1
|
Iqbal A, Sajid M, Abdelkrim G, Riaz A, Farooq U, Bibi S, Albadrani GM, Al-Ghadi MQ, Sayed AA, Abdel-Daim MM. Targeting OPA1 protein for therapeutic intervention in autosomal dominant optic atrophy: In silico drug discovery. J Mol Graph Model 2025; 138:109013. [PMID: 40120379 DOI: 10.1016/j.jmgm.2025.109013] [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: 12/22/2024] [Revised: 02/23/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
Autosomal dominant hereditary optic atrophy (ADOA) is a prevalent hereditary condition characterized by the gradual and simultaneous deterioration of vision. Mutations in Optic atrophy 1 (OPA1) have been linked to ADOA, the prevailing form of inherited optic neuropathy. However, the current therapeutic options are limited. This study aimed to identify a drug-like molecule that can serve as an activator of the OPA1 GTPase domain, using in silico virtual screening and molecular dynamic simulation pipeline. A ligand-based pharmacophore model was generated to identify the important biological entities in natural compounds, followed by virtual screening pipeline. Total 55,96,00 drug-like compounds were screened and then subsequently proceed for molecular docking, molecular dynamics simulation (200ns), MM-PBSA analysis, and ADMET (Swiss ADME server) studies. Virtual screening revealed the top-ranked compound ZINC000009190697 (-8 kcal/mol). Furthermore, the stability of the top hit compound at the active site of OPA1 was demonstrated using molecular dynamics simulations and MM-PBSA calculations. ADMET analysis assisted in the identification of the top hit compound as possible activators of OPA1 with optimal drug-like properties. These results indicated that there is need of further experimental assessment of the top-hit compound ZINC000009190697 in wet lab to confirm its efficacy as a potential OPA1 activator in both in vitro and in vivo studies.
Collapse
Affiliation(s)
- Azhar Iqbal
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, 56300, Punjab, Pakistan
| | - Muhammad Sajid
- Department of Biotechnology, Faculty of Life Sciences, University of Okara, Okara, 56300, Punjab, Pakistan.
| | - Guendouzi Abdelkrim
- Laboratory of Chemistry, Synthesis, Properties and Applications. (LCSPA), Department of Chemistry, University of Saida, Algeria
| | - Ammara Riaz
- Department of Life Sciences, Faculty of Natural and Applied Sciences, Khwaja Fareed University of Engineering and Information Technology, Rahimyar Khan, 64200, Punjab, Pakistan
| | - Umar Farooq
- Department of Poultry Science, University of Agriculture Faisalabad Sub-campus Toba Tek Singh, Pakistan
| | - Shabana Bibi
- Department of Biosciences, Shaifa Tameer e Millat University, Islamabad, 44000, Pakistan.
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 84428, Riyadh, 11671, Saudi Arabia
| | - Muath Q Al-Ghadi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Amany A Sayed
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
2
|
Pei X, Liu J, Wei Y, Zhu Y, Li Y, Wang Q, Zhuang L, Jin G. PINK1 affects mitochondrial oxidative phosphorylation by regulating MFN2 to alleviate diabetic kidney disease. Int Immunopharmacol 2025; 157:114786. [PMID: 40319746 DOI: 10.1016/j.intimp.2025.114786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/17/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is widely recognized as a prevalent and major microvascular complication of diabetes mellitus. Mitofusin 2 (MFN2) has been closely linked to the development of diabetes mellitus, yet its precise role in the pathogenesis of DKD remains uncertain. The objective of our current research was to explore the role of MFN2 in the advanced DKD and its underlying molecular pathway. This research was to examine the involvement and molecular pathways of MFN2 in the advancement of DKD. METHODS In this study, MFN2 was manipulated in high glucose (HG)-treated HK2 cells to investigate its impact on cell proliferation, apoptosis, and mitochondrial oxidative phosphorylation. Models of MFN2 overexpression or silencing were established in db/db mice as a diabetes model. The alterations in kidney morphology, renal fibrosis severity, macrophage polarization, and related inflammatory factors (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]) were evaluated. Furthermore, HK2 cells were co-cultivated with M1-type macrophages to examine the impact of MFN2 expression on macrophage polarization. Subsequently, we delved deeper into the upstream mechanisms utilizing the STRING database. RESULTS Our study identified that MFN2 expression was downregulated in HG-treated HK-2 cells. The overexpression of MFN2 resulted in increased HK2 cell proliferation, improved degree of oxidative phosphorylation, and reduced apoptosis. In db/db mice, MFN2 overexpression exerted a protective effect on DKD-induced nephropathy and fibrosis. Notably, MFN2 overexpression influenced macrophage polarization and modulated expression of related inflammatory factors by promoting mitochondrial oxidative phosphorylation. Additionally, STRING database prediction revealed that PTEN-induced kinase 1 (PINK1) regulated MFN2 expression, which was consistent with MFN2 changes in DKD, and this regulation was associated with DKD progression. CONCLUSION The role of MFN2 in maintaining mitochondrial function in DKD may be regulated by PINK1 and provides a new potential therapeutic target for DKD.
Collapse
Affiliation(s)
- Xiaoyan Pei
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China.
| | - Jie Liu
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Yu Wei
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Yanhui Zhu
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Yu Li
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Qiong Wang
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Langen Zhuang
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Guoxi Jin
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China.
| |
Collapse
|
3
|
Ma Y, Sun Y, Ailikenjiang K, Lv C, Li X, Nie Y, Wang C, Xiong Y, Chen Y. Donafenib Induces Mitochondrial Dysfunction in Liver Cancer Cells via DRP1. Cell Biochem Biophys 2025; 83:2379-2388. [PMID: 39937366 DOI: 10.1007/s12013-024-01648-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2024] [Indexed: 02/13/2025]
Abstract
Hepatocellular carcinoma (HCC) represents a significant global health challenge, characterized by a high incidence rate. Mitochondria have emerged as an important therapeutic target for HCC. Donafenib, a multi-receptor tyrosine kinase inhibitor, has been approved for the treatment of advanced HCC. However, the underlying mechanisms remain to be elucidated. In this study, we aim to investigate the effects of Donafenib on mitochondrial function in HCC cells. Firstly, we show that Donafenib induces mitochondrial oxidative stress in SNU-449 liver cancer cells by increasing mitochondrial ROS while reducing glutathione peroxidase (GPx) activity and the expression of Mn-SOD. We also demonstrate that Donafenib decreases mitochondrial membrane potential (MMP) and induces the opening of the mitochondrial permeability transition pore (mPTP). Furthermore, Donafenib reduces mitochondrial respiratory rate, COX IV activity, and ATP production. Notably, Donafenib induces mitochondrial fragmentation and reduces mitochondrial length by increasing the expression of DRP1, without affecting Mfn1 or Mfn2. Silencing of DRP1 protects against mitochondrial dysfunction induced by Donafenib, indicating that DRP1 plays a key role in mediating Donafenib's effects on mitochondrial function in HCC cells.
Collapse
Affiliation(s)
- Yuhua Ma
- Department of Pathology, Karamay Central Hospital, Karamay, Xinjiang, China
| | - Yougang Sun
- Department of General Surgery, Dushanzi People's Hospital, Karamay, Xinjiang, China
| | - Kayishaer Ailikenjiang
- Department of Hepatobiliary and Pancreatic Surgery, Karamay Central Hospital, Karamay, Xinjiang, China
| | - Chuanjiang Lv
- Department of Hepatobiliary and Pancreatic Surgery, Karamay Central Hospital, Karamay, Xinjiang, China
| | - Xiang Li
- Department of Hepatobiliary and Pancreatic Surgery, Karamay Central Hospital, Karamay, Xinjiang, China
| | - YunQiang Nie
- Department of Hepatobiliary and Pancreatic Surgery, Karamay Central Hospital, Karamay, Xinjiang, China
| | - Chang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Karamay Central Hospital, Karamay, Xinjiang, China
| | - Yan Xiong
- Department of General Medicine, Karamay Central Hospital, Karamay, Xinjiang, China.
| | - Yong Chen
- Department of Hepatobiliary and Pancreatic Surgery, Karamay Central Hospital, Karamay, Xinjiang, China.
| |
Collapse
|
4
|
Zhang J, Tao J, Zhou Z, Pei W, Xiao Y, Guo Y, Gao J, Jiang C, Dai L, Zhang G, Tan C. Current research on mitochondria‑associated membranes in cardiovascular diseases (Review). Mol Med Rep 2025; 31:141. [PMID: 40183396 PMCID: PMC11976516 DOI: 10.3892/mmr.2025.13506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025] Open
Abstract
The present study aimed to explore the role of mitochondria‑associated membranes (MAMs) as a key interface between mitochondria and the endoplasmic reticulum (ER) and to evaluate their importance in maintaining the physiological functions of these two organelles. MAMs not only act as a structural bridge between mitochondria and the ER but also widely participate in the regulation of mitochondrial biosynthesis and function, Ca2+ signal transduction, lipid metabolism, oxidative stress response and autophagy. In addition, the specific protein composition of MAMs is increasingly being recognized as having a profound impact on their function, and these proteins play a central role in regulating intercellular communication. Recently, the scientific community has accumulated a large amount of evidence supporting MAMs as potential targets for cardiovascular disease treatment. The present review focuses on the fine structure and multifunctional properties of MAMs and their mechanisms in the occurrence and development of cardiovascular diseases. The goal is to explore the mechanism of MAMs, therapeutic intervention points directly related to cardiovascular diseases, and feasibility of incorporating MAMs into the diagnostic strategy and treatment plan of cardiovascular diseases to provide novel insights and theoretical support for clinical practice in this field. MAMs have great potential as therapeutic targets for various cardiovascular diseases. This finding not only deepens the understanding of the interaction between organelles but also opens up a promising research path for the development of new therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiaheng Zhang
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Jing Tao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Zijuan Zhou
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Wanjuan Pei
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yili Xiao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yanghongxu Guo
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jian Gao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Chenyv Jiang
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Ling Dai
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Guomin Zhang
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Chao Tan
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Inherit Workroom of Medical Master Professor Xiong Ji-bo's Experiences, First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
5
|
Luo H, Jin M, Hu H, Ying Q, Hu P, Sheng W, Huang Y, Xu K, Lu C, Zhang X. SIRT4 Protects Müller Glial Cells Against Apoptosis by Mediating Mitochondrial Dynamics and Oxidative Stress. Mol Neurobiol 2025; 62:6683-6702. [PMID: 39023793 DOI: 10.1007/s12035-024-04349-4] [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: 01/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
SIRT4 is a member of the sirtuin family, which is related to mitochondrial function and possesses antioxidant and regulatory redox effects. Currently, the roles of SIRT4 in retinal Müller glial cells, oxidative stress, and mitochondrial function are still unclear. We confirmed, by immunofluorescence staining, that SIRT4 is located mainly in the mitochondria of retinal Müller glial cells. Using flow cytometry and Western blotting, we analyzed cell apoptosis, intracellular reactive oxygen species (ROS) levels, apoptotic and proapoptotic proteins, mitochondrial dynamics-related proteins, and mitochondrial morphology and number after the overexpression and downregulation of SIRT4 in rMC-1 cells. Neither the upregulation nor the downregulation of SIRT4 alone affected apoptosis. SIRT4 overexpression reduced intracellular ROS, reduced the BAX/BCL2 protein ratio, and increased the L-OPA/S-OPA1 ratio and the levels of the mitochondrial fusion protein MFN2 and the mitochondrial cleavage protein FIS1, increasing mitochondrial fusion. SIRT4 downregulation had the opposite effect. Mitochondria tend to divide after serum starvation for 24 h, and SIRT4 downregulation increases mitochondrial fragmentation and oxidative stress, leading to aggravated cell damage. The mitochondrial division inhibitor Mdivi-1 reduced oxidative stress levels and thus reduced cell damage caused by serum starvation. The overexpression of SIRT4 in rMC-1 cells reduced mitochondrial fragmentation caused by serum starvation, leading to mitochondrial fusion and reduced expression of cleaved caspase-3, thus alleviating the cellular damage caused by oxidative stress. Thus, we speculate that SIRT4 may protect retinal Müller glial cells against apoptosis by mediating mitochondrial dynamics and oxidative stress.
Collapse
Affiliation(s)
- Hongdou Luo
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Ming Jin
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Qian Ying
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Piaopiao Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Weiwei Sheng
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Yi Huang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Ke Xu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Chuming Lu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China.
| |
Collapse
|
6
|
Du Y, Duan C, Zhu X, Lyu M, Chen J, Wei Y, Hu Y. Visual Analysis of Research Hotspots and Trends on Mitochondria-Associated Membranes in the Past 20 Years-Focused on Neurodegenerative Diseases. Mol Neurobiol 2025; 62:7144-7159. [PMID: 39964584 DOI: 10.1007/s12035-025-04722-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/22/2025] [Indexed: 05/15/2025]
Abstract
The interaction between mitochondria and the endoplasmic reticulum is mediated by mitochondria-associated endoplasmic reticulum membranes (MAMs), which play a crucial role in regulating intracellular signal transduction and molecular interactions. This study employs bibliometric analysis to examine the research progress on MAMS and identify research hotspots and trends. A total of 1406 publications on MAMs were collected from the Web of Science Core Collection. Software such as CiteSpace, VOSviewer, and Scimago Graphica were utilized in the bibliometric analysis process. This study conducted a bibliometric analysis of over 20 years of MAM research, identifying the countries, institutions, authors, journals, and publications involved in the field. The number of publications on MAMs has been increasing annually since 2010, exhibiting a steady upward trend. The main contributors to this field are the USA, China, and Italy, with the journal Frontiers in Cell and Developmental Biology publishing the most publications. Common keywords include "endoplasmic reticulum stress," "Ca2 + ," "mitofusin2," "oxidative stress," "apoptosis," "autophagy," and "Alzheimer's disease." We found that the role of MAMs in neurodegenerative diseases has aroused great interest among researchers. The associations between calcium homeostasis, autophagy, mitochondrial dysfunction, and cell death with MAMs are also considered research hotspots and show broad research prospects. In addition, changes in MAM-resident proteins, including the mitochondria-ER tethering complex, Mfn2, and Sig-1R, have been highlighted as prominent research directions. The findings provide a comprehensive overview of research on MAMs and valuable insights for researchers, which is helpful for exploring future directions and trends in this field.
Collapse
Affiliation(s)
- Yihang Du
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglin Duan
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xueping Zhu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meng Lyu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiafan Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Wei
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yuanhui Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Rozich E, Ozkurede U, Pakkiriswami S, Gemilere R, Azarin SM, Liu JC. Mitochondrial oxidative stress, calcium and dynamics in cardiac ischaemia-reperfusion injury. J Physiol 2025. [PMID: 40448972 DOI: 10.1113/jp287770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 05/07/2025] [Indexed: 06/02/2025] Open
Abstract
Ischaemia-reperfusion injury (IRI) is a major cause of cardiomyocyte damage and death from myocardial infarction. Oxidative stress, dysregulated calcium (Ca2+) handling and disrupted mitochondrial dynamics are all key factors in IRI and can play a role in cell death. Mitochondria are a primary source of oxidative stress, which is generated by electron leak from the respiratory chain complexes and the oxidation of accumulated succinate upon reperfusion. The mitochondrial permeability transition pore (mPTP), a high conductance channel that forms following reperfusion of ischaemic mitochondria, has been implicated in reperfusion-induced cell death. Although factors including mitochondrial Ca2+ overload and oxidative stress that regulate mPTP opening have been well characterized, the composition of the mPTP is still actively investigated. Clinically, mPTP opening and IRI complicate treatment of myocardial infarction. Therefore, many possible therapeutics to reduce the damaging effects of reperfusion are under investigation. Antioxidants, pharmaceutical approaches, postconditioning and synthetic polymers have all been investigated for use in IRI. Still, many of these therapeutics of interest have shown mixed evidence underlying their use in preclinical and clinical research. In this review we discuss our current understanding of the contributions of mitochondrial oxidative stress, mitochondrial Ca2+ and mitochondrial dynamics to cardiomyocyte damage and death in IRI, and where further clarification of these mechanisms is needed to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Emily Rozich
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Ulas Ozkurede
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | | | - Ryan Gemilere
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Julia C Liu
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
8
|
Chen L, He Y, Jiang X, Kow ASF, Lee YZ, Tham CL, Yusof R, Lee MT. Regulation of elevated expression of Mcl-1 in hepatocellular carcinoma - a review. J Recept Signal Transduct Res 2025:1-11. [PMID: 40366802 DOI: 10.1080/10799893.2025.2503393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors worldwide. Mcl-1 (myeloid cell leukemia-1) is highly expressed in HCC cells and plays a critical role in chemotherapy resistance and is a major contributor to chemotherapy failure in HCC. The purpose of this study is to review the recent research progress that explores the key factors in regulating Mcl-1 overexpression in HCC cells, contributing to chemotherapy resistance. The related studies from the past decade on agents targeting Mcl-1 to inhibit HCC were also reviewed to provide insights into overcoming chemotherapy resistance in HCC. Mcl-1 overexpression in HCC is mainly regulated by transcription factors (such as STAT3, p53), non-coding RNAs (such as miRNA, lncRNA), cell cycle proteins, mitochondrial dynamics, and the hypoxic microenvironment. Targeting Mcl-1, alongside multi-target combination therapies, may overcome HCC chemotherapy resistance and improve outcomes. Future research should focus on strategies addressing multiple pathways to minimize monotherapy resistance risks and offer enhanced treatment options for the betterment of human health.
Collapse
Affiliation(s)
- Li Chen
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, PR China
| | - Yuwei He
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, PR China
| | - Xudong Jiang
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, PR China
| | | | - Yu Zhao Lee
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Rohana Yusof
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- Office of Postgraduate Studies, UCSI University, Kuala Lumpur, Malaysia
- UCSI Wellbeing Research Centre, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
9
|
Lu X, Tang L, Wu Y, Hilton T, Chen X, Houck K, Fulton S, Han C, Romero S, Wang Y, Kozar R, Cardenas JC, Fu X, Zhang J, Zhao Z, Dong JF. Metabolically aberrant extracellular mitochondria induce oxidative endotheliopathy in traumatic brain injury. Blood Adv 2025; 9:2079-2090. [PMID: 39913776 PMCID: PMC12052699 DOI: 10.1182/bloodadvances.2024015296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/23/2025] [Indexed: 04/24/2025] Open
Abstract
ABSTRACT Traumatic brain injury (TBI) causes endothelial injuries both at the site of injury and in remote regions of the brain. TBI-induced endotheliopathy also disseminates to extracranial organs, such as the lungs. This TBI-induced systemic endotheliopathy has been reported extensively in clinical studies, but its underlying mechanism remains poorly understood. Here, we report results from a study designed to investigate the role of extracellular mitochondria (exMt) in TBI-induced endotheliopathy. We show that exMt are released from injured brains into the circulation, reaching a maximal level of 1.8 × 104/μL at 3 hours after mice were subjected to severe TBI. These exMt express peroxidized cardiolipin on their surface and generate significant amounts of reactive oxygen species, but they produce less adenosine triphosphate compared with intracellular mitochondria of normal cells. When infused into noninjured mice, exMt induce systemic endotheliopathy defined by increased endothelial permeability, tissue edema, and perivascular bleeding. We further demonstrate that endothelial cells (ECs) endocytose exMt through the lipid-scavenging receptor CD36 and dynamin-driven clathrin-mediated pathway. The endocytosed exMt interacted with the endoplasmic reticulum (ER) of ECs through newly formed mitochondria-associated membranes, leading to ER stress and resultant apoptosis. This study delineates a new pathway of exMt-induced endotheliopathy during acute TBI. It also demonstrates a causal role of exMt in endothelial injuries associated with (poly)trauma, severe infection, and autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoxi Lu
- Bloodworks Research Institute, Seattle, WA
- Department of Pediatric Hematology/Oncology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second Hospital, Sichuan University School of Medicine, Chengdu, China
| | - Lujia Tang
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | - Yingang Wu
- Department of Neurosurgery, The First Affiliated Hospital, School of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | | | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | | | | | - Cha Han
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | | | - Yi Wang
- Bloodworks Research Institute, Seattle, WA
| | - Rosemary Kozar
- Department of Surgery, Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD
| | | | - Xiaoyun Fu
- Bloodworks Research Institute, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | - Zilong Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | - Jing-fei Dong
- Bloodworks Research Institute, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
10
|
Lu L, Huang X, Shi Y, Jiang Y, Han Y, Zhang Y. Mitochondrial dysfunction in pregnancy loss: a review. Mol Cell Biochem 2025; 480:2749-2764. [PMID: 39621222 DOI: 10.1007/s11010-024-05171-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/18/2024] [Indexed: 05/03/2025]
Abstract
A receptive endometrium, a healthy embryo, and harmonious communication between the mother and the embryo/fetus are necessary for a healthy and successful pregnancy. Pregnancy loss (PL) can be the outcome if there is a flaw in any of these critical developmental processes. Multiple risk factors contribute to PL, including genetic predispositions, uterine abnormalities, immune imbalances, endocrine dysfunctions, and environmental exposures, among others. Despite extensive investigations, more than half of women with recurrent pregnancy loss (RPL) lack identifiable risk factors, and causes of RPL remain elusive. To date, an accumulating body of evidence indicates that mitochondrial dysfunction in reproductive organs or cells is a potential underlying factor that may trigger PL. In this comprehensive review, we delve into the intricate relationship between mitochondrial dysfunction and PL, examining studies that focus on this connection in the context of diverse reproductive organs and cells, to unravel the interwoven links between these factors and gain a deeper understanding of their interconnectedness.
Collapse
Affiliation(s)
- Lingjing Lu
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xinyue Huang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yuqian Shi
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yue Jiang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yanhua Han
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yuehui Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530, Gothenburg, Sweden.
| |
Collapse
|
11
|
Pan D, Chen P, Zhang H, Zhao Q, Fang W, Ji S, Chen T. Mitochondrial quality control: A promising target of traditional Chinese medicine in the treatment of cardiovascular disease. Pharmacol Res 2025; 215:107712. [PMID: 40154932 DOI: 10.1016/j.phrs.2025.107712] [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: 01/17/2025] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cardiovascular disease remains the leading cause of death globally, and drugs for new targets are urgently needed. Mitochondria are the primary sources of cellular energy, play crucial roles in regulating cellular homeostasis, and are tightly associated with pathological processes in cardiovascular disease. In response to physiological signals and external stimuli in cardiovascular disease, mitochondrial quality control, which mainly includes mitophagy, mitochondrial dynamics, and mitochondrial biogenesis, is initiated to meet cellular requirements and maintain cellular homeostasis. Traditional Chinese Medicine (TCM) has been shown to have pharmacological effects on alleviating cardiac injury in various cardiovascular diseases, including myocardial ischemia/reperfusion, myocardial infarction, and heart failure, by regulating mitochondrial quality control. Recently, several molecular mechanisms of TCM in the treatment of cardiovascular disease have been elucidated. However, mitochondrial quality control by TCM for treating cardiovascular disease has not been investigated. In this review, we aim to decipher the pharmacological effects and molecular mechanisms of TCM in regulating mitochondrial quality in various cardiovascular diseases. We also present our perspectives regarding future research in this field.
Collapse
Affiliation(s)
- Deng Pan
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| | - Pengfei Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Zhao
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Wei Fang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Siyan Ji
- Stomatology Department of Qiqihar Medical College School, Heilongjiang, China
| | - Tielong Chen
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
12
|
Ge T, Zou R, Zhang M, Hu J, He K, Li G, Zhang T, Fan X. Natural products alleviate atrial fibrillation by modulating mitochondrial quality control. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156555. [PMID: 40056631 DOI: 10.1016/j.phymed.2025.156555] [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: 11/13/2024] [Revised: 02/02/2025] [Accepted: 02/21/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Atrial fibrillation (AF), one of the most common cardiac arrhythmias, is associated with high mortality rates and significant healthcare burdens. Mitochondrial homeostasis has recently emerged as a critical factor in AF pathogenesis but remains at the experimental stage. Current drug and surgical treatments for AF often involve side effects and require ongoing treatment plan evaluation and adjustment. In contrast, natural products (NPs), which have been utilized in China for over 2,000 years, show remarkable efficacy in treating AF and are receiving growing attention. PURPOSE We aimed to investigate the regulatory effects of NPs on mitochondrial quality control (MQC) and their impact on AF occurrence and progression. By constructing a novel NP-mitochondria-AF axis, we propose a framework to translate experimental findings into clinical practice and identify potential therapeutic strategies for AF. METHODS Databases such as PubMed, Web of Science, and China National Knowledge Infrastructure were searched (up to October 2024) using the following keywords: "atrial fibrillation," "traditional Chinese medicine," "mitochondrial biogenesis," "mitochondrial dynamics," "mitophagy," "apoptosis," "oxidative stress," "inflammation," and "Ca2+ concentration." NP targets were identified using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform, while disease targets were retrieved from Online Mendelian Inheritance in Man, GeneCards, and Therapeutic Target Database. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was performed using the Metascape database. Protein-protein interactions were analyzed using the STRING database, and core monomers and hub genes were identified using Cytoscape 3.7.2. RESULTS We found a strong relationship between mitochondrial homeostasis and AF development. KEGG pathway analysis indicated that commonly used NPs regulate mitochondrial homeostasis, affecting AF progression through various hub genes, including protein kinase B-alpha (AKT1), jun proto-oncogene (JUN), and tumor necrosis factor (TNF). Molecular docking analysis revealed that NP core monomers exhibited binding affinities to hub genes below -5 kcal/mol and to transforming growth factor-β (TGF-β) below -7 kcal/mol. CONCLUSION NPs, including traditional Chinese medicine (TCM) compounds, TCM monomers, and traditional Chinese patent medicines, alleviate AF by modulating MQC with minimal side effects and high efficacy. These findings highlight the therapeutic potential of NPs as promising candidates for AF treatment and further underscore the importance of MQC in AF pathogenesis.
Collapse
Affiliation(s)
- Teng Ge
- School of Second Clinical Medical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Rongjun Zou
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Guangdong Provincial Key Laboratory of TCM Emergency Research, Guangzhou 510120, Guangdong, PR China
| | - Miao Zhang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Jinlin Hu
- School of Second Clinical Medical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Kunyang He
- School of Second Clinical Medical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Guanmou Li
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, PR China
| | - Tong Zhang
- Heart Failure Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong, PR China.
| | - Xiaoping Fan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Guangdong Provincial Key Laboratory of TCM Emergency Research, Guangzhou 510120, Guangdong, PR China.
| |
Collapse
|
13
|
Cai H, He J, Zheng W, Cheng H, Ge X, Bao Y, Wei Y, Zhou Y, Liang X, Chen X, Liu C, Wang F, Yang X. Zinc Mitigates the Combined Neurotoxicity of Binary Metal Mixtures via Mitophagy and Mitochondrial Fusion. Mol Neurobiol 2025; 62:5961-5976. [PMID: 39673661 DOI: 10.1007/s12035-024-04648-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: 03/27/2024] [Accepted: 11/25/2024] [Indexed: 12/16/2024]
Abstract
Environmental metal mixtures can cause combined neurotoxicity, but the underlying mechanism remains unclear. Mitochondria are crucial for energy metabolism in the nervous system, and their dysfunction leads to neurodegeneration. Zinc (Zn) is a coenzyme of many mitochondrial enzymes that controls mitochondrial function. This study investigated the role of Zn in the neurotoxicity induced by Mn + Pb and Pb + As mixtures. Zn supplementation improved the survival rate and learning ability of Caenorhabditis elegans following their exposure to mixtures of Mn + Pb and Pb + As by enhancing their mitochondrial morphology, membrane potential, and respiratory chain. Similarly, in HT22 cells, Zn mitigated the decrease in cellular activity and increase in apoptosis induced by the Mn + Pb and Pb + As mixtures by improving mitochondrial morphology and function. Mechanistically, Zn activated the PINK1 and MFN-2/OPA-1 pathways, promoting mitophagy and mitochondrial fusion. However, inhibition of mitophagy reversed the protective effect of Zn, indicating its reliance on mitophagy for neuroprotection. Our study demonstrated that Zn alleviates the combined neurotoxicity of Mn + Pb and Pb + As mixtures by enhancing mitophagy and mitochondrial fusion, suggesting that Zn supplementation is a potential treatment for metal-induced neurotoxicity.
Collapse
Affiliation(s)
- Haiqing Cai
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Junxiu He
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Wanting Zheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoting Ge
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yu Bao
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yue Wei
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanfeng Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Liang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xing Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Chaoqun Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Fei Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
14
|
Wang X, Liu Y, Wang J, Lu X, Guo Z, Lv S, Sun Z, Gao T, Gao F, Yuan J. Mitochondrial Quality Control in Ovarian Function: From Mechanisms to Therapeutic Strategies. Reprod Sci 2025; 32:1399-1413. [PMID: 38981995 DOI: 10.1007/s43032-024-01634-4] [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: 04/02/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Mitochondrial quality control plays a critical role in cytogenetic development by regulating various cell-death pathways and modulating the release of reactive oxygen species (ROS). Dysregulated mitochondrial quality control can lead to a broad spectrum of diseases, including reproductive disorders, particularly female infertility. Ovarian insufficiency is a significant contributor to female infertility, given its high prevalence, complex pathogenesis, and profound impact on women's health. Understanding the pathogenesis of ovarian insufficiency and devising treatment strategies based on this understanding are crucial. Oocytes and granulosa cells (GCs) are the primary ovarian cell types, with GCs regulated by oocytes, fulfilling their specific energy requirements prior to ovulation. Dysregulation of mitochondrial quality control through gene knockout or external stimuli can precipitate apoptosis, inflammatory responses, or ferroptosis in both oocytes and GCs, exacerbating ovarian insufficiency. This review aimed to delineate the regulatory mechanisms of mitochondrial quality control in GCs and oocytes during ovarian development. This study highlights the adverse consequences of dysregulated mitochondrial quality control on GCs and oocyte development and proposes therapeutic interventions for ovarian insufficiency based on mitochondrial quality control. These insights provide a foundation for future clinical approaches for treating ovarian insufficiency.
Collapse
Affiliation(s)
- Xiaomei Wang
- College of Basic Medical, Jining Medical University, Jining, China
| | - Yuxin Liu
- College of Second Clinical Medicine, Jining Medical University, Jining, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
| | - Jinzheng Wang
- College of Second Clinical Medicine, Jining Medical University, Jining, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
| | - Xueyi Lu
- College of Clinical Medicine, Jining Medical University, Jining, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
| | - Zhipeng Guo
- College of Second Clinical Medicine, Jining Medical University, Jining, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
| | - Shenmin Lv
- College of Second Clinical Medicine, Jining Medical University, Jining, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
| | - Zhenyu Sun
- College of Clinical Medicine, Jining Medical University, Jining, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
| | - Tan Gao
- College of Second Clinical Medicine, Jining Medical University, Jining, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
| | - Fei Gao
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China.
| |
Collapse
|
15
|
Shi X, Tian Y, Wang Y, Zhang Y, Yin Y, Tian Q, Li L, Ma B, He X, Zhou L. Mitofusin 1 Drives Preimplantation Development by Enhancing Chromatin Incorporation of Histone H3.3. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414985. [PMID: 40091361 PMCID: PMC12079336 DOI: 10.1002/advs.202414985] [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] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Mitofusin 1 (MFN1) plays a crucial role in mitochondrial fusion and oocyte development. However, its function in preimplantation embryonic development and its potential involvement in epigenetic regulation remain poorly understood. In this study, it is shown that MFN1 interacts with PADI6, a key component of the cytoplasmic lattice in oocytes and early embryos. MFN1 deficiency in mice results in reduced PADI6 levels and decreased expression of translational machinery components, which suppress protein synthesis activity and lower histone H3.3 abundance. These disruptions lead to the failure of male pronucleus formation, aberrant zygotic genome activation, and impaired embryonic development. It is further demonstrated that the MFN1 activator S89 promotes H3.3 incorporation and rescues early development in maternally aged embryos with low MFN1 levels. Additionally, a positive correlation between MFN1 and H3.3 protein levels in early human embryos is observed. Together, these findings provide new insights into MFN1's role in regulating epigenetic reprogramming during preimplantation embryo development.
Collapse
Affiliation(s)
- Xiao‐yan Shi
- Institute of Reproductive HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Yu Tian
- Institute of Reproductive HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Yu‐fan Wang
- Institute of Reproductive HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Yi‐ran Zhang
- Institute of Reproductive HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Ying Yin
- Department of PhysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Qing Tian
- Department of Gynecology and ObstetricsZhongnan Hospital of Wuhan UniversityWuhanHubei430071China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101P. R. China
| | - Bing‐xin Ma
- Reproductive Medicine CenterTongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030P. R. China
| | - Ximiao He
- Department of PhysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic EvaluationHuazhong University of Science and TechnologyWuhanHubei430030China
| | - Li‐quan Zhou
- Institute of Reproductive HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
| |
Collapse
|
16
|
Kirchweger P, Wolf SG, Varsano N, Dadosh T, Resch GP, Elbaum M. Snapshots of mitochondrial fission imaged by cryo-scanning transmission electron tomography. J Cell Sci 2025; 138:jcs263639. [PMID: 40365741 DOI: 10.1242/jcs.263639] [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: 10/28/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondria undergo constant remodeling via fission, fusion, extension and degradation. Fission, in particular, depends on the accumulation of mitochondrial fission factor (MFF) and subsequent recruitment of the dynamin-related protein DRP1 (also known as DNM1L). We used cryo-scanning transmission electron tomography (cryo-STET) to investigate mitochondrial morphologies in MFF mutant (MFF-/-) mouse embryonic fibroblast (MEF) cells in ATP-depleting conditions that normally induce fission. The capability of cryo-STET to image through the cytoplasmic volume to a depth of 1 µm facilitated visualization of intact mitochondria and their surroundings. We imaged changes in mitochondrial morphology and cristae structure, as well as contacts with the endoplasmic reticulum (ER), degradative organelles and the cytoskeleton at stalled fission sites. We found disruption of the outer mitochondrial membrane at contact sites with the ER and degradative organelles at sites of mitophagy. We identified fission sites where the inner mitochondrial membrane is already separated while the outer membrane is still continuous. Although MFF is a general fission factor, these observations demonstrate that mitochondrial fission can proceed to the final stage in its absence. The use of cryo-STET allays concerns about the loss of structures due to sample thinning required for tomography using cryo-transmission electron microscopy.
Collapse
Affiliation(s)
- Peter Kirchweger
- Department of Chemical and Biological Physics, Weizmann Institute of Sciences, 7610001 Rehovot, Israel
- Department of Chemical and Structural Biology, Weizmann Institute of Sciences, 7610001 Rehovot, Israel
| | - Sharon Grayer Wolf
- Department of Chemical Research Support, Weizmann Institute of Sciences, 7610001 Rehovot, Israel
| | - Neta Varsano
- Department of Chemical Research Support, Weizmann Institute of Sciences, 7610001 Rehovot, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Weizmann Institute of Sciences, 7610001 Rehovot, Israel
| | - Guenter P Resch
- Nexperion e.U.-Solutions for Electron Microscopy, 1220 Vienna, Austria
| | - Michael Elbaum
- Department of Chemical and Biological Physics, Weizmann Institute of Sciences, 7610001 Rehovot, Israel
| |
Collapse
|
17
|
Gordaliza-Alaguero I, Sànchez-Fernàndez-de-Landa P, Radivojevikj D, Villarreal L, Arauz-Garofalo G, Gay M, Martinez-Vicente M, Seco J, Martín-Malpartida P, Vilaseca M, Macías MJ, Palacin M, Ivanova S, Zorzano A. Endogenous interactomes of MFN1 and MFN2 provide novel insights into interorganelle communication and autophagy. Autophagy 2025; 21:957-978. [PMID: 39675054 PMCID: PMC12013434 DOI: 10.1080/15548627.2024.2440843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024] Open
Abstract
MFN1 (mitofusin 1) and MFN2 are key players in mitochondrial fusion, endoplasmic reticulum (ER)-mitochondria juxtaposition, and macroautophagy/autophagy. However, the mechanisms by which these proteins participate in these processes are poorly understood. Here, we studied the interactomes of these two proteins by using CRISPR-Cas9 technology to insert an HA-tag at the C terminus of MFN1 and MFN2, and thus generating HeLa cell lines that endogenously expressed MFN1-HA or MFN2-HA. HA-affinity isolation followed by mass spectrometry identified potential interactors of MFN1 and MFN2. A substantial proportion of interactors were common for MFN1 and MFN2 and were regulated by nutrient deprivation. We validated novel ER and endosomal partners of MFN1 and/or MFN2 with a potential role in interorganelle communication. We characterized RAB5C (RAB5C, member RAS oncogene family) as an endosomal modulator of mitochondrial homeostasis, and SLC27A2 (solute carrier family 27 (fatty acid transporter), member 2) as a novel partner of MFN2 relevant in autophagy. We conclude that MFN proteins participate in nutrient-modulated pathways involved in organelle communication and autophagy.Abbreviations: ACTB: actin, beta; ATG2: autophagy related 2; ATG5: autophagy related 5; ATG12: autophagy related 12; ATG14: autophagy related 14; ATG16L1: autophagy related 16 like 1; Baf A1: bafilomycin A1; BECN1: beclin 1, autophagy related; BFDR: Bayesian false discovery rate; Cas9: CRISPR-associated endonuclease Cas9; CRISPR: clustered regularly interspaced short palindromic repeats; DNM1L/DRP1: dynamin 1-like; ER: endoplasmic reticulum; Faa1: fatty acid activation 1; FC: fold change; FDR: false discovery rate; FIS1: fission, mitochondrial 1; GABARAP: gamma-aminobutyric acid receptor associated protein; GABARAPL2: GABA type A receptor associated protein like 2; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HA: hemagglutinin; KO: knockout; LIR: LC3-interacting region; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MARCHF5: membrane associated ring-CH-type finger 5; MDVs: mitochondria-derived vesicles; MFN1: mitofusin 1; MFN2: mitofusin 2; NDFIP2: Nedd4 family interacting protein 2; OMM: outer mitochondrial membrane; OPA1: OPA1, mitochondrial dynamin like GTPase; OXPHOS: oxidative phosphorylation; PE: phosphatidylethanolamine; PINK1: PTEN induced putative kinase 1; PS: phosphatidylserine; RAB5C: RAB5C, member RAS oncogene family; S100A8: S100 calcium binding protein A8 (calgranulin A); S100A9: S100 calcium binding protein A9 (calgranulin B); SLC27A2: solute carrier family 27 (fatty acid transporter), member 2; TIMM44: translocase of inner mitochondrial membrane 44; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like kinase 1; VCL: vinculin; VDAC1: voltage-dependent anion channel 1; WT: wild type.
Collapse
Affiliation(s)
- Isabel Gordaliza-Alaguero
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Paula Sànchez-Fernàndez-de-Landa
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Dragana Radivojevikj
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Villarreal
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
| | - Gianluca Arauz-Garofalo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
| | - Marina Gay
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
| | - Marta Martinez-Vicente
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Jorge Seco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
| | - Pau Martín-Malpartida
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
| | - Marta Vilaseca
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
| | - María J. Macías
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Manuel Palacin
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomedica En Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Saška Ivanova
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Scudese E, Marshall AG, Vue Z, Exil V, Rodriguez BI, Demirci M, Vang L, López EG, Neikirk K, Shao B, Le H, Stephens D, Hall DD, Rostami R, Rodman T, Kabugi K, Shao JQ, Mungai M, AshShareef ST, Hicsasmaz I, Manus S, Wanjalla CN, Whiteside A, Dasari R, Williams CR, Damo SM, Gaddy JA, Glancy B, Dantas EHM, Kinder A, Kadam A, Tomar D, Scartoni F, Baffi M, McReynolds MR, Phillips MA, Cooper A, Murray SA, Quintana AM, Wandira N, Ochayi OM, Ameka M, Kirabo A, Masenga SK, Harris C, Oliver A, Martin P, Gaye A, Korolkova O, Sharma V, Mobley BC, Katti P, Hinton A. 3D Mitochondrial Structure in Aging Human Skeletal Muscle: Insights Into MFN-2-Mediated Changes. Aging Cell 2025:e70054. [PMID: 40285369 DOI: 10.1111/acel.70054] [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: 12/14/2023] [Revised: 02/21/2025] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Age-related skeletal muscle atrophy, known as sarcopenia, is characterized by loss of muscle mass, strength, endurance, and oxidative capacity. Although exercise has been shown to mitigate sarcopenia, the underlying governing mechanisms are poorly understood. Mitochondrial dysfunction is implicated in aging and sarcopenia; however, few studies explore how mitochondrial structure contributes to this dysfunction. In this study, we sought to understand how aging impacts mitochondrial three-dimensional (3D) structure and its regulators in skeletal muscle. We hypothesized that aging leads to remodeling of mitochondrial 3D architecture permissive to dysfunction and is ameliorated by exercise. Using serial block-face scanning electron microscopy (SBF-SEM) and Amira software, mitochondrial 3D reconstructions from patient biopsies were generated and analyzed. Across five human cohorts, we correlate differences in magnetic resonance imaging, mitochondria 3D structure, exercise parameters, and plasma immune markers between young (under 50 years) and old (over 50 years) individuals. We found that mitochondria are less spherical and more complex, indicating age-related declines in contact site capacity. Additionally, aged samples showed a larger volume phenotype in both female and male humans, indicating potential mitochondrial swelling. Concomitantly, muscle area, exercise capacity, and mitochondrial dynamic proteins showed age-related losses. Exercise stimulation restored mitofusin 2 (MFN2), one such of these mitochondrial dynamic proteins, which we show is required for the integrity of mitochondrial structure. Furthermore, we show that this pathway is evolutionarily conserved, as Marf, the MFN2 ortholog in Drosophila, knockdown alters mitochondrial morphology and leads to the downregulation of genes regulating mitochondrial processes. Our results define age-related structural changes in mitochondria and further suggest that exercise may mitigate age-related structural decline through modulation of mitofusin 2.
Collapse
Affiliation(s)
- Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Vernat Exil
- Department of Pediatrics, Div. of Cardiology, St. Louis University School of Medicine, St. Louis, MO, USA
| | - Benjamin I Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Mert Demirci
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Edgar Garza López
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Duane D Hall
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Rahmati Rostami
- Department of Genetic Medicine, Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Taylor Rodman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | | | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | | | - Innes Hicsasmaz
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Sasha Manus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Celestine N Wanjalla
- Division of Infection Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Revathi Dasari
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, AP, India
| | - Clintoria R Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, USA
| | - Jennifer A Gaddy
- Division of Infection Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, USA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- NIAMS, NIH, Bethesda, MD, USA
| | - Estélio Henrique Martin Dantas
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Doctor's Degree Program in Nursing and Biosciences - PpgEnfBio, Federal University of the State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
- Laboratory of Human Motricity Biosciences - LABIMH, Federal University of the State of Rio de Janeiro - UNIRIO, RJ, Brazil
- Brazilian Paralympic Academy - APB, Brazil
- Doctor's Degree Program in Health and Environment - PSA, Tiradentes University - UNIT, Aracaju, SE, Brazil
| | - André Kinder
- Artur Sá Earp Neto University Center - UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Fabiana Scartoni
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Matheus Baffi
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA, USA
| | - Mark A Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, USA
| | - Anthonya Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anita M Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Nelson Wandira
- Institute of Health Sciences Busoga University, Iganga, Uganda
| | - Okwute M Ochayi
- Department of Human Physiology, Baze University, Abuja, Nigeria
| | - Magdalene Ameka
- KAVI Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Sepiso K Masenga
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
| | - Chanel Harris
- Department of Biomedical Sciences, Meharry Medical College, Nashville, US
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, US
| | - Pamela Martin
- Department of Biomedical Sciences, Meharry Medical College, Nashville, US
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, Meharry Medical College, Nashville, TN, USA
| | - Olga Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Vineeta Sharma
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Bret C Mobley
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Prasanna Katti
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, AP, India
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
19
|
Willett BAS, Thompson SB, Chen V, Dareshouri A, Jackson CL, Brunetti T, D'Alessandro A, Klarquist J, Nemkov T, Kedl RM. Mitochondrial protein OPA1 is required for the expansion of effector CD8 T cells. Cell Rep 2025; 44:115610. [PMID: 40261796 DOI: 10.1016/j.celrep.2025.115610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025] Open
Abstract
Short-lived effector cells are characterized metabolically by a highly glycolytic state, driving energy and biomass acquisition, whereas memory-fated T cells have historically been described as meeting these requirements through mitochondrial metabolism. Here, we show that the mitochondrial protein optic atrophy 1 (OPA1) is critical for rapidly dividing CD8 T cells in vivo, the requirement for which is most pronounced in effector CD8 T cells. More specifically, OPA1 supports proper cell cycle initiation and progression and the viability and survival of CD8 T cells during clonal expansion. Use of mice deficient in the mitochondrial membrane fusion proteins Mitofusin 1 and 2 (MFN1/2) in both in vivo proliferation/differentiation assays and ex vivo metabolic analysis indicates that the requirement for OPA1 during cell division supersedes its role in mitochondrial fusion. We conclude that OPA1 is critical for the generation and accumulation of short-lived effector cells that arise during the response to infection.
Collapse
Affiliation(s)
- Benjamin A S Willett
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Scott B Thompson
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vincent Chen
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anza Dareshouri
- Department of Cell and Developmental Biology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Conner L Jackson
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tonya Brunetti
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Travis Nemkov
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
20
|
Lv Y, Tao L, Hu L, Du C, Wang H, Zhang H, Hu Y, Chen L. Postmortem mitochondrial membrane potential dynamics as a temperature-independent biomarker for early postmortem interval estimation. Leg Med (Tokyo) 2025; 75:102626. [PMID: 40267737 DOI: 10.1016/j.legalmed.2025.102626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/02/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Accurate determination of postmortem interval (PMI) and cause of death (COD) is a critical challenge in forensic pathology, with significant implications for criminal investigations. Traditional PMI estimation methods are based on macroscopic changes and are influenced by environmental factors and investigator subjectivity. Recent advances in molecular biology have shown that certain cellular structures, such as mitochondria, retain functionality after death, making them potential biomarkers for forensic assessment. As mitochondria play a central role in cellular metabolism and respond dynamically to post-mortem hypoxia, investigation of mitochondrial membrane potential (ΔΨm) may provide a quantifiable and objective method for estimating PMI. RESULTS We successfully isolated mitochondria from post-mortem tissues and cultured cells, confirming their purity and membrane integrity. Regression analysis showed a strong linear correlation between ΔΨm and PMI in brain, myocardium and skeletal muscle within the first 15-18 h postmortem, with skeletal muscle showing the highest correlation coefficient. ΔΨm values remained stable at different temperatures, suggesting that it is a robust biomarker for estimating PMI. In vitro experiments under hypoxic conditions revealed a transient increase in ΔΨm at 24 h, accompanied by ATP depletion, ROS accumulation and shifts in mitochondrial fission and fusion dynamics, indicating mitochondrial adaptation to oxygen deprivation. CONCLUSIONS These findings highlight ΔΨm as a promising temperature stable biomarker for early assessment of PMI. The observed mitochondrial adaptations suggest that ΔΨm-based models may improve forensic accuracy and provide insights into postmortem metabolic processes. Further validation with human postmortem samples is essential to refine these models and explore their applicability to COD determination.
Collapse
Affiliation(s)
- Yehui Lv
- Institute of Wound Prevention and Treatment, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - Li Tao
- Human Anatomy Teaching and Research Section, School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, China
| | - Luyuyan Hu
- Shanghai Fenglin Forensic Science Center, Shanghai 200231, China
| | - Chengqiang Du
- Institute of Wound Prevention and Treatment, School of Basic Medical Sciences, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - Hui Wang
- Department of Forensic Science, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Heng Zhang
- School of Basic Medical Sciences, Anhui Medical University, Anhui 230032, China
| | - Yikai Hu
- Department of Forensic Science, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Long Chen
- Department of Forensic Science, School of Basic Medical Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
21
|
Liao Y, Octaviani S, Tian Z, Wang SR, Huang C, Huang J. Mitochondrial quality control in hematopoietic stem cells: mechanisms, implications, and therapeutic opportunities. Stem Cell Res Ther 2025; 16:180. [PMID: 40234908 PMCID: PMC12001479 DOI: 10.1186/s13287-025-04304-7] [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: 01/28/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Mitochondrial quality control (MQC) is a critical mechanism for maintaining mitochondrial function and cellular metabolic homeostasis, playing an essential role in the self-renewal, differentiation, and long-term stability of hematopoietic stem cells (HSCs). Recent research highlights the central importance of MQC in HSC biology, particularly the roles of mitophagy, mitochondrial biogenesis, fission, fusion and mitochondrial transfer in regulating HSC function. Mitophagy ensures the removal of damaged mitochondria, maintaining low levels of reactive oxygen species (ROS) in HSCs, thereby preventing premature aging and functional decline. Concurrently, mitochondrial biogenesis adjusts key metabolic regulators such as mitochondrial transcription factor A (TFAM) and peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) to meet environmental demands, ensuring the metabolic needs of HSCs are met. Additionally, mitochondrial transfer, as an essential form of intercellular material exchange, facilitates the transfer of functional mitochondria from bone marrow stromal cells to HSCs, contributing to damage repair and metabolic support. Although existing studies have revealed the significance of MQC in maintaining HSC function, the precise molecular mechanisms and interactions among different regulatory pathways remain to be fully elucidated. Furthermore, the potential role of MQC dysfunction in hematopoietic disorders, including its involvement in disease progression and therapeutic resistance, is not yet fully understood. This review discusses the molecular mechanisms of MQC in HSCs, its functions under physiological and pathological conditions, and its potential therapeutic applications. By summarizing the current progress in this field, we aim to provide insights for further research and the development of innovative treatment strategies.
Collapse
Affiliation(s)
- Yun Liao
- Coriell Institute for Medical Research, Camden, NJ, USA
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | | | - Zhen Tian
- Coriell Institute for Medical Research, Camden, NJ, USA
| | | | - Chunlan Huang
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, Sichuan, China.
| | - Jian Huang
- Coriell Institute for Medical Research, Camden, NJ, USA.
- Cooper Medical School of Rowan University, Camden, NJ, USA.
| |
Collapse
|
22
|
McGill Percy KC, Liu Z, Qi X. Mitochondrial dysfunction in Alzheimer's disease: Guiding the path to targeted therapies. Neurotherapeutics 2025; 22:e00525. [PMID: 39827052 PMCID: PMC12047401 DOI: 10.1016/j.neurot.2025.e00525] [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: 08/31/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration, marked by the accumulation of amyloid-β (Aβ) plaques and tau tangles. Emerging evidence suggests that mitochondrial dysfunction plays a pivotal role in AD pathogenesis, driven by impairments in mitochondrial quality control (MQC) mechanisms. MQC is crucial for maintaining mitochondrial integrity through processes such as proteostasis, mitochondrial dynamics, mitophagy, and precise communication with other subcellular organelles. In AD, disruptions in these processes lead to bioenergetic failure, gene dysregulation, the accumulation of damaged mitochondria, neuroinflammation, and lipid homeostasis impairment, further exacerbating neurodegeneration. This review elucidates the molecular pathways involved in MQC and their pathological relevance in AD, highlighting recent discoveries related to mitochondrial mechanisms underlying neurodegeneration. Furthermore, we explore potential therapeutic strategies targeting mitochondrial dysfunction, including gene therapy and pharmacological interventions, offering new avenues for slowing AD progression. The complex interplay between mitochondrial health and neurodegeneration underscores the need for innovative approaches to restore mitochondrial function and mitigate the onset and progression of AD.
Collapse
Affiliation(s)
- Kyle C McGill Percy
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zunren Liu
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xin Qi
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Center for Mitochondrial Research and Therapeutics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
23
|
Li X, Lin Q, Guan B, Yang M, Huang X, Li L, Chen C, Hong J, Zhang M. Multi-Omics Analysis Links Mitochondrial-Related Genes to Idiopathic Pulmonary Fibrosis and In Vivo Transcriptome Validation. Transl Res 2025; 278:10-21. [PMID: 39952317 DOI: 10.1016/j.trsl.2025.02.002] [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: 01/06/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Mitochondrial dysfunction is closely associated with idiopathic pulmonary fibrosis (IPF). However, the causal association between mitochondria-related genes and IPF remains to be determined. We obtained gene expression, protein abundance, and methylation quantitative trait locus data for mitochondria-related genes from previous studies. Genome-wide association study data for patients with IPF were obtained from the FinnGen study. A two-sample Mendelian randomisation analysis was conducted to assess the association between mitochondria-related genes and IPF. Furthermore, we conducted validation of expression differences utilizing transcriptome data derived from the BLM-induced pulmonary fibrosis mouse model (n=15). Concurrently, multiphoton imaging was utilized to quantify collagen contents and structural assessment. The direction of causality was verified using the Steiger test, and colocalisation analysis was used to better validate causality. Single-cell data were used to explore the localisation and expression of positive genes across different cell types. The study identified significant associations between mitochondria-related genes and IPF, with POLG and NDUFB10 classified as Grade 1; LYRM4, NBR1, and ACSF3 as Grade 2; MCL1, GFER, MFN2, IVD, and SLC25A35 as Grade 3; and METAP1D and MTX1 as Grade 4. Single-cell analysis showed elevated expression of NBR1, MCL1, and MTX1 in pulmonary myofibroblasts of IPF. This study elucidated the causal effects of mitochondria-related genes on IPF, underscoring their significance in pathogenesis. These findings contribute to an improved understanding of the mechanisms underlying IPF, offering new potential therapeutic targets for interventions.
Collapse
Affiliation(s)
- Xiaoxia Li
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Qiaojing Lin
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Bingyue Guan
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Minghuan Yang
- School of Public Health, Fujian Medical University, Fuzhou, PR China
| | - Xingxin Huang
- School of Arts and Sciences, Fujian Medical University, Fuzhou, PR China
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, PR China.
| | - Chun Chen
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China; School of Pharmacy, Fujian Medical University, Fuzhou, PR China.
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China; School of Medical Imaging, Fujian Medical University, Fuzhou, PR China.
| | - Mingwei Zhang
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China; School of Medical Imaging, Fujian Medical University, Fuzhou, PR China.
| |
Collapse
|
24
|
Erdogan CS, Yavuz Y, Ozgun HB, Bilgin VA, Agus S, Kalkan UF, Yilmaz B. Fam163a knockdown and mitochondrial stress in the arcuate nucleus of hypothalamus reduce AgRP neuron activity and differentially regulate mitochondrial dynamics in mice. Acta Physiol (Oxf) 2025; 241:e70020. [PMID: 40071489 PMCID: PMC11897941 DOI: 10.1111/apha.70020] [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: 10/21/2024] [Revised: 01/21/2025] [Accepted: 02/21/2025] [Indexed: 03/15/2025]
Abstract
AIM Mitochondria play key roles in neuronal activity, particularly in modulating agouti-related protein (AgRP) and proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC), which regulates food intake. FAM163A, a newly identified protein, is suggested to be part of the mitochondrial proteome, though its functions remain largely unknown. This study aimed to investigate the effects of Fam163a knockdown and mitochondrial dysfunction on food intake, AgRP neuron activity, and mitochondrial function in the hypothalamus. METHODS Male C57BL/6 and AgRP-Cre mice received intracranial injections of either Fam163a shRNA, rotenone, or appropriate controls. Behavioral assessments included food intake, locomotor activity, and anxiety-like behaviors. qRT-PCR was used to quantify the expression of the genes related to food intake, mitochondrial biogenesis, dynamics, and oxidative stress. Blood glucose, serum insulin, and leptin levels were measured. Electrophysiological patch-clamp recordings were used to assess the AgRP neuronal activity. RESULTS Fam163a knockdown in the ARC increased the cumulative food intake in short term (first 7 days) without altering the 25-day food intake and significantly increased the Pomc mRNA expression. Fam163a silencing significantly reduced leptin levels. Both Fam163a knockdown and rotenone significantly reduced the firing frequency of AgRP neurons. Neither Fam163a silencing nor rotenone altered locomotor or anxiety-like behaviors. Fam163a knockdown and rotenone differentially altered the expression of mitochondrial biogenesis-, mitophagy-, fusion-, and oxidative stress-related genes. CONCLUSION Hypothalamic FAM163A may play a role in modulating AgRP neuronal activity through regulating mitochondrial biogenesis, dynamics, and redox state. These findings provide insights into the role of FAM163A and mitochondrial stress in the central regulation of metabolism.
Collapse
Affiliation(s)
| | - Yavuz Yavuz
- Department of PhysiologyFaculty of Medicine, Yeditepe UniversityIstanbulTurkey
- Department of Neuroscience and PharmacologyThe University of Iowa Carver College of MedicineIowa CityUSA
| | - Huseyin Bugra Ozgun
- Department of PhysiologyFaculty of Medicine, Yeditepe UniversityIstanbulTurkey
| | - Volkan Adem Bilgin
- Department of PhysiologyFaculty of Medicine, Yeditepe UniversityIstanbulTurkey
| | - Sami Agus
- Department of PhysiologyFaculty of Medicine, Yeditepe UniversityIstanbulTurkey
- Department of PhysiologyAugusta UniversityAugustaGeorgiaUSA
| | - Ugur Faruk Kalkan
- Department of PhysiologyFaculty of Medicine, Yeditepe UniversityIstanbulTurkey
| | - Bayram Yilmaz
- Department of PhysiologyFaculty of Medicine, Yeditepe UniversityIstanbulTurkey
- Department of Physiology, Faculty of MedicineDokuz Eylül UniversityIzmirTurkey
- Izmir Biomedicine and Genome CenterIzmirTurkey
| |
Collapse
|
25
|
Fan H, Yuan M, Wang S, Yang X, Shu L, Pu Y, Zou Q, Zhang X, Wang C, Cai Z. Dietary salt promotes cognitive impairment through repression of SIRT3/PINK1-mediated mitophagy and fission. Mol Cell Biochem 2025; 480:2345-2360. [PMID: 38997506 DOI: 10.1007/s11010-024-05069-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/04/2024] [Indexed: 07/14/2024]
Abstract
Dietary salt is increasingly recognized as an independent risk factor for cognitive impairment. However, the exact mechanisms are not yet fully understood. Mitochondria, which play a crucial role in energy metabolism, are implicated in cognitive function through processes such as mitochondrial dynamics and mitophagy. While mitochondrial dysfunction is acknowledged as a significant determinant of cognitive function, the specific relationship between salt-induced cognitive impairment and mitochondrial health has yet to be fully elucidated. Here, we explored the underlying mechanism of cognitive impairment of mice and N2a cells treated with high-salt focusing on the mitochondrial homeostasis with western blotting, immunofluorescence, electron microscopy, RNA sequencing, and more. We further explored the potential role of SIRT3 in salt-induced mitochondrial dysfunction and synaptic alteration through plasmid transfection and siRNA. High salt diet significantly inhibited mitochondrial fission and blocked mitophagy, leading to dysfunctional mitochondria and impaired synaptic plasticity. Our findings demonstrated that SIRT3 not only promote mitochondrial fission by modulating phosphorylated DRP1, but also rescue mitophagy through promoting PINK1/Parkin-dependent pathway. Overall, our data for the first time indicate that mitochondrial homeostasis imbalance is a driver of impaired synaptic plasticity in a cognitive impairment phenotype that is exacerbated by a long-term high-salt diet, and highlight the protective role of SIRT3 in this process.
Collapse
Affiliation(s)
- Haixia Fan
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
- First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Minghao Yuan
- Chongqing Medical University, Chongqing, 400016, China
| | - Shenyuan Wang
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Xu Yang
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Liu Shu
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Yinshuang Pu
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Qian Zou
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Xiaogang Zhang
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Chuanling Wang
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Zhiyou Cai
- Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China.
| |
Collapse
|
26
|
Wu H, Diao H, Zhang F, Jiang W, Pan T, Bian Y. Organelle interplay in cardiovascular diseases: Mechanisms, pathogenesis, and therapeutic perspectives. Biomed Pharmacother 2025; 185:117978. [PMID: 40073746 DOI: 10.1016/j.biopha.2025.117978] [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: 10/31/2024] [Revised: 02/16/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of rising morbidity and mortality among humans worldwide; however, our approach to the pathogenesis, exploration, and management of CVDs still remains limited. As the heart consists of cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, and several types of cells, different types of dysfunction in the interplay between organelles play an important damaging role, resulting in cardiac pathologies. The interplay between cellular organelles is intricate and vital for maintaining cellular homeostasis, as highlighted by multiple diseases linked to the dysfunction of both organelles. Many studies have revealed the potential mechanisms by which organelles communicate with each other and regulate the pathological processes of CVDs together. However, gaps remain in fully understanding the complexity of these interactions and translating these insights into therapeutic approaches. In this review, we summarized how the interplay between cellular organelles in the cardiomyocytes alters in various heart diseases. We find underexplored areas, such as the crucial signaling pathways governing organelle communication, and discuss their implications for disease future progression. Furthermore, we evaluate emerging potential medicines aimed at restoring organelle interactions. Finally, we propose future directions for researching to advance the development of novel medicines and therapies, addressing current gaps and providing a theoretical basis for improved clinical outcomes in CVDs.
Collapse
Affiliation(s)
- Han Wu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hongtao Diao
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Feng Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Weitao Jiang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tengfei Pan
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yu Bian
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
27
|
Xu L, Zhang T, Zhu B, Tao H, Liu Y, Liu X, Zhang Y, Meng X. Mitochondrial quality control disorder in neurodegenerative disorders: Potential and advantages of traditional Chinese medicines. J Pharm Anal 2025; 15:101146. [PMID: 40291018 PMCID: PMC12032916 DOI: 10.1016/j.jpha.2024.101146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 04/30/2025] Open
Abstract
Neurodegenerative disorders (NDDs) are prevalent chronic conditions characterized by progressive synaptic loss and pathological protein alterations. Increasing evidence suggested that mitochondrial quality control (MQC) serves as the key cellular process responsible for clearing misfolded proteins and impaired mitochondria. Herein, we provided a comprehensive analysis of the mechanisms through which MQC mediates the onset and progression of NDDs, emphasizing mitochondrial dynamic stability, the clearance of damaged mitochondria, and the generation of new mitochondria. In addition, traditional Chinese medicines (TCMs) and their active monomers targeting MQC in NDD treatment have been demonstrated. Consequently, we compiled the TCMs that show great potential in the treatment of NDDs by targeting MQC, aiming to offer novel insights and a scientific foundation for the use of MQC stabilizers in NDD prevention and treatment.
Collapse
Affiliation(s)
- Lei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Baojie Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Honglin Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yue Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianfeng Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, Sichuan, 620032, China
| |
Collapse
|
28
|
Sancak B, İnönü D, Alp G, Sağlam FT, Aryan L, Şık S, Akat F, Tuncay E. Cardioprotective role of SIRT1 activation on mitochondrial function in insulin-resistant H9c2 cells. BMC Cardiovasc Disord 2025; 25:232. [PMID: 40155821 PMCID: PMC11954332 DOI: 10.1186/s12872-024-04397-7] [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: 05/06/2024] [Accepted: 12/04/2024] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Insulin-resistance in cardiomyocytes is often associated with metabolic disorders like obesity, and type2 diabetes. Studies demonstrated that sirtuin1 (SIRT1) plays a protective role in cells resistant to insulin by enhancing insulin sensitivity and improving glucose metabolism. Based on these protective functions observed in SIRT1, this study aims to investigate the roles of SIRT1 in palmitate (PA)-induced insulin-resistant H9C2 cells. METHODS Insulin-resistance was induced in H9c2 cells via incubation with palmitic acid (50µM;24 h). Control and Insulin-resistant cells were incubated with SIRT1 inhibitor (EX527;10µM) and SIRT1 activator (SRT1720;2µM) for 24 h, respectively. Mitochondrial membrane potential (MMP), reactive oxygen/nitrogen species (ROS/RNS), total ATP production, intracellular free zinc and calcium levels ([Ca2+]i and [Zn2+]i) were monitored with fluorescence techniques. Protein levels were determined by using western-blot analysis. RESULTS K-acetylation level was increased in PA-induced Insulin-resistant cells and SIRT1 inhibited control cells. ROS/RNS production, [Ca2+]i, and [Zn2+]i levels were elevated, MMP was depolarized and ATP production was decreased in PA and EX527 treated cells compared to control cells. Mfn1 and Fis1 levels were remained unchanged, however Mfn2 protein level was elevated in cells treated with PA and SIRT1 inhibitor. Nevertheless, anti- and pro-apoptotic protein level was reduced and augmented respectively in insulin-resistant and SIRT1 inhibited cells. Activation of SIRT1 in PA-treated cells restored mitochondrial function and intracellular ionic homeostasis, reduced K-acetylation, and mitigated apoptosis. CONCLUSION Therefore, it can be proposed that the activation of SIRT1, acting as a novel regulator, may offer direct cardioprotection by restoring mitochondrial function in the insulin-resistant heart.
Collapse
Affiliation(s)
- Buğrahan Sancak
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Deniz İnönü
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Gülsüm Alp
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Faruk Tuna Sağlam
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Leila Aryan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye
- Institute of Health Sciences, Ankara University, Ankara, Türkiye
- Stem Cell Institute, Ankara University, Ankara, Türkiye
| | - Suatnur Şık
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye
- Department of Biochemistry, Faculty of Medicine, Hatay Mustafa Kemal University, Hatay, Türkiye
| | - Fırat Akat
- Department of Physiology, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Erkan Tuncay
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye.
- Institute of Health Sciences, Ankara University, Ankara, Türkiye.
- Department of Neuroscience, Faculty of Medicine, Ankara University, Ankara, Türkiye.
| |
Collapse
|
29
|
Wu B, Woo JS, Hasiakos S, Pan C, Cokus S, Benincá C, Stiles L, Sun Z, Pellegrini M, Shirihai OS, Lusis AJ, Srikanth S, Gwack Y. Mitochondrial reactive oxygen species regulate acetyl-CoA flux between cytokine production and fatty acid synthesis in effector T cells. Cell Rep 2025; 44:115430. [PMID: 40088449 PMCID: PMC12007815 DOI: 10.1016/j.celrep.2025.115430] [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: 08/08/2024] [Revised: 01/13/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Genetic and environmental factors shape an individual's susceptibility to autoimmunity. To identify genetic variations regulating effector T cell functions, we used a forward genetics screen of inbred mouse strains and uncovered genomic loci linked to cytokine expression. Among the candidate genes, we characterized a mitochondrial inner membrane protein, TMEM11, as an important determinant of Th1 responses. Loss of TMEM11 selectively impairs Th1 cell functions, reducing autoimmune symptoms in mice. Mechanistically, Tmem11-/- Th1 cells exhibit altered cristae architecture, impaired respiration, and increased mitochondrial reactive oxygen species (mtROS) production. Elevated mtROS hindered histone acetylation while promoting neutral lipid accumulation. Further experiments using genetic, biochemical, and pharmacological tools revealed that mtROS regulate acetyl-CoA flux between histone acetylation and fatty acid synthesis. Our findings highlight the role of mitochondrial cristae integrity in directing metabolic pathways that influence chromatin modifications and lipid biosynthesis in Th1 cells, providing new insights into immune cell metabolism.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Spyridon Hasiakos
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shawn Cokus
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cristiane Benincá
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zuoming Sun
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Cellular Integrative Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aldon J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
30
|
Xiu M, Li B, He L, Shi Y, Zhang Y, Zhou S, Liu Y, Wang N, He J. Caffeic Acid Protects Against Ulcerative Colitis via Inhibiting Mitochondrial Apoptosis and Immune Overactivation in Drosophila. Drug Des Devel Ther 2025; 19:2157-2172. [PMID: 40145123 PMCID: PMC11938933 DOI: 10.2147/dddt.s499284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Background Ulcerative colitis (UC) is a chronic intestinal inflammation that is prone to relapse and is difficult to fully recover; therefore, there is a need for safer alternative treatments. Caffeic acid (CA) is a natural polyphenolic compound that has antioxidant and anti-inflammatory properties. However, the beneficial effects and mechanisms of action of CA in UC remain unclear. Purpose This study evaluated the protective effect of CA against dextran sulfate sodium (DSS)-induced intestinal injury in Drosophila melanogaster model. Results Oral administration of CA significantly reduced body damage in UC flies, improved their survival rate, restored damaged digestion, and improved locomotion. CA supplementation significantly alleviated intestinal damage in UC flies by restoring excretion balance, repairing intestinal atrophy, improving acid-base balance imbalance, inhibiting intestinal structural destruction, inhibiting intestinal epithelial cell death and intestinal stem cell (ISC) excessive proliferation, and reducing the number of harmful bacteria. Mechanistic studies found that CA significantly reduced the expression of Toll and Imd pathway genes (including Myd88, Dif, PGRP-LC, Imd, Rel, and Dpt), reduced ROS levels and the expression of apoptosis-related genes (Debcl, Cyt-c-p, DrlCE, Dronc, and Dark), and increased ATP and MFN2 levels. Conclusion CA alleviated intestinal damage mainly by inhibiting the Toll and Imd signaling pathways and inhibiting apoptosis mediated by mitochondrial damage. These findings suggest that CA holds promise as a potential therapeutic for UC treatment.
Collapse
Affiliation(s)
- Minghui Xiu
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou City, Gansu Province, People’s Republic of China
| | - Botong Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
| | - Li He
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
| | - Yan Shi
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
| | - Yongxuan Zhang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
| | - Shihong Zhou
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
| | - Yongqi Liu
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou City, Gansu Province, People’s Republic of China
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
| | - Ningbo Wang
- Tibetan Medical College, Qinghai University, Xining City, Qinghai Province, People’s Republic of China
| | - Jianzheng He
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Lanzhou City, Gansu Province, People’s Republic of China
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
- Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou City, Gansu Province, People’s Republic of China
| |
Collapse
|
31
|
Barsa C, Perrin J, David C, Mourier A, Rojo M. A cellular assay to determine the fusion capacity of MFN2 variants linked to Charcot-Marie-Tooth disease of type 2 A. Sci Rep 2025; 15:9971. [PMID: 40121276 PMCID: PMC11929822 DOI: 10.1038/s41598-025-93702-1] [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: 10/11/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Charcot-Marie-Tooth Disease (CMT) is an inherited peripheral neuropathy with two main forms: demyelinating CMT1 and axonal CMT2. The most frequent subtype of CMT2 (CMT2A) is linked to mutations of MFN2, encoding a ubiquitously expressed GTP-binding protein anchored to the mitochondrial outer membrane and essential for mitochondrial fusion. The use of Next-Generation Sequencing has led to the identification of increasing numbers of MFN2 variants, yet many of them remain of unknown significance, depriving patients of a clear diagnosis. In this work, we establish a cellular assay allowing to assess the impact of 12 known MFN2 variants linked to CMT2A on mitochondrial fusion. The functional analysis revealed that out of the 12 selected MFN2 mutations, only six exhibited reduced fusion activity. The classification of MFN2 variants according to the results of the functional assay revealed a correlation between the fusion capacity, the age at onset of CMT2A and computational variant effect predictions relying on the analysis of the protein sequence. The functional assay and the results obtained will assist and improve the classification of novel MFN2 variants identified in patients.
Collapse
Affiliation(s)
- Chloe Barsa
- CNRS, IBGC, UMR 5095, Institut de Biochimie et Génétique Cellulaires (IBGC), Université de Bordeaux, 33000, Bordeaux, France
| | - Julian Perrin
- CNRS, IBGC, UMR 5095, Institut de Biochimie et Génétique Cellulaires (IBGC), Université de Bordeaux, 33000, Bordeaux, France
| | - Claudine David
- CNRS, IBGC, UMR 5095, Institut de Biochimie et Génétique Cellulaires (IBGC), Université de Bordeaux, 33000, Bordeaux, France
| | - Arnaud Mourier
- CNRS, IBGC, UMR 5095, Institut de Biochimie et Génétique Cellulaires (IBGC), Université de Bordeaux, 33000, Bordeaux, France
| | - Manuel Rojo
- CNRS, IBGC, UMR 5095, Institut de Biochimie et Génétique Cellulaires (IBGC), Université de Bordeaux, 33000, Bordeaux, France.
| |
Collapse
|
32
|
Zanfardino P, Amati A, Perrone M, Petruzzella V. The Balance of MFN2 and OPA1 in Mitochondrial Dynamics, Cellular Homeostasis, and Disease. Biomolecules 2025; 15:433. [PMID: 40149969 PMCID: PMC11940761 DOI: 10.3390/biom15030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondrial dynamics, governed by fusion and fission, are crucial for maintaining cellular homeostasis, energy production, and stress adaptation. MFN2 and OPA1, key regulators of mitochondrial fusion, play essential roles beyond their structural functions, influencing bioenergetics, intracellular signaling, and quality control mechanisms such as mitophagy. Disruptions in these processes, often caused by MFN2 or OPA1 mutations, are linked to neurodegenerative diseases like Charcot-Marie-Tooth disease type 2A (CMT2A) and autosomal dominant optic atrophy (ADOA). This review explores the molecular mechanisms underlying mitochondrial fusion, the impact of MFN2 and OPA1 dysfunction on oxidative phosphorylation and autophagy, and their role in disease progression. Additionally, we discuss the divergent cellular responses to MFN2 and OPA1 mutations, particularly in terms of proliferation, senescence, and metabolic signaling. Finally, we highlight emerging therapeutic strategies to restore mitochondrial integrity, including mTOR modulation and autophagy-targeted approaches, with potential implications for neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Vittoria Petruzzella
- Department of Translational Biomedicine and Neurosciences (DiBraiN), University of Bari Aldo Moro, Piazza Giulio Cesare, 70124 Bari, Italy; (P.Z.); (A.A.); (M.P.)
| |
Collapse
|
33
|
Carbajal C, Rodriguez M, Owens F, Stone N, Veeragoni D, Fan RZ, Tieu K, El-Hage N. Therapeutic Efficacy of Small Extracellular Vesicles Loaded with ROCK Inhibitor in Parkinson's Disease. Pharmaceutics 2025; 17:365. [PMID: 40143028 PMCID: PMC11944340 DOI: 10.3390/pharmaceutics17030365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Parkinson's disease (PD) is a rapidly growing neurological disorder in the developed world, affecting millions over the age of 60. The decline in motor functions occurs due to a progressive loss of midbrain dopaminergic neurons, resulting in lowered dopamine levels and impaired muscle function. Studies show defective mitochondrial autophagy (or "mitophagy") links to PD. Rho-associated coiled-coil containing protein kinases (ROCK) 1 and ROCK2 are serine/threonine kinases, and their inhibition can enhance neuroprotection in PD by promoting mitophagy. Methods: We examine the effects of ROCK inhibitor SR3677, delivered via macrophage-derived small extracellular vesicles (sEVs) to Parkin Q311X(A) PD mouse models. sEVs with SR3677, administered intranasally, increased mitophagy gene expression, reduced inflammatory factors, and elevated dopamine levels in brain tissues. Results: ROCK2 expression decreased, showing the drug's inhibitory effect. sEV-SR3677 treatment was more effective than treatment with the drug alone, although sham EVs showed lower effects. This suggests that EV-SR3677 not only activates mitochondrial processes but also promotes the degradation of damaged mitochondria through autophagy. Mitochondrial functional assays and oxygen consumption in ex vivo glial cultures revealed that sEV-SR3677 significantly improved mitochondrial respiration compared to that in untreated or SR3677-only treated cells. Conclusion: We demonstrated the efficacy of ROCK2 inhibition on mitochondrial function via sEV-SR3677 in the PD mouse model, necessitating further studies to explore design challenges and mechanisms of sEV-SR3677 as mitochondria-targeted therapy for PD.
Collapse
Affiliation(s)
- Candy Carbajal
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (C.C.); (M.R.); (F.O.); (N.S.); (D.V.)
| | - Myosotys Rodriguez
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (C.C.); (M.R.); (F.O.); (N.S.); (D.V.)
| | - Florida Owens
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (C.C.); (M.R.); (F.O.); (N.S.); (D.V.)
| | - Nicole Stone
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (C.C.); (M.R.); (F.O.); (N.S.); (D.V.)
| | - Dileepkumar Veeragoni
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (C.C.); (M.R.); (F.O.); (N.S.); (D.V.)
| | - Rebecca Z. Fan
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA; (R.Z.F.); (K.T.)
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA; (R.Z.F.); (K.T.)
| | - Nazira El-Hage
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (C.C.); (M.R.); (F.O.); (N.S.); (D.V.)
| |
Collapse
|
34
|
Cao G, Li C, Zhang J, Deng L, Li R, Xu C. Procyanidin B2 alleviates damage to mouse testicular tissue after freezing by inhibiting oxidative stress and apoptosis. Cryobiology 2025; 118:105196. [PMID: 39756776 DOI: 10.1016/j.cryobiol.2025.105196] [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: 10/21/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/07/2025]
Abstract
For infertile patients who are unable to obtain sperm or prepubertal boys who require radiotherapy, testicular tissue freezing can be used for later transplantation and is a potentially effective method of preserving male fertility. Oxidative stress caused by the freezing process is an important cause of tissue damage. Procyanidin B2 (PCB2) is a polyphenolic natural compound widely distributed in plants that is known for its anti-inflammatory, anticancer, and neuroprotective properties, and its antioxidant capabilities are particularly noteworthy. Research has indicated that PCB2 exerts a protective effect on the reproductive system. However, its specific role in mitigating testicular tissue cryoinjury and the underlying mechanisms remain unclear. This study investigated whether adding PCB2 to a cryoprotective solution of mouse testicular tissue can alleviate the cryoinjury of testicular tissue and its possible mechanism. Our findings revealed that frozen mouse testicular tissue presented decreased cell viability and induced oxidative stress. Conversely, PCB2 effectively mitigated these adverse effects. In addition, PCB2 improved the tubular structural disorganization caused by freezing and increased the expression of proteins related to the junction function of Sertoli cells. Further experiments indicated that PCB2 activated the nuclear respiratory factor 2 (Nrf2)/heme oxygenase 1 (HO-1) antioxidant signaling pathway, increased the activity of downstream antioxidant enzymes, and improved mitochondrial kinetic homeostasis. Additionally, PCB2 ameliorated apoptosis while increasing the expression levels of key enzymes involved in testosterone synthesis. In summary, these results suggest that PCB2 attenuates damage to mouse testicular tissue during freezing by inhibiting oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Guorui Cao
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Chunyuan Li
- Reproductive Medical Center of Nanning Second People's Hospital, Nanning, Guangxi, 530031, China
| | - Jian Zhang
- Reproductive Medical Center of Nanning Second People's Hospital, Nanning, Guangxi, 530031, China
| | - Liwen Deng
- Reproductive Medical Center of Nanning Second People's Hospital, Nanning, Guangxi, 530031, China
| | - Rong Li
- Reproductive Medical Center of Nanning Second People's Hospital, Nanning, Guangxi, 530031, China
| | - Changlong Xu
- Reproductive Medical Center of Nanning Second People's Hospital, Nanning, Guangxi, 530031, China.
| |
Collapse
|
35
|
Chin MY, Joy DA, Samaddar M, Rana A, Chow J, Miyamoto T, Calvert M. Novel high-content and open-source image analysis tools for profiling mitochondrial morphology in neurological cell models. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100208. [PMID: 39778657 DOI: 10.1016/j.slasd.2025.100208] [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: 10/09/2024] [Revised: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Mitochondria undergo dynamic morphological changes depending on cellular cues, stress, genetic factors, or disease. The structural complexity and disease-relevance of mitochondria have stimulated efforts to generate image analysis tools for describing mitochondrial morphology for therapeutic development. Using high-content analysis, we measured multiple morphological parameters and employed unbiased feature clustering to identify the most robust pair of texture metrics that described mitochondrial state. Here, we introduce a novel image analysis pipeline to enable rapid and accurate profiling of mitochondrial morphology in various cell types and pharmacological perturbations. We applied a high-content adapted implementation of our tool, MitoProfilerHC, to quantify mitochondrial morphology changes in i) a mammalian cell dose response study and ii) compartment-specific drug effects in primary neurons. Next, we expanded the usability of our pipeline by using napari, a Python-powered image analysis tool, to build an open-source version of MitoProfiler and validated its performance and applicability. In conclusion, we introduce MitoProfiler as both a high-content-based and an open-source method to accurately quantify mitochondrial morphology in cells, which we anticipate to greatly facilitate mechanistic discoveries in mitochondrial biology and disease.
Collapse
Affiliation(s)
- Marcus Y Chin
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA.
| | - David A Joy
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA
| | | | - Anil Rana
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA
| | - Johann Chow
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA
| | | | | |
Collapse
|
36
|
Inferrera F, Marino Y, Genovese T, Cuzzocrea S, Fusco R, Di Paola R. Mitochondrial quality control: Biochemical mechanism of cardiovascular disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119906. [PMID: 39837389 DOI: 10.1016/j.bbamcr.2025.119906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/30/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Mitochondria play a key role in the regulation of energy homeostasis and ATP production in cardiac cells. Mitochondrial dysfunction can trigger several pathological events that contribute to the development and progression of cardiovascular diseases. These mechanisms include the induction of oxidative stress, dysregulation of intracellular calcium cycling, activation of the apoptotic pathway, and alteration of lipid metabolism. This review focuses on the role of mitochondria in intracellular signaling associated with cardiovascular diseases, emphasizing the contributions of reactive oxygen species production and mitochondrial dynamics. Indeed, mitochondrial dysfunction has been implicated in every aspect of cardiovascular disease and is currently being evaluated as a potential target for therapeutic interventions. To treat cardiovascular diseases and improve overall heart health, it is important to better understand these biochemical systems. These findings allow the achievement of targeted therapies and preventive measures. Therefore, this review investigates different studies that demonstrate how changes in mitochondrial dynamics like fusion, fission, and mitophagy contribute to the development or worsening of disorders related to heart diseases by summarizing current research on their role.
Collapse
Affiliation(s)
- Francesca Inferrera
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Ylenia Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; Link Campus University, Via del Casale di San Pio V, 4400165 Rome, Italy.
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy.
| |
Collapse
|
37
|
Yamada T, Ikeda A, Murata D, Wang H, Zhang C, Khare P, Adachi Y, Ito F, Quirós PM, Blackshaw S, López-Otín C, Langer T, Chan DC, Le A, Dawson VL, Dawson TM, Iijima M, Sesaki H. Dual regulation of mitochondrial fusion by Parkin-PINK1 and OMA1. Nature 2025; 639:776-783. [PMID: 39972141 DOI: 10.1038/s41586-025-08590-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 01/03/2025] [Indexed: 02/21/2025]
Abstract
Mitochondrial stress pathways protect mitochondrial health from cellular insults1-8. However, their role under physiological conditions is largely unknown. Here, using 18 single, double and triple whole-body and tissue-specific knockout and mutant mice, along with systematic mitochondrial morphology analysis, untargeted metabolomics and RNA sequencing, we discovered that the synergy between two stress-responsive systems-the ubiquitin E3 ligase Parkin and the metalloprotease OMA1-safeguards mitochondrial structure and genome by mitochondrial fusion, mediated by the outer membrane GTPase MFN1 and the inner membrane GTPase OPA1. Whereas the individual loss of Parkin or OMA1 does not affect mitochondrial integrity, their combined loss results in small body size, low locomotor activity, premature death, mitochondrial abnormalities and innate immune responses. Thus, our data show that Parkin and OMA1 maintain a dual regulatory mechanism that controls mitochondrial fusion at the two membranes, even in the absence of extrinsic stress.
Collapse
Affiliation(s)
- Tatsuya Yamada
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Arisa Ikeda
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daisuke Murata
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hu Wang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cissy Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Gigantest, Inc., Baltimore, MD, USA
| | - Pratik Khare
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Gigantest, Inc., Baltimore, MD, USA
| | - Yoshihiro Adachi
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fumiya Ito
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pedro M Quirós
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
- Centre de Recherche des Cordeliers, Inserm U1138, Université Paris Cité, Sorbonne Université, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Anne Le
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Gigantest, Inc., Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA.
| |
Collapse
|
38
|
Kang L, Bao S, Li P, Zhang G, Zhu X, Ji M, Guan H. METTL14-mediated depression of NEIL1 aggravates oxidative damage and mitochondrial dysfunction of lens epithelial cells through regulating KEAP1/NRF2 pathways. Cell Signal 2025; 127:111623. [PMID: 39855533 DOI: 10.1016/j.cellsig.2025.111623] [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: 12/13/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Abnormal base excision repair (BER) pathway and N6-methyladenosine (m6A) of RNA have been proved to be significantly related to age-related cataract (ARC) pathogenesis. However, the relationship between the Nei Endonuclease VIII-Like1 (NEIL1) gene (a representative DNA glycosylase of BER pathway) and its m6A modification remains unclear. Here, we showed that the expression of NEIL1 was decreased in the ARC anterior lens capsules and H2O2-stimulated SRA01/04 cells. Our findings demonstrated that ectopic expression of NEIL1 alleviated DNA oxidative damage, apoptosis and mitochondrial dysfunction through disturbing KEAP1/NRF2 interaction. Furthermore, silencing NEIL1 aggravated H2O2-induced lens opacity, whereas ML334 could mitigate lens cloudy ex vitro in rat lenses. Besides, intravitreal injection of AAV2-NEIL1 alleviated lens opacity in Emory mice in vivo. Mechanistically, the N(6)-Methyladenosine (m6A) methyltransferase-like 14 (METTL14) was identified as a factor in promoting m6A modification of NEIL1, which resulted in the recruitment of YTHDF2 to recognize and impair NEIL1 RNA stability. Collectively, these findings highlight the critical role of the m6A modification in NEIL1 on regulating oxidative stress and mitochondrial homeostasis through KEAP1/NRF2 pathways, providing a new way to explore the pathogenesis of ARC.
Collapse
Affiliation(s)
- Lihua Kang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Sijie Bao
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Pengfei Li
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xi Zhu
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Min Ji
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
39
|
Hemel IMGM, Steen C, Denil SLIJ, Ertaylan G, Kutmon M, Adriaens M, Gerards M. The unusual suspect: A novel role for intermediate filament proteins in mitochondrial morphology. Mitochondrion 2025; 81:102008. [PMID: 39909388 DOI: 10.1016/j.mito.2025.102008] [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: 12/20/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Mitochondrial dynamics is crucial for cellular homeostasis. However, not all proteins involved are known. Using a protein-protein interaction (PPI) approach, we identified ITPRIPL2 for involvement in mitochondrial dynamics. ITPRIPL2 co-localizes with intermediate filament protein vimentin, supported by protein simulations. ITPRIPL2 knockdown reveals mitochondrial elongation, disrupts vimentin processing, intermediate filament formation, and alters vimentin-related pathways. Interestingly, vimentin knockdown also leads to mitochondrial elongation. These findings highlight ITPRIPL2 as vimentin-associated protein essential for intermediate filament structure and suggest a role for intermediate filaments in mitochondrial morphology. Our study demonstrates that PPI analysis is a powerful approach for identifying novel mitochondrial dynamics proteins.
Collapse
Affiliation(s)
- Irene M G M Hemel
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Carlijn Steen
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Simon L I J Denil
- Flemish Institute for Technological Research (VITO) 2400 Mol, Belgium
| | - Gökhan Ertaylan
- Flemish Institute for Technological Research (VITO) 2400 Mol, Belgium
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Michiel Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands
| | - Mike Gerards
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht 6229 EN the Netherlands.
| |
Collapse
|
40
|
Li Z, Ran Q, Qu C, Hu S, Cui S, Zhou Y, Shen B, Yang B. Sigma-1 receptor activation attenuates DOX-induced cardiotoxicity by alleviating endoplasmic reticulum stress and mitochondrial calcium overload via PERK and IP3R-VDAC1-MCU signaling pathways. Biol Direct 2025; 20:23. [PMID: 40001213 PMCID: PMC11853590 DOI: 10.1186/s13062-025-00617-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Doxorubicin (DOX) is an anthracycline with potent antitumor properties and rare yet serious cardiotoxic side effects that limit its clinical application. The sigma-1 receptor is a stress-triggered chaperone often dysregulated in diseases and has known cardioprotective effects. Although its anti-oxidative stress and anti-apoptotic effects have been demonstrated, its effectiveness in DOX-induced cardiotoxicity has never been explored. This study investigated the potential role of the activated sigma-1 receptor in a DOX-induced murine cardiotoxicity model to elucidate the receptor's mechanism of action. METHODS We established the model in C57BL/6 mice by daily intraperitoneal injections of fluvoxamine (Flv) for 4 consecutive weeks to activate the receptor and by weekly intraperitoneal injections of DOX at 5 mg/kg for 3 weeks. We performed in vitro experiments using cardiomyocytes of neonatal Sprague-Dawley rats to verify the protective effect of the sigma-1 receptor. RESULTS We found that sigma-1 expression in the heart decreased in the DOX-treated mice, and activating the receptor with Flv improved cardiac function. Moreover, Flv pretreatment inhibited cardiomyocyte apoptosis and endoplasmic reticulum stress and increased the expression of the Bcl2 apoptosis regulator (Bcl2), effectively alleviating the pathophysiological manifestations in mice. In addition, activating the receptor exerted cardioprotective effects by modulating endoplasmic reticulum stress through the PRKR-like endoplasmic reticulum kinase (PERK) signaling pathway. It also reduced mitochondrial and endoplasmic reticulum contact and alleviated mitochondrial calcium overload through the IP3R-VDAC1-MCU signaling pathway. CONCLUSION In conclusion, our study emphasizes the therapeutic potential of activating sigma-1 receptors against DOX-induced cardiotoxicity, suggesting sigma-1 receptors as potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Qian Ran
- Department of Cardiology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Shan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Shengyu Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - You Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Bo Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| |
Collapse
|
41
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [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: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
42
|
Hemel IMGM, Knoops K, López-Iglesias C, Gerards M. The Curse of the Red Pearl: A Fibroblast-Specific Pearl-Necklace Mitochondrial Phenotype Caused by Phototoxicity. Biomolecules 2025; 15:304. [PMID: 40001607 PMCID: PMC11853634 DOI: 10.3390/biom15020304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
The dynamic nature of mitochondria makes live cell imaging an important tool in mitochondrial research. Although imaging using fluorescent probes is the golden standard in studying mitochondrial morphology, these probes might introduce aspecific features. In this study, live cell fluorescent imaging was applied to investigate a pearl-necklace-shaped mitochondrial phenotype that arises when mitochondrial fission is restricted. In this fibroblast-specific pearl-necklace phenotype, constricted and expanded mitochondrial regions alternate. Imaging studies revealed that the formation time of this pearl-necklace phenotype differs between laser scanning confocal, widefield and spinning disk confocal microscopy. We found that the phenotype formation correlates with the excitation of the fluorescent probe and is the result of phototoxicity. Interestingly, the phenotype only arises in cells stained with red mitochondrial dyes. Serial section electron tomography of the pearl-necklace mitochondria revealed that the mitochondrial membranes remained intact, while the cristae structure was altered. Furthermore, filaments and ER were present at the constricted sites. This study illustrates the importance of considering experimental conditions for live cell imaging to prevent imaging artifacts that can have a major impact on the obtained results.
Collapse
Affiliation(s)
- Irene M. G. M. Hemel
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, 6229 EN Maastricht, The Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab, Maastricht University, 6229 ER Maastricht, The Netherlands (C.L.-I.)
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Maastricht University, 6229 ER Maastricht, The Netherlands (C.L.-I.)
| | - Mike Gerards
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, 6229 EN Maastricht, The Netherlands
| |
Collapse
|
43
|
Pannoni KE, Fischer QS, Tarannum R, Cawley ML, Alsalman MM, Acosta N, Ezigbo C, Gil DV, Campbell LA, Farris S. MCU expression in hippocampal CA2 neurons modulates dendritic mitochondrial morphology and synaptic plasticity. Sci Rep 2025; 15:4540. [PMID: 39915602 PMCID: PMC11802895 DOI: 10.1038/s41598-025-85958-4] [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: 11/01/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Neuronal mitochondria are diverse across cell types and subcellular compartments in order to meet unique energy demands. While mitochondria are essential for synaptic transmission and synaptic plasticity, the mechanisms regulating mitochondria to support normal synapse function are incompletely understood. The mitochondrial calcium uniporter (MCU) is proposed to couple neuronal activity to mitochondrial ATP production, which would allow neurons to rapidly adapt to changing energy demands. MCU is uniquely enriched in hippocampal CA2 distal dendrites compared to proximal dendrites, however, the functional significance of this layer-specific enrichment is not clear. Synapses onto CA2 distal dendrites readily express plasticity, unlike the plasticity-resistant synapses onto CA2 proximal dendrites, but the mechanisms underlying these different plasticity profiles are unknown. Using a CA2-specific MCU knockout (cKO) mouse, we found that MCU deletion impairs plasticity at distal dendrite synapses. However, mitochondria were more fragmented and spine head area was diminished throughout the dendritic layers of MCU cKO mice versus control mice. Fragmented mitochondria might have functional changes, such as altered ATP production, that could explain the structural and functional deficits at cKO synapses. Differences in MCU expression across cell types and circuits might be a general mechanism to tune mitochondrial function to meet distinct synaptic demands.
Collapse
Affiliation(s)
- Katy E Pannoni
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Quentin S Fischer
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Renesa Tarannum
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Mikel L Cawley
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Mayd M Alsalman
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Nicole Acosta
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Chisom Ezigbo
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Daniela V Gil
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Logan A Campbell
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Shannon Farris
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.
| |
Collapse
|
44
|
Tábara LC, Segawa M, Prudent J. Molecular mechanisms of mitochondrial dynamics. Nat Rev Mol Cell Biol 2025; 26:123-146. [PMID: 39420231 DOI: 10.1038/s41580-024-00785-1] [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] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Mitochondria not only synthesize energy required for cellular functions but are also involved in numerous cellular pathways including apoptosis, calcium homoeostasis, inflammation and immunity. Mitochondria are dynamic organelles that undergo cycles of fission and fusion, and these transitions between fragmented and hyperfused networks ensure mitochondrial function, enabling adaptations to metabolic changes or cellular stress. Defects in mitochondrial morphology have been associated with numerous diseases, highlighting the importance of elucidating the molecular mechanisms regulating mitochondrial morphology. Here, we discuss recent structural insights into the assembly and mechanism of action of the core mitochondrial dynamics proteins, such as the dynamin-related protein 1 (DRP1) that controls division, and the mitofusins (MFN1 and MFN2) and optic atrophy 1 (OPA1) driving membrane fusion. Furthermore, we provide an updated view of the complex interplay between different proteins, lipids and organelles during the processes of mitochondrial membrane fusion and fission. Overall, we aim to present a valuable framework reflecting current perspectives on how mitochondrial membrane remodelling is regulated.
Collapse
Affiliation(s)
- Luis-Carlos Tábara
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Mayuko Segawa
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
45
|
Crabtree A, Le H, Harris C, Oliver A, Barillas A, Moore J, Nguyen DN, Rabago IM, Rodriguez B, Stephens DC, Beasley HK, Garza‐Lopez E, Neikirk K, Mungai M, Vang L, Vue Z, Vue N, Marshall AG, Turner K, Shao J, Murray S, Gaddy JA, Wanjalla C, Davis J, Damo S, Hinton AO. Optimizing In Situ Proximity Ligation Assays for Mitochondria, ER, or MERC Markers in Skeletal Muscle Tissue and Cells. Curr Protoc 2025; 5:e70043. [PMID: 39906960 PMCID: PMC11795341 DOI: 10.1002/cpz1.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Proximity ligation assays (PLAs) use specific antibodies to detect endogenous protein-protein interactions. PLAs are a highly useful biochemical technique that allow two proteins within proximity to be visualized with fluorescent probes amplified by PCR. While this technique has gained prominence, the use of a PLA in mouse skeletal muscle (SkM) is novel. In this article, we discuss how the PLA method can be used in SkM to study the protein-protein interactions within mitochondria-endoplasmic reticulum contact sites (MERCs). © 2025 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Proximity ligation assay for skeletal muscle tissue and myoblast for MERC proteins.
Collapse
Affiliation(s)
- Amber Crabtree
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
- These authors contributed equally to this work
| | - Han Le
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
- These authors contributed equally to this work
| | - Chanel Harris
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
- These authors contributed equally to this work
| | - Ashton Oliver
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
- These authors contributed equally to this work
| | - Andy Barillas
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Johnathan Moore
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Desiree Ngoc‐ha Nguyen
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Izabella Marie Rabago
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Benjamin Rodriguez
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Dominique C. Stephens
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
- Department of Life and Physical SciencesFisk UniversityNashvilleTennessee
| | - Heather K. Beasley
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
- These authors contributed equally to this work
| | | | - Kit Neikirk
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Margaret Mungai
- Department of Internal MedicineUniversity of IowaIowa CityIowa
| | - Larry Vang
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Zer Vue
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Neng Vue
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Andrea G. Marshall
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| | - Kyrin Turner
- Department of Life and Physical SciencesFisk UniversityNashvilleTennessee
| | - Jianqiang Shao
- Central Microscopy Research FacilityUniversity of IowaIowa CityIowa
| | - Sandra Murray
- Department of Cell Biology, College of MedicineUniversity of PittsburghPittsburghPennsylvania
| | - Jennifer A. Gaddy
- Division of Infectious DiseasesVanderbilt University School of MedicineNashvilleTennessee
- Tennessee Valley Healthcare SystemsUS Department of Veterans AffairsNashvilleTennessee
| | - Celestine Wanjalla
- Vanderbilt University Medical Center, Department of MedicineDivision of Infectious DiseaseNashvilleTennessee
| | - Jamaine Davis
- Department of Biochemistry and Cancer BiologyMeharry Medical CollegeNashvilleTennessee
| | - Steven Damo
- Department of Life and Physical SciencesFisk UniversityNashvilleTennessee
| | - Antentor O. Hinton
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennessee
| |
Collapse
|
46
|
Shi SM, Hu B, Chi ZC, Qu LY, Liu LY, He YY, Jia GL, Li JH, Kong I, Jin YX, Yu XF. Role of MFN2 in bovine embryonic development and the mitigation of ER stress. Anim Reprod Sci 2025; 273:107664. [PMID: 39675305 DOI: 10.1016/j.anireprosci.2024.107664] [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: 08/19/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
This study investigated the role of mitochondrial fusion protein-2 (MFN2) in bovine embryonic development and its relationship with endoplasmic reticulum (ER) stress, aiming to increase the efficiency of in vitro embryo culture. Western blot analysis revealed that MFN2 expression peaked at the 2-cell stage, decreased at the 4-cell stage, and gradually increased from the 6-8-cell stage to the blastocyst stage. Inhibiting MFN2 at the zygote stage reduced blastocyst formation and proliferation, and this damage was partially reversed by the ER stress protective agent TUDCA. MFN2 inhibition also led to the decreased formation of the inner cell mass (ICM) and reduced expression of the totipotent genes CDX2 and SOX2. Additionally, reactive oxygen species (ROS) levels increased following MFN2 inhibition but decreased after TUDCA treatment. The expression of antioxidative stress-related genes (SOD and CAT) was downregulated after MFN2 inhibition but upregulated following TUDCA treatment. Furthermore, MFN2 inhibition reduced ER fluorescence intensity and increased the expression of UPR signaling markers (GRP78, XBP1, CHOP, IRE1, and ATF6), indicating increased ER stress. TUDCA administration reversed these effects, restoring MFN2 levels and reducing apoptosis. In conclusion, MFN2 is essential for bovine embryonic development because it regulates ER stress and maintains cell function, with MFN2 deficiency leading to developmental disorders and cell damage. ER stress protectors such as TUDCA can effectively mitigate these negative effects, highlighting a potential strategy for improving in vitro embryo culture efficiency.
Collapse
Affiliation(s)
- Shu-Ming Shi
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China
| | - Bing Hu
- Animal Genome Editing Technology Innovation Center, Jilin Province, College of Animal Science, Jilin University, Changchun 130062, China
| | - Zhi-Chao Chi
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China
| | - Lin-Yi Qu
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China
| | - Li-Ying Liu
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China
| | - Yu-Yan He
- Animal Genome Editing Technology Innovation Center, Jilin Province, College of Animal Science, Jilin University, Changchun 130062, China
| | - Guan-Lin Jia
- Animal Genome Editing Technology Innovation Center, Jilin Province, College of Animal Science, Jilin University, Changchun 130062, China
| | - Jing-Hang Li
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China
| | - Ilkeun Kong
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China; Department of Animal Science, Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Yong-Xun Jin
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China.
| | - Xian-Feng Yu
- Jilin Provincial Key Laboratory of Animal Model, College of Animal Science, Jilin University, Changchun 130062, China.
| |
Collapse
|
47
|
K Soman S, Swain M, Dagda RK. BDNF-TrkB Signaling in Mitochondria: Implications for Neurodegenerative Diseases. Mol Neurobiol 2025; 62:1756-1769. [PMID: 39030441 PMCID: PMC11909598 DOI: 10.1007/s12035-024-04357-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a pivotal role in neuronal development, synaptic plasticity, and overall neuronal health by binding to its receptor, tyrosine receptor kinase B (TrkB). This review delves into the intricate mechanisms through which BDNF-TrkB signaling influences mitochondrial function and potentially influences pathology in neurodegenerative diseases. This review highlights the BDNF-TrkB signaling pathway which regulates mitochondrial bioenergetics, biogenesis, and dynamics, mitochondrial processes vital for synaptic transmission and plasticity. Furthermore, we explore how the BDNF-TrkB-PKA signaling in the cytosol and in mitochondria affects mitochondrial transport and distribution and mitochondrial content, which is crucial for supporting the energy demands of synapses. The dysregulation of this signaling pathway is linked to various neurodegenerative diseases, including Alzheimer's and Parkinson's disease, which are characterized by mitochondrial dysfunction and reduced BDNF expression. By examining seminal studies that have characterized this signaling pathway in health and disease, the present review underscores the potential of enhancing BDNF-TrkB signaling to mitigate mitochondrial dysfunction in neurodegenerative diseases, offering insights into therapeutic strategies to enhance neuronal resilience and function.
Collapse
Affiliation(s)
- Smijin K Soman
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Maryann Swain
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Ruben K Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA.
| |
Collapse
|
48
|
Ritenis EJ, Padilha CS, Cooke MB, Stathis CG, Philp A, Camera DM. The acute and chronic influence of exercise on mitochondrial dynamics in skeletal muscle. Am J Physiol Endocrinol Metab 2025; 328:E198-E209. [PMID: 39441237 DOI: 10.1152/ajpendo.00311.2024] [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: 08/13/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Exercise and nutritional modulation are potent stimuli for eliciting increases in mitochondrial mass and function. Collectively, these beneficial adaptations are increasingly recognized to coincide with improvements in skeletal muscle health. Mitochondrial dynamics of fission and fusion are increasingly implicated as having a central role in mediating aspects of key organelle adaptations that are seen with exercise. Exercise-induced mitochondrial adaptation dynamics that have been implicated are 1) increases to mitochondrial turnover, resulting from elevated rates of mitochondrial synthesis (biogenesis) and degradative (mitophagy) processes and 2) morphological changes to the three-dimensional (3-D) tubular network, known as the mitochondrial reticulum, that mitochondria form in skeletal muscle. Notably, mitochondrial fission has also been implicated in coordinating increases in mitophagy, following acute exercise. Furthermore, increased fusion following exercise training promotes increased connectivity of the mitochondrial reticulum and is associated with improved metabolism and mitochondrial function. However, the molecular basis and fashion in which exercise infers beneficial mitochondrial adaptations through mitochondrial dynamics remains to be fully elucidated. This review attempts to highlight recent developments investigating the effects of exercise on mitochondrial dynamics, while attempting to offer a perspective of the methodological refinements and potential variables, such as substrate/glycogen availability, which should be considered going forward.
Collapse
Affiliation(s)
- Elya J Ritenis
- Department of Health Sciences and Biostatistics, Swinburne University of Technology, Melbourne, Victoria, Australia
- Centre for Healthy Ageing, Centenary Institute of Cancer Medicine and Cell Biology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Camila S Padilha
- Centre for Healthy Ageing, Centenary Institute of Cancer Medicine and Cell Biology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Matthew B Cooke
- Sport, Performance, and Nutrition Research Group, School of Allied Health, Human Services and Sport, La Trobe University, Melbourne, Victoria, Australia
| | - Christos G Stathis
- College of Sport, Health and Engineering, Victoria University, Melbourne, Victoria, Australia
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Andrew Philp
- Centre for Healthy Ageing, Centenary Institute of Cancer Medicine and Cell Biology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Donny M Camera
- Department of Health Sciences and Biostatistics, Swinburne University of Technology, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Meng K, Jia H, Hou X, Zhu Z, Lu Y, Feng Y, Feng J, Xia Y, Tan R, Cui F, Yuan J. Mitochondrial Dysfunction in Neurodegenerative Diseases: Mechanisms and Corresponding Therapeutic Strategies. Biomedicines 2025; 13:327. [PMID: 40002740 PMCID: PMC11852430 DOI: 10.3390/biomedicines13020327] [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: 12/04/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Neurodegenerative disease (ND) refers to the progressive loss and morphological abnormalities of neurons in the central nervous system (CNS) or peripheral nervous system (PNS). Examples of neurodegenerative diseases include Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Recent studies have shown that mitochondria play a broad role in cell signaling, immune response, and metabolic regulation. For example, mitochondrial dysfunction is closely associated with the onset and progression of a variety of diseases, including ND, cardiovascular diseases, diabetes, and cancer. The dysfunction of energy metabolism, imbalance of mitochondrial dynamics, or abnormal mitophagy can lead to the imbalance of mitochondrial homeostasis, which can induce pathological reactions such as oxidative stress, apoptosis, and inflammation, damage the nervous system, and participate in the occurrence and development of degenerative nervous system diseases such as AD, PD, and ALS. In this paper, the latest research progress of this subject is detailed. The mechanisms of oxidative stress, mitochondrial homeostasis, and mitophagy-mediated ND are reviewed from the perspectives of β-amyloid (Aβ) accumulation, dopamine neuron damage, and superoxide dismutase 1 (SOD1) mutation. Based on the mechanism research, new ideas and methods for the treatment and prevention of ND are proposed.
Collapse
Affiliation(s)
- Kai Meng
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining 272067, China;
| | - Haocheng Jia
- College of Clinical Medicine, Jining Medical University, Jining 272067, China; (H.J.); (X.H.); (Z.Z.); (Y.L.); (Y.F.)
| | - Xiaoqing Hou
- College of Clinical Medicine, Jining Medical University, Jining 272067, China; (H.J.); (X.H.); (Z.Z.); (Y.L.); (Y.F.)
| | - Ziming Zhu
- College of Clinical Medicine, Jining Medical University, Jining 272067, China; (H.J.); (X.H.); (Z.Z.); (Y.L.); (Y.F.)
| | - Yuguang Lu
- College of Clinical Medicine, Jining Medical University, Jining 272067, China; (H.J.); (X.H.); (Z.Z.); (Y.L.); (Y.F.)
| | - Yingying Feng
- College of Clinical Medicine, Jining Medical University, Jining 272067, China; (H.J.); (X.H.); (Z.Z.); (Y.L.); (Y.F.)
| | - Jingwen Feng
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China;
| | - Yong Xia
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining 272067, China;
| | - Rubin Tan
- College of Basic Medical, Xuzhou Medical University, Xuzhou 221004, China;
| | - Fen Cui
- Educational Institute of Behavioral Medicine, Jining Medical University, Jining 272067, China
| | - Jinxiang Yuan
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining 272067, China;
| |
Collapse
|
50
|
Serano M, Perni S, Pierantozzi E, Laurino A, Sorrentino V, Rossi D. Intracellular Membrane Contact Sites in Skeletal Muscle Cells. MEMBRANES 2025; 15:29. [PMID: 39852269 PMCID: PMC11767089 DOI: 10.3390/membranes15010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
Intracellular organelles are common to eukaryotic cells and provide physical support for the assembly of specialized compartments. In skeletal muscle fibers, the largest intracellular organelle is the sarcoplasmic reticulum, a specialized form of the endoplasmic reticulum primarily devoted to Ca2+ storage and release for muscle contraction. Occupying about 10% of the total cell volume, the sarcoplasmic reticulum forms multiple membrane contact sites, some of which are unique to skeletal muscle. These contact sites primarily involve the plasma membrane; among these, specialized membrane contact sites between the transverse tubules and the terminal cisternae of the sarcoplasmic reticulum form triads. Triads are skeletal muscle-specific contact sites where Ca2+ channels and regulatory proteins assemble to form the so-called calcium release complex. Additionally, the sarcoplasmic reticulum contacts mitochondria to enable a more precise regulation of Ca2+ homeostasis and energy metabolism. The sarcoplasmic reticulum and the plasma membrane also undergo dynamic remodeling to allow Ca2+ entry from the extracellular space and replenish the stores. This process involves the formation of dynamic membrane contact sites called Ca2+ Entry Units. This review explores the key processes in biogenesis and assembly of intracellular membrane contact sites as well as the membrane remodeling that occurs in response to muscle fatigue.
Collapse
Affiliation(s)
- Matteo Serano
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Stefano Perni
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Annunziatina Laurino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|