1
|
Fang M, Yang S, Liu R, Wu X, Jiang L, Yang J, Liu X, Wang G, Mu C, Wang X, Song Y. DUT (p.Y116C)-Mutation-Induced Thrombocytopenia in Rabbits. Int J Mol Sci 2025; 26:4169. [PMID: 40362420 PMCID: PMC12072063 DOI: 10.3390/ijms26094169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Thrombocytopenia is a hematologic disorder characterized by an abnormally low platelet count in peripheral blood. Recent studies have identified mutations in DUT as the primary cause of bone marrow failure and diabetes mellitus syndrome (BMFDMS), a condition commonly associated with thrombocytopenia. In this study, a novel rabbit model of thrombocytopenia carrying the DUT c.3020A>G (p.Y116C) mutation was established using SpRY-ABEmax-mediated base editing. This model accurately recapitulates the clinical manifestations of human thrombocytopenia. Phenotypic analysis has revealed that mutant rabbits exhibited significant reductions in megakaryocyte numbers, platelet counts, and survival rates when compared to wild-type controls. Mechanistic investigations showed that the DUT mutation leads to mitochondrial structural abnormalities and functional impairments. Notably, platelets from DUT (p.Y116C)-mutant rabbits exhibited markedly reduced DUT protein expression and enhanced mitophagy, potentially mediated through the Park2 pathway. This study presents the first genetic model of thrombocytopenia that closely mimics the human DUT (p.Y116C) mutation, offering new insights into the relationship between DUT mutations and platelet function, and highlighting potential therapeutic targets for human thrombocytopenia.
Collapse
Affiliation(s)
- Mengmeng Fang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Shujun Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Ruonan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Xinyu Wu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Liqiang Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Jie Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Xin Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Gerong Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Chao Mu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| | - Xiuwen Wang
- College of Life Sciences, Jilin Normal University, No. 1301, Hai Feng Street, Tiexi District, Siping 136000, China;
| | - Yuning Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.F.); (S.Y.); (R.L.); (X.W.); (L.J.); (J.Y.); (X.L.); (G.W.); (C.M.)
| |
Collapse
|
2
|
Li JY, Chen HK, Huang YH, Zhi YP, Li YE, Lin KY, Chen C, Guo YS. Identification of mitophagy-related genes in patients with acute myocardial infarction. Hereditas 2025; 162:70. [PMID: 40287718 PMCID: PMC12034215 DOI: 10.1186/s41065-025-00424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Mitophagy is involved in acute myocardial infarction (AMI) process. However, the role of mitophagy-related genes (MRGs) in the AMI process is not well illustrated. We identified MRGs involved in AMI by bioinformatics analysis. The external datasets were employed for the validation of the MRGs, alongside the execution of cellular and animal experiments. Forty-five MRGs were detected, and machine learning identified the top four hub genes, namely ALDH2, ACSL1, IL1B, and GABARAPL1. Additionally, an external validation set was used to screen for three diagnostic markers (ACSL1, IL1B, and GABARAPL1) among these hub genes. Immune infiltration analysis revealed changes in the immune microenvironment among patients with AMI. Finally, the significant upregulation of ACSL1, IL1B, and GABARAPL1 in both cellular and animal models was confirmed. The occurrence of mitophagy was observed in the cell model through transmission electron microscopy (TEM). Our study demonstrated that ACSL1, IL1B, and GABARAPL1 possess potential biomarkers for AMI.
Collapse
Affiliation(s)
- Ju-Ying Li
- The First People's Hospital of Yibin, Yibin, Sichuan Province, 644000, China
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Hong-Kui Chen
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Yi-Hao Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Yu-Peng Zhi
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Yue-E Li
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Kai-Yang Lin
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Chun Chen
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China.
| | - Yan-Song Guo
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China.
| |
Collapse
|
3
|
Zhao K, Chan ITC, Tse EHY, Xie Z, Cheung TH, Zeng YA. Autophagy in adult stem cell homeostasis, aging, and disease therapy. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:14. [PMID: 40208372 PMCID: PMC11985830 DOI: 10.1186/s13619-025-00224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/16/2025] [Accepted: 02/23/2025] [Indexed: 04/11/2025]
Abstract
Autophagy is a crucial cellular process that facilitates the degradation of damaged organelles and protein aggregates, and the recycling of cellular components for the energy production and macromolecule synthesis. It plays an indispensable role in maintaining cellular homeostasis. Over recent decades, research has increasingly focused on the role of autophagy in regulating adult stem cells (SCs). Studies suggest that autophagy modulates various cellular processes and states of adult SCs, including quiescence, proliferation, self-renewal, and differentiation. The primary role of autophagy in these contexts is to sustain homeostasis, withstand stressors, and supply energy. Notably, the dysfunction of adult SCs during aging is correlated with a decline in autophagic activity, suggesting that autophagy is also involved in SC- and aging-associated disorders. Given the diverse cellular processes mediated by autophagy and the intricate mechanisms governing adult SCs, further research is essential to elucidate both universal and cell type-specific regulatory pathways of autophagy. This review discusses the role of autophagy in regulating adult SCs during quiescence, proliferation, self-renewal, and differentiation. Additionally, it summarizes the relationship between SC aging and autophagy, providing therapeutical insights into treating and ameliorating aging-associated diseases and cancers, and ultimately promoting longevity.
Collapse
Affiliation(s)
- Ke Zhao
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Indigo T C Chan
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, HKUST-Nan Fung Life Sciences Joint Laboratory, the Hong Kong University of Science and Technology, Hong Kong, China
| | - Erin H Y Tse
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, HKUST-Nan Fung Life Sciences Joint Laboratory, the Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Zhiyao Xie
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, HKUST-Nan Fung Life Sciences Joint Laboratory, the Hong Kong University of Science and Technology, Hong Kong, China.
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China.
| | - Yi Arial Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
4
|
Balachandar Thendral S, Bacot S, Morton KS, Chi Q, Kenny-Ganzert IW, Meyer JN, Sherwood DR. Programmed mitophagy at the oocyte-to-zygote transition promotes species immortality. RESEARCH SQUARE 2025:rs.3.rs-6330979. [PMID: 40297685 PMCID: PMC12036463 DOI: 10.21203/rs.3.rs-6330979/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The quality of mitochondria inherited from the oocyte determines embryonic viability, metabolic health throughout progeny lifetime, and future generation endurance. High levels of endogenous reactive oxygen species and exogenous toxicants are threats to mitochondrial DNA (mtDNA) in fully developed oocytes. Deleterious mtDNA is commonly detected in developed oocytes, but is absent in embryos, suggesting the existence of a cryptic purifying selection mechanism. Here we discover that in C. elegans, the onset of oocyte-to-zygote transition (OZT) developmentally triggers a rapid mitophagy event. We show that mitophagy at OZT (MOZT) requires mitochondrial fragmentation, the macroautophagy pathway, and the mitophagy receptor FUNDC1, but not the prevalent mitophagy factors PINK1 and BNIP3. Impaired MOZT leads to increased deleterious mtDNA inheritance and decreases embryonic survival. Inherited mtDNA damage accumulates across generations, leading to the extinction of descendent populations. Thus, MOZT represents a strategy that preserves mitochondrial health during the mother-to-offspring transmission and promotes species continuity.
Collapse
Affiliation(s)
| | - Sasha Bacot
- Nicholas School of the Environment, Duke University, Durham, NC 27710, USA
| | | | - Qiuyi Chi
- Department of Biology, Duke University, Durham, NC 27710, USA
| | | | - Joel N. Meyer
- Nicholas School of the Environment, Duke University, Durham, NC 27710, USA
| | | |
Collapse
|
5
|
Stefanatos R, Robertson F, Castejon-Vega B, Yu Y, Uribe AH, Myers K, Kataura T, Korolchuk VI, Maddocks ODK, Martins LM, Sanz A. Developmental mitochondrial complex I activity determines lifespan. EMBO Rep 2025; 26:1957-1983. [PMID: 40097814 PMCID: PMC12019323 DOI: 10.1038/s44319-025-00416-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Aberrant mitochondrial function has been associated with an increasingly large number of human disease states. Observations from in vivo models where mitochondrial function is altered suggest that maladaptations to mitochondrial dysfunction may underpin disease pathology. We hypothesized that the severity of this maladaptation could be shaped by the plasticity of the system when mitochondrial dysfunction manifests. To investigate this, we have used inducible fly models of mitochondrial complex I (CI) dysfunction to reduce mitochondrial function at two stages of the fly lifecycle, from early development and adult eclosion. Here, we show that in early life (developmental) mitochondrial dysfunction results in severe reductions in survival and stress resistance in adulthood, while flies where mitochondrial function is perturbed from adulthood, are long-lived and stress resistant despite having up to a 75% reduction in CI activity. After excluding developmental defects as a cause, we went on to molecularly characterize these two populations of mitochondrially compromised flies, short- and long-lived. We find that our short-lived flies have unique transcriptomic, proteomic and metabolomic responses, which overlap significantly in discrete models of CI dysfunction. Our data demonstrate that early mitochondrial dysfunction via CI depletion elicits a maladaptive response, which severely reduces survival, while CI depletion from adulthood is insufficient to reduce survival and stress resistance.
Collapse
Affiliation(s)
- Rhoda Stefanatos
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, NE4 5PL, Newcastle upon Tyne, UK.
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, NE2 4HH, Newcastle upon Tyne, UK.
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, Glasgow, UK.
| | - Fiona Robertson
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, Glasgow, UK
| | - Beatriz Castejon-Vega
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, Glasgow, UK
| | - Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, CB2 1QR, Cambridge, UK
| | - Alejandro Huerta Uribe
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, G61 1QH, Glasgow, UK
| | - Kevin Myers
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, Glasgow, UK
| | - Tetsushi Kataura
- Department of Neurology, Institute of Medicine, University of Tsukuba, 305-8575, Ibaraki, Japan
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, NE4 5PL, Newcastle upon Tyne, UK
| | - Oliver D K Maddocks
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, G61 1QH, Glasgow, UK
| | - L Miguel Martins
- MRC Toxicology Unit, University of Cambridge, CB2 1QR, Cambridge, UK
| | - Alberto Sanz
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, G12 8QQ, Glasgow, UK.
| |
Collapse
|
6
|
Zhang X, Xiao J, Jiang M, Phillips CJC, Shi B. Thermogenesis and Energy Metabolism in Brown Adipose Tissue in Animals Experiencing Cold Stress. Int J Mol Sci 2025; 26:3233. [PMID: 40244078 PMCID: PMC11989373 DOI: 10.3390/ijms26073233] [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: 03/01/2025] [Revised: 03/29/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Cold exposure is a regulatory biological functions in animals. The interaction of thermogenesis and energy metabolism in brown adipose tissue (BAT) is important for metabolic regulation in cold stress. Brown adipocytes (BAs) produce uncoupling protein 1 (UCP1) in mitochondria, activating non-shivering thermogenesis (NST) by uncoupling fuel combustion from ATP production in response to cold stimuli. To elucidate the mechanisms underlying thermogenesis and energy metabolism in BAT under cold stress, we explored how cold exposure triggers the activation of BAT thermogenesis and regulates overall energy metabolism. First, we briefly outline the precursor composition and function of BA. Second, we explore the roles of the cAMP- protein kinase A (PKA) and adenosine monophosphate-activated protein kinase (AMPK) signaling pathways in thermogenesis and energy metabolism in BA during cold stress. Then, we analyze the mechanism by which BA regulates mitochondria homeostasis and energy balance during cold stress. This research reveals potential therapeutic targets, such as PKA, AMPK, UCP1 and PGC-1α, which can be used to develop innovative strategies for treating metabolic diseases. Furthermore, it provides theoretical support for optimizing cold stress response strategies, including the pharmacological activation of BAT and the genetic modulation of thermogenic pathways, to improve energy homeostasis in livestock.
Collapse
Affiliation(s)
- Xuekai Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| | - Jin Xiao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| | - Min Jiang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| | - Clive J. C. Phillips
- Curtin University Sustainability Policy (CUSP) Institute, Curtin University, Perth, WA 6845, Australia;
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51006 Tartu, Estonia
| | - Binlin Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (X.Z.); (M.J.); (B.S.)
| |
Collapse
|
7
|
Santos F, Sum H, Yan DCL, Brewer AC. Metaboloepigenetics: Role in the Regulation of Flow-Mediated Endothelial (Dys)Function and Atherosclerosis. Cells 2025; 14:378. [PMID: 40072106 PMCID: PMC11898952 DOI: 10.3390/cells14050378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Endothelial dysfunction is the main initiating factor in atherosclerosis. Through mechanotransduction, shear stress regulates endothelial cell function in both homeostatic and diseased states. Accumulating evidence reveals that epigenetic changes play critical roles in the etiology of cardiovascular diseases, including atherosclerosis. The metabolic regulation of epigenetics has emerged as an important factor in the control of gene expression in diseased states, but to the best of our knowledge, this connection remains largely unexplored in endothelial dysfunction and atherosclerosis. In this review, we (1) summarize how shear stress (or flow) regulates endothelial (dys)function; (2) explore the epigenetic alterations that occur in the endothelium in response to disturbed flow; (3) review endothelial cell metabolism under different shear stress conditions; and (4) suggest mechanisms which may link this altered metabolism to the regulation of the endothelial epigenome by modulations in metabolite availability. We believe that metabolic regulation plays an important role in endothelial epigenetic reprogramming and could pave the way for novel metabolism-based therapeutic strategies.
Collapse
Affiliation(s)
- Francisco Santos
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| | - Hashum Sum
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| | | | - Alison C. Brewer
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK; (F.S.); (H.S.)
| |
Collapse
|
8
|
Inferrera F, Marino Y, Genovese T, Cuzzocrea S, Fusco R, Di Paola R. Mitochondrial quality control: Biochemical mechanism of cardiovascular disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119906. [PMID: 39837389 DOI: 10.1016/j.bbamcr.2025.119906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/30/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Mitochondria play a key role in the regulation of energy homeostasis and ATP production in cardiac cells. Mitochondrial dysfunction can trigger several pathological events that contribute to the development and progression of cardiovascular diseases. These mechanisms include the induction of oxidative stress, dysregulation of intracellular calcium cycling, activation of the apoptotic pathway, and alteration of lipid metabolism. This review focuses on the role of mitochondria in intracellular signaling associated with cardiovascular diseases, emphasizing the contributions of reactive oxygen species production and mitochondrial dynamics. Indeed, mitochondrial dysfunction has been implicated in every aspect of cardiovascular disease and is currently being evaluated as a potential target for therapeutic interventions. To treat cardiovascular diseases and improve overall heart health, it is important to better understand these biochemical systems. These findings allow the achievement of targeted therapies and preventive measures. Therefore, this review investigates different studies that demonstrate how changes in mitochondrial dynamics like fusion, fission, and mitophagy contribute to the development or worsening of disorders related to heart diseases by summarizing current research on their role.
Collapse
Affiliation(s)
- Francesca Inferrera
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Ylenia Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; Link Campus University, Via del Casale di San Pio V, 4400165 Rome, Italy.
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy.
| |
Collapse
|
9
|
Xiao D, Chang W, Ao X, Ye L, Wu W, Song L, Yuan X, Feng L, Wang P, Wang Y, Jia Y, Tang X, Wang J. Parkin inhibits iron overload-induced cardiomyocyte ferroptosis by ubiquitinating ACSL4 and modulating PUFA-phospholipids metabolism. Acta Pharm Sin B 2025; 15:1589-1607. [PMID: 40370554 PMCID: PMC12069115 DOI: 10.1016/j.apsb.2024.12.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 05/16/2025] Open
Abstract
Iron overload is strongly associated with heart disease. Ferroptosis is a new form of regulated cell death indicated in cardiac ischemia-reperfusion (I/R) injury. However, the specific molecular mechanism of myocardial injury caused by iron overload in the heart is still unclear, and the involvement of ferroptosis in iron overload-induced myocardial injury is not fully understood. In this study, we observed that ferroptosis participated in developing of iron overload and I/R-induced cardiomyopathy. Mechanistically, we discovered that Parkin inhibited iron overload-induced ferroptosis in cardiomyocytes by promoting the ubiquitination of long-chain acyl-CoA synthetase 4 (ACSL4), a crucial protein involved in ferroptosis-related lipid metabolism pathways. Additionally, we identified p53 as a transcription factor that transcriptionally suppressed Parkin expression in iron-overloaded cardiomyocytes, thereby regulating iron overload-induced ferroptosis. In animal studies, cardiac-specific Parkin knockout mice (Myh6-CreER T2 /Parkin fl/fl ) fed a high-iron diet presented more severe myocardial damage, and the high iron levels exacerbated myocardial I/R injury. However, the ferroptosis inhibitor Fer-1 significantly suppressed iron overload-induced ferroptosis and myocardial I/R injury. Moreover, Parkin effectively protected against impaired mitochondrial function and prevented iron overload-induced mitochondrial lipid peroxidation. These findings unveil a novel regulatory pathway involving p53-Parkin-ACSL4 in heart disease by inhibiting of ferroptosis.
Collapse
Affiliation(s)
- Dandan Xiao
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Wenguang Chang
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
| | - Xiang Ao
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Lin Ye
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
| | - Weiwei Wu
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Lin Song
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xiaosu Yuan
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Luxin Feng
- Department of Cardiovascular Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Peiyan Wang
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yu Wang
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yi Jia
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xiaopeng Tang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Jianxun Wang
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266021, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| |
Collapse
|
10
|
Wei J, Peng MY, Lu HX. Functional transformation of macrophage mitochondria in cardiovascular diseases. Mol Cell Biochem 2025; 480:747-757. [PMID: 38884847 DOI: 10.1007/s11010-024-05049-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024]
Abstract
Mitochondria are pivotal in the modulation of macrophage activation, differentiation, and survival. Furthermore, macrophages are instrumental in the onset and progression of cardiovascular diseases. Hence, it is imperative to investigate the role of mitochondria within macrophages in the context of cardiovascular disease. In this review, we provide an updated description of the origin and classification of cardiac macrophages and also focused on the relationship between macrophages and mitochondria in cardiovascular diseases with respect to (1) proinflammatory or anti-inflammatory macrophages, (2) macrophage apoptosis, (3) macrophage pyroptosis, and (4) macrophage efferocytosis. Clarifying the relationship between mitochondria and macrophages can aid the exploration of novel therapeutic strategies for cardiovascular disease.
Collapse
Affiliation(s)
- Jing Wei
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing, 211100, China
| | - Ming-Yu Peng
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing, 211100, China
| | - Hong-Xiang Lu
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing, 211100, China.
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing, 211100, China.
| |
Collapse
|
11
|
Santos F, Correia M, Dias R, Bola B, Noberini R, Ferreira RS, Trigo D, Domingues P, Teixeira J, Bonaldi T, Oliveira PJ, Bär C, de Jesus BB, Nóbrega‐Pereira S. Age-associated metabolic and epigenetic barriers during direct reprogramming of mouse fibroblasts into induced cardiomyocytes. Aging Cell 2025; 24:e14371. [PMID: 39540462 PMCID: PMC11822649 DOI: 10.1111/acel.14371] [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: 11/16/2023] [Revised: 08/28/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
Heart disease is the leading cause of mortality in developed countries, and novel regenerative procedures are warranted. Direct cardiac conversion (DCC) of adult fibroblasts can create induced cardiomyocytes (iCMs) for gene and cell-based heart therapy, and in addition to holding great promise, still lacks effectiveness as metabolic and age-associated barriers remain elusive. Here, by employing MGT (Mef2c, Gata4, Tbx5) transduction of mouse embryonic fibroblasts (MEFs) and adult (dermal and cardiac) fibroblasts from animals of different ages, we provide evidence that the direct reprogramming of fibroblasts into iCMs decreases with age. Analyses of histone posttranslational modifications and ChIP-qPCR revealed age-dependent alterations in the epigenetic landscape of DCC. Moreover, DCC is accompanied by profound mitochondrial metabolic adaptations, including a lower abundance of anabolic metabolites, network remodeling, and reliance on mitochondrial respiration. In vitro metabolic modulation and dietary manipulation in vivo improve DCC efficiency and are accompanied by significant alterations in histone marks and mitochondrial homeostasis. Importantly, adult-derived iCMs exhibit increased accumulation of oxidative stress in the mitochondria and activation of mitophagy or dietary lipids; they improve DCC and revert mitochondrial oxidative damage. Our study provides evidence that metaboloepigenetics plays a direct role in cell fate transitions driving DCC, highlighting the potential use of metabolic modulation to improve cardiac regenerative strategies.
Collapse
Affiliation(s)
- Francisco Santos
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Magda Correia
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Rafaela Dias
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Bárbara Bola
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Roberta Noberini
- Department of Experimental OncologyEuropean Institute of Oncology (IEO), IRCCSMilanItaly
| | - Rita S. Ferreira
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Diogo Trigo
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Pedro Domingues
- Mass Spectrometry Center, Department of ChemistryUniversity of AveiroAveiroPortugal
- LAQV/REQUIMTEUniversity of AveiroAveiroPortugal
| | - José Teixeira
- CNC‐UC, Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB, Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCantanhedePortugal
| | - Tiziana Bonaldi
- Department of Experimental OncologyEuropean Institute of Oncology (IEO), IRCCSMilanItaly
- Department of Oncology and Hematology‐OncologyUniversity of MilanoMilanItaly
| | - Paulo J. Oliveira
- CNC‐UC, Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB, Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCantanhedePortugal
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS)Hannover Medical School (MHH)HannoverGermany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM)HannoverGermany
| | - Bruno Bernardes de Jesus
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| | - Sandrina Nóbrega‐Pereira
- Department of Medical Sciences and Institute of Biomedicine – iBiMEDUniversity of AveiroAveiroPortugal
| |
Collapse
|
12
|
Xu M, Feng P, Yan J, Li L. Mitochondrial quality control: a pathophysiological mechanism and potential therapeutic target for chronic obstructive pulmonary disease. Front Pharmacol 2025; 15:1474310. [PMID: 39830343 PMCID: PMC11739169 DOI: 10.3389/fphar.2024.1474310] [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: 08/01/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent chronic respiratory disease worldwide. Mitochondrial quality control mechanisms encompass processes such as mitochondrial biogenesis, fusion, fission, and autophagy, which collectively maintain the quantity, morphology, and function of mitochondria, ensuring cellular energy supply and the progression of normal physiological activities. However, in COPD, due to the persistent stimulation of harmful factors such as smoking and air pollution, mitochondrial quality control mechanisms often become deregulated, leading to mitochondrial dysfunction. Mitochondrial dysfunction plays a pivotal role in the pathogenesis of COPD, contributing toinflammatory response, oxidative stress, cellular senescence. However, therapeutic strategies targeting mitochondria remain underexplored. This review highlights recent advances in mitochondrial dysfunction in COPD, focusing on the role of mitochondrial quality control mechanisms and their dysregulation in disease progression. We emphasize the significance of mitochondria in the pathophysiological processes of COPD and explore potential strategies to regulate mitochondrial quality and improve mitochondrial function through mitochondrial interventions, aiming to treat COPD effectively. Additionally, we analyze the limitations and challenges of existing therapeutic strategies, aiming to provide new insights and methods for COPD treatment.
Collapse
Affiliation(s)
- Mengjiao Xu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Feng
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Ferguson Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jun Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Jiang X, Wang J, Ma F, Li Y. FOXO3 Activates MFN2 Expression to Maintain the Autophagy Response in Cancer Cells Under Amino Acid Deprivation. J Cell Biochem 2025; 126:e30641. [PMID: 39175152 DOI: 10.1002/jcb.30641] [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: 05/29/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
The lack of amino acids triggers the autophagic response. Some studies have shown such starvation conditions also induce mitochondrial fusion, revealing a close correlation between the two processes. Although Mitofusin-2 (MFN2) has been demonstrated to play a role in fusion regulation, its role in the autophagic response and the variables that activate MFN2 under stress remain unknown. In this investigation, we screened and confirmed that forkhead box protein O3 (FOXO3) participates in MFN2's expression during short periods of starvation. Luciferase reporter test proved that FOXO3 facilitates MFN2's transcription by binding to its promoter region, and FOXO3 downregulation directly depresses MFN2's expression. Consequently, inhibiting the FOXO3-MFN2 axis results in the loss of mitochondrial fusion, disrupting the normal morphology of mitochondria, impairing the degradation of substrates, and reducing autophagosome accumulation, ultimately leading to the blockage of the autophagy. In conclusion, our work demonstrates that the FOXO3-MFN2 pathway is essential for adaptive changes in mitochondrial morphology and cellular autophagy response under nutritional constraints.
Collapse
Affiliation(s)
- Xu Jiang
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Jing Wang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, China
| | - Fang Ma
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Yuyun Li
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
14
|
Xiong Z, Yang L, Zhang C, Huang W, Zhong W, Yi J, Feng J, Zouxu X, Song L, Wang X. MANF facilitates breast cancer cell survival under glucose-starvation conditions via PRKN-mediated mitophagy regulation. Autophagy 2025; 21:80-101. [PMID: 39147386 DOI: 10.1080/15548627.2024.2392415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024] Open
Abstract
During tumor expansion, breast cancer (BC) cells often experience reactive oxygen species accumulation and mitochondrial damage because of glucose shortage. However, the mechanism by which BC cells deal with the glucose-shortage-induced oxidative stress remains unclear. Here, we showed that MANF (mesencephalic astrocyte derived neurotrophic factor)-mediated mitophagy facilitates BC cell survival under glucose-starvation conditions. MANF-mediated mitophagy also promotes fatty acid oxidation in glucose-starved BC cells. Moreover, during glucose starvation, SENP1-mediated de-SUMOylation of MANF increases cytoplasmic MANF expression through the inhibition of MANF's nuclear translocation and hence renders mitochondrial distribution of MANF. MANF mediates mitophagy by binding to PRKN (parkin RBR E3 ubiquitin protein ligase), a key mitophagy regulator, in the mitochondria. Under conditions of glucose starvation, protein oxidation inhibits PRKN activity; nevertheless, the CXXC motif of MANF alleviates protein oxidation in RING II-domain of PRKN and restores its E3 ligase activity. Furthermore, MANF-PRKN interactions are essential for BC tumor growth and metastasis. High MANF expression predicts poor outcomes in patients with BC. Our results highlight the prosurvival role of MANF-mediated mitophagy in BC cells during glucose starvation, suggesting MANF as a potential therapeutic target.Abbreviation: 2DG, 2-deoxy-D-glucose; 5TG, 5-thio-D-glucose; ACSL4/FACL4, acyl-CoA synthetase long chain family member 4; Baf A1, bafilomycin A1; BRCA, breast cancer; CHX, cycloheximide; DMF, distant metastasis-free; DMFS, distant metastasis-free survival; ECM, extracellular matrix; ER, endoplasmic reticulum; ERS, endoplasmic reticulum stress; F-1,6-BP, fructose-1,6-bisphosphate; FAO, fatty acid oxidation; GSH, reduced glutathione; GSVA, gene set variation analysis; HCC, hepatocellular carcinoma; ICC, intrahepatic cholangiocarcinoma; IF, immunofluorescence; MANF, mesencephalic astrocyte derived neurotrophic factor; Mdivi-1, mitochondrial division inhibitor 1; MFI, mean fluorescence intensity; NAC, N-acetyl-L-cysteine; OCR, oxygen-consumption rate; OS, overall survival; PMI, SQSTM1/p62-mediated mitophagy inducer; PPP, pentose phosphate pathway; PRKN, parkin RBR E3 ubiquitin protein ligase; RBR, RING in between RING; RFS, relapse-free survival; ROS, reactive oxygen species; SAPLIPs, saposin-like proteins; TCGA, The Cancer Genome Atlas; TNBC, triple-negative breast cancer; WT, wild type.
Collapse
Affiliation(s)
- Zhenchong Xiong
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lin Yang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chao Zhang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Weiling Huang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenjing Zhong
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiarong Yi
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jikun Feng
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiazi Zouxu
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Libing Song
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xi Wang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
15
|
Zuo B, Li X, Xu D, Zhao L, Yang Y, Luan Y, Zhang B. Targeting mitochondrial transfer: a new horizon in cardiovascular disease treatment. J Transl Med 2024; 22:1160. [PMID: 39741312 PMCID: PMC11687156 DOI: 10.1186/s12967-024-05979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality among individuals with noncommunicable diseases worldwide. Obesity is associated with an increased risk of developing cardiovascular disease (CVD). Mitochondria are integral to the cardiovascular system, and it has been reported that mitochondrial transfer is associated with the pathogenesis of multiple CVDs and obesity. This review offers a comprehensive examination of the relevance of mitochondrial transfer to cardiovascular health and disease, emphasizing the critical functions of mitochondria in energy metabolism and signal transduction within the cardiovascular system. This highlights how disruptions in mitochondrial transfer contribute to various CVDs, such as myocardial infarction, cardiomyopathies, and hypertension. Additionally, we provide an overview of the molecular mechanisms governing mitochondrial transfer and its potential implications for CVD treatment. This finding underscores the therapeutic potential of mitochondrial transfer and addresses the various mechanisms and challenges in its implementation. By delving into mitochondrial transfer and its targeted modulation, this review aims to advance our understanding of cardiovascular disease treatment, presenting new insights and potential therapeutic strategies in this evolving field.
Collapse
Affiliation(s)
- Baile Zuo
- Molecular Immunology and Immunotherapy Laboratory, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
- Department of Clinical Laboratory, Heping Branch, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Dawei Xu
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Liping Zhao
- Department of Pathology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Bi Zhang
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| |
Collapse
|
16
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
17
|
Ouyang F, Li Y, Wang H, Liu X, Tan X, Xie G, Zeng J, Zeng G, Luo Q, Zhou H, Chen S, Hou K, Fang J, Zhang X, Zhou L, Li Y, Gao A. Aloe Emodin Alleviates Radiation-Induced Heart Disease via Blocking P4HB Lactylation and Mitigating Kynurenine Metabolic Disruption. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406026. [PMID: 39494721 PMCID: PMC11653682 DOI: 10.1002/advs.202406026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/22/2024] [Indexed: 11/05/2024]
Abstract
Aloe emodin is an anthraquinone of traditional Chinese medicine monomer, which plays a protective action in cardiovascular diseases. However, the regulatory mechanisms of aloe emodin in the protection of radiation-induced heart damage (RIHD) are unclear. As a novel post-translational modification, lactylation is considered as a critical mediator in inflammatory cascade and cardiac injury. Here, using a cross of differential omics and 4D label-free lactylation omics, protein disulfide-isomerase (P4HB) is identified as a novel target for lactylation, and aloe emodin inhibits the binding of lactate to the K311 site of P4HB. Aloe emodin stabilizes kynurenine metabolism through inhibition of aspartate aminotransferase (GOT2) accumulation on damaged mitochondria. Mechanistically, aloe emodin inhibits phosphorylated glycogen synthase kinase 3B (p-GSK3B) transcription in the nucleus to repress the interaction of prostaglandin G/H synthase 2 (PTGS2) with SH3 domain of SH3 domain-containing GRB2-like protein B1 (SH3GLB1), thereby disrupting the functions of mitochondrial complexes and reducing SH3GLB1-mediated mitoROS accumulation, eventually suppressing calcium-binding and coiled-coil domain-containing protein 2 (NDP52)-induced mitophagy. This study unveils the regulatory role of aloe emodin in RIHD alleviation through PTGS2/SH3GLB1/NDP52 axis, indicates aloe emodin stabilizes GOT2-mediated kynurenine metabolism through P4HB lactylation. Collectively, this study provides novel insights into the regulatory mechanisms underlying the protective role of aloe emodin in cardiac injury, and opens new avenues for therapeutic strategies of aloe emodin in preventing RIHD by regulating lactylation.
Collapse
Affiliation(s)
- Fan Ouyang
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Yaling Li
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Haoming Wang
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Xiangyang Liu
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Xiaoli Tan
- Zhuzhou Clinical CollegeJishou UniversityJishouHunan416000P. R. China
| | - Genyuan Xie
- Zhuzhou Clinical CollegeJishou UniversityJishouHunan416000P. R. China
| | - Junfa Zeng
- Department of Critical Care MedicineHengyang Medical SchoolThe Second Affiliated HospitalUniversity of South ChinaHengyangHunan421001P. R. China
| | - Gaofeng Zeng
- Clinical Research InstituteThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Assisted Reproductive CentreZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Qiong Luo
- Clinical Research Center for Arteriosclerotic Disease in Hunan ProvinceHengyangHunan421001P. R. China
| | - Hong Zhou
- Department of RadiologyHengyang Medical SchoolThe First Affiliated HospitalUniversity of South ChinaHengyangHunan421001P. R. China
| | - Siming Chen
- Clinical Research Center for Arteriosclerotic Disease in Hunan ProvinceHengyangHunan421001P. R. China
| | - Kai Hou
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Jinren Fang
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Xia Zhang
- Department of Ultrasound MedicineHengyang Medical SchoolThe Second Affiliated HospitalUniversity of South ChinaHengyangHunan421001P. R. China
| | - Linlin Zhou
- Zhuzhou Clinical CollegeJishou UniversityJishouHunan416000P. R. China
| | - Yukun Li
- Department of Assisted Reproductive CentreZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Anbo Gao
- Clinical Research InstituteThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Assisted Reproductive CentreZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal CancerHengyangHunan421001P. R. China
| |
Collapse
|
18
|
Pines O, Horwitz M, Herrmann JM. Privileged proteins with a second residence: dual targeting and conditional re-routing of mitochondrial proteins. FEBS J 2024; 291:5379-5393. [PMID: 38857249 PMCID: PMC11653698 DOI: 10.1111/febs.17191] [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: 02/20/2024] [Revised: 04/15/2024] [Accepted: 05/22/2024] [Indexed: 06/12/2024]
Abstract
Almost all mitochondrial proteins are encoded by nuclear genes and synthesized in the cytosol as precursor proteins. Signals in the amino acid sequence of these precursors ensure their targeting and translocation into mitochondria. However, in many cases, only a certain fraction of a specific protein is transported into mitochondria, while the rest either remains in the cytosol or undergoes reverse translocation to the cytosol, and can populate other cellular compartments. This phenomenon is called dual localization which can be instigated by different mechanisms. These include alternative start or stop codons, differential transcripts, and ambiguous or competing targeting sequences. In many cases, dual localization might serve as an economic strategy to reduce the number of required genes; for example, when the same groups of enzymes are required both in mitochondria and chloroplasts or both in mitochondria and the nucleus/cytoplasm. Such cases frequently employ ambiguous targeting sequences to distribute proteins between both organelles. However, alternative localizations can also be used for signaling, for example when non-imported precursors serve as mitophagy signals or when they represent transcription factors in the nucleus to induce the mitochondrial unfolded stress response. This review provides an overview regarding the mechanisms and the physiological consequences of dual targeting.
Collapse
Affiliation(s)
- Ophry Pines
- Microbiology and Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Margalit Horwitz
- Microbiology and Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | |
Collapse
|
19
|
Zhang R, Yang H, Guo M, Niu S, Xue Y. Mitophagy and its regulatory mechanisms in the biological effects of nanomaterials. J Appl Toxicol 2024; 44:1834-1853. [PMID: 38642013 DOI: 10.1002/jat.4609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/22/2024]
Abstract
Mitophagy is a selective cellular process critical for the removal of damaged mitochondria. It is essential in regulating mitochondrial number, ensuring mitochondrial functionality, and maintaining cellular equilibrium, ultimately influencing cell destiny. Numerous pathologies, such as neurodegenerative diseases, cardiovascular disorders, cancers, and various other conditions, are associated with mitochondrial dysfunctions. Thus, a detailed exploration of the regulatory mechanisms of mitophagy is pivotal for enhancing our understanding and for the discovery of novel preventive and therapeutic options for these diseases. Nanomaterials have become integral in biomedicine and various other sectors, offering advanced solutions for medical uses including biological imaging, drug delivery, and disease diagnostics and therapy. Mitophagy is vital in managing the cellular effects elicited by nanomaterials. This review provides a comprehensive analysis of the molecular mechanisms underpinning mitophagy, underscoring its significant influence on the biological responses of cells to nanomaterials. Nanoparticles can initiate mitophagy via various pathways, among which the PINK1-Parkin pathway is critical for cellular defense against nanomaterial-induced damage by promoting mitophagy. The role of mitophagy in biological effects was induced by nanomaterials, which are associated with alterations in Ca2+ levels, the production of reactive oxygen species, endoplasmic reticulum stress, and lysosomal damage.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Haitao Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Menghao Guo
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
20
|
Rawnsley DR, Islam M, Zhao C, Kargar Gaz Kooh Y, Mendoza A, Navid H, Kumari M, Guan X, Murphy JT, Nigro J, Kovacs A, Mani K, Huebsch N, Ma X, Diwan A. Mitophagy Facilitates Cytosolic Proteostasis to Preserve Cardiac Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.24.624947. [PMID: 39651239 PMCID: PMC11623534 DOI: 10.1101/2024.11.24.624947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background Protein quality control (PQC) is critical for maintaining sarcomere structure and function in cardiac myocytes, and mutations in PQC pathway proteins, such as CRYAB (arginine to glycine at position 120, R120G) and BAG3 (proline to lysine at position 209, P209L) induce protein aggregate pathology with cardiomyopathy in humans. Novel observations in yeast and mammalian cells demonstrate mitochondrial uptake of cytosolic protein aggregates. We hypothesized that mitochondrial uptake of cytosolic protein aggregates and their removal by mitophagy, a lysosomal degradative pathway essential for myocardial homeostasis, facilitates cytosolic protein quality control in cardiac myocytes. Methods Mice with inducible cardiac myocyte specific ablation of TRAF2 (TRAF2icKO), which impairs mitophagy, were assessed for protein aggregates with biochemical fractionation and super-resolution imaging in comparison to floxed controls. Induced pluripotent stem cell (iPSC)-derived cardiac myocytes with R120G knock-in to the CRYAB locus were assessed for localization of the CRYAB protein. Transgenic mice expressing R120G CRYAB protein (R120G-TG) were subjected to both TRAF2 gain-of-function (with AAV9-cardiac Troponin T promoter-driven TRAF2 transduction) and TRAF2 loss-of-function (with tamoxifen-inducible ablation of one Traf2 allele) in cardiac myocytes to determine the effect of mitophagy modulation on cardiac structure, function, and protein aggregate pathology. Results Cardiomyocyte-specific ablation of TRAF2 results accumulation of mitochondrial and cytosolic protein aggregates and DESMIN mis-localization to protein aggregates. Isolated mitochondria take up cardiomyopathy-associated aggregate-prone cytosolic chaperone proteins, namely arginine to glycine (R120G) CRYAB mutant and proline to lysine (P209L) BAG3 mutant. R120G-CRYAB mutant protein increasingly localizes to mitochondria in human and mouse cardiomyocytes. R120G-TG mice demonstrate upregulation of TRAF2 in the mitochondrial fraction with increased mitophagy as compared with wild type. Adult-onset inducible haplo-insufficiency of TRAF2 resulted in accelerated mortality, impaired left ventricular systolic function and increased protein aggregates in R120G-TG mice as compared with controls. Conversely, AAV9-mediated TRAF2 transduction in R120G-TG mice reduced mortality and attenuated left ventricular systolic dysfunction, with reduced protein aggregates and restoration of normal localization of DESMIN, a cytosolic scaffolding protein chaperoned by CRYAB, as compared with control AAV9-GFP group. Conclusions TRAF2-mediated mitophagy in cardiac myocytes facilitates removal of cytosolic protein aggregates and can be stimulated to ameliorate proteotoxic cardiomyopathy.
Collapse
|
21
|
Leinheiser AK, Mitchell CC, Rooke E, Strack S, Grueter CE. A dynamical systems model for the total fission rate in Drp1-dependent mitochondrial fission. PLoS Comput Biol 2024; 20:e1012596. [PMID: 39556606 DOI: 10.1371/journal.pcbi.1012596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
Mitochondrial hyperfission in response to cellular insult is associated with reduced energy production and programmed cell death. Thus, there is a critical need to understand the molecular mechanisms coordinating and regulating the complex process of mitochondrial fission. We develop a nonlinear dynamical systems model of dynamin related protein one (Drp1)-dependent mitochondrial fission and use it to identify parameters which can regulate the total fission rate (TFR) as a function of time. The TFR defined from a nondimensionalization of the model undergoes a Hopf bifurcation with bifurcation parameter [Formula: see text] where [Formula: see text] is the total concentration of mitochondrial fission factor (Mff) and k+ and k- are the association and dissociation rate constants between oligomers on the outer mitochondrial membrane. The variable μ can be thought of as the maximum build rate over the disassembling rate of oligomers. Though the nondimensionalization of the system results in four dimensionless parameters, we found the TFR and the cumulative total fission (TF) depend strongly on only one, μ. Interestingly, the cumulative TF does not monotonically increase as μ increases. Instead it increases with μ to a certain point and then begins to decrease as μ continues to increase. This non-monotone dependence on μ suggests interventions targeting k+, k-, or [Formula: see text] may have a non-intuitive impact on the fission mechanism. Thus understanding the impact of regulatory parameters, such as μ, may assist future therapeutic target selection.
Collapse
Affiliation(s)
- Anna K Leinheiser
- Department of Mathematics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States of America
| | - Colleen C Mitchell
- Department of Mathematics, University of Iowa, Iowa City, Iowa, United States of America
| | - Ethan Rooke
- Department of Mathematics, University of Iowa, Iowa City, Iowa, United States of America
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, United States of America
| | - Chad E Grueter
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
22
|
Li H, Dai X, Zhou J, Wang Y, Zhang S, Guo J, Shen L, Yan H, Jiang H. Mitochondrial dynamics in pulmonary disease: Implications for the potential therapeutics. J Cell Physiol 2024; 239:e31370. [PMID: 38988059 DOI: 10.1002/jcp.31370] [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: 02/26/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Mitochondria are dynamic organelles that continuously undergo fusion/fission to maintain normal cell physiological activities and energy metabolism. When mitochondrial dynamics is unbalanced, mitochondrial homeostasis is broken, thus damaging mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to lung tissue injury and pulmonary disease progression in a variety of disease models, including inflammatory responses, apoptosis, and barrier breakdown, and that the role of mitochondrial dynamics varies among pulmonary diseases. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in pulmonary diseases. In this review, we discuss the current evidence on the role of mitochondrial dynamics in pulmonary diseases, with a particular focus on its underlying mechanisms in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis (PF), pulmonary arterial hypertension (PAH), lung cancer and bronchopulmonary dysplasia (BPD), and outline effective drugs targeting mitochondrial dynamics-related proteins, highlighting the great potential of targeting mitochondrial dynamics in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Hui Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinyan Dai
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
23
|
Ravindran S, Rau CD. The multifaceted role of mitochondria in cardiac function: insights and approaches. Cell Commun Signal 2024; 22:525. [PMID: 39472951 PMCID: PMC11523909 DOI: 10.1186/s12964-024-01899-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
Cardiovascular disease (CVD) remains a global economic burden even in the 21st century with 85% of deaths resulting from heart attacks. Despite efforts in reducing the risk factors, and enhancing pharmacotherapeutic strategies, challenges persist in early identification of disease progression and functional recovery of damaged hearts. Targeting mitochondrial dysfunction, a key player in the pathogenesis of CVD has been less successful due to its role in other coexisting diseases. Additionally, it is the only organelle with an agathokakological function that is a remedy and a poison for the cell. In this review, we describe the origins of cardiac mitochondria and the role of heteroplasmy and mitochondrial subpopulations namely the interfibrillar, subsarcolemmal, perinuclear, and intranuclear mitochondria in maintaining cardiac function and in disease-associated remodeling. The cumulative evidence of mitochondrial retrograde communication with the nucleus is addressed, highlighting the need to study the genotype-phenotype relationships of specific organelle functions with CVD by using approaches like genome-wide association study (GWAS). Finally, we discuss the practicality of computational methods combined with single-cell sequencing technologies to address the challenges of genetic screening in the identification of heteroplasmy and contributory genes towards CVD.
Collapse
Affiliation(s)
- Sriram Ravindran
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA
| | - Christoph D Rau
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA.
| |
Collapse
|
24
|
Liu X, Jiang S, Jiang T, Lan Z, Zhang X, Zhong Z, Wu X, Xu C, Du Y, Zhang S. Bioenergetic-active exosomes for cartilage regeneration and homeostasis maintenance. SCIENCE ADVANCES 2024; 10:eadp7872. [PMID: 39423269 PMCID: PMC11488572 DOI: 10.1126/sciadv.adp7872] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
Cartilage regeneration relies on adequate and continuous bioenergy supply to facilitate cellular differentiation and extracellular matrix synthesis. Chondrocytes frequently undergo energy stress under pathological conditions, characterized by disrupted cellular metabolism and reduced adenosine triphosphate (ATP) levels. However, there has limited progress in modulating energy metabolism for cartilage regeneration thus far. Here, we developed bioenergetic-active exosomes (Suc-EXO) to promote cartilage regeneration and homeostasis maintenance. Suc-EXO exhibited a 5.42-fold increase in ATP content, enabling the manipulation of cellular energy metabolism by fueling the TCA cycle. With continuous energy supply, Suc-EXO promoted BMSC chondrogenic differentiation via the P2X7-mediated PI3K-AKT pathway. Moreover, Suc-EXO improved chondrocyte anabolism and mitochondrial homeostasis via the P2X7-mediated SIRT3 pathway. In a rabbit cartilage defect model, the Suc-EXO-encapsulated hydrogel notably promoted cartilage regeneration and maintained neocartilage homeostasis, leading to 2.26 and 1.53 times increase in Col2 and ACAN abundance, respectively. These findings make a remarkable breakthrough in modulating energy metabolism for cartilage regeneration, offering immense potential for clinical translation.
Collapse
Affiliation(s)
- Xulong Liu
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shangtong Jiang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ting Jiang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ziyang Lan
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Zhang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhenyu Zhong
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaodan Wu
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Cunjing Xu
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yingying Du
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
- Research Base of Regulatory Science for Medical Devices, National Medical Products Administration, Wuhan 430074, China
- Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shengmin Zhang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, China
- Research Base of Regulatory Science for Medical Devices, National Medical Products Administration, Wuhan 430074, China
- Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
25
|
Shi J, Jin Y, Lin S, Li X, Zhang D, Wu J, Qi Y, Li Y. Mitochondrial non-energetic function and embryonic cardiac development. Front Cell Dev Biol 2024; 12:1475603. [PMID: 39435335 PMCID: PMC11491369 DOI: 10.3389/fcell.2024.1475603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/20/2024] [Indexed: 10/23/2024] Open
Abstract
The initial contraction of the heart during the embryonic stage necessitates a substantial energy supply, predominantly derived from mitochondrial function. However, during embryonic heart development, mitochondria influence beyond energy supplementation. Increasing evidence suggests that mitochondrial permeability transition pore opening and closing, mitochondrial fusion and fission, mitophagy, reactive oxygen species production, apoptosis regulation, Ca2+ homeostasis, and cellular redox state also play critical roles in early cardiac development. Therefore, this review aims to describe the essential roles of mitochondrial non-energetic function embryonic cardiac development.
Collapse
Affiliation(s)
- Jingxian Shi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuxi Jin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Du Z, Zhou Y, Li Q, Xie Y, Zhu T, Qiao J, Zhang R, Bao Y, Wang L, Xie Y, Quan J, Lin M, Zhang N, Jin Q, Liang W, Wu L, Yin T, Xie Y. SIRT1 Ameliorates Lamin A/C Deficiency-Induced Cardiac Dysfunction by Promoting Mitochondrial Bioenergetics. JACC Basic Transl Sci 2024; 9:1211-1230. [PMID: 39534638 PMCID: PMC11551877 DOI: 10.1016/j.jacbts.2024.05.011] [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: 10/08/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 11/16/2024]
Abstract
Dilated cardiomyopathy (DCM) is associated with high mortality despite advanced therapies. The LMNA gene encodes lamin A/C and is the second most frequently mutated gene associated with DCM, for which therapeutic options are limited. Here we generated Lmna -/- mice and found they exhibited cardiac dysfunction at the age of 1 month but not at 2 weeks. Proteomics showed down-regulation of mitochondrial function-related pathways in Lmna -/- hearts. Moreover, early injured mitochondria with decreased cristae density and sirtuin 1 (SIRT1) down-regulation were observed in 2-week-old Lmna -/- hearts. Adenoviral overexpression of SIRT1 in lamin A/C knockdown neonatal rat ventricular myocytes improved mitochondrial oxidative respiration capacity. Adeno-associated virus-mediated SIRT1 overexpression alleviated mitochondrial injury, cardiac systolic dysfunction, ventricular dilation, and fibrosis, and prolonged lifespan in Lmna -/- mice. Mechanistically, LMNA maintains mitochondrial bioenergetics through the SIRT1-PARKIN axis. Our results suggest that targeting the SIRT1 signaling pathway is expected to be a novel therapeutic strategy for LMNA mutation-associated DCM.
Collapse
Affiliation(s)
- Zunhui Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanting Zhou
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiheng Li
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Xie
- College of Osteopathic Medicine, Kansas City University, Kansas City, Missouri, USA
| | - Tingfang Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruihong Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Bao
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingjie Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinyin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinwei Quan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Menglu Lin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbin Liang
- University of Ottawa Heart Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Liqun Wu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Yin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yucai Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Brogyanyi T, Kejík Z, Veselá K, Dytrych P, Hoskovec D, Masařik M, Babula P, Kaplánek R, Přibyl T, Zelenka J, Ruml T, Vokurka M, Martásek P, Jakubek M. Iron chelators as mitophagy agents: Potential and limitations. Biomed Pharmacother 2024; 179:117407. [PMID: 39265234 DOI: 10.1016/j.biopha.2024.117407] [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: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Mitochondrial autophagy (mitophagy) is very important process for the maintenance of cellular homeostasis, functionality and survival. Its dysregulation is associated with high risk and progression numerous serious diseases (e.g., oncological, neurodegenerative and cardiovascular ones). Therefore, targeting mitophagy mechanisms is very hot topic in the biological and medicinal research. The interrelationships between the regulation of mitophagy and iron homeostasis are now becoming apparent. In short, mitochondria are central point for the regulation of iron homeostasis, but change in intracellular cheatable iron level can induce/repress mitophagy. In this review, relationships between iron homeostasis and mitophagy are thoroughly discussed and described. Also, therapeutic applicability of mitophagy chelators in the context of individual diseases is comprehensively and critically evaluated.
Collapse
Affiliation(s)
- Tereza Brogyanyi
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - Michal Masařik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Babula
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Tomáš Přibyl
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic.
| |
Collapse
|
28
|
Kaur B, Miglioranza Scavuzzi B, Yang M, Yao J, Jia L, Abcouwer SF, Zacks DN. ER Stress and Mitochondrial Perturbations Regulate Cell Death in Retinal Detachment: Exploring the Role of HIF1α. Invest Ophthalmol Vis Sci 2024; 65:39. [PMID: 39325470 PMCID: PMC11437674 DOI: 10.1167/iovs.65.11.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
Purpose Retinal detachment (RD) leads to photoreceptor (PR) hypoxia due to separation from the retinal pigment epithelium (RPE). Hypoxia stabilizes retinal hypoxia-inducible factor 1-alpha (HIF1α), crucial for PR survival during RD. This study explores the regulatory role of HIF1α in PR cell survival pathways during RD. Methods Experimental RD was created in C57BL/6J and HIF1αΔrod mice by injecting 1% hyaluronic acid into the subretinal space. The 661W photoreceptor cells were exposed to hypoxic conditions. Markers of endoplasmic reticulum stress (ERS), mitophagy, and accumulation of polyubiquinated proteins were evaluated using RT-PCR and western blot analyses. Cell death of PR cells was quantified using trypan blue exclusion assay and TUNEL staining. Retinal cell death was assessed using a DNA fragmentation assay. Results In C57BL/6J mice and 661W cells, there were increases in HIF1α protein levels: 2.2-fold after RD (P = 0.04) and threefold after hypoxia (P = 0.057). Both the in vivo and in vitro RD models showed increased protein expression of ERS markers (including BIP, CHOP, and IRE1α), mitophagy markers (Parkin, PINK, and FUNDC1), and polyubiquitinated proteins. In 661W cells, hypoxia resulted in a loss of mitochondrial membrane potential, an increase in mitochondrial reactive oxygen species, and a decrease in intracellular adenosine triphosphate levels. Lack of HIF1α in rods blocked the upregulation of mitophagy markers after RD. Conclusions RD results in the activation of ERS, mitophagy, mitochondrial dysfunction, and accumulation of polyubiquitinated proteins. Results suggest a role for HIF1α in activation of the mitophagy pathway after RD, which may serve to protect the PR cells.
Collapse
Affiliation(s)
- Bhavneet Kaur
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Bruna Miglioranza Scavuzzi
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Mengling Yang
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Jingyu Yao
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Lin Jia
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Steven F Abcouwer
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - David N Zacks
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
29
|
Lacombe A, Scorrano L. The interplay between mitochondrial dynamics and autophagy: From a key homeostatic mechanism to a driver of pathology. Semin Cell Dev Biol 2024; 161-162:1-19. [PMID: 38430721 DOI: 10.1016/j.semcdb.2024.02.001] [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: 11/06/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
The complex relationship between mitochondrial dynamics and autophagy illustrates how two cellular housekeeping processes are intimately linked, illuminating fundamental principles of cellular homeostasis and shedding light on disparate pathological conditions including several neurodegenerative disorders. Here we review the basic tenets of mitochondrial dynamics i.e., the concerted balance between fusion and fission of the organelle, and its interplay with macroautophagy and selective mitochondrial autophagy, also dubbed mitophagy, in the maintenance of mitochondrial quality control and ultimately in cell viability. We illustrate how conditions of altered mitochondrial dynamics reverberate on autophagy and vice versa. Finally, we illustrate how altered interplay between these two key cellular processes participates in the pathogenesis of human disorders affecting multiple organs and systems.
Collapse
Affiliation(s)
- Alice Lacombe
- Dept. of Biology, University of Padova, Padova, Italy
| | - Luca Scorrano
- Dept. of Biology, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
30
|
Qiu Y, Xu J, Chen Y, Wu Y, Lin YN, Liu W, Wang Z, Wu Y, Qian X, Li YC. Parkin plays a crucial role in acute viral myocarditis by regulating mitophagy activity. Theranostics 2024; 14:5303-5315. [PMID: 39267792 PMCID: PMC11388078 DOI: 10.7150/thno.97675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Rationale: Parkin (an E3 ubiquitin protein ligase) is an important regulator of mitophagy. However, the role of Parkin in viral myocarditis (VMC) remains unclear. Methods: Coxsackievirus B3 (CVB3) infection was induced in mice to create VMC. Cardiac function and inflammatory response were evaluated by echocardiography, histological assessment, and molecular analyses. AAV9 (adeno-associated virus 9), transmission electron microscopy (TEM) and western blotting were used to investigate the mechanisms by which Parkin regulates mitophagy and cardiac inflammation. Results: Our data indicated that Parkin- and BNIP3 (BCL2 interacting protein 3 like)-mediated mitophagy was activated in VMC mice and neonatal rat cardiac myocytes (NRCMs) infected with CVB3, which blocked autophagic flux by inhibiting autophagosome-lysosome fusion. Parkin silencing aggravated mortality and accelerated the development of cardiac dysfunction in CVB3-treated mice. While silencing of Parkin did not significantly increase inflammatory response through activating NF-κB pathway and production of inflammatory cytokines post-VMC, the mitophagy activity were reduced, which stimulated the accumulation of damaged mitochondria. Moreover, Parkin silencing exacerbated VMC-induced apoptosis. We consistently found that Parkin knockdown disrupted mitophagy activity and inflammatory response in NRCMs. Conclusion: This study elucidated the important role of Parkin in maintaining cardiac function and inflammatory response by regulating mitophagy activity and the NF-κB pathway during acute VMC. Although the functional impact of mitophagy remains unclear, our findings suggest that Parkin silencing may accelerate VMC development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yue-Chun Li
- From the Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
31
|
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: 5] [Impact Index Per Article: 5.0] [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
|
32
|
Ahola S, Pazurek LA, Mayer F, Lampe P, Hermans S, Becker L, Amarie OV, Fuchs H, Gailus-Durner V, de Angelis MH, Riedel D, Nolte H, Langer T. Opa1 processing is dispensable in mouse development but is protective in mitochondrial cardiomyopathy. SCIENCE ADVANCES 2024; 10:eadp0443. [PMID: 39093974 PMCID: PMC11296347 DOI: 10.1126/sciadv.adp0443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
Mitochondrial fusion and fission accompany adaptive responses to stress and altered metabolic demands. Inner membrane fusion and cristae morphogenesis depends on optic atrophy 1 (Opa1), which is expressed in different isoforms and is cleaved from a membrane-bound, long to a soluble, short form. Here, we have analyzed the physiological role of Opa1 isoforms and Opa1 processing by generating mouse lines expressing only one cleavable Opa1 isoform or a non-cleavable variant thereof. Our results show that expression of a single cleavable or non-cleavable Opa1 isoform preserves embryonic development and the health of adult mice. Opa1 processing is dispensable under metabolic and thermal stress but prolongs life span and protects against mitochondrial cardiomyopathy in OXPHOS-deficient Cox10-/- mice. Mechanistically, loss of Opa1 processing disturbs the balance between mitochondrial biogenesis and mitophagy, suppressing cardiac hypertrophic growth in Cox10-/- hearts. Our results highlight the critical regulatory role of Opa1 processing, mitochondrial dynamics, and metabolism for cardiac hypertrophy.
Collapse
Affiliation(s)
- Sofia Ahola
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Fiona Mayer
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Philipp Lampe
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Steffen Hermans
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Oana V Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg
| | - Dietmar Riedel
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
33
|
Forte M, D'Ambrosio L, Schiattarella GG, Salerno N, Perrone MA, Loffredo FS, Bertero E, Pilichou K, Manno G, Valenti V, Spadafora L, Bernardi M, Simeone B, Sarto G, Frati G, Perrino C, Sciarretta S. Mitophagy modulation for the treatment of cardiovascular diseases. Eur J Clin Invest 2024; 54:e14199. [PMID: 38530070 DOI: 10.1111/eci.14199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Defects of mitophagy, the selective form of autophagy for mitochondria, are commonly observed in several cardiovascular diseases and represent the main cause of mitochondrial dysfunction. For this reason, mitophagy has emerged as a novel and potential therapeutic target. METHODS In this review, we discuss current evidence about the biological significance of mitophagy in relevant preclinical models of cardiac and vascular diseases, such as heart failure, ischemia/reperfusion injury, metabolic cardiomyopathy and atherosclerosis. RESULTS Multiple studies have shown that cardiac and vascular mitophagy is an adaptive mechanism in response to stress, contributing to cardiovascular homeostasis. Mitophagy defects lead to cell death, ultimately impairing cardiac and vascular function, whereas restoration of mitophagy by specific compounds delays disease progression. CONCLUSIONS Despite previous efforts, the molecular mechanisms underlying mitophagy activation in response to stress are not fully characterized. A comprehensive understanding of different forms of mitophagy active in the cardiovascular system is extremely important for the development of new drugs targeting this process. Human studies evaluating mitophagy abnormalities in patients at high cardiovascular risk also represent a future challenge.
Collapse
Affiliation(s)
| | - Luca D'Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nadia Salerno
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francesco S Loffredo
- Division of Cardiology, Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino-Italian IRCCS Cardiology Network, Genoa, Italy
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Valenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- ICOT Istituto Marco Pasquali, Latina, Italy
| | | | - Marco Bernardi
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | | | | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
34
|
Schiavon CR, Wang Y, Feng JW, Garrett S, Sung TC, Dayn Y, Wang C, Youle RJ, Quintero-Carmona OA, Shadel GS, Manor U. INF2-mediated actin polymerization at ER-organelle contacts regulates organelle size and movement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.06.602365. [PMID: 39005402 PMCID: PMC11245118 DOI: 10.1101/2024.07.06.602365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Proper regulation of organelle dynamics is critical for cellular function, but the mechanisms coordinating multiple organelles remain poorly understood. Here we show that actin polymerization mediated by the endoplasmic reticulum (ER)-anchored formin INF2 acts as a master regulator of organelle morphology and movement. Using high-resolution imaging, we demonstrate that INF2-polymerized actin filaments assemble at ER contact sites on mitochondria, endosomes, and lysosomes just prior to their fission. Genetic manipulation of INF2 activity alters the size, shape and motility of all three organelles. Our findings reveal a conserved mechanism by which the ER uses actin polymerization to control diverse organelles, with implications for understanding organelle dysfunction in neurodegenerative and other diseases. This work establishes INF2-mediated actin assembly as a central coordinator of organelle dynamics and inter-organelle communication.
Collapse
Affiliation(s)
- Cara R. Schiavon
- Department of Cell & Developmental Biology, University of California, San Diego
| | - Yuning Wang
- Department of Cell & Developmental Biology, University of California, San Diego
| | | | - Stephanie Garrett
- Department of Cell & Developmental Biology, University of California, San Diego
| | | | | | - Chunxin Wang
- National Institute of Neurological Disorders and Stroke
| | | | | | | | - Uri Manor
- Department of Cell & Developmental Biology, University of California, San Diego
| |
Collapse
|
35
|
Saha B, Olsvik H, Williams GL, Oh S, Evjen G, Sjøttem E, Mandell MA. TBK1 is ubiquitinated by TRIM5α to assemble mitophagy machinery. Cell Rep 2024; 43:114294. [PMID: 38814780 PMCID: PMC11216866 DOI: 10.1016/j.celrep.2024.114294] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/05/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024] Open
Abstract
Ubiquitination of mitochondrial proteins provides a basis for the downstream recruitment of mitophagy machinery, yet whether ubiquitination of the machinery itself contributes to mitophagy is unknown. Here, we show that K63-linked polyubiquitination of the key mitophagy regulator TBK1 is essential for its mitophagy functions. This modification is catalyzed by the ubiquitin ligase TRIM5α and is required for TBK1 to interact with and activate a set of ubiquitin-binding autophagy adaptors including NDP52, p62/SQSTM1, and NBR1. Autophagy adaptors, along with TRIM27, enable TRIM5α to engage with TBK1 following mitochondrial damage. TRIM5α's ubiquitin ligase activity is required for the accumulation of active TBK1 on damaged mitochondria in Parkin-dependent and Parkin-independent mitophagy pathways. Our data support a model in which TRIM5α provides a mitochondria-localized, ubiquitin-based, self-amplifying assembly platform for TBK1 and mitophagy adaptors that is ultimately necessary for the recruitment of the core autophagy machinery.
Collapse
Affiliation(s)
- Bhaskar Saha
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Hallvard Olsvik
- Autophagy Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Geneva L Williams
- Biomedical Sciences Graduate Program, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Seeun Oh
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Gry Evjen
- Autophagy Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Eva Sjøttem
- Autophagy Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Michael A Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
36
|
Yildirim RM, Seli E. The role of mitochondrial dynamics in oocyte and early embryo development. Semin Cell Dev Biol 2024; 159-160:52-61. [PMID: 38330625 DOI: 10.1016/j.semcdb.2024.01.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
Mitochondrial dysfunction is widely implicated in various human diseases, through mechanisms that go beyond mitochondria's well-established role in energy generation. These dynamic organelles exert vital control over numerous cellular processes, including calcium regulation, phospholipid synthesis, innate immunity, and apoptosis. While mitochondria's importance is acknowledged in all cell types, research has revealed the exceptionally dynamic nature of the mitochondrial network in oocytes and embryos, finely tuned to meet unique needs during gamete and pre-implantation embryo development. Within oocytes, both the quantity and morphology of mitochondria can significantly change during maturation and post-fertilization. These changes are orchestrated by fusion and fission processes (collectively known as mitochondrial dynamics), crucial for energy production, content exchange, and quality control as mitochondria adjust to the shifting energy demands of oocytes and embryos. The roles of proteins that regulate mitochondrial dynamics in reproductive processes have been primarily elucidated through targeted deletion studies in animal models. Notably, impaired mitochondrial dynamics have been linked to female reproductive health, affecting oocyte quality, fertilization, and embryo development. Dysfunctional mitochondria can lead to fertility problems and can have an impact on the success of pregnancy, particularly in older reproductive age women.
Collapse
Affiliation(s)
- Raziye Melike Yildirim
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Emre Seli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
37
|
ZHU W, GUO C, DU M, MA Y, CUI Y, CHEN X, GUO C. Acupotomy alleviates knee osteoarthritis in rabbit by regulating chondrocyte mitophagy Pink1-Parkin pathway. J TRADIT CHIN MED 2024; 44:468-477. [PMID: 38767630 PMCID: PMC11077155 DOI: 10.19852/j.cnki.jtcm.20240402.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/19/2023] [Indexed: 05/22/2024]
Abstract
OBJECTIVE To investigate the effect of acupotomy, on mitophagy and the Pink1-Parkin pathway in chondrocytes from rabbits with knee osteoarthritis (KOA). METHODS A KOA model was established via the modified Videman method. Rabbits were randomly divided into a control group (CON), KOA group and KOA + acupotomy group (Acu). Rabbits in the acupotomy group were subjected to acupotomy for 4 weeks after model establishment. The behavior of the rabbits before and after intervention was recorded. Cartilage degeneration was evaluated by optical microscopy and fluorescence microscopy. The level of mitophagy was evaluated by transmission electron microscopy, immunofluorescence and enzyme-linked immunosorbent assay (ELISA). The expression of phosphatase and tensin homolog (PTEN)-induced kinase 1 (Pink1)-Parkin mitophagy pathway components was evaluated by immunofluorescence, Western blotting and real-time polymerase chain reaction. RESULTS In rabbits with KOA, joint pain, mobility disorders and cartilage degeneration were observed, the Mankin score was increased, collagen type Ⅱ (Col-Ⅱ) expression was significantly decreased, mitophagy was inhibited, mitochondrial function was impaired, and factors associated with the Pink1-Parkin pathway were inhibited. Acupotomy regulated the expression of Pink1-Parkin pathway-related proteins, the mitophagy-related protein microtubule-associated protein-1 light chain-3, the translocase of the outer membrane, and the inner mitochondrial membrane 23; increased the colocalization of mitochondria and autophagosomes; promoted the removal of damaged mitochondria; restored mitochondrial adenosine-triphosphate (ATP) production; and alleviated cartilage degeneration in rabbits with KOA. CONCLUSIONS Acupotomy played a role in alleviating KOA in rabbits by activating mitophagy in chondrocytes via the regulation of proteins that are related to the Pink1-Parkin pathway.
Collapse
Affiliation(s)
- Wenting ZHU
- 1 the Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Changqing GUO
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mei DU
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yunxuan MA
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yongqi CUI
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xilin CHEN
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Changqing GUO
- 3 Department of Medical Technology, Shijiazhuang Medical College, Hebei 050599, China
| |
Collapse
|
38
|
Baninameh Z, Watzlawik JO, Hou X, Richardson T, Kurchaba NW, Yan T, Di Florio DN, Fairweather D, Kang L, Nguyen JH, Kanekiyo T, Dickson DW, Noda S, Sato S, Hattori N, Goldberg MS, Ganley IG, Stauch KL, Fiesel FC, Springer W. Alterations of PINK1-PRKN signaling in mice during normal aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591753. [PMID: 38746191 PMCID: PMC11092476 DOI: 10.1101/2024.04.29.591753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The ubiquitin kinase-ligase pair PINK1-PRKN identifies and selectively marks damaged mitochondria for elimination via the autophagy-lysosome system (mitophagy). While this cytoprotective pathway has been extensively studied in vitro upon acute and complete depolarization of mitochondria, the significance of PINK1-PRKN mitophagy in vivo is less well established. Here we used a novel approach to study PINK1-PRKN signaling in different energetically demanding tissues of mice during normal aging. We demonstrate a generally increased expression of both genes and enhanced enzymatic activity with aging across tissue types. Collectively our data suggest a distinct regulation of PINK1-PRKN signaling under basal conditions with the most pronounced activation and flux of the pathway in mouse heart compared to brain or skeletal muscle. Our biochemical analyses complement existing mitophagy reporter readouts and provide an important baseline assessment in vivo, setting the stage for further investigations of the PINK1-PRKN pathway during stress and in relevant disease conditions.
Collapse
|
39
|
Parvaresh H, Paczek K, Al-Bari MAA, Eid N. Mechanistic insights into fasting-induced autophagy in the aging heart. World J Cardiol 2024; 16:109-117. [PMID: 38576517 PMCID: PMC10989221 DOI: 10.4330/wjc.v16.i3.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/01/2024] [Accepted: 03/04/2024] [Indexed: 03/21/2024] Open
Abstract
Autophagy is a prosurvival mechanism for the clearance of accumulated abnormal proteins, damaged organelles, and excessive lipids within mammalian cells. A growing body of data indicates that autophagy is reduced in aging cells. This reduction leads to various diseases, such as myocardial hypertrophy, infarction, and atherosclerosis. Recent studies in animal models of an aging heart showed that fasting-induced autophagy improved cardiac function and longevity. This improvement is related to autophagic clearance of damaged cellular components via either bulk or selective autophagy (such as mitophagy). In this editorial, we summarize the mechanisms of autophagy in normal and aging hearts. In addition, the protective effect of fasting-induced autophagy in cardiac aging has been highlighted.
Collapse
Affiliation(s)
- Hannaneh Parvaresh
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Katarzyna Paczek
- Department of Chiropractic, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
40
|
Beutner G, Burris JR, Collins MP, Kulkarni CA, Nadtochiy SM, de Mesy Bentley KL, Cohen ED, Brookes PS, Porter GA. Coordinated metabolic responses to cyclophilin D deletion in the developing heart. iScience 2024; 27:109157. [PMID: 38414851 PMCID: PMC10897919 DOI: 10.1016/j.isci.2024.109157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/02/2023] [Accepted: 02/03/2024] [Indexed: 02/29/2024] Open
Abstract
In the embryonic heart, the activation of the mitochondrial electron transport chain (ETC) coincides with the closure of the cyclophilin D (CypD) regulated mitochondrial permeability transition pore (mPTP). However, it remains to be established whether the absence of CypD has a regulatory effect on mitochondria during cardiac development. Using a variety of assays to analyze cardiac tissue from wildtype and CypD knockout mice from embryonic day (E)9.5 to adult, we found that mitochondrial structure, function, and metabolism show distinct transitions. Deletion of CypD altered the timing of these transitions as the mPTP was closed at all ages, leading to coupled ETC activity in the early embryo, decreased citrate synthase activity, and an altered metabolome particularly after birth. Our results suggest that manipulating CypD activity may control myocyte proliferation and differentiation and could be a tool to increase ATP production and cardiac function in immature hearts.
Collapse
Affiliation(s)
- Gisela Beutner
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jonathan Ryan Burris
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pediatrics, Division of Neonatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael P. Collins
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chaitanya A. Kulkarni
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sergiy M. Nadtochiy
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Karen L. de Mesy Bentley
- Department of Pathology & Laboratory Medicine and the Electron Microscope Resource, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ethan D. Cohen
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Paul S. Brookes
- Department of Anesthesiology & Perioperative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - George A. Porter
- Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Departments of Medicine (Aab Cardiovascular Research Institute) and Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
41
|
Wang H, Luo W, Chen H, Cai Z, Xu G. Mitochondrial dynamics and mitochondrial autophagy: Molecular structure, orchestrating mechanism and related disorders. Mitochondrion 2024; 75:101847. [PMID: 38246334 DOI: 10.1016/j.mito.2024.101847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
Mitochondrial dynamics and autophagy play essential roles in normal cellular physiological activities, while abnormal mitochondrial dynamics and mitochondrial autophagy can cause cancer and related disorders. Abnormal mitochondrial dynamics usually occur in parallel with mitochondrial autophagy. Both have been reported to have a synergistic effect and can therefore complement or inhibit each other. Progress has been made in understanding the classical mitochondrial PINK1/Parkin pathway and mitochondrial dynamical abnormalities. Still, the mechanisms and regulatory pathways underlying the interaction between mitophagy and mitochondrial dynamics remain unexplored. Like other existing reviews, we review the molecular structure of proteins involved in mitochondrial dynamics and mitochondrial autophagy, and how their abnormalities can lead to the development of related diseases. We will also review the individual or synergistic effects of abnormal mitochondrial dynamics and mitophagy leading to cellular proliferation, differentiation and invasion. In addition, we explore the mechanisms underlying mitochondrial dynamics and mitochondrial autophagy to contribute to targeted and precise regulation of mitochondrial function. Through the study of abnormal mitochondrial dynamics and mitochondrial autophagy regulation mechanisms, as well as the role of early disease development, effective targets for mitochondrial function regulation can be proposed to enable accurate diagnosis and treatment of the associated disorders.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Urology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China; Guangzhou Medical University, Guangzhou 511495, China
| | - Wenjun Luo
- Department of Urology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
| | - Haoyu Chen
- Department of Urology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
| | - Zhiduan Cai
- Department of Urology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China.
| | - Guibin Xu
- Department of Urology, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510230, China.
| |
Collapse
|
42
|
Nakano H, Nakano A. The role of metabolism in cardiac development. Curr Top Dev Biol 2024; 156:201-243. [PMID: 38556424 DOI: 10.1016/bs.ctdb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Metabolism is the fundamental process that sustains life. The heart, in particular, is an organ of high energy demand, and its energy substrates have been studied for more than a century. In recent years, there has been a growing interest in understanding the role of metabolism in the early differentiation of pluripotent stem cells and in cancer research. Studies have revealed that metabolic intermediates from glycolysis and the tricarboxylic acid cycle act as co-factors for intracellular signal transduction, playing crucial roles in regulating cell behaviors. Mitochondria, as the central hub of metabolism, are also under intensive investigation regarding the regulation of their dynamics. The metabolic environment of the fetus is intricately linked to the maternal metabolic status, and the impact of the mother's nutrition and metabolic health on fetal development is significant. For instance, it is well known that maternal diabetes increases the risk of cardiac and nervous system malformations in the fetus. Another notable example is the decrease in the risk of neural tube defects when pregnant women are supplemented with folic acid. These examples highlight the profound influence of the maternal metabolic environment on the fetal organ development program. Therefore, gaining insights into the metabolic environment within developing fetal organs is critical for deepening our understanding of normal organ development. This review aims to summarize recent findings that build upon the historical recognition of the environmental and metabolic factors involved in the developing embryo.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States; Cardiology Division, Department of Medicine, UCLA, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States; Molecular Biology Institute, UCLA, Los Angeles, CA, United States; Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
43
|
Sakamoto T, Kelly DP. Cardiac maturation. J Mol Cell Cardiol 2024; 187:38-50. [PMID: 38160640 PMCID: PMC10923079 DOI: 10.1016/j.yjmcc.2023.12.008] [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: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The heart undergoes a dynamic maturation process following birth, in response to a wide range of stimuli, including both physiological and pathological cues. This process entails substantial re-programming of mitochondrial energy metabolism coincident with the emergence of specialized structural and contractile machinery to meet the demands of the adult heart. Many components of this program revert to a more "fetal" format during development of pathological cardiac hypertrophy and heart failure. In this review, emphasis is placed on recent progress in our understanding of the transcriptional control of cardiac maturation, encompassing the results of studies spanning from in vivo models to cardiomyocytes derived from human stem cells. The potential applications of this current state of knowledge to new translational avenues aimed at the treatment of heart failure is also addressed.
Collapse
Affiliation(s)
- Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
44
|
Zhang Q, Li Z, Li Q, Trammell SA, Schmidt MS, Pires KM, Cai J, Zhang Y, Kenny H, Boudina S, Brenner C, Abel ED. Control of NAD + homeostasis by autophagic flux modulates mitochondrial and cardiac function. EMBO J 2024; 43:362-390. [PMID: 38212381 PMCID: PMC10897141 DOI: 10.1038/s44318-023-00009-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: 01/20/2023] [Revised: 10/31/2023] [Accepted: 11/08/2023] [Indexed: 01/13/2024] Open
Abstract
Impaired autophagy is known to cause mitochondrial dysfunction and heart failure, in part due to altered mitophagy and protein quality control. However, whether additional mechanisms are involved in the development of mitochondrial dysfunction and heart failure in the setting of deficient autophagic flux remains poorly explored. Here, we show that impaired autophagic flux reduces nicotinamide adenine dinucleotide (NAD+) availability in cardiomyocytes. NAD+ deficiency upon autophagic impairment is attributable to the induction of nicotinamide N-methyltransferase (NNMT), which methylates the NAD+ precursor nicotinamide (NAM) to generate N-methyl-nicotinamide (MeNAM). The administration of nicotinamide mononucleotide (NMN) or inhibition of NNMT activity in autophagy-deficient hearts and cardiomyocytes restores NAD+ levels and ameliorates cardiac and mitochondrial dysfunction. Mechanistically, autophagic inhibition causes the accumulation of SQSTM1, which activates NF-κB signaling and promotes NNMT transcription. In summary, we describe a novel mechanism illustrating how autophagic flux maintains mitochondrial and cardiac function by mediating SQSTM1-NF-κB-NNMT signaling and controlling the cellular levels of NAD+.
Collapse
Affiliation(s)
- Quanjiang Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Zhonggang Li
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Qiuxia Li
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Samuel Aj Trammell
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Karla Maria Pires
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jinjin Cai
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yuan Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Helena Kenny
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sihem Boudina
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, 84112, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - E Dale Abel
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
45
|
Carvalho RA. The glycolytic pathway to heart failure. GLYCOLYSIS 2024:235-266. [DOI: 10.1016/b978-0-323-91704-9.00010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
46
|
Gao A, Wang M, Tang X, Shi G, Hou K, Fang J, Zhou L, Zhou H, Jiang W, Li Y, Ouyang F. NDP52 SUMOylation contributes to low-dose X-rays-induced cardiac hypertrophy through PINK1/Parkin-mediated mitophagy via MUL1/SUMO2 signalling. J Cell Physiol 2024; 239:79-96. [PMID: 37942585 DOI: 10.1002/jcp.31145] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023]
Abstract
Radiation-induced heart damage caused by low-dose X-rays has a significant impact on tumour patients' prognosis, with cardiac hypertrophy being the most severe noncarcinogenic adverse effect. Our previous study demonstrated that mitophagy activation promoted cardiac hypertrophy, but the underlying mechanisms remained unclear. In the present study, PARL-IN-1 enhanced excessive hypertrophy of cardiomyocytes and exacerbated mitochondrial damage. Isobaric tags for relative and absolute quantification-based quantitative proteomics identified NDP52 as a crucial target mediating cardiac hypertrophy induced by low-dose X-rays. SUMOylation proteomics revealed that the SUMO E3 ligase MUL1 facilitated NDP52 SUMOylation through SUMO2. Co-IP coupled with LC-MS/MS identified a critical lysine residue at position 262 of NDP52 as the key site for SUMO2-mediated SUMOylation of NDP52. The point mutation plasmid NDP52K262R inhibited mitophagy under MUL1 overexpression, as evidenced by inhibition of LC3 interaction with NDP52, PINK1 and LAMP2A. A mitochondrial dissociation study revealed that NDP52K262R inhibited PINK1 targeting to endosomes early endosomal marker (EEA1), late/lysosome endosomal marker (LAMP2A) and recycling endosomal marker (RAB11), and laser confocal microscopy confirmed that NDP52K262R impaired the recruitment of mitochondria to the autophagic pathway through EEA1/RAB11 and ATG3, ATG5, ATG16L1 and STX17, but did not affect mitochondrial delivery to lysosomes via LAMP2A for degradation. In conclusion, our findings suggest that MUL1-mediated SUMOylation of NDP52 plays a crucial role in regulating mitophagy in the context of low-dose X-ray-induced cardiac hypertrophy. Two hundred sixty-second lysine of NDP52 is identified as a key SUMOylation site for low-dose X-ray promoting mitophagy activation and cardiac hypertrophy. Collectively, this study provides novel implications for the development of therapeutic strategies aimed at preventing the progression of cardiac hypertrophy induced by low-dose X-rays.
Collapse
Affiliation(s)
- Anbo Gao
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Mengjie Wang
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Gangqing Shi
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Kai Hou
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Jinren Fang
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Linlin Zhou
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Hong Zhou
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weimin Jiang
- Department of Cardiology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Fan Ouyang
- Department of Cardiology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| |
Collapse
|
47
|
Li A, Qin Y, Zhang Y, Zhen X, Gong G. Evaluation of Oxygen Consumption Rates In Situ. Methods Mol Biol 2024; 2755:215-226. [PMID: 38319581 DOI: 10.1007/978-1-0716-3633-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
An analysis of the mitochondrial respiration function represented by the oxygen consumption rate is necessary to assess mitochondrial bioenergetics and redox function. This protocol describes two alternative techniques to evaluate mitochondrial respiration function in situ: (1) measure oxygen consumption rates via an electrode; (2) measure oxygen consumption rates via a seahorse instrument. These in situ approaches provide more physiological access to mitochondria to evaluate mitochondrial respiration function in a relatively integrated cellular system.
Collapse
Affiliation(s)
- Anqi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuan Qin
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Ying Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoqun Zhen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guohua Gong
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
48
|
Santos EW, Khatoon S, Di Mise A, Zheng YM, Wang YX. Mitochondrial Dynamics in Pulmonary Hypertension. Biomedicines 2023; 12:53. [PMID: 38255160 PMCID: PMC10813473 DOI: 10.3390/biomedicines12010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Mitochondria are essential organelles for energy production, calcium homeostasis, redox signaling, and other cellular responses involved in pulmonary vascular biology and disease processes. Mitochondrial homeostasis depends on a balance in mitochondrial fusion and fission (dynamics). Mitochondrial dynamics are regulated by a viable circadian clock. Hypoxia and nicotine exposure can cause dysfunctions in mitochondrial dynamics, increases in mitochondrial reactive oxygen species generation and calcium concentration, and decreases in ATP production. These mitochondrial changes contribute significantly to pulmonary vascular oxidative stress, inflammatory responses, contractile dysfunction, pathologic remodeling, and eventually pulmonary hypertension. In this review article, therefore, we primarily summarize recent advances in basic, translational, and clinical studies of circadian roles in mitochondrial metabolism in the pulmonary vasculature. This knowledge may not only be crucial to fully understanding the development of pulmonary hypertension, but also greatly help to create new therapeutic strategies for treating this devastating disease and other related pulmonary disorders.
Collapse
Affiliation(s)
- Ed Wilson Santos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Subika Khatoon
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70125 Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| |
Collapse
|
49
|
Zhu Q, Combs ME, Liu J, Bai X, Wang WB, Herring LE, Liu J, Locasale JW, Bowles DE, Gross RT, Pla MM, Mack CP, Taylor JM. GRAF1 integrates PINK1-Parkin signaling and actin dynamics to mediate cardiac mitochondrial homeostasis. Nat Commun 2023; 14:8187. [PMID: 38081847 PMCID: PMC10713658 DOI: 10.1038/s41467-023-43889-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The serine/threonine kinase, PINK1, and the E3 ubiquitin ligase, Parkin, are known to facilitate LC3-dependent autophagosomal encasement and lysosomal clearance of dysfunctional mitochondria, and defects in this process contribute to a variety of cardiometabolic and neurological diseases. Although recent evidence indicates that dynamic actin remodeling plays an important role in PINK1/Parkin-mediated mitochondrial autophagy (mitophagy), the underlying signaling mechanisms remain unknown. Here, we identify the RhoGAP GRAF1 (Arhgap26) as a PINK1 substrate that regulates mitophagy. GRAF1 promotes the release of damaged mitochondria from F-actin anchors, regulates mitochondrial-associated Arp2/3-mediated actin remodeling and facilitates Parkin-LC3 interactions to enhance mitochondria capture by autophagosomes. Graf1 phosphorylation on PINK1-dependent sites is dysregulated in human heart failure, and cardiomyocyte-restricted Graf1 depletion in mice blunts mitochondrial clearance and attenuates compensatory metabolic adaptations to stress. Overall, we identify GRAF1 as an enzyme that coordinates cytoskeletal and metabolic remodeling to promote cardioprotection.
Collapse
Affiliation(s)
- Qiang Zhu
- Department of Pathology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Matthew E Combs
- Department of Pathology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Xue Bai
- Department of Pathology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Wenbo B Wang
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Laura E Herring
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jiandong Liu
- Department of Pathology, University of North Carolina, Chapel Hill, NC, 27599, USA
- McAllister Heart Institute University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Dawn E Bowles
- Division of Surgical Sciences, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ryan T Gross
- Division of Surgical Sciences, Duke University Medical Center, Durham, NC, 27710, USA
| | - Michelle Mendiola Pla
- Division of Surgical Sciences, Duke University Medical Center, Durham, NC, 27710, USA
| | - Christopher P Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC, 27599, USA
- McAllister Heart Institute University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Joan M Taylor
- Department of Pathology, University of North Carolina, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
50
|
Wang Y, Dai X, Li H, Jiang H, Zhou J, Zhang S, Guo J, Shen L, Yang H, Lin J, Yan H. The role of mitochondrial dynamics in disease. MedComm (Beijing) 2023; 4:e462. [PMID: 38156294 PMCID: PMC10753647 DOI: 10.1002/mco2.462] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/14/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023] Open
Abstract
Mitochondria are multifaceted and dynamic organelles regulating various important cellular processes from signal transduction to determining cell fate. As dynamic properties of mitochondria, fusion and fission accompanied with mitophagy, undergo constant changes in number and morphology to sustain mitochondrial homeostasis in response to cell context changes. Thus, the dysregulation of mitochondrial dynamics and mitophagy is unsurprisingly related with various diseases, but the unclear underlying mechanism hinders their clinical application. In this review, we summarize the recent developments in the molecular mechanism of mitochondrial dynamics and mitophagy, particularly the different roles of key components in mitochondrial dynamics in different context. We also summarize the roles of mitochondrial dynamics and target treatment in diseases related to the cardiovascular system, nervous system, respiratory system, and tumor cell metabolism demanding high-energy. In these diseases, it is common that excessive mitochondrial fission is dominant and accompanied by impaired fusion and mitophagy. But there have been many conflicting findings about them recently, which are specifically highlighted in this view. We look forward that these findings will help broaden our understanding of the roles of the mitochondrial dynamics in diseases and will be beneficial to the discovery of novel selective therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Xinyan Dai
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Hui Li
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huiling Jiang
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Junfu Zhou
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Shiying Zhang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jiacheng Guo
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Lidu Shen
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huantao Yang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jie Lin
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Hengxiu Yan
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| |
Collapse
|