1
|
Bai J, He S, Wang X, Zhang L, Ma C, Gao D, Yuan H, Mei J, Guan X, Yu H, Wan K, Zhu D. Mitochondrial Genome-Encoded lncND5 Regulates Mitophagy in Hypoxic Pulmonary Artery Smooth Muscle Cell. FASEB J 2025; 39:e70618. [PMID: 40364724 DOI: 10.1096/fj.202500389r] [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: 02/09/2025] [Revised: 04/17/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
Long noncoding RNAs (lncRNAs) are implicated in pulmonary hypertension (PH) progression. However, the underlying mechanisms remain largely unknown. Although mitophagy plays a crucial role in hypoxia-induced PH pathogenesis, the role of lncRNAs in mitophagy remains unclear. Especially, the mechanism of lncRNA encoded by the mitochondrial genome in regulating mitophagy needs to be elucidated. We explored the role of lncND5 in human pulmonary artery smooth muscle cells (PASMCs) and Sugen5416 plus hypoxia (SuHx)-induced PH mouse model in vitro and in vivo. LncND5 expression and localization were detected using real-time quantitative polymerase chain reaction (RT-qPCR) and fluorescence in situ hybridization (FISH). We investigated the molecular mechanism of lncND5 using western blotting, flow cytometry, RNA immunoprecipitation, RNA pulldown, transmission electron microscopy (TEM), immunofluorescence (IF), and echocardiography. Mitochondrial lncND5 expression was decreased under hypoxia in human PASMCs. Mechanistically, in the mitochondria, lncND5 maintains complex I activity by binding with mitochondrial ADH-ubiquinone oxidoreductase chain 5 (MT-ND5) at nucleotides 1086-1159 bp, thereby regulating mitochondrial reactive oxygen species (mROS) release and alleviating mitophagy. Additionally, lncND5 regulates mitophagy via cardiolipin (CL), which regulates complex I activity, inhibiting ROS release then relieving mitophagy. In the cytoplasm, lncND5 inhibits mitophagy by directly interacting with hydroxymethylglutaryl-CoA synthase 1 (HMGCS1). Notably, lncND5 is transported from the mitochondria to the cytoplasm and is mediated by TAR DNA-binding protein 43 (TDP-43). Our findings, for the first time, reveal that lncND5 may be a potential therapeutic approach for PH.
Collapse
MESH Headings
- Mitophagy/genetics
- Mitophagy/physiology
- Humans
- Animals
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/cytology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Male
- Hypoxia/metabolism
- Hypoxia/genetics
- Mitochondria/metabolism
- Mitochondria/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Cell Hypoxia
- Mice, Inbred C57BL
- Reactive Oxygen Species/metabolism
- Muscle, Smooth, Vascular/metabolism
- Cells, Cultured
Collapse
Affiliation(s)
- June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Siyu He
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Xiaoying Wang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Pharmacy, Harbin Medical University (Daqing), Daqing, P. R. China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, P. R. China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, P. R. China
| | - Danni Gao
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Hao Yuan
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Jian Mei
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, P. R. China
| | - Xiaoyu Guan
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Hang Yu
- Department of Physiology, Harbin Medical University (Daqing), Daqing, P. R. China
| | - Kuiyu Wan
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, P. R. China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, P. R. China
- College of Pharmacy, Harbin Medical University, Harbin, P. R. China
- Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin, P. R. China
| |
Collapse
|
2
|
Li J, Lin Q, Ren C, Li X, Li X, Li H, Li S. The perspective of modern transplant science - transplant arteriosclerosis: inspiration derived from mitochondria associated endoplasmic reticulum membrane dysfunction in arterial diseases. Int J Surg 2025; 111:3430-3440. [PMID: 40146783 DOI: 10.1097/js9.0000000000002362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
The mitochondria-associated endoplasmic reticulum membrane (MAM) is a crucial structure connecting mitochondria and the endoplasmic reticulum (ER), regulating intracellular calcium homeostasis, lipid metabolism, and various signaling pathways essential for arterial health. Recent studies highlight MAM's significant role in modulating vascular endothelial cells (EC) and vascular smooth muscle cells (VSMC), establishing it as a key regulator of arterial health and a contributor to vascular disease pathogenesis. Organ transplantation is the preferred treatment for end-stage organ failure, but transplant arteriosclerosis (TA) can lead to chronic transplant dysfunction, significantly impacting patient survival. TA, like other vascular diseases, features endothelial dysfunction and abnormal proliferation and migration of VSMC. Previous research on TA has focused on immune factors; the pathological and physiological changes in grafts following immune system attacks have garnered insufficient attention. For example, the potential roles of MAM in TA have not been thoroughly investigated. Investigating the relationship between MAM and TA, as well as the mechanisms behind TA progression, is essential. This review aims to outline the fundamental structure and the primary functions of MAM, summarize its key molecular regulators of vascular health, and explore future prospects for MAM in the context of TA research, providing insights for both basic research and clinical management of TA.
Collapse
Affiliation(s)
- Jingyi Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qian Lin
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Ren
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaodong Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaowei Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Haofeng Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Shadan Li
- Department of Urology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Michalak KP, Michalak AZ. Understanding chronic inflammation: couplings between cytokines, ROS, NO, Ca i 2+, HIF-1α, Nrf2 and autophagy. Front Immunol 2025; 16:1558263. [PMID: 40264757 PMCID: PMC12012389 DOI: 10.3389/fimmu.2025.1558263] [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: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Chronic inflammation is an important component of many diseases, including autoimmune diseases, intracellular infections, dysbiosis and degenerative diseases. An important element of this state is the mainly positive feedback between inflammatory cytokines, reactive oxygen species (ROS), nitric oxide (NO), increased intracellular calcium, hypoxia-inducible factor 1-alpha (HIF-1α) stabilisation and mitochondrial oxidative stress, which, under normal conditions, enhance the response against pathogens. Autophagy and the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response are mainly negatively coupled with the above-mentioned elements to maintain the defence response at a level appropriate to the severity of the infection. The current review is the first attempt to build a multidimensional model of cellular self-regulation of chronic inflammation. It describes the feedbacks involved in the inflammatory response and explains the possible pathways by which inflammation becomes chronic. The multiplicity of positive feedbacks suggests that symptomatic treatment of chronic inflammation should focus on inhibiting multiple positive feedbacks to effectively suppress all dysregulated elements including inflammation, oxidative stress, calcium stress, mito-stress and other metabolic disturbances.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
4
|
Hindmarch CCT, Potus F, Al‐Qazazi R, Ott BP, Nichols WC, Rauh MJ, Archer SL. Tet Methylcytosine Dioxygenase 2 (TET2) Mutation Drives a Global Hypermethylation Signature in Patients With Pulmonary Arterial Hypertension (PAH): Correlation With Altered Gene Expression Relevant to a Common T Cell Phenotype. Compr Physiol 2025; 15:e70011. [PMID: 40274312 PMCID: PMC12021535 DOI: 10.1002/cph4.70011] [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: 11/10/2024] [Revised: 03/17/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
Epigenetic changes in gene expression due to DNA methylation regulate pulmonary vascular structure and function. Genetic or acquired alterations in DNA methylation/demethylation can promote the development of pulmonary arterial hypertension (PAH). Here, we performed epigenome-wide mapping of DNA methylation in whole blood from 10 healthy people and 19 age/sex-matched PAH patients from the PAH Biobank. Exome sequencing confirmed the absence of known mutations in PAH-associated gene variants identifying subjects with or without mutations of TET2, a putative PAH gene encoding the demethylating enzyme, TET2. DNA of patients with PAH and no TET2 mutation was hypermethylated compared to healthy controls. Patients with PAH and a TET2 mutation had greater DNA CpG methylation than mutation-free PAH patients. Unique Differentially Methylated Regions (DMR) were more common in patients with PAH with TET2 mutations (1164) than in PAH without mutations (262). We correlated methylome findings with a public PAH transcriptomic RNA dataset, prioritizing targets that are both hypermethylated in our cohort and downregulated at the RNA level. Relative to controls, functional analysis reveals enriched functions related to T cell differentiation in PAH patients with a TET2 mutation. We identified genes with downregulated expression that were hypermethylated in PAH patients (with or without a TET2 mutation). In both cases, a conserved T cell phenotype emerged. Pan-chromosomal hypermethylation in PAH is greatest in patients with TET2 mutations. Observed hypermethylation of genes involved in the pathogenesis of PAH, such as EIF2AK4, and transcription factors that regulate T cell development, such as TCF7, merit further study and may contribute to the inflammation in PAH.
Collapse
Affiliation(s)
- Charles C. T. Hindmarch
- Department of Biomedical and Molecular Science (DBMS)Queen's UniversityKingstonOntarioCanada
- Department of MedicineQueen's UniversityKingstonOntarioCanada
- Queen's CardioPulmonary Unit, Translational Institute of Medicine (TIME), Department of MedicineQueen's UniversityKingstonOntarioCanada
| | - François Potus
- Pulmonary Hypertension Research GroupCenter de Recherche de L'institut Universitaire de Cardiologie et de Pneumologie de QuébecQuebecCanada
| | - Ruaa Al‐Qazazi
- Department of MedicineQueen's UniversityKingstonOntarioCanada
| | - Benjamin P. Ott
- Department of MedicineQueen's UniversityKingstonOntarioCanada
- Queen's CardioPulmonary Unit, Translational Institute of Medicine (TIME), Department of MedicineQueen's UniversityKingstonOntarioCanada
| | - William C. Nichols
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical CenterUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Michael J. Rauh
- Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
| | - Stephen L. Archer
- Department of MedicineQueen's UniversityKingstonOntarioCanada
- Queen's CardioPulmonary Unit, Translational Institute of Medicine (TIME), Department of MedicineQueen's UniversityKingstonOntarioCanada
| |
Collapse
|
5
|
Sun X, Yegambaram M, Lu Q, Garcia Flores AE, Pokharel MD, Soto J, Aggarwal S, Wang T, Fineman JR, Black SM. Mitochondrial fission produces a Warburg effect via the oxidative inhibition of prolyl hydroxylase domain-2. Redox Biol 2025; 81:103529. [PMID: 39978304 PMCID: PMC11889635 DOI: 10.1016/j.redox.2025.103529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025] Open
Abstract
Excessive mitochondrial fission and a shift to a Warburg phenotype are hallmarks of pulmonary hypertension (PH), although the mechanistic link between these processes remains unclear. We show that in pulmonary arterial endothelial cells (PAEC), Drp1 overexpression induces mitochondrial fission and increases glycolytic ATP production and glycolysis. This is due to mitochondrial reactive oxygen species (mito-ROS)-mediated activation of hypoxia-inducible factor-1α (HIF-1α) signaling, and this is linked to hydrogen peroxide (H2O2)-mediated inhibition of prolyl hydroxylase domain-2 (PHD2) due to its cysteine 326 oxidation and dimerization. Furthermore, these findings are validated in PAEC isolated from a lamb model of PH, which are glycolytic (Shunt PAEC), exhibit increases in both H2O2 and PHD2 dimer levels. The overexpression of catalase reversed the PHD2 dimerization, decreased HIF-1α levels, and attenuated glycolysis in Shunt PAEC. Our data suggest that reducing PHD2 dimerization could be a potential therapeutic target for PH.
Collapse
Affiliation(s)
- Xutong Sun
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Manivannan Yegambaram
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Qing Lu
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Alejandro E Garcia Flores
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Marissa D Pokharel
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jamie Soto
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Saurabh Aggarwal
- The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jeffrey R Fineman
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; The Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA; The Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
6
|
Peng TY, Lu JM, Zheng XL, Zeng C, He YH. The role of lactate metabolism and lactylation in pulmonary arterial hypertension. Respir Res 2025; 26:99. [PMID: 40075458 PMCID: PMC11905457 DOI: 10.1186/s12931-025-03163-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex and progressive disease characterized by elevated pulmonary artery pressure and vascular remodeling. Recent studies have underscored the pivotal role of metabolic dysregulation and epigenetic modifications in the pathogenesis of PAH. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that links cellular metabolism with activity regulation. Recent findings indicate that, in addition to altered glycolytic activity and dysregulated. Lactate homeostasis and lactylation-a novel epigenetic modification-also play a significant role in the development of PAH. This review synthesizes current knowledge regarding the relationship between altered glycolytic activity and PAH, with a particular focus on the cumulative effects of lactate in pulmonary vascular cells. Furthermore, lactylation, an emerging epigenetic modification, is discussed in the context of PAH. By elucidating the complex interplay between lactate metabolism and lactylation in PAH, this review aims to provide insights into potential therapeutic targets. Understanding these metabolic pathways may lead to innovative strategies for managing PAH and improving patient outcomes. Future research should focus on the underlying mechanisms through which lactylation influences the pathophysiology of PAH, thereby aiding in the development of targeted interventions.
Collapse
Affiliation(s)
- Tong-Yu Peng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jun-Mi Lu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xia-Lei Zheng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Cheng Zeng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yu-Hu He
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
7
|
Chen ZH, Cao SH, Ren ZY, Zhang T, Jiang HM, Hu ZK, Dong LH. Lactate Dehydrogenase A Crotonylation and Mono-Ubiquitination Maintains Vascular Smooth Muscle Cell Growth and Migration and Promotes Neointima Hyperplasia. J Am Heart Assoc 2025; 14:e036377. [PMID: 40028887 DOI: 10.1161/jaha.124.036377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Phenotypic plasticity of vascular smooth muscle cells (VSMCs) is believed to be a key factor in neointima hyperplasia, which is the pathological basis of vascular remodeling diseases. LDHA (lactate dehydrogenase A) has been demonstrated to promote the proliferation and migration of VSMCs. However, the mechanism is still unclear. METHODS AND RESULTS LDHA ubiquitination and crotonylation in VSMCs were predicted by modified omics and proteomic analysis and were verified by immunoprecipitation. Lysine mutants of LDHA were conducted to determine the specific modified sites. Immunofluorescent staining, cell growth and migration assays, lactate production, immunobloting, adenovirus transduction, LDHA tetramerization, and mitochondrial extraction assays were performed to determine the molecular mechanism. LDHA expression, crotonylation, and ubiquitination in vivo were observed in the carotid arteries of ligation injury mice. We showed that the expression, crotonylation, and mono-ubiquitination of LDHA is upregulated in PDGF-BB (platelet-derived growth factor-BB)-induced proliferative VSMCs and ligation-induced neointima. LDHA is crotonylated at lysine 5 and is mono-ubiquitinated at K76. Crotonylation at lysine 5 activates LDHA through tetramer formation to enhance lactate production and VSMC growth. Mono-ubiquitination at K76 induces the translocation of LDHA into mitochondria, which promotes mitochondria fission and subsequent formation of lamellipodia and podosomes, thereby enhancing VSMC migration and growth. Furthermore, deletion of LDHA K5 crotonylation or K76 mono-ubiquitination decreases ligation-induced neointima formation. CONCLUSIONS Our study reveals a novel mechanism that combines VSMC metabolic reprogramming and vascular remodeling. Inhibition of LDHA K5 crotonylation or K76 mono-ubiquitination may be a promising approach for the therapy of vascular remodeling diseases.
Collapse
Affiliation(s)
- Zhi-Huan Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
- Hebei Special Education Collaborative Innovation Center School of Special Education, Handan University Handan China
| | - Shan-Hu Cao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Zhi-Yan Ren
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Ting Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Han-Mei Jiang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Zhao-Kun Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Li-Hua Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| |
Collapse
|
8
|
Wang Y, Wang Y, Zhang W. Dysregulation of Mitochondrial in Pulmonary Hypertension-Related Right Ventricular Remodeling: Pathophysiological Features and Targeting Drugs. Rev Cardiovasc Med 2025; 26:25781. [PMID: 40160582 PMCID: PMC11951289 DOI: 10.31083/rcm25781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 04/02/2025] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening condition characterized by right ventricular (RV) remodeling, which is a major determinant of patient survival. The progression of right ventricular remodeling is significantly influenced by mitochondrial dysfunction, providing profound insights into vascular health and cardiovascular risk. In this review, we discuss the molecular targets, pathophysiological characteristics, and potential mechanisms underlying mitochondrial dysfunction in PH, encompassing disturbances in mitochondrial dynamics, inflammation, and dysregulation of mitochondrial energy metabolism. Finally, we review the primary therapeutic targets currently utilized to address cardiac dysfunction resulting from mitochondrial damage. Hopefully, this might inspire novel approaches to the management of cardiovascular disorders.
Collapse
Affiliation(s)
- Yuehan Wang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Yingzhuo Wang
- The First Clinical Medical College, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Weifang Zhang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Zhang J, Yan H, Wang Y, Yue X, Wang M, Liu L, Qiao P, Zhu Y, Li Z. Emerging insights into pulmonary hypertension: the potential role of mitochondrial dysfunction and redox homeostasis. Mol Cell Biochem 2025; 480:1407-1429. [PMID: 39254871 DOI: 10.1007/s11010-024-05096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Pulmonary hypertension (PH) is heterogeneous diseases that can lead to death due to progressive right heart failure. Emerging evidence suggests that, in addition to its role in ATP production, changes in mitochondrial play a central role in their pathogenesis, regulating integrated metabolic and signal transduction pathways. This review focuses on the basic principles of mitochondrial redox status in pulmonary vascular and right ventricular disorders, a series of dysfunctional processes including mitochondrial quality control (mitochondrial biogenesis, mitophagy, mitochondrial dynamics, mitochondrial unfolded protein response) and mitochondrial redox homeostasis. In addition, we will summarize how mitochondrial renewal and dynamic changes provide innovative insights for studying and evaluating PH. This will provide us with a clearer understanding of the initial signal transmission of mitochondria in PH, which would further improve our understanding of the pathogenesis of PH.
Collapse
Affiliation(s)
- Junming Zhang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Huimin Yan
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yan Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Xian Yue
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Meng Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Limin Liu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Pengfei Qiao
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yixuan Zhu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Zhichao Li
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China.
| |
Collapse
|
10
|
Wang Z, Sun W, Zhang K, Ke X, Wang Z. New insights into the relationship of mitochondrial metabolism and atherosclerosis. Cell Signal 2025; 127:111580. [PMID: 39732307 DOI: 10.1016/j.cellsig.2024.111580] [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: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Atherosclerotic cardiovascular and cerebrovascular diseases are the number one killer of human health. In view of the important role of mitochondria in the formation and evolution of atherosclerosis, our manuscript aims to comprehensively elaborate the relationship between mitochondria and the formation and evolution of atherosclerosis from the aspects of mitochondrial dynamics, mitochondria-organelle interaction (communication), mitochondria and cell death, mitochondria and vascular smooth muscle cell phenotypic switch, etc., which is combined with genome, transcriptome and proteome, in order to provide new ideas for the pathogenesis of atherosclerosis and the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Zexun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China
| | - Wangqing Sun
- Department of Radiology, Yixing Tumor Hospital, Yixing 214200, China
| | - Kai Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
11
|
Rahimi A, Geraiely B, Vahidi H, Eftekhari M, Rahimi Darehbagh R, Mohammadi S, Ghaffari G, Khoshavi M. Augmentation index as a predictor of right ventricular dysfunction in coronary artery disease: a cross-sectional study. BMC Cardiovasc Disord 2025; 25:71. [PMID: 39893481 PMCID: PMC11786491 DOI: 10.1186/s12872-024-04444-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Arterial stiffness is a well-established predictor of cardiovascular events and mortality. However, its relationship with right ventricular (RV) function in patients with coronary artery disease (CAD) remains unclear. We aimed to investigate the association between aortic augmentation index (AIx), a marker of arterial stiffness, and RV dysfunction in CAD patients. METHODS In this cross-sectional study, 121 patients with stable CAD or acute coronary syndrome who underwent coronary angiography were enrolled. AIx was measured using radial artery applanation tonometry. Comprehensive echocardiography was performed to assess RV function using conventional and speckle-tracking derived parameters. Multivariable linear and logistic regression analyses were used to evaluate the relationship between AIx and RV function, adjusting for potential confounders. RESULTS Patients with high AIx (> 80%, n = 53) had significantly worse RV systolic function compared to those with normal AIx (≤ 80%, n = 68), as evidenced by lower tricuspid annular plane systolic excursion (TAPSE), fractional area change (FAC), RV free wall longitudinal strain (RVLS), and RV systolic velocity (RV S') (all P < 0.05). AIx correlated negatively with TAPSE (r=-0.421), FAC (r=-0.376), RVLS (r=-0.428), and RV S' (r=-0.355) (all P < 0.001), and positively with pulmonary artery systolic pressure (r = 0.467, P < 0.001) and pulmonary vascular resistance (r = 0.297, P = 0.001). In multivariable analyses, AIx remained an independent predictor of RV dysfunction (adjusted odds ratio 3.42, 95% confidence interval 1.56-7.51, P = 0.002) after adjusting for age, sex, hypertension, diabetes, dyslipidemia, smoking, left ventricular ejection fraction, and Gensini score. CONCLUSIONS Increased aortic stiffness assessed by AIx is independently associated with RV dysfunction in patients with CAD. This association is evident across multiple echocardiographic parameters of RV systolic function and is independent of traditional cardiovascular risk factors, left ventricular systolic function, and the extent of coronary artery disease. Our findings suggest that arterial stiffness may play a role in the development of RV dysfunction in CAD patients and highlight the potential importance of assessing and targeting arterial stiffness in this population.
Collapse
Affiliation(s)
- Arash Rahimi
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Geraiely
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Vahidi
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Eftekhari
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramyar Rahimi Darehbagh
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Universal Scientific Education and Research Network (USERN), Sanandaj, Kurdistan, Iran
| | - Somayeh Mohammadi
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Golrokh Ghaffari
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Meysam Khoshavi
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Xie S, Zhao J, Zhang F, Li X, Yu X, Shu Z, Cheng H, Liu S, Shi S. Dehydrodiisoeugenol inhibits PDGF-BB-induced proliferation and migration of human pulmonary artery smooth muscle cells via the mTOR/HIF1-α/HK2 signaling pathway. Toxicol Appl Pharmacol 2025; 495:117212. [PMID: 39719250 DOI: 10.1016/j.taap.2024.117212] [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: 11/05/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
Abnormal proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) leading to pulmonary vascular remodeling are critical factors in the development of pulmonary hypertension (pH). Dehydrodiisoeugenol (DEH), a natural phenolic compound, is renowned for its antioxidant and anti-inflammatory properties. However, the precise role and mechanisms of DEH in PH remain unclear. In this study, human PASMCs were exposed to PDGF-BB for 48 h to establish an in vitro model. Subsequently, cells were treated with DEH, and assessments of cell proliferation, migration, and apoptosis were performed using CCK-8/EdU assays, scratch/transwell assays, and flow cytometry. The results showed that PDGF-BB induced phenotypic modulation, proliferation, and migration of PASMCs while reducing apoptosis. Treatment with DEH effectively reversed these effects. Bioinformatics analysis identified mTOR as a target of DEH action. Western blot experiments were conducted to evaluate the expression of proteins involved in the mTOR/HIF1-α/HK2 signaling pathway, suggesting that DEH modulates this pathway by targeting and inhibiting mTOR. After treating cells with mTOR inhibitors, cellular glycolysis was assessed using the extracellular acidification rate (ECAR) assay. The results indicated that inhibition of mTOR phosphorylation decreased aerobic glycolysis in PASMCs and suppressed cell proliferation, migration, and apoptosis resistance, regardless of PDGF-BB treatment. Activation of mTOR reversed the inhibition of PDGF-BB-induced PASMC-related protein expression by DEH. These findings suggest that DEH inhibits aerobic glycolysis in PDGF-BB-induced PASMCs through the mTOR/HIF1-α/HK2 signaling pathway, thereby suppressing cell proliferation, migration, and resistance to apoptosis. Consequently, DEH holds promise as a novel therapeutic agent for treating pulmonary arterial hypertension.
Collapse
MESH Headings
- Humans
- Pulmonary Artery/drug effects
- Pulmonary Artery/cytology
- Pulmonary Artery/pathology
- Pulmonary Artery/enzymology
- Cell Proliferation/drug effects
- TOR Serine-Threonine Kinases/metabolism
- Cell Movement/drug effects
- Signal Transduction/drug effects
- Becaplermin/pharmacology
- Becaplermin/toxicity
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Eugenol/pharmacology
- Eugenol/analogs & derivatives
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Cells, Cultured
- Apoptosis/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
Collapse
Affiliation(s)
- Shishun Xie
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun 130000, China; Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jianjun Zhao
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Fan Zhang
- Qingdao Municipal Hospital, Qingdao 266000, China
| | - Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun 130000, China
| | - Xiaoyan Yu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun 130000, China
| | - Zhiyun Shu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun 130000, China
| | - Hongyuan Cheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun 130000, China
| | - Siyao Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun 130000, China
| | - Shaomin Shi
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
13
|
Fujiwara-Tani R, Nakashima C, Ohmori H, Fujii K, Luo Y, Sasaki T, Ogata R, Kuniyasu H. Significance of Malic Enzyme 1 in Cancer: A Review. Curr Issues Mol Biol 2025; 47:83. [PMID: 39996805 PMCID: PMC11854147 DOI: 10.3390/cimb47020083] [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: 01/02/2025] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Malic enzyme 1 (ME1) plays a key role in promoting malignant phenotypes in various types of cancer. ME1 promotes epithelial-mesenchymal transition (EMT) and enhances stemness via glutaminolysis, energy metabolism reprogramming from oxidative phosphorylation to glycolysis. As a result, ME1 promotes the malignant phenotypes of cancer cells and poor patient prognosis. In particular, ME1 expression is promoted in hypoxic environments associated with hypoxia-inducible factor (HIF1) α. ME1 is overexpressed in budding cells at the cancer invasive front, promoting cancer invasion and metastasis. ME1 also generates nicotinamide adenine dinucleotide (NADPH), which, together with glucose-6-phosphate dehydrogenase (G6PD) and isocitrate dehydrogenase (IDH1), expands the NADPH pool, maintaining the redox balance in cancer cells, suppressing cell death by neutralizing mitochondrial reactive oxygen species (ROS), and promoting stemness. This review summarizes the latest research insights into the mechanisms by which ME1 contributes to cancer progression. Because ME1 is involved in various aspects of cancer and promotes many of its malignant phenotypes, it is expected that ME1 will become a novel drug target in the near future.
Collapse
Affiliation(s)
- Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara 634-8521, Japan; (C.N.); (H.O.); (K.F.); (Y.L.); (T.S.); (R.O.)
| | | | | | | | | | | | | | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara 634-8521, Japan; (C.N.); (H.O.); (K.F.); (Y.L.); (T.S.); (R.O.)
| |
Collapse
|
14
|
Zhang X, Li J, Fu M, Geng X, Hu J, Tang KJ, Chen P, Zou J, Liu X, Zeng B. Dysfunction in mitochondrial electron transport chain drives the pathogenesis of pulmonary arterial hypertension: insights from a multi-omics investigation. Respir Res 2025; 26:29. [PMID: 39833797 PMCID: PMC11749457 DOI: 10.1186/s12931-025-03099-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive disorder that can lead to right ventricular failure and severe consequences. Despite extensive efforts, limited progress has been made in preventing the progression of PAH. Mitochondrial dysfunction is implicated in the development of PAH, but the key mitochondrial functional alterations in the pathogenesis have yet to be elucidated. METHODS We integrated three microarray datasets from the Gene Expression Omnibus (GEO), including 222 lung samples (164 PAH, 58 controls), for differential expression and functional enrichment analyses. Machine learning identified key mitochondria-related signaling pathways. PAH and control lung tissue samples were collected, and transcriptomic and metabolomic profiling were performed. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis investigated shared pathways, and canonical correlation analysis assessed gene-metabolite relationships. RESULTS In the GEO datasets, mitochondria-related signaling pathways were significantly enriched in PAH samples, in particular the electron transport chain (ETC) in mitochondrial oxidative phosphorylation system. Notably, the electron transport from cytochrome c to oxygen in ETC was identified as the most crucial mitochondria-related pathway, which was down-regulated in PAH samples. Transcriptomic profiling of the clinical lung tissue analysis identified 14 differentially expressed genes (DEGs) related to mitochondrial function. Metabolomic analysis revealed three differential metabolites in PAH samples: increased 3-phenyllactic acid and ADP, and decreased citric acid. Mitochondria-related genes highly correlated with these metabolites included KIT, OTC, CAMK2A, and CHRNA1. CONCLUSIONS Down-regulation of electron transport from cytochrome c to oxygen in mitochondrial ETC and disruption of the citric acid cycle homeostasis may contribute to PAH pathogenesis. 3-phenyllactic acid emerges as a potential novel diagnostic biomarker for PAH. These findings offer insights for developing novel PAH therapies and diagnostics.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jieling Li
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Minyi Fu
- Surgical and Anesthesia Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xijie Geng
- Surgical and Anesthesia Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junjie Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ke-Jing Tang
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pan Chen
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianyong Zou
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Xiaoman Liu
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Bo Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
15
|
Belenichev I, Popazova O, Bukhtiyarova N, Ryzhenko V, Pavlov S, Suprun E, Oksenych V, Kamyshnyi O. Targeting Mitochondrial Dysfunction in Cerebral Ischemia: Advances in Pharmacological Interventions. Antioxidants (Basel) 2025; 14:108. [PMID: 39857442 PMCID: PMC11760872 DOI: 10.3390/antiox14010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
The study of mitochondrial dysfunction has become increasingly pivotal in elucidating the pathophysiology of various cerebral pathologies, particularly neurodegenerative disorders. Mitochondria are essential for cellular energy metabolism, regulation of reactive oxygen species (ROS), calcium homeostasis, and the execution of apoptotic processes. Disruptions in mitochondrial function, driven by factors such as oxidative stress, excitotoxicity, and altered ion balance, lead to neuronal death and contribute to cognitive impairments in several brain diseases. Mitochondrial dysfunction can arise from genetic mutations, ischemic events, hypoxia, and other environmental factors. This article highlights the critical role of mitochondrial dysfunction in the progression of neurodegenerative diseases and discusses the need for targeted therapeutic strategies to attenuate cellular damage, restore mitochondrial function, and enhance neuroprotection.
Collapse
Affiliation(s)
- Igor Belenichev
- Department of Pharmacology and Medical Formulation with Course of Normal Physiology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine;
| | - Olena Popazova
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Nina Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Victor Ryzhenko
- Department of Medical and Pharmaceutical Informatics and Advanced Technologies, Zaporizhzhia State Medical University, 69000 Zaporizhzhia, Ukraine
| | - Sergii Pavlov
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical and Pharmaceutical University, 69000 Zaporizhzhia, Ukraine
| | - Elina Suprun
- The State Institute of Neurology, Psychiatry and Narcology of the National Academy of Medical Sciences of Ukraine, 46 Academician Pavlov Street, 61076 Kharkov, Ukraine
| | | | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, I. Horbachevsky Ternopil State Medical University, 46001 Ternopil, Ukraine;
| |
Collapse
|
16
|
Fujiwara-Tani R, Luo Y, Ogata R, Fujii K, Sasaki T, Sasaki R, Nishiguchi Y, Mori S, Ohmori H, Kuniyasu H. Energy Metabolism and Stemness and the Role of Lauric Acid in Reversing 5-Fluorouracil Resistance in Colorectal Cancer Cells. Int J Mol Sci 2025; 26:664. [PMID: 39859378 PMCID: PMC11766121 DOI: 10.3390/ijms26020664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
While 5-fluorouracil (5FU) plays a central role in chemotherapy for colorectal cancer (CRC), resistance to 5FU remains a major challenge in CRC treatment, and its underlying mechanisms remain unclear. In this study, we investigated the relationship between 5FU resistance acquisition, stemness, and energy metabolism. Among the two CRC cell lines, HT29 cells exhibited glycolytic and quiescent properties, while CT26 cells relied on oxidative phosphorylation (OXPHOS) for energy. In contrast, the 5FU-resistant sublines (HT29R and CT26R), developed through continuous exposure to low concentrations of 5FU, demonstrated enhanced stemness. This was associated with glycolytic dominance, low proliferation, and reduced reactive oxygen species (ROS) production. However, treatment with the medium-chain fatty acid lauric acid shifted the cells to OXPHOS, reducing stemness, increasing ROS levels, and inducing cell death, therefore reversing 5FU resistance. These findings suggest that an enhancement in stemness and the reprogramming of energy metabolism play key roles in acquiring 5FU resistance in CRC. While lauric acid reversed 5FU resistance, further clinical studies are required.
Collapse
Grants
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 23K19900 Ministry of Education, Culture, Sports, Science and Technology
- 23K10481 Ministry of Education, Culture, Sports, Science and Technology
- 21K11223 Ministry of Education, Culture, Sports, Science and Technology
- 22K16497 Ministry of Education, Culture, Sports, Science and Technology
- 21K06926 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Yi Luo
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Ruiko Ogata
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Rika Sasaki
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka 573-1010, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan; (Y.L.); (R.O.); (K.F.); (T.S.); (R.S.); (Y.N.); (S.M.); (H.O.)
| |
Collapse
|
17
|
You Q, Song H, Zhu Z, Wang J, Wang R, Du M, Fu Y, Yuan J, Tan R. Decoding the enigmatic estrogen paradox in pulmonary hypertension: delving into estrogen metabolites and metabolic enzymes. Cell Mol Biol Lett 2024; 29:155. [PMID: 39695964 DOI: 10.1186/s11658-024-00671-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: 06/07/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Pulmonary hypertension (PH) presents a puzzling sex bias, being more prevalent in women yet often less severe than in men, and the underlying reasons remain unclear. Studies using animal models, and limited clinical data have revealed a protective influence of exogenous estrogens, known as the estrogen paradox. Research suggests that beyond its receptor-mediated effects, estrogen acts through metabolites such as 2-ME2, 4-OHE2, and 16-OHE2, which are capable of exhibiting protective or detrimental effects in PH, prompting the need to explore their roles in PH to untangle sex differences and the estrogen paradox. Hypoxia disrupts the balance of estrogen metabolites by affecting the enzymes responsible for estrogen metabolism. Delving into the role of these metabolic enzymes not only illuminates the sex difference in PH but also provides a potential rationale for the estrogen paradox. This review delves into the intricate interplay between estrogen metabolites, metabolic enzymes, and PH, offering a deeper understanding of sex-specific differences and the perplexing estrogen paradox in the context of this condition.
Collapse
Affiliation(s)
- Qiang You
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Hequn Song
- First Clinical Medical School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ziming Zhu
- College of Second Clinical Medical, Jining Medical University, Jining, 272067, Shandong, China
| | - Jinzheng Wang
- College of Second Clinical Medical, Jining Medical University, Jining, 272067, Shandong, China
| | - Ruixin Wang
- School of Nursing, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Mingjia Du
- School of Nursing, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yingjie Fu
- School of Pharmacy, Jining Medical University, Rizhao, 276826, Shandong, China.
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, 272067, Shandong, China.
| | - Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
18
|
Hou D, Liu R, Hao S, Dou Y, Chen G, Liu L, Li T, Cao Y, Huang H, Duan C. Notoginsenoside R1 improves intestinal microvascular functioning in sepsis by targeting Drp1-mediated mitochondrial quality imbalance. PHARMACEUTICAL BIOLOGY 2024; 62:250-260. [PMID: 38389274 PMCID: PMC10896147 DOI: 10.1080/13880209.2024.2318349] [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: 03/07/2023] [Accepted: 02/08/2024] [Indexed: 02/24/2024]
Abstract
CONTEXT Sepsis can result in critical organ failure, and notoginsenoside R1 (NGR1) offers mitochondrial protection. OBJECTIVE To determine whether NGR1 improves organ function and prognosis after sepsis by protecting mitochondrial quality. MATERIALS AND METHODS A sepsis model was established in C57BL/6 mice using cecum ligation puncture (CLP) and an in vitro model with lipopolysaccharide (LPS, 10 µg/mL)-stimulated primary intestinal microvascular endothelial cells (IMVECs) and then determine NGR1's safe dosage. Groups for each model were: in vivo-a control group, a CLP-induced sepsis group, and a CLP + NGR1 treatment group (30 mg/kg/d for 3 d); in vitro-a control group, a LPS-induced sepsis group, and a LPS + NGR1 treatment group (4 μM for 30 min). NGR1's effects on survival, intestinal function, mitochondrial quality, and mitochondrial dynamic-related protein (Drp1) were evaluated. RESULTS Sepsis resulted in approximately 60% mortality within 7 days post-CLP, with significant reductions in intestinal microvascular perfusion and increases in vascular leakage. Severe mitochondrial quality imbalance was observed in IMVECs. NGR1 (IC50 is 854.1 μM at 30 min) targeted Drp1, inhibiting mitochondrial translocation, preventing mitochondrial fragmentation and restoring IMVEC morphology and function, thus protecting against intestinal barrier dysfunction, vascular permeability, microcirculatory flow, and improving sepsis prognosis. DISCUSSION AND CONCLUSIONS Drp1-mediated mitochondrial quality imbalance is a potential therapeutic target for sepsis. Small molecule natural drugs like NGR1 targeting Drp1 may offer new directions for organ protection following sepsis. Future research should focus on clinical trials to evaluate NGR1's efficacy across various patient populations, potentially leading to novel treatments for sepsis.
Collapse
Affiliation(s)
- Dongyao Hou
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Ruixue Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Shuai Hao
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| | - Yong Dou
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Guizhen Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Liangming Liu
- Department of Shock and Transfusion, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Tao Li
- Department of Shock and Transfusion, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Yunxing Cao
- Department of Intensive Care Unit, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
19
|
Archer SL, Dunham-Snary KJ, Bentley R, Alizadeh E, Weir EK. Hypoxic Pulmonary Vasoconstriction: An Important Component of the Homeostatic Oxygen Sensing System. Physiol Res 2024; 73:S493-S510. [PMID: 39589299 PMCID: PMC11627260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/20/2024] [Indexed: 11/27/2024] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) rapidly and reversibly matches lung ventilation (V) and perfusion (Q), optimizing oxygen uptake and systemic oxygen delivery. HPV occurs in small pulmonary arteries (PA), which uniquely constrict to hypoxia. Although HPV is modulated by the endothelium the core mechanism of HPV resides in PA smooth muscle cells (PASMC). The PASMC's mitochondrial oxygen sensor lies within the electron transport chain (ETC) and includes NDUFS2 in ETC Complex-I. PASMC mitochondria respond to hypoxia by varying production of reactive oxygen species (ROS) and hydrogen peroxide in proportion to alveolar oxygen tension. Hypoxic ROS inhibition results in a state of reduction which triggers a redox-mediated inhibition of oxygen-sensitive, voltage-gated, potassium channels, including Kv1.5 and Kv2.1. Kv channel inhibition depolarizes the PASMC, opening of large-conductance calcium channels (CaL), elevating cytosolic calcium and activating the contractile apparatus. HPV is strongest in small PAs where sensors (hypoxia-responsive mitochondria) and effectors (oxygen-sensitive K+ channels) are enriched. Oxygenation at birth reverses fetal HPV, contributing to the rapid neonatal drop in pulmonary vascular resistance (PVR). A similar mitochon-drial-K+ channel sensor-effector mechanism exists in the ductus arteriosus (DA), however in DASMC it is oxygen-induced increases in mitochondrial ROS that inhibit DASMC K+ channels, causing DA constriction. Atelectasis and pneumonia elicit HPV, which optimises V/Q matching, increasing systemic oxygenation. Whilst HPV in response to localized hypoxia in a single lung lobe does not increase PA pressure; global airway hypoxia, as occurs with altitude or sleep apnea, causes pulmonary hypertension. HPV can be inhibited by drugs, including calcium channel blockers, or used to maintain a dry operative field during single lung anesthesia for lung surgery. HPV does not normally cause lung edema but excessive, heterogenous HPV contributes to high altitude pulmonary edema. HPV is suppressed in COVID-19 pneumonia by a SARS-CoV-2 mitochondriopathy. HPV is a component of the body's homeostatic oxygen sensing system. Keywords: Ductus arteriosus, Redox, NDUFS2, Oxygen sensitive potassium, Channels, High altitude pulmonary edema (HAPE), Mitochondrial electron transport chain, COVID-19 pneumonia, Atelectasis.
Collapse
Affiliation(s)
- S L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| | | | | | | | | |
Collapse
|
20
|
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
|
21
|
Lu X, Wang Y, Geng N, Zou Z, Feng X, Wang Y, Xu Z, Zhang N, Pu J. Dysregulated Mitochondrial Calcium Causes Spiral Artery Remodeling Failure in Preeclampsia. Hypertension 2024; 81:2368-2382. [PMID: 39291377 DOI: 10.1161/hypertensionaha.124.23046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Calcium deficiency in women is strongly linked to an increased risk of developing preeclampsia. Mitochondrial calcium ([Ca2+]m) homeostasis is essential to regulate vascular smooth muscle cell (VSMC) function. However, the role of [Ca2+]m in preeclampsia development remains largely unknown. METHODS To investigate this, human spiral arteries obtained from normotensive and preeclamptic women were collected for vascular function, RNA sequencing, and VSMC studies. N(ω)-nitro-L-arginine methyl ester-induced preeclampsia animal experiments were established to investigate the effects of intervening in [Ca2+]m to improve the outcome for preeclamptic mothers or their infants. RESULTS Our initial findings revealed compromised vessel function in spiral arteries derived from patients with preeclampsia, as evidenced by diminished vasoconstriction and vasodilation responses to angiotensin II and sodium nitroprusside, respectively. Moreover, the spiral artery VSMCs from patients with preeclampsia exhibited phenotypic transformation and proliferation associated with the disrupted regulatory mechanisms of [Ca2+]m uptake. Subsequent in vitro experiments employing gain- and loss-of-function approaches demonstrated that the mitochondrial Na+/Ca2+ exchanger played a role in promoting phenotypic switching and impaired mitochondrial functions in VSMCs. Furthermore, mtNCLX (mitochondrial Na+/Ca2+ exchanger) inhibitor CGP37157 significantly improved VSMC phenotypic changes and restored mitochondrial function in both patients with preeclampsia-derived VSMCs and the preeclampsia rat model. CONCLUSIONS This study provides comprehensive evidence supporting the disrupted regulatory mechanisms of [Ca2+]m uptake in VSMCs of spiral arteries of patients with preeclampsia and further elucidates its correlation with VSMC phenotypic switching and defective spiral artery remodeling. The findings suggest that targeting mtNCLX holds promise as a novel therapeutic approach for managing preeclampsia.
Collapse
Affiliation(s)
- Xiyuan Lu
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yifan Wang
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Na Geng
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zhiguo Zou
- Department of Cardiology (X.L., Yifan Wang, N.G., Z.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xueqing Feng
- Department of Obstetrics Affiliated Hospital of Jining Medical University, China (X.F.)
| | - Yuehong Wang
- State Key Laboratory of Systems Medicine for Cancer (Yuehong Wang), School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, China
| | - Zhice Xu
- Wuxi Maternity and Child Health Care Hospital, China (Z.X.)
| | - Ning Zhang
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology (N.Z.), Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital State Key Laboratory of Systems Medicine for Cancer (J.P.), School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, China
| |
Collapse
|
22
|
Xiao W, Lee LY, Loscalzo J. Metabolic Responses to Redox Stress in Vascular Cells. Antioxid Redox Signal 2024; 41:793-817. [PMID: 38985660 PMCID: PMC11876825 DOI: 10.1089/ars.2023.0476] [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: 10/19/2023] [Accepted: 11/11/2023] [Indexed: 07/12/2024]
Abstract
Significance: Redox stress underlies numerous vascular disease mechanisms. Metabolic adaptability is essential for vascular cells to preserve energy and redox homeostasis. Recent Advances: Single-cell technologies and multiomic studies demonstrate significant metabolic heterogeneity among vascular cells in health and disease. Increasing evidence shows that reductive or oxidative stress can induce metabolic reprogramming of vascular cells. A recent example is intracellular L-2-hydroxyglutarate accumulation in response to hypoxic reductive stress, which attenuates the glucose flux through glycolysis and mitochondrial respiration in pulmonary vascular cells and provides protection against further reductive stress. Critical Issues: Regulation of cellular redox homeostasis is highly compartmentalized and complex. Vascular cells rely on multiple metabolic pathways, but the precise connectivity among these pathways and their regulatory mechanisms is only partially defined. There is also a critical need to understand better the cross-regulatory mechanisms between the redox system and metabolic pathways as perturbations in either systems or their cross talk can be detrimental. Future Directions: Future studies are needed to define further how multiple metabolic pathways are wired in vascular cells individually and as a network of closely intertwined processes given that a perturbation in one metabolic compartment often affects others. There also needs to be a comprehensive understanding of how different types of redox perturbations are sensed by and regulate different cellular metabolic pathways with specific attention to subcellular compartmentalization. Lastly, integration of dynamic changes occurring in multiple metabolic pathways and their cross talk with the redox system is an important goal in this multiomics era. Antioxid. Redox Signal. 41,793-817.
Collapse
Affiliation(s)
- Wusheng Xiao
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Laurel Y. Lee
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Pan Z, Yao Y, Liu X, Wang Y, Zhang X, Zha S, Hu K. Nr1d1 inhibition mitigates intermittent hypoxia-induced pulmonary hypertension via Dusp1-mediated Erk1/2 deactivation and mitochondrial fission attenuation. Cell Death Discov 2024; 10:459. [PMID: 39472573 PMCID: PMC11522549 DOI: 10.1038/s41420-024-02219-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 09/30/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Intermittent hypoxia (IH) precipitates pulmonary vasoconstriction, culminating in the onset of pulmonary hypertension (PH) among individuals afflicted with sleep apnea. While Nuclear receptor subfamily 1 group D member 1 (Nr1d1) is progressively recognized as pivotal regulator of cellular physiology, the role in the pathogenesis of IH-induced PH remains largely uncharted. The expression of Nr1d1 was examined in IH-induced rodent PH and in IH-treated PASMCs. To elucidate the contribution of Nr1d1 to the development of IH-induced PH, we employed siRNA to modulate Nr1d1 expression in vitro and employed serotype 1 adeno-associated virus (AAV1) in vivo. Nr1d1 levels were elevated in IH-induced rodents PH lung tissues and IH-treated PASMCs. Knocking down Nr1d1 by AAV1 effectively inhibited PH progression in chronic IH-induced PH models. Mechanistic investigations identified dual specificity phosphatase 1 (Dusp1), as a direct target that Nr1d1 trans-repressed, mediating Nr1d1's regulatory influence on Erk1/2/Drp1 signaling. Nr1d1 deficiency ameliorates mitochondrial dysfunction and fission by restoring Dusp1 dysregulation and Drp1 phosphorylation. Activation of Erk1/2 with PMA reversed the Dusp1-mediated regulation of Drp1 phosphorylation, indicating the involvement of the Erk1/2 pathway in Drp1 phosphorylation controlled by Dusp1. Meanwhile, intermittent hypoxia induced more severe PH in Dusp1 knockout mice compared with wild-type mice. Our data unveil a novel role for Nr1d1 in IH-induced PH pathogenesis and an undisclosed Nr1d1-Dusp1 axis in PASMCs mitochondrial fission regulation.
Collapse
Affiliation(s)
- Zhou Pan
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Yao
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xu Liu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yixuan Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyue Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiqian Zha
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
24
|
Song Y, Jia H, Ma Q, Zhang L, Lai X, Wang Y. The causes of pulmonary hypertension and the benefits of aerobic exercise for pulmonary hypertension from an integrated perspective. Front Physiol 2024; 15:1461519. [PMID: 39483752 PMCID: PMC11525220 DOI: 10.3389/fphys.2024.1461519] [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: 07/08/2024] [Accepted: 09/26/2024] [Indexed: 11/03/2024] Open
Abstract
Pulmonary hypertension is a progressive disease of the pulmonary arteries that begins with increased pulmonary artery pressure, driven by progressive remodeling of the small pulmonary arteries, and ultimately leads to right heart failure and death. Vascular remodeling is the main pathological feature of pulmonary hypertension, but treatments for pulmonary hypertension are lacking. Determining the process of vascular proliferation and dysfunction may be a way to decipher the pathogenesis of pulmonary hypertension. In this review, we summarize the important pathways of pulmonary hypertension pathogenesis. We show how these processes are integrated and emphasize the benign role of aerobic exercise, which, as an adjunctive therapy, may be able to modify vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Yinping Song
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Hao Jia
- School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Qing Ma
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Lulu Zhang
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Xiangyi Lai
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Youhua Wang
- School of Physical Education, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
25
|
Xie Y, Liu X, Xie D, Zhang W, Zhao H, Guan H, Zhou PK. Voltage-dependent anion channel 1 mediates mitochondrial fission and glucose metabolic reprogramming in response to ionizing radiation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174246. [PMID: 38955266 DOI: 10.1016/j.scitotenv.2024.174246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
The ionizing radiation (IR) represents a formidable challenge as an environmental factor to mitochondria, leading to disrupt cellular energy metabolism and posing health risks. Although the deleterious impacts of IR on mitochondrial function are recognized, the specific molecular targets remain incompletely elucidated. In this study, HeLa cells subjected to γ-rays exhibited concomitant oxidative stress, mitochondrial structural alterations, and diminished ATP production capacity. The γ-rays induced a dose-dependent induction of mitochondrial fission, simultaneously manifested by an elevated S616/S637 phosphorylation ratio of the dynamin-related protein 1 (DRP1) and a reduction in the expression of the mitochondrial fusion protein mitofusin 2 (MFN2). Knockdown of DRP1 effectively mitigated γ-rays-induced mitochondrial network damage, implying that DRP1 phosphorylation may act as an effector of radiation-induced mitochondrial damage. The mitochondrial outer membrane protein voltage-dependent anion channel 1 (VDAC1) was identified as a crucial player in IR-induced mitochondrial damage. The VDAC1 inhibitor 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS), counteracts the excessive mitochondrial fission induced by γ-rays, consequently rebalancing the glycolytic and oxidative phosphorylation equilibrium. This metabolic shift was uncovered to enhance glycolytic capacity, thus fortifying cellular resilience and elevating the radiosensitivity of cancer cells. These findings elucidate the intricate regulatory mechanisms governing mitochondrial morphology under radiation response. It is anticipated that the development of targeted drugs directed against VDAC1 may hold promise in augmenting the sensitivity of tumor cells to radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Ying Xie
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, PR China; Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081, PR China
| | - Xiaochang Liu
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Dafei Xie
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Wen Zhang
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Hongling Zhao
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Hua Guan
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China.
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China.
| |
Collapse
|
26
|
Mitra A, Yi D, Dai Z, de Jesus Perez V. Unraveling the role of HIF and epigenetic regulation in pulmonary arterial hypertension: implications for clinical research and its therapeutic approach. Front Med (Lausanne) 2024; 11:1460376. [PMID: 39450110 PMCID: PMC11499164 DOI: 10.3389/fmed.2024.1460376] [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: 07/06/2024] [Accepted: 09/12/2024] [Indexed: 10/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling with high pulmonary pressure, which ultimately leads to right heart failure and premature death. Emerging evidence suggests that both hypoxia and epigenetics play a pivotal role in the pathogenesis of PAH development. In this review article, we summarize the current developments in regulation of hypoxia inducible factor (HIF) isoforms in PAH vascular remodeling and the development of suitable animal models for discovery and testing of HIF pathway-targeting PAH therapeutics. In addition, we also discuss the epigenetic regulation of HIF-dependent isoforms in PAH and its therapeutic potential from a new perspective which highlights the importance of HIF isoform-specific targeting as a novel salutary strategy for PAH treatment.
Collapse
Affiliation(s)
- Ankita Mitra
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, United States
| | - Dan Yi
- Department of Internal Medicine, University of Arizona College of Medicine Phoenix, Phoenix, AZ, United States
| | - Zhiyu Dai
- Department of Internal Medicine, University of Arizona College of Medicine Phoenix, Phoenix, AZ, United States
- Department of Medicine, Washington University School of Medicine in St. Louis (WashU), St. Louis, MO, United States
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
27
|
Wang G, Wang C, Zhu P, Tian J, Yang H. The protective mechanism of sevoflurane in pulmonary arterial hypertension via downregulation of TRAF6. Toxicol Appl Pharmacol 2024; 491:117065. [PMID: 39127353 DOI: 10.1016/j.taap.2024.117065] [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: 06/06/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Pulmonary arterial hypertension (PAH) is an obstructive vasculopathy that, if not promptly treated, culminates in right heart failure. Therefore, pre-clinical studies are needed to support and optimize therapeutic approaches of PAH. Here, we explore a prospective function of sevoflurane in experimental PAH through regulating TRAF6. Monocrotaline (MCT)-induced PAH rats were subjected to sevoflurane inhalation and intratracheal instillation of lentivirus overexpressing TRAF6. Platelet-derived growth factor (PDGF)-treated pulmonary artery smooth muscle cells (PASMCs) were exposed to sevoflurane and genetically manipulated for TRAF6 overexpression. It was found that MCT and PDGF challenge upregulated the levels of TRAF6 in rat lung tissues and PASMCs, but sevoflurane treatment led to reduced TRAF6 expression. Sevoflurane inhalation in MCT-induced rats resulted in alleviative pulmonary vascular remodeling, mitigated right ventricular dysfunction and hypertrophy, improved mitochondrial function and dynamics, and inactivation of NF-κB pathway. In vitro studies confirmed that exposure to sevoflurane repressed PDGF-induced proliferation, migration, and phenotype switching of PASMCs, and suppressed mitochondrial dysfunction and NF-κB activation in PDGF-stimulated PASMCs. The beneficial impact of sevoflurane on pathological changes of lung and cell phenotype of PASMCs were reversed by overexpression of TRAF6. In summary, our study suggested the protective properties of sevoflurane in targeting PAH by downregulating TRAF6 expression, providing a novel avenue for the management of PAH.
Collapse
Affiliation(s)
- Guan Wang
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning, China
| | - Chun Wang
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning, China
| | - Pengcheng Zhu
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning, China
| | - Jiaxin Tian
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning, China.
| | - Haitao Yang
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning, China.
| |
Collapse
|
28
|
Wu D, Jansen-van Vuuren RD, Dasgupta A, Al-Qazazi R, Chen KH, Martin A, Mewburn JD, Alizadeh E, Lima PD, Jones O, Colpman P, Bentley RE, VandenBroek MM, Breault NM, Emon IM, Yerramilli VS, Jedlovčnik L, Zhao YY, Wells M, Sutendra G, Ormiston ML, Archer SL. Novel Drp1 GTPase Inhibitor, Drpitor1a: Efficacy in Pulmonary Hypertension. Hypertension 2024; 81:2189-2201. [PMID: 39162036 PMCID: PMC11410511 DOI: 10.1161/hypertensionaha.124.22822] [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: 02/02/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Drp1 (dynamin-related protein 1), a large GTPase, mediates the increased mitochondrial fission, which contributes to hyperproliferation of pulmonary artery smooth muscle cells in pulmonary arterial hypertension (PAH). We developed a potent Drp1 GTPase inhibitor, Drpitor1a, but its specificity, pharmacokinetics, and efficacy in PAH are unknown. METHODS Drpitor1a's ability to inhibit recombinant and endogenous Drp1 GTPase was assessed. Drpitor1a's effects on fission were studied in control and PAH human pulmonary artery smooth muscle cells (hPASMC) and blood outgrowth endothelial cells (BOEC). Cell proliferation and apoptosis were studied in hPASMC. Pharmacokinetics and tissue concentrations were measured following intravenous and oral drug administration. Drpitor1a's efficacy in regressing monocrotaline-PAH was assessed in rats. In a pilot study, Drpitor1a reduced PA remodeling only in females. Subsequently, we compared Drpitor1a to vehicles in control and monocrotaline-PAH females. RESULTS Drp1 GTPase activity was increased in PAH hPASMC. Drpitor1a inhibited the GTPase activity of recombinant and endogenous Drp1 and reversed the increased fission, seen in PAH hPASMC and PAH BOEC. Drpitor1a inhibited proliferation and induced apoptosis in PAH hPASMC without affecting electron transport chain activity, respiration, fission/fusion mediator expression, or mitochondrial Drp1 translocation. Drpitor1a did not inhibit proliferation or alter mitochondrial dynamics in normal hPASMC. Drpitor1a regressed monocrotaline-PAH without systemic vascular effects or toxicity. CONCLUSIONS Drpitor1a is a specific Drp1 GTPase inhibitor that reduces mitochondrial fission in PAH hPASMC and PAH BOEC. Drpitor1a reduces proliferation and induces apoptosis in PAH hPASMC and regresses monocrotaline-PAH. Drp1 is a therapeutic target in PAH, and Drpitor1a is a potential therapy with an interesting therapeutic sexual dimorphism.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Rats
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dynamins/antagonists & inhibitors
- Dynamins/metabolism
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- GTP Phosphohydrolases/metabolism
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Mitochondrial Dynamics/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Pulmonary Artery/drug effects
- Rats, Sprague-Dawley
- Middle Aged
Collapse
Affiliation(s)
- Danchen Wu
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Ross D. Jansen-van Vuuren
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia
| | - Asish Dasgupta
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Ruaa Al-Qazazi
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Kuang-Hueih Chen
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Ashley Martin
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Jeffrey D. Mewburn
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Elahe Alizadeh
- Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Patricia D.A. Lima
- Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Oliver Jones
- Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Pierce Colpman
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | | | | | - Nolan M. Breault
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Isaac M. Emon
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | | | - Luka Jedlovčnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia
| | - Yuan Yuan Zhao
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Michael Wells
- Partnerships and Innovation, Queen's University, Kingston, ON K7K 3B5, Canada
| | - Gopinath Sutendra
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Mark L. Ormiston
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
- Departments of Biomedical and Molecular Sciences and Surgery, Queen's University, Kingston, K7L 3N6, Canada
| | - Stephen L. Archer
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
- Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
- Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
29
|
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
|
30
|
Wai T. Is mitochondrial morphology important for cellular physiology? Trends Endocrinol Metab 2024; 35:854-871. [PMID: 38866638 DOI: 10.1016/j.tem.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
Mitochondria are double membrane-bound organelles the network morphology of which in cells is shaped by opposing events of fusion and fission executed by dynamin-like GTPases. Mutations in these genes can perturb the form and functions of mitochondria in cell and animal models of mitochondrial diseases. An expanding array of chemical, mechanical, and genetic stressors can converge on mitochondrial-shaping proteins and disrupt mitochondrial morphology. In recent years, studies aimed at disentangling the multiple roles of mitochondrial-shaping proteins beyond fission or fusion have provided insights into the homeostatic relevance of mitochondrial morphology. Here, I review the pleiotropy of mitochondrial fusion and fission proteins with the aim of understanding whether mitochondrial morphology is important for cell and tissue physiology.
Collapse
Affiliation(s)
- Timothy Wai
- Institut Pasteur, Mitochondrial Biology, CNRS UMR 3691, Université Paris Cité, Paris, France.
| |
Collapse
|
31
|
Cappelletti M, Pallotta L, Vona R, Tinari A, Pisano A, Casella G, Crocetti D, Carlomagno D, Tattoli I, Giordano C, Matarrese P, Severi C. The Unexplored Role of Mitochondria-Related Oxidative Stress in Diverticular Disease. Int J Mol Sci 2024; 25:9680. [PMID: 39273627 PMCID: PMC11395029 DOI: 10.3390/ijms25179680] [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: 08/01/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
The pathophysiology of diverticular disease (DD) is not well outlined. Recent studies performed on the DD human ex vivo model have shown the presence of a predominant transmural oxidative imbalance whose origin remains unknown. Considering the central role of mitochondria in oxidative stress, the present study evaluates their involvement in the alterations of DD clinical phenotypes. Colonic surgical samples of patients with asymptomatic diverticulosis, complicated DD, and controls were analyzed. Electron microscopy, protein expression, and cytofluorimetric analyses were performed to assess the contribution of mitochondrial oxidative stress. Functional muscle activity was tested on cells in response to contractile and relaxant agents. To assess the possibility of reverting oxidative damages, N-acetylcysteine was tested on an in vitro model. Compared with the controls, DD tissues showed a marketed increase in mitochondrial number and fusion accompanied by the altered mitochondrial electron transport chain complexes. In SMCs, the mitochondrial mass increase was accompanied by altered mitochondrial metabolic activity supported by a membrane potential decrease. Ulteriorly, a decrease in antioxidant content and altered contraction-relaxation dynamics reverted by N-acetylcysteine were observed. Therefore, the oxidative stress-driven alterations resulted in mitochondrial impairment. The beneficial effects of antioxidant treatments open new possibilities for tailored therapeutic strategies that have not been tested for this disease.
Collapse
Affiliation(s)
- Martina Cappelletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Lucia Pallotta
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Rosa Vona
- Center for Gender-Specific Medicine, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Antonella Tinari
- Center for Gender-Specific Medicine, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Annalinda Pisano
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Giovanni Casella
- Department of Surgical Science, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Daniele Crocetti
- Department of Surgical Science, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Dominga Carlomagno
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Ivan Tattoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Carla Giordano
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Carola Severi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| |
Collapse
|
32
|
Kaur S, Khullar N, Navik U, Bali A, Bhatti GK, Bhatti JS. Multifaceted role of dynamin-related protein 1 in cardiovascular disease: From mitochondrial fission to therapeutic interventions. Mitochondrion 2024; 78:101904. [PMID: 38763184 DOI: 10.1016/j.mito.2024.101904] [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: 12/06/2023] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Mitochondria are central to cellular energy production and metabolic regulation, particularly in cardiomyocytes. These organelles constantly undergo cycles of fusion and fission, orchestrated by key proteins like Dynamin-related Protein 1 (Drp-1). This review focuses on the intricate roles of Drp-1 in regulating mitochondrial dynamics, its implications in cardiovascular health, and particularly in myocardial infarction. Drp-1 is not merely a mediator of mitochondrial fission; it also plays pivotal roles in autophagy, mitophagy, apoptosis, and necrosis in cardiac cells. This multifaceted functionality is often modulated through various post-translational alterations, and Drp-1's interaction with intracellular calcium (Ca2 + ) adds another layer of complexity. We also explore the pathological consequences of Drp-1 dysregulation, including increased reactive oxygen species (ROS) production and endothelial dysfunction. Furthermore, this review delves into the potential therapeutic interventions targeting Drp-1 to modulate mitochondrial dynamics and improve cardiovascular outcomes. We highlight recent findings on the interaction between Drp-1 and sirtuin-3 and suggest that understanding this interaction may open new avenues for therapeutically modulating endothelial cells, fibroblasts, and cardiomyocytes. As the cardiovascular system increasingly becomes the focal point of aging and chronic disease research, understanding the nuances of Drp-1's functionality can lead to innovative therapeutic approaches.
Collapse
Affiliation(s)
- Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India.
| |
Collapse
|
33
|
Wan JJ, Yi J, Wang FY, Li X, Zhang C, Song L, Dai AG. Role of mitophagy in pulmonary hypertension: Targeting the mechanism and pharmacological intervention. Mitochondrion 2024; 78:101928. [PMID: 38992857 DOI: 10.1016/j.mito.2024.101928] [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: 11/20/2023] [Revised: 05/29/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
Mitophagy, a crucial pathway in eukaryotic cells, selectively eliminates dysfunctional mitochondria, thereby maintaining cellular homeostasis via mitochondrial quality control. Pulmonary hypertension (PH) refers to a pathological condition where pulmonary arterial pressure is abnormally elevated due to various reasons, and the underlying pathogenesis remains elusive. This article examines the molecular mechanisms underlying mitophagy, emphasizing its role in PH and the progress in elucidating related molecular signaling pathways. Additionally, it highlights current drug regulatory pathways, aiming to provide novel insights into the prevention and treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Jia-Jing Wan
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Jian Yi
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Fei-Ying Wang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Xia Li
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Chao Zhang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Lan Song
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Ai-Guo Dai
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China.
| |
Collapse
|
34
|
Zhang Y, Wang J, Zhang M, Li X, Zhang F, Zhou M, Yang K, Chen W, Ding H, Tan X, Zhang Q, Qiao Z. Study on the Regulatory Mechanism of the PDK1-Mediated TGF-β/Smad Signaling Pathway in Hypoxia-Induced Yak Lungs. Animals (Basel) 2024; 14:2422. [PMID: 39199957 PMCID: PMC11350703 DOI: 10.3390/ani14162422] [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: 07/14/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
The aim of this study was to investigate the effects of hypoxia-induced phenotype, glucose metabolism, ROS levels, and the PDK1-mediated regulation of TGF-β/Smad signaling in yellow cattles, yaks, and those overexpressing PDK1 PASMCs using growth curves, flow cytometry, scratch experiments, glucose and lactic acid assays, RT-qPCR, and Western blotting. The results showed that hypoxia significantly promoted proliferation, migration, antiapoptosis, ROS levels, glucose consumption, and lactate production in yellow cattle PASMCs (p < 0.05), and the cells were dedifferentiated from the contractile phenotype; conversely, hypoxia had no significant effect on yak PASMCs (p > 0.05). PDK1 overexpression significantly promoted proliferation, antiapoptosis, glucose consumption, and lactate production in yak PASMCs under normoxia and hypoxia (p < 0.05), decreased their migration levels under hypoxia (p < 0.05), and dedifferentiated the contractile phenotype of the cells. Overexpression of PDK1 in yak PASMCs is detrimental to their adaptation to hypoxic environments. Yak PASMCs adapted to the effects of hypoxia on lung tissue by downregulating the expression of genes related to the PDK1 and TGF-β/Smad signaling pathways. Taken together, the regulation of PDK1-mediated TGF-β/Smad signaling may be involved in the process of yaks' adaptation to the hypoxic environment of the plateau, reflecting the good adaptive ability of yaks. The present study provides basic information to further elucidate the mechanism of PDK1-mediated TGF-β/Smad signaling induced by hypoxia in the lungs of yaks, as well as target genes for the treatment of plateau diseases in humans and animals.
Collapse
Affiliation(s)
- Yiyang Zhang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Jun Wang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Meng Zhang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Xiaoyun Li
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Fan Zhang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Manlin Zhou
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Kun Yang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Weiji Chen
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Haie Ding
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Xiao Tan
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Qian Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| | - Zilin Qiao
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| |
Collapse
|
35
|
Hong WL, Huang H, Zeng X, Duan CY. Targeting mitochondrial quality control: new therapeutic strategies for major diseases. Mil Med Res 2024; 11:59. [PMID: 39164792 PMCID: PMC11337860 DOI: 10.1186/s40779-024-00556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 07/13/2024] [Indexed: 08/22/2024] Open
Abstract
Mitochondria play a crucial role in maintaining the normal physiological state of cells. Hence, ensuring mitochondrial quality control is imperative for the prevention and treatment of numerous diseases. Previous reviews on this topic have however been inconsistencies and lack of systematic organization. Therefore, this review aims to provide a comprehensive and systematic overview of mitochondrial quality control and explore the possibility of targeting the same for the treatment of major diseases. This review systematically summarizes three fundamental characteristics of mitochondrial quality control, including mitochondrial morphology and dynamics, function and metabolism, and protein expression and regulation. It also extensively examines how imbalances in mitochondrial quality are linked to major diseases, such as ischemia-hypoxia, inflammatory disorders, viral infections, metabolic dysregulations, degenerative conditions, and tumors. Additionally, the review explores innovative approaches to target mitochondrial quality control, including using small molecule drugs that regulate critical steps in maintaining mitochondrial quality, nanomolecular materials designed for precise targeting of mitochondria, and novel cellular therapies, such as vesicle therapy and mitochondrial transplantation. This review offers a novel perspective on comprehending the shared mechanisms underlying the occurrence and progression of major diseases and provides theoretical support and practical guidance for the clinical implementation of innovative therapeutic strategies that target mitochondrial quality control for treating major diseases.
Collapse
Affiliation(s)
- Wei-Long Hong
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - He Huang
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xue Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Chen-Yang Duan
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
36
|
Mialet-Perez J, Belaidi E. Interplay between hypoxia inducible Factor-1 and mitochondria in cardiac diseases. Free Radic Biol Med 2024; 221:13-22. [PMID: 38697490 DOI: 10.1016/j.freeradbiomed.2024.04.239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Ischemic heart diseases and cardiomyopathies are characterized by hypoxia, energy starvation and mitochondrial dysfunction. HIF-1 acts as a cellular oxygen sensor, tuning the balance of metabolic and oxidative stress pathways to provide ATP and sustain cell survival. Acting on mitochondria, HIF-1 regulates different processes such as energy substrate utilization, oxidative phosphorylation and mitochondrial dynamics. In turn, mitochondrial homeostasis modifications impact HIF-1 activity. This underlies that HIF-1 and mitochondria are tightly interconnected to maintain cell homeostasis. Despite many evidences linking HIF-1 and mitochondria, the mechanistic insights are far from being understood, particularly in the context of cardiac diseases. Here, we explore the current understanding of how HIF-1, reactive oxygen species and cell metabolism are interconnected, with a specific focus on mitochondrial function and dynamics. We also discuss the divergent roles of HIF in acute and chronic cardiac diseases in order to highlight that HIF-1, mitochondria and oxidative stress interaction deserves to be deeply investigated. While the strategies aiming at stabilizing HIF-1 have provided beneficial effects in acute ischemic injury, some deleterious effects were observed during prolonged HIF-1 activation. Thus, deciphering the link between HIF-1 and mitochondria will help to optimize HIF-1 modulation and provide new therapeutic perspectives for the treatment of cardiovascular pathologies.
Collapse
Affiliation(s)
- Jeanne Mialet-Perez
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, France
| | - Elise Belaidi
- Univ. Lyon 1, Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, LBTI UMR 5305, 69367, Lyon, France.
| |
Collapse
|
37
|
Rezaei Adariani S, Agne D, Koska S, Burhop A, Seitz C, Warmers J, Janning P, Metz M, Pahl A, Sievers S, Waldmann H, Ziegler S. Detection of a Mitochondrial Fragmentation and Integrated Stress Response Using the Cell Painting Assay. J Med Chem 2024; 67:13252-13270. [PMID: 39018123 PMCID: PMC11320566 DOI: 10.1021/acs.jmedchem.4c01183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Mitochondria are cellular powerhouses and are crucial for cell function. However, they are vulnerable to internal and external perturbagens that may impair mitochondrial function and eventually lead to cell death. In particular, small molecules may impact mitochondrial function, and therefore, their influence on mitochondrial homeostasis is at best assessed early on in the characterization of biologically active small molecules and drug discovery. We demonstrate that unbiased morphological profiling by means of the cell painting assay (CPA) can detect mitochondrial stress coupled with the induction of an integrated stress response. This activity is common for compounds addressing different targets, is not shared by direct inhibitors of the electron transport chain, and enables prediction of mitochondrial stress induction for small molecules that are profiled using CPA.
Collapse
Affiliation(s)
- Soheila Rezaei Adariani
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical
University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Daya Agne
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Sandra Koska
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Annina Burhop
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Carina Seitz
- Compound
Management and Screening Center, Max Planck
Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Jens Warmers
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical
University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Petra Janning
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Malte Metz
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Axel Pahl
- Compound
Management and Screening Center, Max Planck
Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Sonja Sievers
- Compound
Management and Screening Center, Max Planck
Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Herbert Waldmann
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical
University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Slava Ziegler
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| |
Collapse
|
38
|
Zhang H, Chen L, Li J, Sun J, Zhao Q, Wang S, Li G. STAT3 phosphorylation at Tyr705 affects DRP1 (dynamin-related protein 1) controlled-mitochondrial fission during the development of apoptotic-resistance in pulmonary arterial endothelial cells. Genes Genomics 2024; 46:751-762. [PMID: 38733520 PMCID: PMC11208226 DOI: 10.1007/s13258-024-01522-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The apoptosis-resistant pulmonary arterial endothelial cells (PAECs) are known to be major players in the pulmonary remodeling of pulmonary arterial hypertension (PAH) and exhibit an abnormal metabolic profile with mitochondrial dysfunction. Mitochondrial fission has been shown to regulate the apoptosis of several cell types, but this is largely unexplored in the PAECs. OBJECTIVE The roles of mitochondrial fission control by Dynamin related protein-1 (DRP1) in the development of PAECs apoptosis suppression were investigated in present study and the potential mechanisms behind this were furtherly explored. METHODS The mitochondrial morphology was investigated in PAECs from PAH rats with the pulmonary plexiform lesions, and the relations of it with DRP1 expression and apoptosis were furtherly identified in apoptosis-resistant PAECs induced by hypoxia. PAECs were isolated from rats with severe PAH and from normal subjects, the apoptotic-resistant PAECs were induced by hypoxia. DRP1 gene knockdown was achieved via DRP1-siRNA, DRP1 and STAT3 phosphorylation were blocked using its inhibitors, respectively. Apoptosis was analyzed by flow cytometry, and mitochondrial morphology was investigated by transmission electron microscope and confocal microscopy. RESULTS The PAECs isolated from PAH rats with the pulmonary plexiform-like lesions and displayed lower apoptotic rate with increased DRP1 expression and mitochondrial fragmentation. In addition, similar observations were achieved in apoptosis-resistant PAECs induced by hypoxia. Targeting DRP1 using siRNA and pharmacologic blockade prevented the mitochondrial fission and subsequent apoptotic resistance in PAECs under hypoxia. Mechanistically, STAT3 phosphorylation at Tyr705 was shown to be activated in both PAH and hypoxia-treated PAECs, leading to the regulation of DRP1 expression. Of importance, targeting STAT3Tyr705 phosphorylation prevented DRP1 disruption on apoptosis in PAECs under hypoxia. CONCLUSIONS These data indicated that STAT3 phosphorylation at Tyr705 impacted DRP1-controlled mitochondrial fission during the development of apoptosis-resistance in PAECs, suggesting mitochondrial dynamics may represent a therapeutic target for PAH.
Collapse
Affiliation(s)
- Han Zhang
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Li Chen
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Jiachen Li
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Jiashu Sun
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Qixu Zhao
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Sheng Wang
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Gang Li
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China.
| |
Collapse
|
39
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
40
|
Wang Z, Zhang YX, Shi JZ, Yan Y, Zhao LL, Kou JJ, He YY, Xie XM, Zhang SJ, Pang XB. RNA m6A methylation and regulatory proteins in pulmonary arterial hypertension. Hypertens Res 2024; 47:1273-1287. [PMID: 38438725 DOI: 10.1038/s41440-024-01607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/12/2023] [Accepted: 01/27/2024] [Indexed: 03/06/2024]
Abstract
m6A (N6‑methyladenosine) is the most common and abundant apparent modification in mRNA of eukaryotes. The modification of m6A is regulated dynamically and reversibly by methyltransferase (writer), demethylase (eraser), and binding protein (reader). It plays a significant role in various processes of mRNA metabolism, including regulation of transcription, maturation, translation, degradation, and stability. Pulmonary arterial hypertension (PAH) is a malignant cardiopulmonary vascular disease characterized by abnormal proliferation of pulmonary artery smooth muscle cells. Despite the existence of several effective and targeted therapies, there is currently no cure for PAH and the prognosis remains poor. Recent studies have highlighted the crucial role of m6A modification in cardiovascular diseases. Investigating the role of RNA m6A methylation in PAH could provide valuable insights for drug development. This review aims to explore the mechanism and function of m6A in the pathogenesis of PAH and discuss the potential targeting of RNA m6A methylation modification as a treatment for PAH.
Collapse
Affiliation(s)
- Zhe Wang
- School of Pharmacy, Henan University, Henan, China
| | - Yi-Xuan Zhang
- Department of Anesthesiology, Huaihe Hospital of Henan University, Henan, China
| | - Jun-Zhuo Shi
- School of Pharmacy, Henan University, Henan, China
| | - Yi Yan
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu-Ling Zhao
- School of Pharmacy, Henan University, Henan, China
| | - Jie-Jian Kou
- Department of Pharmacy, Huaihe Hospital of Henan University, Henan, China
| | - Yang-Yang He
- School of Pharmacy, Henan University, Henan, China
| | - Xin-Mei Xie
- School of Pharmacy, Henan University, Henan, China.
| | - Si-Jin Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | | |
Collapse
|
41
|
Tong Z, Du X, Zhou Y, Jing F, Ma J, Feng Y, Lou S, Wang Q, Dong Z. Drp1-mediated mitochondrial fission promotes pulmonary fibrosis progression through the regulation of lipid metabolic reprogramming by ROS/HIF-1α. Cell Signal 2024; 117:111075. [PMID: 38311302 DOI: 10.1016/j.cellsig.2024.111075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVE To confirm the mechanism of dynamic-related protein 1 (Drp1)-mediated mitochondrial fission through ROS/HIF-1α-mediated regulation of lipid metabolic reprogramming in the progression of pulmonary fibrosis (PF). METHODS A mouse model of PF was established by intratracheal instillation of bleomycin (BLM) (2.5 mg/kg). A PF cell model was constructed by stimulating MRC-5 cells with TGF-β (10 ng/mL). Pathological changes in the lung tissue and related protein levels were observed via tissue staining. The indicators related to lipid oxidation were detected by a kit, and lipid production was confirmed through oil red O staining. Inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA). RT-qPCR, Western blotting and immunofluorescence staining were used to detect the expression of genes and proteins related to the disease. We used CCK-8 and EdU staining to confirm cell proliferation, flow cytometry was used to confirm apoptosis and ROS levels, α-SMA expression was detected by immunofluorescence staining, and mitochondria were observed by MitoTracker staining. RESULTS The BLM induced lung tissue structure and alveolar wall thickening in mice. Mitochondrial fission was observed in MRC-5 cells induced by TGF-β, which led to increased cell proliferation; decreased apoptosis; increased expression of collagen, α-SMA and Drp1; and increased lipid oxidation and inflammation. Treatment with the Drp1 inhibitor mdivi-1 or transfection with si-Drp1 attenuated the induction of BLM and TGF-β. For lipid metabolism, lipid droplets were formed in BLM-induced lung tissue and in TGF-β-induced cells, fatty acid oxidation genes and lipogenesis-related genes were upregulated, ROS levels in cells were increased, and the expression of HIF-1α was upregulated. Mdivi-1 treatment reversed TGF-β induction, while H2O2 treatment or OE-HIF-1α transfection reversed the effect of mdivi-1. CONCLUSION In PF, inhibition of Drp1 can prevent mitochondrial fission in fibroblasts and regulate lipid metabolism reprogramming through ROS/HIF-1α; thus, fibroblast activation was inhibited, alleviating the progression of PF.
Collapse
Affiliation(s)
- Zhongkai Tong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Xuekui Du
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Ying Zhou
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Fangxue Jing
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Health Science Center, Ningbo University, Ningbo 315211, China
| | - JiangPo Ma
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325000, China
| | - Yingying Feng
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Health Science Center, Ningbo University, Ningbo 315211, China
| | - Saiyun Lou
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China; Second Clinical Medicine Faculty of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiong Wang
- Department of Respiratory Infection, Zhenhai Hospital of Traditional Chinese Medicine, Ningbo 315200, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China.
| |
Collapse
|
42
|
Zhang W, Li M, Ye X, Jiang M, Wu X, Tang Z, Hu L, Zhang H, Li Y, Pan J. Disturbance of mitochondrial dynamics in myocardium of broilers with pulmonary hypertension syndrome. Br Poult Sci 2024; 65:154-164. [PMID: 38380624 DOI: 10.1080/00071668.2024.2308277] [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: 08/03/2023] [Accepted: 12/05/2023] [Indexed: 02/22/2024]
Abstract
1. The following study investigated the relationship between pulmonary hypertension syndrome (PHS) and mitochondrial dynamics in broiler cardiomyocytes.2. An animal model for PHS was established by injecting broiler chickens with CM-32 cellulose particles. Broiler myocardial cells were cultured under hypoxic conditions to establish an in vitro model. The ascites heart index, histomorphology, mitochondrial ultrastructure, and mitochondrial dynamic-related gene and protein expression were evaluated.3. The myocardial fibres from PHS broilers had wider spaces and were wavy and twisted and the number of mitochondria increased. Compared with the control group, the gene and protein expression levels were decreased for Opa1, Mfn1, and Mfn2 in the myocardium of PHS broilers. The gene and protein expression was significantly increased for Drp1 and Mff.4. This study showed that PHS in broilers may cause myocardial mitochondrial dysfunction, specifically by diminishing mitochondrial fusion and enhancing fission, causing disturbances in the mitochondrial dynamics of the heart.
Collapse
Affiliation(s)
- W Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - M Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - X Ye
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - M Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - X Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - Z Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - L Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - H Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - Y Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - J Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
43
|
Pokharel MD, Garcia-Flores A, Marciano D, Franco MC, Fineman JR, Aggarwal S, Wang T, Black SM. Mitochondrial network dynamics in pulmonary disease: Bridging the gap between inflammation, oxidative stress, and bioenergetics. Redox Biol 2024; 70:103049. [PMID: 38295575 PMCID: PMC10844980 DOI: 10.1016/j.redox.2024.103049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/16/2024] [Indexed: 02/02/2024] Open
Abstract
Once thought of in terms of bioenergetics, mitochondria are now widely accepted as both the orchestrator of cellular health and the gatekeeper of cell death. The pulmonary disease field has performed extensive efforts to explore the role of mitochondria in regulating inflammation, cellular metabolism, apoptosis, and oxidative stress. However, a critical component of these processes needs to be more studied: mitochondrial network dynamics. Mitochondria morphologically change in response to their environment to regulate these processes through fusion, fission, and mitophagy. This allows mitochondria to adapt their function to respond to cellular requirements, a critical component in maintaining cellular homeostasis. For that reason, mitochondrial network dynamics can be considered a bridge that brings multiple cellular processes together, revealing a potential pathway for therapeutic intervention. In this review, we discuss the critical modulators of mitochondrial dynamics and how they are affected in pulmonary diseases, including chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), and pulmonary arterial hypertension (PAH). A dysregulated mitochondrial network plays a crucial role in lung disease pathobiology, and aberrant fission/fusion/mitophagy pathways are druggable processes that warrant further exploration. Thus, we also discuss the candidates for lung disease therapeutics that regulate mitochondrial network dynamics.
Collapse
Affiliation(s)
- Marissa D Pokharel
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Alejandro Garcia-Flores
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | - David Marciano
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Maria C Franco
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, UC San Francisco, San Francisco, CA, 94143, USA
| | - Saurabh Aggarwal
- Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
44
|
Zhang X, Zhang Z, Wan S, Qi J, Hao Y, An P, Luo Y, Luo J. Ameliorative Effect of Coenzyme Q10 on Phenotypic Transformation in Human Smooth Muscle Cells with FBN1 Knockdown. Int J Mol Sci 2024; 25:2662. [PMID: 38473909 PMCID: PMC10931635 DOI: 10.3390/ijms25052662] [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: 01/12/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Mutations of the FBN1 gene lead to Marfan syndrome (MFS), which is an autosomal dominant connective tissue disorder featured by thoracic aortic aneurysm risk. There is currently no effective treatment for MFS. Here, we studied the role of mitochondrial dysfunction in the phenotypic transformation of human smooth muscle cells (SMCs) and whether a mitochondrial boosting strategy can be a potential treatment. We knocked down FBN1 in SMCs to create an MFS cell model and used rotenone to induce mitochondrial dysfunction. Furthermore, we incubated the shFBN1 SMCs with Coenzyme Q10 (CoQ10) to assess whether restoring mitochondrial function can reverse the phenotypic transformation. The results showed that shFBN1 SMCs had decreased TFAM (mitochondrial transcription factor A), mtDNA levels and mitochondrial mass, lost their contractile capacity and had increased synthetic phenotype markers. Inhibiting the mitochondrial function of SMCs can decrease the expression of contractile markers and increase the expression of synthetic genes. Imposing mitochondrial stress causes a double-hit effect on the TFAM level, oxidative phosphorylation and phenotypic transformation of FBN1-knockdown SMCs while restoring mitochondrial metabolism with CoQ10 can rapidly reverse the synthetic phenotype. Our results suggest that mitochondria function is a potential therapeutic target for the phenotypic transformation of SMCs in MFS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yongting Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (X.Z.); (Z.Z.); (S.W.); (J.Q.); (Y.H.); (P.A.)
| | - Junjie Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (X.Z.); (Z.Z.); (S.W.); (J.Q.); (Y.H.); (P.A.)
| |
Collapse
|
45
|
Kobayashi H, Yoshimoto C, Matsubara S, Shigetomi H, Imanaka S. An integral role of mitochondrial function in the pathophysiology of preeclampsia. Mol Biol Rep 2024; 51:330. [PMID: 38393449 DOI: 10.1007/s11033-024-09285-z] [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: 11/12/2023] [Accepted: 01/24/2024] [Indexed: 02/25/2024]
Abstract
Preeclampsia (PE) is associated with high maternal and perinatal morbidity and mortality. The development of effective treatment strategies remains a major challenge due to the limited understanding of the pathogenesis. In this review, we summarize the current understanding of PE research, focusing on the molecular basis of mitochondrial function in normal and PE placentas, and discuss perspectives on future research directions. Mitochondria integrate numerous physiological processes such as energy production, cellular redox homeostasis, mitochondrial dynamics, and mitophagy, a selective autophagic clearance of damaged or dysfunctional mitochondria. Normal placental mitochondria have evolved innovative survival strategies to cope with uncertain environments (e.g., hypoxia and nutrient starvation). Cytotrophoblasts, extravillous trophoblast cells, and syncytiotrophoblasts all have distinct mitochondrial morphology and function. Recent advances in molecular studies on the spatial and temporal changes in normal mitochondrial function are providing valuable insight into PE pathogenesis. In PE placentas, hypoxia-mediated mitochondrial fission may induce activation of mitophagy machinery, leading to increased mitochondrial fragmentation and placental tissue damage over time. Repair mechanisms in mitochondrial function restore placental function, but disruption of compensatory mechanisms can induce apoptotic death of trophoblast cells. Additionally, molecular markers associated with repair or compensatory mechanisms that may influence the development and progression of PE are beginning to be identified. However, contradictory results have been obtained regarding some of the molecules that control mitochondrial biogenesis, dynamics, and mitophagy in PE placentas. In conclusion, understanding how the mitochondrial morphology and function influence cell fate decisions of trophoblast cells is an important issue in normal as well as pathological placentation biology. Research focusing on mitochondrial function will become increasingly important for elucidating the pathogenesis and effective treatment strategies of PE.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara, 634-0813, Japan.
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan.
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, 2-897-5 Shichijyonishi-machi, Nara, 630-8581, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
- Department of Medicine, Kei Oushin Clinic, 5-2-6, Naruo-cho, Nishinomiya, 663-8184, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, 3-3-17 Kitatomigaoka-cho, Nara, 634- 0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara, 634-0813, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
| |
Collapse
|
46
|
Chen LJ, Li JYS, Nguyen P, He M, Chen ZB, Subramaniam S, Shyy JYJ, Chien S. Single-cell RNA sequencing unveils unique transcriptomic signatures of endothelial cells and role of ENO1 in response to disturbed flow. Proc Natl Acad Sci U S A 2024; 121:e2318904121. [PMID: 38261622 PMCID: PMC10835041 DOI: 10.1073/pnas.2318904121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Flow patterns exert significant effects on vascular endothelial cells (ECs) to lead to the focal nature of atherosclerosis. Using a step flow chamber to investigate the effects of disturbed shear (DS) and pulsatile shear (PS) on ECs in the same flow channel, we conducted single-cell RNA sequencing analyses to explore the distinct transcriptomic profiles regulated by DS vs. PS. Integrated analysis identified eight cell clusters and demonstrated that DS induces EC transition from atheroprotective to proatherogenic phenotypes. Using an automated cell type annotation algorithm (SingleR), we showed that DS promoted endothelial-to-mesenchymal transition (EndMT) by inducing the transcriptional phenotypes for inflammation, hypoxia responses, transforming growth factor-beta (TGF-β) signaling, glycolysis, and fatty acid synthesis. Enolase 1 (ENO1), a key gene in glycolysis, was one of the top-ranked genes in the DS-induced EndMT cluster. Pseudotime trajectory analysis revealed that the kinetic expression of ENO1 was significantly associated with EndMT and that ENO1 silencing repressed the DS- and TGF-β-induced EC inflammation and EndMT. Consistent with these findings, ENO1 was highly expressed in ECs at the inner curvature of the mouse aortic arch (which is exposed to DS) and atherosclerotic lesions, suggesting its proatherogenic role in vivo. In summary, we present a comprehensive single-cell atlas of ECs in response to different flow patterns within the same flow channel. Among the DS-regulated genes, ENO1 plays an important role in DS-induced EC inflammation and EndMT. These results provide insights into how hemodynamic forces regulate vascular endothelium in health and disease.
Collapse
Affiliation(s)
- Li-Jing Chen
- Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Julie Yi-Shuan Li
- Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Phu Nguyen
- Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ming He
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Shankar Subramaniam
- Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
- San Diego Supercomputer Center, University of California at San Diego, La Jolla, CA 92093
| | - John Y-J Shyy
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Shu Chien
- Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
47
|
Mocumbi A, Humbert M, Saxena A, Jing ZC, Sliwa K, Thienemann F, Archer SL, Stewart S. Pulmonary hypertension. Nat Rev Dis Primers 2024; 10:1. [PMID: 38177157 DOI: 10.1038/s41572-023-00486-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 01/06/2024]
Abstract
Pulmonary hypertension encompasses a range of conditions directly or indirectly leading to elevated pressures within the pulmonary arteries. Five main groups of pulmonary hypertension are recognized, all defined by a mean pulmonary artery pressure of >20 mmHg: pulmonary arterial hypertension (rare), pulmonary hypertension associated with left-sided heart disease (very common), pulmonary hypertension associated with lung disease (common), pulmonary hypertension associated with pulmonary artery obstructions, usually related to thromboembolic disease (rare), and pulmonary hypertension with unclear and/or multifactorial mechanisms (rare). At least 1% of the world's population is affected, with a greater burden more likely in low-income and middle-income countries. Across all its forms, pulmonary hypertension is associated with adverse vascular remodelling with obstruction, stiffening and vasoconstriction of the pulmonary vasculature. Without proactive management this leads to hypertrophy and ultimately failure of the right ventricle, the main cause of death. In older individuals, dyspnoea is the most common symptom. Stepwise investigation precedes definitive diagnosis with right heart catheterization. Medical and surgical treatments are approved for pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. There are emerging treatments for other forms of pulmonary hypertension; but current therapy primarily targets the underlying cause. There are still major gaps in basic, clinical and translational knowledge; thus, further research, with a focus on vulnerable populations, is needed to better characterize, detect and effectively treat all forms of pulmonary hypertension.
Collapse
Affiliation(s)
- Ana Mocumbi
- Faculdade de Medicina, Universidade Eduardo Mondlane, Maputo, Moçambique.
- Instituto Nacional de Saúde, EN 1, Marracuene, Moçambique.
| | - Marc Humbert
- Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre (Assistance Publique Hôpitaux de Paris), Université Paris-Saclay, INSERM UMR_S 999, Paris, France
- ERN-LUNG, Le Kremlin Bicêtre, Paris, France
| | - Anita Saxena
- Sharma University of Health Sciences, Haryana, New Delhi, India
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Karen Sliwa
- Cape Heart Institute, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Friedrich Thienemann
- Department of Medicine, Groote Schuur Hospital, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
- Department of Internal Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Simon Stewart
- Institute of Health Research, University of Notre Dame, Fremantle, Western Australia, Australia
| |
Collapse
|
48
|
Wan JJ, Yi J, Wang FY, Zhang C, Dai AG. Expression and regulation of HIF-1a in hypoxic pulmonary hypertension: Focus on pathological mechanism and Pharmacological Treatment. Int J Med Sci 2024; 21:45-60. [PMID: 38164358 PMCID: PMC10750340 DOI: 10.7150/ijms.88216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/20/2023] [Indexed: 01/03/2024] Open
Abstract
Hypoxia inducible factor-1(HIF-1), a heterodimeric transcription factor, is composed of two subunits (HIF-1α and HIF-1β). It is considered as an important transcription factor for regulating oxygen changes in hypoxic environment, which can regulate the expression of various hypoxia-related target genes and play a role in acute and chronic hypoxia pulmonary vascular reactions. In this paper, the function and mechanism of HIF-1a expression and regulation in hypoxic pulmonary hypertension (HPH) were reviewed, and current candidate schemes for treating pulmonary hypertension by using HIF-1a as the target were introduced, so as to provide reference for studying the pathogenesis of HPH and screening effective treatment methods.
Collapse
Affiliation(s)
- Jia-Jing Wan
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| | - Jian Yi
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, People's Republic of China
| | - Fei-Ying Wang
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| | - Chao Zhang
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| | - Ai-Guo Dai
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
- Department of Respiratory Medicine, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha 410021, Hunan, People's Republic of China
| |
Collapse
|
49
|
Yegambaram M, Sun X, Lu Q, Jin Y, Ornatowski W, Soto J, Aggarwal S, Wang T, Tieu K, Gu H, Fineman JR, Black SM. Mitochondrial hyperfusion induces metabolic remodeling in lung endothelial cells by modifying the activities of electron transport chain complexes I and III. Free Radic Biol Med 2024; 210:183-194. [PMID: 37979892 PMCID: PMC12051485 DOI: 10.1016/j.freeradbiomed.2023.11.008] [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: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is a progressive disease with vascular remodeling as a critical structural alteration. We have previously shown that metabolic reprogramming is an early initiating mechanism in animal models of PH. This metabolic dysregulation has been linked to remodeling the mitochondrial network to favor fission. However, whether the mitochondrial fission/fusion balance underlies the metabolic reprogramming found early in PH development is unknown. METHODS Utilizing a rat early model of PH, in conjunction with cultured pulmonary endothelial cells (PECs), we utilized metabolic flux assays, Seahorse Bioassays, measurements of electron transport chain (ETC) complex activity, fluorescent microscopy, and molecular approaches to investigate the link between the disruption of mitochondrial dynamics and the early metabolic changes that occur in PH. RESULTS We observed increased fusion mediators, including Mfn1, Mfn2, and Opa1, and unchanged fission mediators, including Drp1 and Fis1, in a two-week monocrotaline-induced PH animal model (early-stage PH). We were able to establish a connection between increases in fusion mediator Mfn1 and metabolic reprogramming. Using an adenoviral expression system to enhance Mfn1 levels in pulmonary endothelial cells and utilizing 13C-glucose labeled substrate, we found increased production of 13C lactate and decreased TCA cycle metabolites, revealing a Warburg phenotype. The use of a 13C5-glutamine substrate showed evidence that hyperfusion also induces oxidative carboxylation. The increase in glycolysis was linked to increased hypoxia-inducible factor 1α (HIF-1α) protein levels secondary to the disruption of cellular bioenergetics and higher levels of mitochondrial reactive oxygen species (mt-ROS). The elevation in mt-ROS correlated with attenuated ETC complexes I and III activities. Utilizing a mitochondrial-targeted antioxidant to suppress mt-ROS, limited HIF-1α protein levels, which reduced cellular glycolysis and reestablished mitochondrial membrane potential. CONCLUSIONS Our data connects mitochondrial fusion-mediated mt-ROS to the Warburg phenotype in early-stage PH development.
Collapse
Affiliation(s)
- Manivannan Yegambaram
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | | | - Jamie Soto
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | - Saurabh Aggarwal
- Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
50
|
Zhao Q, Liu Z, Song P, Yuan Z, Zou MH. Mitochondria-derived Vesicle Packaging as a Novel Therapeutic Mechanism in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2024; 70:39-49. [PMID: 37713305 PMCID: PMC10768832 DOI: 10.1165/rcmb.2023-0010oc] [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: 01/06/2023] [Accepted: 09/14/2023] [Indexed: 09/17/2023] Open
Abstract
Increasing evidence suggests that mitochondrial dysfunction in pulmonary endothelial cells (ECs) plays a causative role in the initiation and progression of pulmonary hypertension (PH); how mitochondria become dysfunctional in PH remains elusive. Mitochondria-derived vesicles (MDVs) are small subcellular vesicles that excise from mitochondria. Whether MDV deregulation causes mitochondrial dysfunction in PH is unknown. The aim of this study was to determine MDV regulation in ECs and to elucidate how MDV deregulation in ECs leads to PH. MDV formation and mitochondrial morphology/dynamics were examined in ECs of EC-specific liver kinase B1 (LKB1) knockout mice (LKB1ec-/-), in monocrotaline-induced PH rats, and in lungs of patients with PH. Pulmonary ECs of patients with PH and hypoxia-treated pulmonary ECs exhibited increased mitochondrial fragmentation and disorganized mitochondrial ultrastructure characterized by electron lucent-swelling matrix compartments and concentric layering of the cristae network, together with defective MDV shedding. MDVs actively regulated mitochondrial membrane dynamics and mitochondrial ultrastructure via removing mitofission-related cargoes. The shedding of MDVs from parental mitochondria required LKB1-mediated mitochondrial recruitment of Rab9 GTPase. LKB1ec-/- mice spontaneously developed PH with decreased mitochondrial pools of Rab9 GTPase, defective MDV shedding, and disequilibrium of the mitochondrial fusion-fission cycle in pulmonary ECs. Aerosol intratracheal delivery of adeno-associated virus LKB1 reversed PH, together with improved MDV shedding and mitochondrial function in rats in vivo. We conclude that LKB1 regulates MDV shedding and mitochondrial dynamics in pulmonary ECs by enhancing mitochondrial recruitment of Rab9 GTPase. Defects of LKB1-mediated MDV shedding from parental mitochondria instigate EC dysfunction and PH.
Collapse
Affiliation(s)
- Qiang Zhao
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
- Department of Cardiology, The First Affiliated Hospital of Xian Jiaotong University, Xi’an, Shaanxi, China
| | - Zhixue Liu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
| | - Zuyi Yuan
- Department of Cardiology, The First Affiliated Hospital of Xian Jiaotong University, Xi’an, Shaanxi, China
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
| |
Collapse
|