steps of BCAA oxidation and demonstrated that beiging of subcutaneous adipose tissue in MMA could readily be induced with small molecules.
Collapse
Affiliation(s)
- Irini Manoli
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Justin R Sysol
- Metabolic Medicine Branch, National Human Genome Research Institute
| | | | | | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Melissa K Crocker
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | - Jennifer L Sloan
- Metabolic Medicine Branch, National Human Genome Research Institute
| | | | - Cindy Wang
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Yiouli P Ktena
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Sophia Mendelson
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | - Alexandra R Pass
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Patricia M Zerfas
- Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA
| | - Victoria Hoffmann
- Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Laura A Fletcher
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | | | - Maria G Tsokos
- Ultrastructural Pathology Section, Center for Cancer Research; and
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Stephan D Voss
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Rebecca J Brown
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Ada Hamosh
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gerard T Berry
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoyuan Shawn Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, Maryland, USA
| | - Jack A Yanovski
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | | |
Collapse
|
26
|
Eggelbusch M, Charlton BT, Bosutti A, Ganse B, Giakoumaki I, Grootemaat AE, Hendrickse PW, Jaspers Y, Kemp S, Kerkhoff TJ, Noort W, van Weeghel M, van der Wel NN, Wesseling JR, Frings-Meuthen P, Rittweger J, Mulder ER, Jaspers RT, Degens H, Wüst RCI. The impact of bed rest on human skeletal muscle metabolism. Cell Rep Med 2024; 5:101372. [PMID: 38232697 PMCID: PMC10829795 DOI: 10.1016/j.xcrm.2023.101372] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/19/2023] [Accepted: 12/14/2023] [Indexed: 01/19/2024]
Abstract
Insulin sensitivity and metabolic flexibility decrease in response to bed rest, but the temporal and causal adaptations in human skeletal muscle metabolism are not fully defined. Here, we use an integrative approach to assess human skeletal muscle metabolism during bed rest and provide a multi-system analysis of how skeletal muscle and the circulatory system adapt to short- and long-term bed rest (German Clinical Trials: DRKS00015677). We uncover that intracellular glycogen accumulation after short-term bed rest accompanies a rapid reduction in systemic insulin sensitivity and less GLUT4 localization at the muscle cell membrane, preventing further intracellular glycogen deposition after long-term bed rest. We provide evidence of a temporal link between the accumulation of intracellular triglycerides, lipotoxic ceramides, and sphingomyelins and an altered skeletal muscle mitochondrial structure and function after long-term bed rest. An intracellular nutrient overload therefore represents a crucial determinant for rapid skeletal muscle insulin insensitivity and mitochondrial alterations after prolonged bed rest.
Collapse
Affiliation(s)
- Moritz Eggelbusch
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands; Department of Nutrition and Dietetics, Amsterdam University Medical Centers, Amsterdam Movement Sciences, Amsterdam, the Netherlands; Faculty of Sports and Nutrition, Center of Expertise Urban Vitality, Amsterdam University of Applied Sciences, Amsterdam, the Netherlands
| | - Braeden T Charlton
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | | | - Bergita Ganse
- Research Centre for Musculoskeletal Science and Sports Medicine, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK; Clinics and Institutes of Surgery, Saarland University, Homburg, Germany
| | - Ifigenia Giakoumaki
- Research Centre for Musculoskeletal Science and Sports Medicine, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Anita E Grootemaat
- Electron Microscopy Center Amsterdam, Department of Medical Biology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Paul W Hendrickse
- Research Centre for Musculoskeletal Science and Sports Medicine, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK; Lancaster Medical School, Lancaster University, Lancaster, UK
| | - Yorrick Jaspers
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Tom J Kerkhoff
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Wendy Noort
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Nicole N van der Wel
- Electron Microscopy Center Amsterdam, Department of Medical Biology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Julia R Wesseling
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Petra Frings-Meuthen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Jörn Rittweger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Department of Pediatrics and Adolescent Medicine, University Hospital Cologne, Cologne, Germany
| | - Edwin R Mulder
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Richard T Jaspers
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Hans Degens
- Research Centre for Musculoskeletal Science and Sports Medicine, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK; Lithuanian Sports University, Kaunas, Lithuania
| | - Rob C I Wüst
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
27
|
Di Leo V, Bernardino Gomes TM, Vincent AE. Interactions of mitochondrial and skeletal muscle biology in mitochondrial myopathy. Biochem J 2023; 480:1767-1789. [PMID: 37965929 PMCID: PMC10657187 DOI: 10.1042/bcj20220233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
Mitochondrial dysfunction in skeletal muscle fibres occurs with both healthy aging and a range of neuromuscular diseases. The impact of mitochondrial dysfunction in skeletal muscle and the way muscle fibres adapt to this dysfunction is important to understand disease mechanisms and to develop therapeutic interventions. Furthermore, interactions between mitochondrial dysfunction and skeletal muscle biology, in mitochondrial myopathy, likely have important implications for normal muscle function and physiology. In this review, we will try to give an overview of what is known to date about these interactions including metabolic remodelling, mitochondrial morphology, mitochondrial turnover, cellular processes and muscle cell structure and function. Each of these topics is at a different stage of understanding, with some being well researched and understood, and others in their infancy. Furthermore, some of what we know comes from disease models. Whilst some findings are confirmed in humans, where this is not yet the case, we must be cautious in interpreting findings in the context of human muscle and disease. Here, our goal is to discuss what is known, highlight what is unknown and give a perspective on the future direction of research in this area.
Collapse
Affiliation(s)
- Valeria Di Leo
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
| | - Tiago M. Bernardino Gomes
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, U.K
| | - Amy E. Vincent
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
| |
Collapse
|
28
|
Glynos A, Bozhilova LV, Frison M, Burr S, Stewart JB, Chinnery PF. High-throughput single-cell analysis reveals progressive mitochondrial DNA mosaicism throughout life. SCIENCE ADVANCES 2023; 9:eadi4038. [PMID: 37878704 PMCID: PMC10599618 DOI: 10.1126/sciadv.adi4038] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/21/2023] [Indexed: 10/27/2023]
Abstract
Heteroplasmic mitochondrial DNA (mtDNA) mutations are a major cause of inherited disease and contribute to common late-onset human disorders. The late onset and clinical progression of mtDNA-associated disease is thought to be due to changing heteroplasmy levels, but it is not known how and when this occurs. Performing high-throughput single-cell genotyping in two mouse models of human mtDNA disease, we saw unanticipated cell-to-cell differences in mtDNA heteroplasmy levels that emerged prenatally and progressively increased throughout life. Proliferating spleen cells and nondividing brain cells had a similar single-cell heteroplasmy variance, implicating mtDNA or organelle turnover as the major force determining cell heteroplasmy levels. The two different mtDNA mutations segregated at different rates with no evidence of selection, consistent with different rates of random genetic drift in vivo, leading to the accumulation of cells with a very high mutation burden at different rates. This provides an explanation for differences in severity seen in human diseases caused by similar mtDNA mutations.
Collapse
Affiliation(s)
- Angelos Glynos
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Lyuba V. Bozhilova
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Michele Frison
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Stephen Burr
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - James B. Stewart
- Biosciences Institute, Faculty of Medical Sciences, Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK
| | - Patrick F. Chinnery
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
29
|
Lee MJ, Cho Y, Hwang Y, Jo Y, Kim YG, Lee SH, Lee JH. Kaempferol Alleviates Mitochondrial Damage by Reducing Mitochondrial Reactive Oxygen Species Production in Lipopolysaccharide-Induced Prostate Organoids. Foods 2023; 12:3836. [PMID: 37893729 PMCID: PMC10606128 DOI: 10.3390/foods12203836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Common prostate diseases such as prostatitis and benign prostatic hyperplasia (BPH) have a high incidence at any age. Cellular stresses, such as reactive oxygen species (ROS) and chronic inflammation, are implicated in prostate enlargement and cancer progression and development. Kaempferol is a flavonoid found in abundance in various plants, including broccoli and spinach, and has been reported to exhibit positive biological activities, such as antioxidant and anti-inflammatory properties. In the present study, we introduced prostate organoids to investigate the protective effects of kaempferol against various cellular stresses. The levels of COX-2, iNOS, p-IκB, a pro-inflammatory cytokine, and ROS were increased by LPS treatment but reversed by kaempferol treatment. Kaempferol activated the nuclear factor erythroid 2-related factor 2(Nrf2)-related pathway and enhanced the mitochondrial quality control proteins PGC-1α, PINK1, Parkin, and Beclin. The increase in mitochondrial ROS and oxygen consumption induced by LPS was stabilized by kaempferol treatment. First, our study used prostate organoids as a novel evaluation platform. Secondly, it was demonstrated that kaempferol could alleviate the mitochondrial damage in LPS-induced induced prostate organoids by reducing the production of mitochondrial ROS.
Collapse
Affiliation(s)
- Myeong Joon Lee
- Department of Food Science and Biotechnology, Gachon University, Seongnam 13120, Republic of Korea; (M.J.L.); (Y.C.); (Y.H.)
| | - Yeonoh Cho
- Department of Food Science and Biotechnology, Gachon University, Seongnam 13120, Republic of Korea; (M.J.L.); (Y.C.); (Y.H.)
| | - Yujin Hwang
- Department of Food Science and Biotechnology, Gachon University, Seongnam 13120, Republic of Korea; (M.J.L.); (Y.C.); (Y.H.)
| | - Youngheun Jo
- Department of Urology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea;
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Seung Hwan Lee
- Department of Urology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jong Hun Lee
- Department of Food Science and Biotechnology, Gachon University, Seongnam 13120, Republic of Korea; (M.J.L.); (Y.C.); (Y.H.)
| |
Collapse
|
30
|
Averina OA, Kuznetsova SA, Permyakov OA, Sergiev PV. Animal Models of Mitochondrial Diseases Associated with Nuclear Gene Mutations. Acta Naturae 2023; 15:4-22. [PMID: 38234606 PMCID: PMC10790356 DOI: 10.32607/actanaturae.25442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 10/05/2023] [Indexed: 01/19/2024] Open
Abstract
Mitochondrial diseases (MDs) associated with nuclear gene mutations are part of a large group of inherited diseases caused by the suppression of energy metabolism. These diseases are of particular interest, because nuclear genes encode not only most of the structural proteins of the oxidative phosphorylation system (OXPHOS), but also all the proteins involved in the OXPHOS protein import from the cytoplasm and their assembly in mitochondria. Defects in any of these proteins can lead to functional impairment of the respiratory chain, including dysfunction of complex I that plays a central role in cellular respiration and oxidative phosphorylation, which is the most common cause of mitopathologies. Mitochondrial diseases are characterized by an early age of onset and a progressive course and affect primarily energy-consuming tissues and organs. The treatment of MDs should be initiated as soon as possible, but the diagnosis of mitopathologies is extremely difficult because of their heterogeneity and overlapping clinical features. The molecular pathogenesis of mitochondrial diseases is investigated using animal models: i.e. animals carrying mutations causing MD symptoms in humans. The use of mutant animal models opens new opportunities in the study of genes encoding mitochondrial proteins, as well as the molecular mechanisms of mitopathology development, which is necessary for improving diagnosis and developing approaches to drug therapy. In this review, we present the most recent information on mitochondrial diseases associated with nuclear gene mutations and animal models developed to investigate them.
Collapse
Affiliation(s)
- O. A. Averina
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - S. A. Kuznetsova
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - O. A. Permyakov
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - P. V. Sergiev
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| |
Collapse
|
31
|
Chen L, Zhou M, Li H, Liu D, Liao P, Zong Y, Zhang C, Zou W, Gao J. Mitochondrial heterogeneity in diseases. Signal Transduct Target Ther 2023; 8:311. [PMID: 37607925 PMCID: PMC10444818 DOI: 10.1038/s41392-023-01546-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 08/24/2023] Open
Abstract
As key organelles involved in cellular metabolism, mitochondria frequently undergo adaptive changes in morphology, components and functions in response to various environmental stresses and cellular demands. Previous studies of mitochondria research have gradually evolved, from focusing on morphological change analysis to systematic multiomics, thereby revealing the mitochondrial variation between cells or within the mitochondrial population within a single cell. The phenomenon of mitochondrial variation features is defined as mitochondrial heterogeneity. Moreover, mitochondrial heterogeneity has been reported to influence a variety of physiological processes, including tissue homeostasis, tissue repair, immunoregulation, and tumor progression. Here, we comprehensively review the mitochondrial heterogeneity in different tissues under pathological states, involving variant features of mitochondrial DNA, RNA, protein and lipid components. Then, the mechanisms that contribute to mitochondrial heterogeneity are also summarized, such as the mutation of the mitochondrial genome and the import of mitochondrial proteins that result in the heterogeneity of mitochondrial DNA and protein components. Additionally, multiple perspectives are investigated to better comprehend the mysteries of mitochondrial heterogeneity between cells. Finally, we summarize the prospective mitochondrial heterogeneity-targeting therapies in terms of alleviating mitochondrial oxidative damage, reducing mitochondrial carbon stress and enhancing mitochondrial biogenesis to relieve various pathological conditions. The possibility of recent technological advances in targeted mitochondrial gene editing is also discussed.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengnan Zhou
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, 110001, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China.
| |
Collapse
|
32
|
Meng M, Li X, Huo R, Ma N, Chang G, Shen X. A high-concentrate diet induces mitochondrial dysfunction by activating the MAPK signaling pathway in the mammary gland of dairy cows. J Dairy Sci 2023; 106:5775-5787. [PMID: 37296051 DOI: 10.3168/jds.2022-22907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/10/2023] [Indexed: 06/12/2023]
Abstract
Subacute rumen acidosis can lead to mastitis in dairy cows. Mitochondrial dysfunction is closely related to the inflammatory response. This experiment was conducted to investigate the effects of a high-concentrate diet on mammary gland inflammation and mitochondrial damage in dairy cows. Twelve Holstein dairy cows in mid-lactation were randomly divided into 2 groups and fed a 40% concentrate (low concentrate, LC) diet or a 60% concentrate (high concentrate, HC) diet. Cows were fed individually, and the experiment lasted for 3 wk. After the experiment, mammary gland tissue, blood, and rumen fluid were collected. Compared with the LC diet, the HC diet significantly decreased rumen pH; the pH was <5.6 for more than 3 h. The HC diet also increased the concentration of LPS in the blood (7.17 ± 1.25 µg/mL vs. 12.12 ± 1.26 µg/mL), which indicated that feeding the HC diet successfully induced subacute rumen acidosis. The HC diet also increased the concentration of Ca2+ (34.80 ± 4.23 µg/g vs. 46.87 ± 7.24 µg/g) in the mammary gland and upregulated the expression of inflammatory factors IL-6 (1,128.31 ± 147.53 pg/g vs. 1,538.42 ± 241.38 pg/g), IL-1β (69.67 ± 5.86 pg/g vs. 90.13 ± 4.78 pg/g), and tumor necrosis factor-α (91.99 ± 10.43 pg/g vs. 131.75 ± 17.89 pg/g) in mammary venous blood. The HC diet also increased the activity of myeloperoxidase (0.41 ± 0.05 U/g vs. 0.71 ± 0.11 U/g) and decreased the content of ATP (0.47 ± 0.10 µg/mL vs. 0.32 ± 0.11 µg/mL) in the mammary gland. In addition, phosphorylation of JNK (1.00 ± 0.21 vs. 2.84 ± 0.75), ERK (1.00 ± 0.20 vs. 1.53 ± 0.31), and p38 (1.00 ± 0.13 vs. 1.47 ± 0.41) and protein expression of IL-6 (1.00 ± 0.22 vs. 2.21 ± 0.27) and IL-8 (1.00 ± 0.17 vs. 1.96 ± 0.26) were enhanced in cows of the HC group, indicating that the mitogen-activated protein kinase (MAPK) signaling pathway was activated. Compared with the LC diet, the HC diet reduced the protein expression of mitochondrial biogenesis-related proteins PGC-1α (1.00 ± 0.17 vs. 0.55 ± 0.12), NRF1 (1.00 ± 0.17 vs. 0.60 ± 0.10), TFAM (1.00 ± 0.10 vs. 0.73 ± 0.09), and SIRTI (1.00 ± 0.44 vs. 0.40 ± 0.10). The HC diet promoted mitochondrial fission and inhibited mitochondrial fusion by reducing protein expression of MFN1 (1.00 ± 0.31 vs. 0.49 ± 0.09), MFN2 (1.00 ± 0.19 vs. 0.69 ± 0.13), and OPA1 (1.00 ± 0.08 vs. 0.72 ± 0.07), and by increasing that of DRP1 (1.00 ± 0.09 vs. 1.39 ± 0.10), MFF (1.00 ± 0.15 vs. 1.89 ± 0.12), and TTC1/FIS1 (1.00 ± 0.08 vs. 1.76 ± 0.14), leading to mitochondrial dysfunction. The HC diet increased mitochondrial permeability by upregulating the protein expression of VDAC1 (1.00 ± 0.42 vs. 1.90 ± 0.44), ANT (1.00 ± 0.22 vs. 1.27 ± 0.17), and CYPD (1.00 ± 0.41 vs. 1.82 ± 0.43). Taken together, these results indicated that feeding the HC diet induced mitochondrial damage via the MAPK signaling pathway in the mammary gland of dairy cows.
Collapse
Affiliation(s)
- Meijuan Meng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, P. R. China
| | - Xuerui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, P. R. China
| | - Ran Huo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, P. R. China
| | - Nana Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, P. R. China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, P. R. China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, P. R. China.
| |
Collapse
|
33
|
Misra SK, Rosenholm JM, Pathak K. Functionalized and Nonfunctionalized Nanosystems for Mitochondrial Drug Delivery with Metallic Nanoparticles. Molecules 2023; 28:4701. [PMID: 37375256 DOI: 10.3390/molecules28124701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/04/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Background: The application of metallic nanoparticles as a novel therapeutic tool has significant potential to facilitate the treatment and diagnosis of mitochondria-based disorders. Recently, subcellular mitochondria have been trialed to cure pathologies that depend on their dysfunction. Nanoparticles made from metals and their oxides (including gold, iron, silver, platinum, zinc oxide, and titanium dioxide) have unique modi operandi that can competently rectify mitochondrial disorders. Materials: This review presents insight into the recent research reports on exposure to a myriad of metallic nanoparticles that can alter the dynamic ultrastructure of mitochondria (via altering metabolic homeostasis), as well as pause ATP production, and trigger oxidative stress. The facts and figures have been compiled from more than a hundred PubMed, Web of Science, and Scopus indexed articles that describe the essential functions of mitochondria for the management of human diseases. Result: Nanoengineered metals and their oxide nanoparticles are targeted at the mitochondrial architecture that partakes in the management of a myriad of health issues, including different cancers. These nanosystems not only act as antioxidants but are also fabricated for the delivery of chemotherapeutic agents. However, the biocompatibility, safety, and efficacy of using metal nanoparticles is contested among researchers, which will be discussed further in this review.
Collapse
Affiliation(s)
- Shashi Kiran Misra
- School of Pharmaceutical Sciences, CSJM University Kanpur, Kanpur 208024, India
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity (3rd Floor), Tykistökatu, 6A, 20520 Turku, Finland
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India
| |
Collapse
|
34
|
Xenotopic expression of alternative oxidase (AOX) to study mechanisms of mitochondrial disease. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2023; 1864:148947. [PMID: 36481273 DOI: 10.1016/j.bbabio.2022.148947] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
The mitochondrial respiratory chain or electron transport chain (ETC) facilitates redox reactions which ultimately lead to the reduction of oxygen to water (respiration). Energy released by this process is used to establish a proton electrochemical gradient which drives ATP formation (oxidative phosphorylation, OXPHOS). It also plays an important role in vital processes beyond ATP formation and cellular metabolism, such as heat production, redox and ion homeostasis. Dysfunction of the ETC can thus impair cellular and organismal viability and is thought to be the underlying cause of a heterogeneous group of so-called mitochondrial diseases. Plants, yeasts, and many lower organisms, but not insects and vertebrates, possess an enzymatic mechanism that confers resistance to respiratory stress conditions, i.e., the alternative oxidase (AOX). Even in cells that naturally lack AOX, it is autonomously imported into the mitochondrial compartment upon xenotopic expression, where it refolds and becomes catalytically engaged when the cytochrome segment of the ETC is blocked. AOX was therefore proposed as a tool to study disease etiologies. To this end, AOX has been xenotopically expressed in mammalian cells and disease models of the fruit fly and mouse. Surprisingly, AOX showed remarkable rescue effects in some cases, whilst in others it had no effect or even exacerbated a condition. Here we summarize what has been learnt from the use of AOX in various disease models and discuss issues which still need to be addressed in order to understand the role of the ETC in health and disease.
Collapse
|
35
|
Seminotti B, Grings M, Glänzel NM, Vockley J, Leipnitz G. Peroxisome proliferator-activated receptor (PPAR) agonists as a potential therapy for inherited metabolic disorders. Biochem Pharmacol 2023; 209:115433. [PMID: 36709926 DOI: 10.1016/j.bcp.2023.115433] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023]
Abstract
Inherited metabolic disorders (IMDs) are genetic disorders that cause a disruption of a specific metabolic pathway leading to biochemical, clinical and pathophysiological sequelae. While the metabolite abnormalities in body fluids and tissues can usually be defined by directed or broad-spectrum metabolomic analysis, the pathophysiology of these changes is often not obvious. Mounting evidence has revealed that secondary mitochondrial dysfunction, mainly oxidative phosphorylation impairment and elevated reactive oxygen species, plays a pivotal role in many disorders. Peroxisomal proliferator-activated receptors (PPARs) consist of a group of nuclear hormone receptors (PPARα, PPARβ/δ, and PPARγ) that regulate multiple cellular functions and processes, including response to oxidative stress, inflammation, lipid metabolism, and mitochondrial bioenergetics and biogenesis. In this context, the activation of PPARs has been shown to stimulate oxidative phosphorylation and reduce reactive species levels. Thus, pharmacological treatment with PPAR activators, such as fibrates, has gained much attention in the last 15 years. This review summarizes preclinical (animal models and patient-derived cells) and clinical data on the effect of PPARs in IMDs.
Collapse
Affiliation(s)
- Bianca Seminotti
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil; Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mateus Grings
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Nícolas Manzke Glänzel
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Jerry Vockley
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, CEP 90035-190, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, Porto Alegre, RS, Brazil.
| |
Collapse
|
36
|
Karaa A, Klopstock T. Clinical trials in mitochondrial diseases. HANDBOOK OF CLINICAL NEUROLOGY 2023; 194:229-250. [PMID: 36813315 DOI: 10.1016/b978-0-12-821751-1.00002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Primary mitochondrial diseases are some of the most common and complex inherited inborn errors of metabolism. Their molecular and phenotypic diversity has led to difficulties in finding disease-modifying therapies and clinical trial efforts have been slow due to multiple significant challenges. Lack of robust natural history data, difficulties in finding specific biomarkers, absence of well-validated outcome measures, and small patient numbers have made clinical trial design and conduct difficult. Encouragingly, new interest in treating mitochondrial dysfunction in common diseases and regulatory incentives to develop therapies for rare conditions have led to significant interest and efforts to develop drugs for primary mitochondrial diseases. Here, we review past and present clinical trials and future strategies of drug development in primary mitochondrial diseases.
Collapse
Affiliation(s)
- Amel Karaa
- Mitochondrial Disease Program, Division of Medical Genetics and Metabolism, Massachusetts General Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institute, University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Network for mitochondrial disorders (mitoNET), Munich, Germany
| |
Collapse
|
37
|
Viscomi C, Zeviani M. Experimental therapy for mitochondrial diseases. HANDBOOK OF CLINICAL NEUROLOGY 2023; 194:259-277. [PMID: 36813318 DOI: 10.1016/b978-0-12-821751-1.00013-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Mitochondrial diseases are extremely heterogeneous genetic disorders due to faulty oxidative phosphorylation (OxPhos). No cure is currently available for these conditions, beside supportive interventions aimed at relieving complications. Mitochondria are under a double genetic control carried out by the mitochondrial DNA (mtDNA) and by nuclear DNA. Thus, not surprisingly, mutations in either genome can cause mitochondrial disease. Although mitochondria are usually associated with respiration and ATP synthesis, they play fundamental roles in a large number of other biochemical, signaling, and execution pathways, each being a potential target for therapeutic interventions. These can be classified as general therapies, i.e., potentially applicable to a number of different mitochondrial conditions, or therapies tailored to a single disease, i.e., personalized approaches, such as gene therapy, cell therapy, and organ replacement. Mitochondrial medicine is a particularly lively research field, and the last few years witnessed a steady increase in the number of clinical applications. This chapter will present the most recent therapeutic attempts emerged from preclinical work and an update of the currently ongoing clinical applications. We think that we are starting a new era in which the etiologic treatment of these conditions is becoming a realistic option.
Collapse
Affiliation(s)
- Carlo Viscomi
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | - Massimo Zeviani
- Department of Neurosciences, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
38
|
Chen Z, Bordieanu B, Kesavan R, Lesner NP, Venigalla SSK, Shelton SD, DeBerardinis RJ, Mishra P. Lactate metabolism is essential in early-onset mitochondrial myopathy. SCIENCE ADVANCES 2023; 9:eadd3216. [PMID: 36598990 PMCID: PMC9812384 DOI: 10.1126/sciadv.add3216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/30/2022] [Indexed: 06/17/2023]
Abstract
Myopathies secondary to mitochondrial electron transport chain (ETC) dysfunction can result in devastating disease. While the consequences of ETC defects have been extensively studied in culture, little in vivo data are available. Using a mouse model of severe, early-onset mitochondrial myopathy, we characterized the proteomic, transcriptomic, and metabolic characteristics of disease progression. Unexpectedly, ETC dysfunction in muscle results in reduced expression of glycolytic enzymes in our animal model and patient muscle biopsies. The decrease in glycolysis was mediated by loss of constitutive Hif1α signaling, down-regulation of the purine nucleotide cycle enzyme AMPD1, and activation of AMPK. In vivo isotope tracing experiments indicated that myopathic muscle relies on lactate import to supply central carbon metabolites. Inhibition of lactate import reduced steady-state levels of tricarboxylic acid cycle intermediates and compromised the life span of myopathic mice. These data indicate an unexpected mode of metabolic reprogramming in severe mitochondrial myopathy that regulates disease progression.
Collapse
Affiliation(s)
- Zhenkang Chen
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bogdan Bordieanu
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rushendhiran Kesavan
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nicholas P. Lesner
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Siva Sai Krishna Venigalla
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Spencer D. Shelton
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Prashant Mishra
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
39
|
Mowat FM. Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1Alpha (PGC-1α): A Transcriptional Regulator at the Interface of Aging and Age-Related Macular Degeneration? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:49-53. [PMID: 37440013 DOI: 10.1007/978-3-031-27681-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Human age-related macular degeneration (AMD) is a prevalent age-related disease which causes retinal dysfunction and disability. Genetic and cell culture studies from AMD patients have implicated impaired activity of peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1α). PGC-1α is a transcriptional co-regulator that acts to control a plethora of metabolic processes relevant to AMD pathophysiology including gluconeogenesis, oxidative phosphorylation, and response to oxidative injury. Perturbation of PGC-1α activity in mice causes AMD-like RPE and retinal pathology. There is potential for therapeutic modulation of the PGC-1α pathway in AMD treatment.
Collapse
Affiliation(s)
- Freya M Mowat
- Department of Ophthalmology and Visual Sciences (School of Medicine and Public Health), Department of Surgical Sciences (School of Veterinary Medicine), Medical Sciences Center, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
40
|
Pedriali G, Ramaccini D, Bouhamida E, Wieckowski MR, Giorgi C, Tremoli E, Pinton P. Perspectives on mitochondrial relevance in cardiac ischemia/reperfusion injury. Front Cell Dev Biol 2022; 10:1082095. [PMID: 36561366 PMCID: PMC9763599 DOI: 10.3389/fcell.2022.1082095] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the most common cause of death worldwide and in particular, ischemic heart disease holds the most considerable position. Even if it has been deeply studied, myocardial ischemia-reperfusion injury (IRI) is still a side-effect of the clinical treatment for several heart diseases: ischemia process itself leads to temporary damage to heart tissue and obviously the recovery of blood flow is promptly required even if it worsens the ischemic injury. There is no doubt that mitochondria play a key role in pathogenesis of IRI: dysfunctions of these important organelles alter cell homeostasis and survival. It has been demonstrated that during IRI the system of mitochondrial quality control undergoes alterations with the disruption of the complex balance between the processes of mitochondrial fusion, fission, biogenesis and mitophagy. The fundamental role of mitochondria is carried out thanks to the finely regulated connection to other organelles such as plasma membrane, endoplasmic reticulum and nucleus, therefore impairments of these inter-organelle communications exacerbate IRI. This review pointed to enhance the importance of the mitochondrial network in the pathogenesis of IRI with the aim to focus on potential mitochondria-targeting therapies as new approach to control heart tissue damage after ischemia and reperfusion process.
Collapse
Affiliation(s)
- Gaia Pedriali
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | | | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| |
Collapse
|
41
|
Bennett CF, Ronayne CT, Puigserver P. Targeting adaptive cellular responses to mitochondrial bioenergetic deficiencies in human disease. FEBS J 2022; 289:6969-6993. [PMID: 34510753 PMCID: PMC8917243 DOI: 10.1111/febs.16195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/25/2021] [Accepted: 09/10/2021] [Indexed: 01/13/2023]
Abstract
Mitochondrial dysfunction is increasingly appreciated as a central contributor to human disease. Oxidative metabolism at the mitochondrial respiratory chain produces ATP and is intricately tied to redox homeostasis and biosynthetic pathways. Metabolic stress arising from genetic mutations in mitochondrial genes and environmental factors such as malnutrition or overnutrition is perceived by the cell and leads to adaptive and maladaptive responses that can underlie pathology. Here, we will outline cellular sensors that react to alterations in energy production, organellar redox, and metabolites stemming from mitochondrial disease (MD) mutations. MD is a heterogeneous group of disorders primarily defined by defects in mitochondrial oxidative phosphorylation from nuclear or mitochondrial-encoded gene mutations. Preclinical therapies that improve fitness of MD mouse models have been recently identified. Targeting metabolic/energetic deficiencies, maladaptive signaling processes, and hyper-oxygenation of tissues are all strategies aside from direct genetic approaches that hold therapeutic promise. A further mechanistic understanding of these curative processes as well as the identification of novel targets will significantly impact mitochondrial biology and disease research.
Collapse
Affiliation(s)
- Christopher F Bennett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Conor T Ronayne
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Li H, Zhang L, Zhang L, Han R. Autophagy in striated muscle diseases. Front Cardiovasc Med 2022; 9:1000067. [PMID: 36312227 PMCID: PMC9606591 DOI: 10.3389/fcvm.2022.1000067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Impaired biomolecules and cellular organelles are gradually built up during the development and aging of organisms, and this deteriorating process is expedited under stress conditions. As a major lysosome-mediated catabolic process, autophagy has evolved to eradicate these damaged cellular components and recycle nutrients to restore cellular homeostasis and fitness. The autophagic activities are altered under various disease conditions such as ischemia-reperfusion cardiac injury, sarcopenia, and genetic myopathies, which impact multiple cellular processes related to cellular growth and survival in cardiac and skeletal muscles. Thus, autophagy has been the focus for therapeutic development to treat these muscle diseases. To develop the specific and effective interventions targeting autophagy, it is essential to understand the molecular mechanisms by which autophagy is altered in heart and skeletal muscle disorders. Herein, we summarize how autophagy alterations are linked to cardiac and skeletal muscle defects and how these alterations occur. We further discuss potential pharmacological and genetic interventions to regulate autophagy activities and their applications in cardiac and skeletal muscle diseases.
Collapse
Affiliation(s)
- Haiwen Li
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Lei Zhang
- Department of Anatomy and Neurobiology, Shanghai Yangzhi Rehabilitation Hospital, Shanghai Sunshine Rehabilitation Center, School of Medicine, Tongji University, Shanghai, China
| | - Renzhi Han
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
43
|
Bajia D, Bottani E, Derwich K. Effects of Noonan Syndrome-Germline Mutations on Mitochondria and Energy Metabolism. Cells 2022; 11:cells11193099. [PMID: 36231062 PMCID: PMC9563972 DOI: 10.3390/cells11193099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Noonan syndrome (NS) and related Noonan syndrome with multiple lentigines (NSML) contribute to the pathogenesis of human diseases in the RASopathy family. This family of genetic disorders constitute one of the largest groups of developmental disorders with variable penetrance and severity, associated with distinctive congenital disabilities, including facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was first clinically described decades ago, and several genes have since been identified, providing a molecular foundation to understand their physiopathology and identify targets for therapeutic strategies. These genes encode proteins that participate in, or regulate, RAS/MAPK signalling. The RAS pathway regulates cellular metabolism by controlling mitochondrial homeostasis, dynamics, and energy production; however, little is known about the role of mitochondrial metabolism in NS and NSML. This manuscript comprehensively reviews the most frequently mutated genes responsible for NS and NSML, covering their role in the current knowledge of cellular signalling pathways, and focuses on the pathophysiological outcomes on mitochondria and energy metabolism.
Collapse
Affiliation(s)
- Donald Bajia
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
| | - Emanuela Bottani
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Piazzale L. A. Scuro 10, 37134 Verona, Italy
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| | - Katarzyna Derwich
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| |
Collapse
|
44
|
Wu YX, Yang XY, Han BS, Hu YY, An T, Lv BH, Lian J, Wang TY, Bao XL, Gao L, Jiang GJ. Naringenin regulates gut microbiota and SIRT1/ PGC-1ɑ signaling pathway in rats with letrozole-induced polycystic ovary syndrome. Biomed Pharmacother 2022; 153:113286. [PMID: 35724506 DOI: 10.1016/j.biopha.2022.113286] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/31/2022] [Accepted: 06/09/2022] [Indexed: 11/02/2022] Open
Abstract
PURPOSE To evaluate the effect of naringenin on improving PCOS and explore the mechanism. METHODS Firstly, we carried out differential gene expression analysis from transcriptome sequencing data of human oocyte to screen the KEGG pathway, then the PCOS-like rat model was induced by letrozole. They were randomly divided into four groups: Normal group (N), PCOS group (P), Diane-35 group (D), and Naringenin group (Nar). The changes of estrus cycle, body weight, ovarian function, serum hormone levels, glucose metabolism, along with the expression of SIRT1, PGC-1ɑ, claudin-1 and occludin of the ovary and colon were investigated. Furthermore, the composition of the gut microbiome of fecal was tested. RESULTS By searching the KEGG pathway in target genes, we found that at least 15 KEGG pathways are significantly enriched in the ovarian function, such as AMPK signaling pathway, insulin secretion, and ovarian steroidogenesis. Interestingly, naringenin supplementation significantly reduced body weight, ameliorated hormone levels, improved insulin resistance, and mitigated pathological changes in ovarian tissue, up-regulated the expression of PGC-1ɑ, SIRT1, occludin and claudin-1 in colon. In addition, we also found that the abundance of Prevotella and Gemella was down-regulated, while the abundance of Butyricimonas, Lachnospira, Parabacteroides, Butyricicoccus, Streptococcus, Coprococcus was up-regulated. CONCLUSION Our data suggest that naringenin exerts a treatment PCOS effect, which may be related to the modulation of the gut microbiota and SIRT1/PGC-1ɑ signaling pathway. Our research may provide a new perspective for the treatment of PCOS and related diseases.
Collapse
Affiliation(s)
- Yan-Xiang Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiu-Yan Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bao-Sheng Han
- Maternity and Child Care Hospital of North China University of Science and Technology, Tangshan, China
| | - Yuan-Yuan Hu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tian An
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bo-Han Lv
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Juan Lian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting-Ye Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xue-Li Bao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lin Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Guang-Jian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
45
|
Dogan SA, Giacchin G, Zito E, Viscomi C. Redox Signaling and Stress in Inherited Myopathies. Antioxid Redox Signal 2022; 37:301-323. [PMID: 35081731 DOI: 10.1089/ars.2021.0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Reactive oxygen species (ROS) are highly reactive compounds that behave like a double-edged sword; they damage cellular structures and act as second messengers in signal transduction. Mitochondria and endoplasmic reticulum (ER) are interconnected organelles with a central role in ROS production, detoxification, and oxidative stress response. Skeletal muscle is the most abundant tissue in mammals and one of the most metabolically active ones and thus relies mainly on oxidative phosphorylation (OxPhos) to synthesize adenosine triphosphate. The impairment of OxPhos leads to myopathy and increased ROS production, thus affecting both redox poise and signaling. In addition, ROS enter the ER and trigger ER stress and its maladaptive response, which also lead to a myopathic phenotype with mitochondrial involvement. Here, we review the role of ROS signaling in myopathies due to either mitochondrial or ER dysfunction. Recent Advances: Relevant advances have been evolving over the last 10 years on the intricate ROS-dependent pathways that act as modifiers of the disease course in several myopathies. To this end, pathways related to mitochondrial biogenesis, satellite cell differentiation, and ER stress have been studied extensively in myopathies. Critical Issues: The analysis of the chemistry and the exact quantitation, as well as the localization of ROS, are still challenging due to the intrinsic labile nature of ROS and the technical limitations of their sensors. Future Directions: The mechanistic studies of the pathogenesis of mitochondrial and ER-related myopathies offer a unique possibility to discover novel ROS-dependent pathways. Antioxid. Redox Signal. 37, 301-323.
Collapse
Affiliation(s)
- Sukru Anil Dogan
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Istanbul, Turkey
| | - Giacomo Giacchin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ester Zito
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.,Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carlo Viscomi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
46
|
Adipocyte-Specific Expression of PGC1α Promotes Adipocyte Browning and Alleviates Obesity-Induced Metabolic Dysfunction in an HO-1-Dependent Fashion. Antioxidants (Basel) 2022; 11:antiox11061147. [PMID: 35740043 PMCID: PMC9220759 DOI: 10.3390/antiox11061147] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/11/2022] Open
Abstract
Recent studies suggest that PGC1-α plays a crucial role in mitochondrial and vascular function, yet the physiological significance of PGC1α and HO expression in adipose tissues in the context of obesity-linked vascular dysfunction remains unclear. We studied three groups of six-week-old C57BL/6J male mice: (1) mice fed a normal chow diet; (2) mice fed a high-fat diet (H.F.D.) for 28 weeks, and (3) mice fed a high-fat diet (H.F.D.) for 28 weeks, treated with adipose-specific overexpression of PGC-1α (transgenic-adipocyte-PGC-1α) at week 20, and continued on H.F.D. for weeks 20–28. R.N.A. arrays examined 88 genes involved in adipocyte proliferation and maturation. Blood pressure, tissue fibrosis, fasting glucose, and oxygen consumption were measured, as well as liver steatosis, and the expression levels of metabolic and mitochondrial markers. Obese mice exhibited a marked reduction of PGC1α and developed adipocyte hypertrophy, fibrosis, hepatic steatosis, and decreased mitochondrial respiration. Mice with adipose-specific overexpression of PGC1-α exhibited improvement in HO-1, mitochondrial biogenesis and respiration, with a decrease in fasting glucose, reduced blood pressure and fibrosis, and increased oxygen consumption. PGC-1α led to the upregulated expression of processes associated with the browning of fat tissue, including UCP1, FGF21, and pAMPK signaling, with a reduction in inflammatory adipokines, NOV/CCN3 expression, and TGFβ. These changes required HO-1 expression. The R.N.A. array analysis identified subgroups of genes positively correlated with contributions to the browning of adipose tissue, all dependent on HO-1. Our observations reveal a positive impact of adipose-PGC1-α on distal organ systems, with beneficial effects on HO-1 levels, reversing obesity-linked cardiometabolic disturbances.
Collapse
|
47
|
Roy A, Kandettu A, Ray S, Chakrabarty S. Mitochondrial DNA replication and repair defects: Clinical phenotypes and therapeutic interventions. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148554. [PMID: 35341749 DOI: 10.1016/j.bbabio.2022.148554] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/06/2022] [Accepted: 03/16/2022] [Indexed: 12/15/2022]
Abstract
Mitochondria is a unique cellular organelle involved in multiple cellular processes and is critical for maintaining cellular homeostasis. This semi-autonomous organelle contains its circular genome - mtDNA (mitochondrial DNA), that undergoes continuous cycles of replication and repair to maintain the mitochondrial genome integrity. The majority of the mitochondrial genes, including mitochondrial replisome and repair genes, are nuclear-encoded. Although the repair machinery of mitochondria is quite efficient, the mitochondrial genome is highly susceptible to oxidative damage and other types of exogenous and endogenous agent-induced DNA damage, due to the absence of protective histones and their proximity to the main ROS production sites. Mutations in replication and repair genes of mitochondria can result in mtDNA depletion and deletions subsequently leading to mitochondrial genome instability. The combined action of mutations and deletions can result in compromised mitochondrial genome maintenance and lead to various mitochondrial disorders. Here, we review the mechanism of mitochondrial DNA replication and repair process, key proteins involved, and their altered function in mitochondrial disorders. The focus of this review will be on the key genes of mitochondrial DNA replication and repair machinery and the clinical phenotypes associated with mutations in these genes.
Collapse
Affiliation(s)
- Abhipsa Roy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Amoolya Kandettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Swagat Ray
- Department of Life Sciences, School of Life and Environmental Sciences, University of Lincoln, Lincoln LN6 7TS, United Kingdom
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
48
|
da Rosa-Junior NT, Parmeggiani B, Glänzel NM, de Moura Alvorcem L, Brondani M, Britto R, Grings M, Ortiz VD, Turck P, da Rosa Araujo AS, Wajner M, Leipnitz G. Antioxidant system disturbances and mitochondrial dysfunction induced by 3-methyglutaric acid in rat heart are prevented by bezafibrate. Eur J Pharmacol 2022; 924:174950. [DOI: 10.1016/j.ejphar.2022.174950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 03/25/2022] [Accepted: 04/05/2022] [Indexed: 11/30/2022]
|
49
|
Lyu J, Zhao Y, Zhang N, Xu X, Zheng R, Yu W, Xin W, Yan C, Ji K. Bezafibrate Rescues Mitochondrial Encephalopathy in Mice via Induction of Daily Torpor and Hypometabolic State. Neurotherapeutics 2022; 19:994-1006. [PMID: 35334081 PMCID: PMC9294104 DOI: 10.1007/s13311-022-01216-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 02/07/2023] Open
Abstract
Leigh syndrome (LS) is one of the most common mitochondrial encephalopathy diseases in infants. To date, there is still an absence of effective therapy. Bezafibrate (BEZ), a pan-peroxisome proliferator-activated receptor (PPAR) agonist, ameliorates the phenotype of the mouse model of mitochondrial disease via an unclear mechanism. Here, we applied it to Ndufs4 knockout (KO) mice, a widely used LS animal model, to observe the therapeutic effects and metabolic changes associated with BEZ treatment to explore the therapeutic strategies for mitochondrial diseases. Administration of BEZ significantly enhances survival and attenuates disease progression in Ndufs4 KO mice. Decreased oxidative stress and stunted growth were also observed. As a PPAR agonist, we did not find mitochondrial biogenesis or enhanced metabolism upon BEZ treatment. On the contrary, mice with dietary BEZ showed daily torpor bouts and lower metabolic rates. We speculate that activating energy-saving metabolism in mice may be associated with the therapeutic effects of BEZ, but the exact mechanism of action requires further study.
Collapse
Affiliation(s)
- Jingwei Lyu
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yuying Zhao
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
- Department of Neurology, Qilu Hospital of Shandong University, No.107 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Na Zhang
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Xuebi Xu
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Nanbaixiang Street, Wenzhou, 325000, China
| | - Rui Zheng
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
- Department of Neurology Qilu Hospital, Qingdao of Shandong University, Qingdao, 266035, Shandong, China
| | - Wenfei Yu
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Wang Xin
- College of Chemistry, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Chuanzhu Yan
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
- Department of Neurology, Qilu Hospital of Shandong University, No.107 West Wenhua Road, Jinan, 250012, Shandong, China
- Department of Neurology Qilu Hospital, Qingdao of Shandong University, Qingdao, 266035, Shandong, China
- Brain Science Research Institute, Shandong University, Jinan, 250012, Shandong, China
| | - Kunqian Ji
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China.
- Department of Neurology, Qilu Hospital of Shandong University, No.107 West Wenhua Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
50
|
Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, Sinclair DA. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol 2022; 18:243-258. [PMID: 35145250 PMCID: PMC9059418 DOI: 10.1038/s41574-021-00626-7] [Citation(s) in RCA: 452] [Impact Index Per Article: 150.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/11/2022]
Abstract
Organismal ageing is accompanied by progressive loss of cellular function and systemic deterioration of multiple tissues, leading to impaired function and increased vulnerability to death. Mitochondria have become recognized not merely as being energy suppliers but also as having an essential role in the development of diseases associated with ageing, such as neurodegenerative and cardiovascular diseases. A growing body of evidence suggests that ageing and age-related diseases are tightly related to an energy supply and demand imbalance, which might be alleviated by a variety of interventions, including physical activity and calorie restriction, as well as naturally occurring molecules targeting conserved longevity pathways. Here, we review key historical advances and progress from the past few years in our understanding of the role of mitochondria in ageing and age-related metabolic diseases. We also highlight emerging scientific innovations using mitochondria-targeted therapeutic approaches.
Collapse
Affiliation(s)
- João A Amorim
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- IIIUC, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Giuseppe Coppotelli
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- George and Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Anabela P Rolo
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- Department of Life Sciences of the University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- Department of Life Sciences of the University of Coimbra, Coimbra, Portugal
| | - Jaime M Ross
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- George and Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - David A Sinclair
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|