1
|
Mendelsohn DH, Walter N, Cheung WH, Wong RMY, Schönmehl R, Winter L, El Khassawna T, Heiss C, Brochhausen C, Rupp M. Targeting mitochondria in bone and cartilage diseases: A narrative review. Redox Biol 2025; 83:103667. [PMID: 40354767 DOI: 10.1016/j.redox.2025.103667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Mitochondria are essential regulators of bone health, controlling cell differentiation, cellular energy production, immune function, osteogenesis, and osteoclast activity. Their dysfunction is linked to orthopedic disorders such as osteoporosis, osteoarthritis, and osteomyelitis, contributing to impaired bone homeostasis and increased fracture risk. While mitochondrial research has been more advanced in fields such as cardiology and neurology, emerging therapeutic strategies from these areas are beginning to show potential for translation into orthopedics. These include mitochondrial biogenesis stimulation, mitochondrial fission inhibition, antioxidant therapies, mitochondrial transplantation, and photobiomodulation, which have demonstrated success in enhancing tissue repair, reducing oxidative stress, and improving overall cellular function in non-orthopedic applications. The novel inhibitor of mitochondrial fission and accumulation of reactive oxygen species Mdivi-1 offers potential to improve clinical outcomes of bone diseases by alleviating cellular dysfunction and preventing bone loss. While these treatments are still in the developmental phase, they present innovative approaches to address mitochondrial dysfunction in orthopedic conditions, potentially transforming bone disease management and enhancing patient outcomes. This report explores research regarding the involvement of mitochondrial health in bone and joint function and discusses possible future treatment strategies targeting mitochondria in orthopedic conditions.
Collapse
Affiliation(s)
- Daniel H Mendelsohn
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Friedrich-Baur-Institute, Department of Neurology, LMU Clinic Munich, Germany
| | - Nike Walter
- Department of Psychosomatic Medicine, University Medical Center Regensburg, Regensburg, Germany
| | - Wing-Hoi Cheung
- Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Ronald Man Yeung Wong
- Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Rebecca Schönmehl
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lina Winter
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Christian Heiss
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen, Germany; Biruni University, Istanbul, Türkiye
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus Rupp
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen, Germany.
| |
Collapse
|
2
|
He Y, Liu T, Peng X, Yao C, Zhou D, Song C, Wei Z, Chen J, Liu Z, Jiang F. Molecular mechanism of mitochondrial autophagy mediating impaired energy metabolism leading to osteoporosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167685. [PMID: 39842521 DOI: 10.1016/j.bbadis.2025.167685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Osteoporosis (OP) is a bone metabolic disease caused by decreased bone mass leading to destruction of bone microstructure. Treatment of OP is characterized by a lifelong nature, causing extreme financial and psychological burdens to patients. Hormonal abnormalities, cellular autophagy, Ferroptosis, and oxidative stress are all part of the intricate and varied pathophysiology of OP. Recent research has revealed that mitochondrial dysfunction is a significant factor in the onset and progression of OP. By regulating bone marrow mesenchymal stem cell differentiation through various signaling pathways and cytokines, abnormal mitochondrial energy metabolism brought on by oxidative stress processes impacts osteoblast and osteoclast proliferation and differentiation, causing an imbalance in bone metabolism that ultimately results in OP. Therefore, one possible method to prevent and manage OP may be to use mitochondria as a carrier to trigger osteogenic differentiation of bone marrow mesenchymal stem cells from mitochondrial energy consumption, oxidative stress, autophagy, and osteoclast death. In order to offer some theoretical references and therapeutic approaches for the clinical prevention and treatment of OP, we will examine the pathophysiology of OP from mitochondrial dysfunction in this work.
Collapse
Affiliation(s)
- Yuheng He
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Tao Liu
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xin Peng
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chaorui Yao
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Daqian Zhou
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chao Song
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Department of Orthopedics, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Zhangchao Wei
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jinwen Chen
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Zongchao Liu
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Luzhou Longmatan District People's Hospital, Luzhou 646000, Sichuan Province, China.
| | - Feng Jiang
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
3
|
Shirvani P, Shirvani A, Holick MF. Mitochondrial Dysfunction and Its Potential Molecular Interplay in Hypermobile Ehlers-Danlos Syndrome: A Scoping Review Bridging Cellular Energetics and Genetic Pathways. Curr Issues Mol Biol 2025; 47:134. [PMID: 39996855 PMCID: PMC11854588 DOI: 10.3390/cimb47020134] [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: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Hypermobile Ehlers-Danlos Syndrome (hEDS) is a hereditary connective tissue disorder characterized by joint hypermobility, skin hyperextensibility, and systemic manifestations such as chronic fatigue, gastrointestinal dysfunction, and neurological symptoms. Unlike other EDS subtypes with known genetic mutations, hEDS lacks definitive markers, suggesting a multifactorial etiology involving both mitochondrial dysfunction and non-mitochondrial pathways. This scoping review, conducted in accordance with the PRISMA-ScR guidelines, highlights mitochondrial dysfunction as a potential unifying mechanism in hEDS pathophysiology. Impaired oxidative phosphorylation (OXPHOS), elevated reactive oxygen species (ROS) levels, and calcium dysregulation disrupt cellular energetics and extracellular matrix (ECM) homeostasis, contributing to the hallmark features of hEDS. We reviewed candidate genes associated with ECM remodeling, signaling pathways, and immune regulation. Protein-protein interaction (PPI) network analyses revealed interconnected pathways linking mitochondrial dysfunction with these candidate genes. Comparative insights from Fabry disease and fragile X premutation carriers underscore shared mechanisms such as RNA toxicity, matrix metalloproteinases (MMP) activation, and ECM degradation. These findings may suggest that mitochondrial dysfunction amplifies systemic manifestations through its interplay with non-mitochondrial molecular pathways. By integrating these perspectives, this review provides a potential framework for understanding hEDS pathogenesis while highlighting latent avenues for future research into its molecular basis. Understanding the potential role of mitochondrial dysfunction in hEDS not only sheds light on its complex molecular etiology but also opens new paths for targeted interventions.
Collapse
Affiliation(s)
| | - Arash Shirvani
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Michael F. Holick
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
4
|
Chen Y, Xiao H, Teng F, Yang A, Yang F, Chen C, Chen R, Geng B, Xia Y. IL-16 Mediates the Effect of Circulating Metabolites on Postmenopausal Osteoporosis: A Two-Step, Multivariable Mendelian Randomization Study. Biol Res Nurs 2025; 27:91-100. [PMID: 39212665 DOI: 10.1177/10998004241279934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Objectives: This study aimed to explore the relationship between circulating metabolites and postmenopausal osteoporosis (PMOP) and to assess the mediating role of inflammatory factors. Methods: Utilizing summary-level data from genome-wide association studies (GWAS) and employing a Mendelian Randomization approach, a two-sample MR analysis was conducted to assess the relationship between circulating metabolites and PMOP. Additionally, a two-step MR was used to quantify the mediating impact of inflammatory factors on the effect of circulating metabolites on PMOP. Results: The results revealed a significant association between certain metabolites and the risk of PMOP, notably the ratio of free cholesterol to total lipids in very large VLDL particles (OR: 1.399, 95% CI: 1.002-1.954, p = 0.048) and IL-16 (OR: 0.773, 95% CI: 0.608-0.983, p = 0.036). IL-16 was found to partially mediate the impact of circulating metabolites on PMOP, with a mediation effect of 10.4%. Conclusion: This study underscores the crucial role of circulating metabolites and inflammatory factors in PMOP pathogenesis. A causal relationship between circulating metabolites and PMOP was established, with IL-16 mediating some effects. These findings hold promise for clinical applications in early detection, personalized medicine, and the identification of therapeutic targets for PMOP.
Collapse
Affiliation(s)
- Yi Chen
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Hefang Xiao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Fei Teng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Ao Yang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Fei Yang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Changshun Chen
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Rongjin Chen
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Orthopaedic Clinical Medicine Research Center, Lanzhou, China
- Gansu Province Intelligent Orthopedics Industry Technology Center, Lanzhou, China
| |
Collapse
|
5
|
Liu J, Zhang X, Hou H, Ouyang J, Dai J. Advances in Osteoblast and Mitochondrial Dynamics and Their Transfer in Osteoporosis. J Cell Mol Med 2024; 28:e70299. [PMID: 39700051 DOI: 10.1111/jcmm.70299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
Mitochondria are important organelles in the human body and play a major role in providing cellular energy, maintaining tissue homeostasis and apoptosis. Osteoporosis, characterised by a decrease in the amount of bone tissue per unit volume, is a metabolic bone pathology with multiple causes. Under pathological conditions, mitochondrial dysfunction leads to an imbalance in mitochondrial homeostasis, resulting in a disruption of osteoblast-osteoclast homeostasis, which in turn disrupts bone homeostasis, and this disruption of homeostasis is an important pathogenetic mechanism underlying chronic metabolic bone disease in osteoporosis. Numerous studies have shown that bone homeostasis is closely related to mitochondrial dynamics and mitochondrial translocation in the mitochondrial quality control system, and the balance between osteoblasts and osteoclasts is closely related to osteoporosis. In this review, we describe the progress of osteoblast and osteoclast research and mitochondrial dynamics in osteoporosis, and the role of mitochondrial translocation in bone homeostasis, in the hope that it can stimulate new research in osteoporotic metabolic bone disease and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Jiaxuan Liu
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | | | - Honghao Hou
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics & Guangdong Engineering Research Center for Translation of Medical 3D Printing Application & National Virtual & Reality Experimental Education Center for Medical Morphology (Southern Medical University) & National key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Jiang L, He H, Tang Y, Li J, Reilly S, Xin H, Li Z, Cai H, Zhang X. Activation of BK channels prevents diabetes-induced osteopenia by regulating mitochondrial Ca 2+ and SLC25A5/ANT2-PINK1-PRKN-mediated mitophagy. Autophagy 2024; 20:2388-2404. [PMID: 38873928 PMCID: PMC11572260 DOI: 10.1080/15548627.2024.2367184] [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: 07/18/2023] [Revised: 05/23/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024] Open
Abstract
Osteopenia and osteoporosis are among the most common metabolic bone diseases and represent major public health problems, with sufferers having an increased fracture risk. Diabetes is one of the most common diseases contributing to osteopenia and osteoporosis. However, the mechanisms underlying diabetes-induced osteopenia and osteoporosis remain unclear. Bone reconstruction, including bone formation and absorption, is a dynamic process. Large-conductance Ca2+-activated K+ channels (BK channels) regulate the function of bone marrow-derived mesenchymal stem cells, osteoblasts, and osteoclasts. Our previous studies revealed the relationship between BK channels and the function of osteoblasts via various pathways under physiological conditions. In this study, we reported a decrease in the expression of BK channels in mice with diabetes-induced osteopenia. BK deficiency enhanced mitochondrial Ca2+ and activated classical PINK1 (PTEN induced putative kinase 1)-PRKN/Parkin (parkin RBR E3 ubiquitin protein ligase)-dependent mitophagy, whereas the upregulation of BK channels inhibited mitophagy in osteoblasts. Moreover, SLC25A5/ANT2 (solute carrier family 25 (mitochondrial carrier, adenine nucleotide translocator), member 5), a critical inner mitochondrial membrane protein participating in PINK1-PRKN-dependent mitophagy, was also regulated by BK channels. Overall, these data identified a novel role of BK channels in regulating mitophagy in osteoblasts, which might be a potential target for diabetes-induced bone diseases.Abbreviations: AGE, advanced glycation end products; Baf A1, bafilomycin A1; BK channels, big-conductance Ca2+-activated K+ channels; BMSCs, bone marrow-derived mesenchymal stem cells; BSA, bovine serum albumin; FBG, fasting blood glucose; IMM, inner mitochondrial membrane; ITPR1, inositol 1,4,5-trisphosphate receptor 1; MAM, mitochondria-associated ER membrane; OMM, outer mitochondrial membrane; PINK1, PTEN induced putative kinase 1; PPID/CyP-D, peptidylprolyl isomerase D (cyclophilin D); PRKN/PARK2, parkin RBR E3 ubiquitin protein ligase; ROS, reactive oxygen species; SLC25A5/ANT2, solute carrier family 25 (mitochondrial carrier, adenine nucleotide translocator), member 5; STZ, streptozotocin.
Collapse
Affiliation(s)
- Lan Jiang
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Haidong He
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Yuyan Tang
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Jiawei Li
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhiping Li
- Department of Clinical Pharmacy, National Children’s Medical Center, Children’s Hospital of Fudan University, Shanghai, China
| | - Hui Cai
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, GA, USA
- Section of Nephrology, Atlanta Veteran Administration Medical Center, Decatur, GA, USA
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Suh J, Lee YS. The multifaceted roles of mitochondria in osteoblasts: from energy production to mitochondrial-derived vesicle secretion. J Bone Miner Res 2024; 39:1205-1214. [PMID: 38907370 PMCID: PMC11371665 DOI: 10.1093/jbmr/zjae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/03/2024] [Indexed: 06/24/2024]
Abstract
Mitochondria in osteoblasts have been demonstrated to play multiple crucial functions in bone formation from intracellular adenosine triphosphate production to extracellular secretion of mitochondrial components. The present review explores the current knowledge about mitochondrial biology in osteoblasts, including mitochondrial biogenesis, bioenergetics, oxidative stress generation, and dynamic changes in morphology. Special attention is given to recent findings, including mitochondrial donut formation in osteoblasts, which actively generates mitochondrial-derived vesicles (MDVs), followed by extracellular secretion of small mitochondria and MDVs. We also discuss the therapeutic effects of targeting osteoblast mitochondria, highlighting their potential applications in improving bone health.
Collapse
Affiliation(s)
- Joonho Suh
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
8
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Zheng H, Liu J, Sun L, Meng Z. The role of N-acetylcysteine in osteogenic microenvironment for bone tissue engineering. Front Cell Dev Biol 2024; 12:1435125. [PMID: 39055649 PMCID: PMC11269162 DOI: 10.3389/fcell.2024.1435125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Bone defect is a common clinical symptom which can arise from various causes. Currently, bone tissue engineering has demonstrated positive therapeutic effects for bone defect repair by using seeding cells such as mesenchymal stem cells and precursor cells. N-acetylcysteine (NAC) is a stable, safe and highly bioavailable antioxidant that shows promising prospects in bone tissue engineering due to the ability to attenuate oxidative stress and enhance the osteogenic potential and immune regulatory function of cells. This review systematically introduces the antioxidant mechanism of NAC, analyzes the advancements in NAC-related research involving mesenchymal stem cells, precursor cells, innate immune cells and animal models, discusses its function using the classic oral microenvironment as an example, and places particular emphasis on the innovative applications of NAC-modified tissue engineering biomaterials. Finally, current limitations and future prospects are proposed, with the aim of providing inspiration for targeted readers in the field.
Collapse
Affiliation(s)
- Haowen Zheng
- School of Dentistry, Tianjin Medical University, Tianjin, China
| | - Jiacheng Liu
- School of Dentistry, Tianjin Medical University, Tianjin, China
- Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin, China
| | - Lanxin Sun
- School of Dentistry, Tianjin Medical University, Tianjin, China
| | - Zhaosong Meng
- Department of Oral and Maxillofacial Surgery, Tianjin Medical University School and Hospital of Stomatology, Tianjin, China
| |
Collapse
|
10
|
Zhou QY, Ren C, Li JY, Wang L, Duan Y, Yao RQ, Tian YP, Yao YM. The crosstalk between mitochondrial quality control and metal-dependent cell death. Cell Death Dis 2024; 15:299. [PMID: 38678018 PMCID: PMC11055915 DOI: 10.1038/s41419-024-06691-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: 10/02/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Mitochondria are the centers of energy and material metabolism, and they also serve as the storage and dispatch hubs of metal ions. Damage to mitochondrial structure and function can cause abnormal levels and distribution of metal ions, leading to cell dysfunction and even death. For a long time, mitochondrial quality control pathways such as mitochondrial dynamics and mitophagy have been considered to inhibit metal-induced cell death. However, with the discovery of new metal-dependent cell death including ferroptosis and cuproptosis, increasing evidence shows that there is a complex relationship between mitochondrial quality control and metal-dependent cell death. This article reviews the latest research results and mechanisms of crosstalk between mitochondrial quality control and metal-dependent cell death in recent years, as well as their involvement in neurodegenerative diseases, tumors and other diseases, in order to provide new ideas for the research and treatment of related diseases.
Collapse
Affiliation(s)
- Qi-Yuan Zhou
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Chao Ren
- Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing-Yan Li
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lu Wang
- Department of Critical Care Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Ren-Qi Yao
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Ying-Ping Tian
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Yong-Ming Yao
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
11
|
Wang Y, Dai X, Li H, Jiang H, Zhou J, Zhang S, Guo J, Shen L, Yang H, Lin J, Yan H. The role of mitochondrial dynamics in disease. MedComm (Beijing) 2023; 4:e462. [PMID: 38156294 PMCID: PMC10753647 DOI: 10.1002/mco2.462] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/14/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023] Open
Abstract
Mitochondria are multifaceted and dynamic organelles regulating various important cellular processes from signal transduction to determining cell fate. As dynamic properties of mitochondria, fusion and fission accompanied with mitophagy, undergo constant changes in number and morphology to sustain mitochondrial homeostasis in response to cell context changes. Thus, the dysregulation of mitochondrial dynamics and mitophagy is unsurprisingly related with various diseases, but the unclear underlying mechanism hinders their clinical application. In this review, we summarize the recent developments in the molecular mechanism of mitochondrial dynamics and mitophagy, particularly the different roles of key components in mitochondrial dynamics in different context. We also summarize the roles of mitochondrial dynamics and target treatment in diseases related to the cardiovascular system, nervous system, respiratory system, and tumor cell metabolism demanding high-energy. In these diseases, it is common that excessive mitochondrial fission is dominant and accompanied by impaired fusion and mitophagy. But there have been many conflicting findings about them recently, which are specifically highlighted in this view. We look forward that these findings will help broaden our understanding of the roles of the mitochondrial dynamics in diseases and will be beneficial to the discovery of novel selective therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Xinyan Dai
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Hui Li
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huiling Jiang
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Junfu Zhou
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Shiying Zhang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jiacheng Guo
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Lidu Shen
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huantao Yang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jie Lin
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Hengxiu Yan
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| |
Collapse
|
12
|
Mendelsohn DH, Niedermair T, Walter N, Alt V, Rupp M, Brochhausen C. Ultrastructural Evidence of Mitochondrial Dysfunction in Osteomyelitis Patients. Int J Mol Sci 2023; 24:5709. [PMID: 36982790 PMCID: PMC10053973 DOI: 10.3390/ijms24065709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
Osteomyelitis is a difficult-to-treat disease with high chronification rates. First studies suggest increases in mitochondrial fission and mitochondrial dysfunction as possible contributors to the accumulation of intracellular reactive oxygen species and thereby to the cell death of infected bone cells. The aim of the present study is to analyze the ultrastructural impact of bacterial infection on osteocytic and osteoblastic mitochondria. Human infected bone tissue samples were visualized via light microscopy and transmission electron microscopy. Osteoblasts, osteocytes and their mitochondria were analyzed histomorphometrically and compared with the control group of noninfectious human bone tissue samples. The results depicted swollen hydropic mitochondria including depleted cristae and a decrease in matrix density in the infected samples. Furthermore, perinuclear clustering of mitochondria could also be observed regularly. Additionally, increases in relative mitochondrial area and number were found as a correlate for increased mitochondrial fission. In conclusion, mitochondrial morphology is altered during osteomyelitis in a comparable way to mitochondria from hypoxic tissues. This gives new perspectives on the treatment strategies since the manipulation of mitochondrial dynamics may improve bone cell survival as a potential new target for the therapy of osteomyelitis.
Collapse
Affiliation(s)
- Daniel H. Mendelsohn
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University Regensburg, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Tanja Niedermair
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University Regensburg, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Nike Walter
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Volker Alt
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Markus Rupp
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University Regensburg, University Hospital Regensburg, 93053 Regensburg, Germany
- Institute of Pathology, University Medical Centre Mannheim, 68167 Mannheim, Germany
| |
Collapse
|
13
|
Briones-Suarez L, Cifuentes M, Bravo-Sagua R. Secretory Factors from Calcium-Sensing Receptor-Activated SW872 Pre-Adipocytes Induce Cellular Senescence and A Mitochondrial Fragmentation-Mediated Inflammatory Response in HepG2 Cells. Int J Mol Sci 2023; 24:ijms24065217. [PMID: 36982291 PMCID: PMC10049719 DOI: 10.3390/ijms24065217] [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: 10/17/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 03/11/2023] Open
Abstract
Adipose tissue inflammation in obesity has a deleterious impact on organs such as the liver, ultimately leading to their dysfunction. We have previously shown that activation of the calcium-sensing receptor (CaSR) in pre-adipocytes induces TNF-α and IL-1β expression and secretion; however, it is unknown whether these factors promote hepatocyte alterations, particularly promoting cell senescence and/or mitochondrial dysfunction. We generated conditioned medium (CM) from the pre-adipocyte cell line SW872 treated with either vehicle (CMveh) or the CaSR activator cinacalcet 2 µM (CMcin), in the absence or presence of the CaSR inhibitor calhex 231 10 µM (CMcin+cal). HepG2 cells were cultured with these CM for 120 h and then assessed for cell senescence and mitochondrial dysfunction. CMcin-treated cells showed increased SA-β-GAL staining, which was absent in TNF-α- and IL-1β-depleted CM. Compared to CMveh, CMcin arrested cell cycle, increased IL-1β and CCL2 mRNA, and induced p16 and p53 senescence markers, which was prevented by CMcin+cal. Crucial proteins for mitochondrial function, PGC-1α and OPA1, were decreased with CMcin treatment, concomitant with fragmentation of the mitochondrial network and decreased mitochondrial transmembrane potential. We conclude that pro-inflammatory cytokines TNF-α and IL-1β secreted by SW872 cells after CaSR activation promote cell senescence and mitochondrial dysfunction, which is mediated by mitochondrial fragmentation in HepG2 cells and whose effects were reversed with Mdivi-1. This investigation provides new evidence about the deleterious CaSR-induced communication between pre-adipocytes and liver cells, incorporating the mechanisms involved in cellular senescence.
Collapse
Affiliation(s)
- Lautaro Briones-Suarez
- Laboratory of Obesity and Metabolism (OMEGA), Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Department of Nutrition and Public Health, Faculty of Health and Food Sciences, University of Bío-Bío, Chillán 3800708, Chile
| | - Mariana Cifuentes
- Laboratory of Obesity and Metabolism (OMEGA), Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Center for Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
- Correspondence: (M.C.); (R.B.-S.); Tel.: +56-229781428 (M.C.); +56-229781563 (R.B.-S.)
| | - Roberto Bravo-Sagua
- Laboratory of Obesity and Metabolism (OMEGA), Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago 7830490, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380492, Chile
- Interuniversity Center for Healthy Aging (CIES), Consortium of Universities of the State of Chile (CUECH), Santiago 8320216, Chile
- Correspondence: (M.C.); (R.B.-S.); Tel.: +56-229781428 (M.C.); +56-229781563 (R.B.-S.)
| |
Collapse
|
14
|
Yan C, Shi Y, Yuan L, Lv D, Sun B, Wang J, Liu X, An F. Mitochondrial quality control and its role in osteoporosis. Front Endocrinol (Lausanne) 2023; 14:1077058. [PMID: 36793284 PMCID: PMC9922754 DOI: 10.3389/fendo.2023.1077058] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Mitochondria are important organelles that provide cellular energy and play a vital role in cell differentiation and apoptosis. Osteoporosis is a chronic metabolic bone disease mainly caused by an imbalance in osteoblast and osteoclast activity. Under physiological conditions, mitochondria regulate the balance between osteogenesis and osteoclast activity and maintain bone homeostasis. Under pathological conditions, mitochondrial dysfunction alters this balance; this disruption is important in the pathogenesis of osteoporosis. Because of the role of mitochondrial dysfunction in osteoporosis, mitochondrial function can be targeted therapeutically in osteoporosis-related diseases. This article reviews different aspects of the pathological mechanism of mitochondrial dysfunction in osteoporosis, including mitochondrial fusion and fission, mitochondrial biogenesis, and mitophagy, and highlights targeted therapy of mitochondria in osteoporosis (diabetes induced osteoporosis and postmenopausal osteoporosis) to provide novel targets and prevention strategies for the prevention and treatment of osteoporosis and other chronic bone diseases.
Collapse
Affiliation(s)
- Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Research Center of Traditional Chinese Medicine of Gansu, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yao Shi
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Lingqing Yuan
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Donghui Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Bai Sun
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jiayu Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiyan Liu
- Internal Medicine, Northwestern University, Xian, Shanxi, China
- *Correspondence: Xiyan Liu, ; Fangyu An,
| | - Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- *Correspondence: Xiyan Liu, ; Fangyu An,
| |
Collapse
|
15
|
Mohammadi E, Nikbakht F, Barati M, Roghani M, Vazifekhah S, Khanizadeh AM, Heidari Z. Protective effect of N-acetyl cysteine on the mitochondrial dynamic imbalance in temporal lobe epilepsy: Possible role of mTOR. Neuropeptides 2022; 96:102294. [PMID: 36270032 DOI: 10.1016/j.npep.2022.102294] [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: 07/27/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022]
Abstract
Understanding the underlying molecular mechanisms involved in epilepsy is critical for the development of more effective therapies. It is believed that mTOR (Mechanistic Target of Rapamycin kinases) activity and the mitochondrial dynamic balance change during epilepsy. mTOR affects mitochondrial fission by stimulating the translation of mitochondrial fission process 1 (MTFP1). In This study, the protective role of N-acetylcysteine was studied in temporal lobe epilepsy (TLE) through the regulation of mTOR and mitochondrial dynamic proteins. Rats received N-acetylcysteine (oral administration) seven days before induction of epilepsy, followed by one day after epilepsy. TLE was induced by microinjection of kainite into the left lateral ventricle. The total mTOR and Drp1 levels in the hippocampus were evaluated by western blotting. MFN1 was assessed using immunohistochemistry, and the expression of Fis.1 and MTFP1 (fission-related proteins) and OPA (fusion-related protein) were detected by real-time PCR. The mitochondrial membrane potential was measured by Rhodamin 123. The results showed that 72 h after induction of epilepsy, the mTOR protein level increased, and the balance of the mitochondrial dynamic was disturbed; however, oral administration of NAC decreased the mTOR protein level and improved the mitochondrial dynamic. These findings indicate that NAC plays a neuroprotective role in temporal lobe epilepsy, probably through decreasing the mTOR protein level, which can improve the imbalance in the mitochondrial dynamic.
Collapse
Affiliation(s)
- Ekram Mohammadi
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farnaz Nikbakht
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahmoud Barati
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| | - Somayeh Vazifekhah
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Khanizadeh
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Heidari
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Lin AJ, Joshi AU, Mukherjee R, Tompkins CA, Vijayan V, Mochly-Rosen D, Haileselassie B. δPKC-Mediated DRP1 Phosphorylation Impacts Macrophage Mitochondrial Function and Inflammatory Response to Endotoxin. Shock 2022; 57:435-443. [PMID: 34738957 PMCID: PMC8885892 DOI: 10.1097/shk.0000000000001885] [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: 11/25/2022]
Abstract
BACKGROUND Recent studies have demonstrated that alterations in mitochondrial dynamics can impact innate immune function. However, the upstream mechanisms that link mitochondrial dynamics to innate immune phenotypes have not been completely elucidated. This study asks if Protein Kinase C, subunit delta (δPKC)-mediated phosphorylation of dynamin-related protein 1 (Drp1), a key driver of mitochondrial fission, impacts macrophage pro-inflammatory response following bacterial-derived lipopolysaccharide (LPS) stimulation. METHODS Using RAW 264.7 cells, bone marrow-derived macrophages from C57BL/6J mice, as well as human monocyte-derived macrophages, we first characterized changes in δPKC-mediated phosphorylation of Drp1 following LPS stimulation. Next, using rationally designed peptides that inhibit δPKC activation (δV1-1) and δPKC-Drp1 interaction (ψDrp1), we determined whether δPKC-mediated phosphorylation of Drp1 impacts LPS-induced changes in mitochondrial morphology, mitochondrial function, and inflammatory response. RESULTS Our results demonstrated that δPKC-dependent Drp1 activation is associated with increased mitochondrial fission, impaired cellular respiration, and increased mitochondrial reactive oxygen species in LPS-treated macrophages. This is reversed using a rationally designed peptide that selectively inhibits δPKC phosphorylation of Drp1 (ψDrp1). Interestingly, limiting excessive mitochondrial fission using ψDrp1 reduced LPS-triggered pro-inflammatory response, including a decrease in NF-κB nuclear localization, decreased iNOS induction, and a reduction in pro-inflammatory cytokines (IL-1β, TNFα, IL-6). CONCLUSION These data suggest that inhibiting Drp1 phosphorylation by δPKC abates the excessive mitochondrial fragmentation and mitochondrial dysfunction that is seen following LPS treatment. Furthermore, these data suggest that limiting δPKC-dependent Drp1 activation decreases the pro-inflammatory response following LPS treatment. Altogether, δPKC-dependent Drp1 phosphorylation might be an upstream mechanistic link between alterations in mitochondrial dynamics and innate immune phenotypes, and may have therapeutic potential.
Collapse
Affiliation(s)
- Amanda J Lin
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amit U Joshi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Riddhita Mukherjee
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics Division of Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Carly A Tompkins
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics Division of Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vijith Vijayan
- Department of Pediatrics Division of Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bereketeab Haileselassie
- Department of Pediatrics Division of Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
17
|
Afroz SF, Condon ND, Sweet MJ, Kapetanovic R. Quantifying Regulated Mitochondrial Fission in Macrophages. Methods Mol Biol 2022; 2523:281-301. [PMID: 35759204 DOI: 10.1007/978-1-0716-2449-4_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Mitochondria have co-evolved with eukaryotic cells for more than a billion years, becoming an important cog in their machinery. They are best known for being tasked with energy generation through the production of adenosine triphosphate, but they also have roles in several other cellular processes, for example, immune and inflammatory responses. Mitochondria have important functions in macrophages, key innate immune cells that detect pathogens and drive inflammation. Mitochondrial activity is influenced by the highly dynamic nature of the mitochondrial network, which alternates between interconnected tubular and fragmented forms. The dynamic balance between this interconnected fused network and fission-mediated mitochondrial fragmentation modulates inflammatory responses such as production of cytokines and mitochondrial reactive oxygen species. Here we describe methods to differentiate mouse bone marrow cells into macrophages and the use of light microscopy, electron microscopy, flow cytometry, and Western blotting to quantify regulated mitochondrial dynamics in these differentiated macrophages.
Collapse
Affiliation(s)
- Syeda Farhana Afroz
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Nicholas D Condon
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, QLD, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, QLD, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
18
|
Mitochondrial Transplantation Modulates Inflammation and Apoptosis, Alleviating Tendinopathy Both In Vivo and In Vitro. Antioxidants (Basel) 2021; 10:antiox10050696. [PMID: 33925007 PMCID: PMC8146308 DOI: 10.3390/antiox10050696] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022] Open
Abstract
Tendinopathy is a common musculoskeletal condition causing pain and dysfunction. Conventional treatment and surgical procedures for tendinopathy are insufficient; accordingly, recent research has focused on tendon-healing regenerative approaches. Tendon injuries usually occur in the hypoxic critical zone, characterized by increased oxidative stress and mitochondrial dysfunction; thus, exogenous intact mitochondria may be therapeutic. We aimed to assess whether mitochondrial transplantation could induce anti-inflammatory activity and modulate the metabolic state of a tendinopathy model. Exogenous mitochondria were successfully delivered into damaged tenocytes by centrifugation. Levels of Tenomodulin and Collagen I in damaged tenocytes were restored with reductions in nuclear factor-κB and matrix metalloproteinase 1. The dysregulation of oxidative stress and mitochondrial membrane potential was attenuated by mitochondrial transplantation. Activated mitochondrial fission markers, such as fission 1 and dynamin-related protein 1, were dose-dependently downregulated. Apoptosis signaling pathway proteins were restored to the pre-damage levels. Similar changes were observed in a collagenase injection-induced rat model of tendinopathy. Exogenous mitochondria incorporated into the Achilles tendon reduced inflammatory and fission marker levels. Notably, collagen production was restored. Our results demonstrate the therapeutic effects of direct mitochondrial transplantation in tendinopathy. These effects may be explained by alterations in anti-inflammatory and apoptotic processes via changes in mitochondrial dynamics.
Collapse
|
19
|
Recombinant Klotho Protects Human Periodontal Ligament Stem Cells by Regulating Mitochondrial Function and the Antioxidant System during H 2O 2-Induced Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9261565. [PMID: 31885825 PMCID: PMC6914990 DOI: 10.1155/2019/9261565] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023]
Abstract
Human periodontal ligament stem cells (hPDLSCs) are a favourable source for tissue engineering, but oxidative stress conditions during cell culture and transplantation could affect stem cell viability and stemness, leading to failed regeneration. The aim of this study was to evaluate the antioxidant and protective effects of Klotho, an antiageing protein, against cell damage and the loss of osteogenesis in hPDLSCs in H2O2-induced oxidative environments. H2O2 was used as an exogenous reactive oxygen species (ROS) to induce oxidative stress. Recombinant human Klotho protein was administered before H2O2 treatment. Multitechniques were used to assess antioxidant activity, cell damage, and osteogenic ability of hPDLSCs in oxidative stress and the effects of Klotho on hPDLSCs. Mitochondrial function was analyzed by an electron microscopy scan of cellular structure, mitochondrial DNA copy number, and cellular oxygen consumption rate (OCR). Furthermore, we explored the pathway by which Klotho may function to regulate the antioxidant system. We found that pretreatment with recombinant human Klotho protein could enhance SOD activity and reduce intracellular oxidative stress levels. Klotho reduced H2O2-induced cellular damage and eventually maintained the osteogenic differentiation potential of hPDLSCs. Notably, Klotho promoted mitochondrial function and activated antioxidants by negatively regulating the PI3K/AKT/FoxO1 pathway. The findings suggest that Klotho protein enhanced the antioxidative ability of hPDLSCs and protected stem cell viability and stemness from H2O2-induced oxidative stress by restoring mitochondrial functions and the antioxidant system.
Collapse
|
20
|
FoxO1 Overexpression Ameliorates TNF- α-Induced Oxidative Damage and Promotes Osteogenesis of Human Periodontal Ligament Stem Cells via Antioxidant Defense Activation. Stem Cells Int 2019; 2019:2120453. [PMID: 31781234 PMCID: PMC6875375 DOI: 10.1155/2019/2120453] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
Periodontitis is a chronic disease that includes the pathologic loss of periodontal tissue and alveolar bone. The inflammatory environment in periodontitis impairs the osteogenic differentiation potential and depresses the regeneration capacity of human periodontal ligament stem cells (hPDLSCs). Since Forkhead box protein O1 (FoxO1) plays an important role in redox balance and bone formation, we investigated the role of FoxO1 in oxidative stress resistance and osteogenic differentiation in an inflammatory environment by overexpressing FoxO1 in hPDLSCs. First, we found that FoxO1 overexpression reduced reactive oxygen species (ROS) accumulation, decreased malondialdehyde (MDA) levels, and elevated antioxidant potential under oxidative condition. Next, the overexpression of FoxO1 protected hPDLSCs against oxidative damage, which involved stabilization of the mitochondrial membrane potential. Third, overexpressed FoxO1 promoted extracellular matrix (ECM) mineralization and increased the expression of the osteogenic markers Runx2 and SP7 in the inflammatory environment. These results indicated that FoxO1 overexpression in hPDLSCs has an anti-inflammatory effect, increases antioxidative capacity, and positively regulates osteogenesis in a mimicked inflammatory environment.
Collapse
|
21
|
Zhang Q, Hu C, Huang J, Liu W, Lai W, Leng F, Tang Q, Liu Y, Wang Q, Zhou M, Sheng F, Li G, Zhang R. ROCK1 induces dopaminergic nerve cell apoptosis via the activation of Drp1-mediated aberrant mitochondrial fission in Parkinson's disease. Exp Mol Med 2019; 51:1-13. [PMID: 31578315 PMCID: PMC6802738 DOI: 10.1038/s12276-019-0318-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/22/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
Dopamine deficiency is mainly caused by apoptosis of dopaminergic nerve cells in the substantia nigra of the midbrain and the striatum and is an important pathologic basis of Parkinson’s disease (PD). Recent research has shown that dynamin-related protein 1 (Drp1)-mediated aberrant mitochondrial fission plays a crucial role in dopaminergic nerve cell apoptosis. However, the upstream regulatory mechanism remains unclear. Our study showed that Drp1 knockdown inhibited aberrant mitochondrial fission and apoptosis. Importantly, we found that ROCK1 was activated in an MPP+-induced PD cell model and that ROCK1 knockdown and the specific ROCK1 activation inhibitor Y-27632 blocked Drp1-mediated aberrant mitochondrial fission and apoptosis of dopaminergic nerve cells by suppressing Drp1 dephosphorylation/activation. Our in vivo study confirmed that Y-27632 significantly improved symptoms in a PD mouse model by inhibiting Drp1-mediated aberrant mitochondrial fission and apoptosis. Collectively, our findings suggest an important molecular mechanism of PD pathogenesis involving ROCK1-regulated dopaminergic nerve cell apoptosis via the activation of Drp1-induced aberrant mitochondrial fission. Researchers in China have revealed how a protein molecule plays an early part in the molecular steps that can lead to Parkinson’s disease, which is caused by the death of nerve cells that make the neurotransmitter dopamine. Disruption of mitochondria, the energy-generating bodies inside cells, was already known to lead to the death of dopamine-producing cells. Rong Zhang, Guobing Li and colleagues at The Second Affiliated Hospital of Army Medical University in Chongqing, China traced the chain of cause and effect back to a protein called ROCK-1. Using a mouse model of Parkinson’s disease, they found that ROCK-1 activates another protein previously shown to trigger the disruption of mitochondria. ROCK-1’s early role in the sequence might make it a suitable target for treatment using drugs that inhibit its activity.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Jingbin Huang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Faning Leng
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Qin Tang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Yali Liu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Qing Wang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Min Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Fangfang Sheng
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China.
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, 400037, Chongqing, China.
| |
Collapse
|
22
|
Klimova N, Long A, Kristian T. Nicotinamide mononucleotide alters mitochondrial dynamics by SIRT3-dependent mechanism in male mice. J Neurosci Res 2019; 97:975-990. [PMID: 30801823 PMCID: PMC6565489 DOI: 10.1002/jnr.24397] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 01/02/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+ ) is a central signaling molecule and enzyme cofactor that is involved in a variety of fundamental biological processes. NAD+ levels decline with age, neurodegenerative conditions, acute brain injury, and in obesity or diabetes. Loss of NAD+ results in impaired mitochondrial and cellular functions. Administration of NAD+ precursor, nicotinamide mononucleotide (NMN), has shown to improve mitochondrial bioenergetics, reverse age-associated physiological decline, and inhibit postischemic NAD+ degradation and cellular death. In this study, we identified a novel link between NAD+ metabolism and mitochondrial dynamics. A single dose (62.5 mg/kg) of NMN, administered to male mice, increases hippocampal mitochondria NAD+ pools for up to 24 hr posttreatment and drives a sirtuin 3 (SIRT3)-mediated global decrease in mitochondrial protein acetylation. This results in a reduction of hippocampal reactive oxygen species levels via SIRT3-driven deacetylation of mitochondrial manganese superoxide dismutase. Consequently, mitochondria in neurons become less fragmented due to lower interaction of phosphorylated fission protein, dynamin-related protein 1 (pDrp1 [S616]), with mitochondria. In conclusion, manipulation of mitochondrial NAD+ levels by NMN results in metabolic changes that protect mitochondria against reactive oxygen species and excessive fragmentation, offering therapeutic approaches for pathophysiologic stress conditions.
Collapse
Affiliation(s)
- Nina Klimova
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), Baltimore, Maryland 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Aaron Long
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), Baltimore, Maryland 21201, USA
| |
Collapse
|
23
|
Mitochondrial dynamics and their potential as a therapeutic target. Mitochondrion 2019; 49:269-283. [PMID: 31228566 DOI: 10.1016/j.mito.2019.06.002] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/02/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022]
Abstract
Mitochondrial dynamics shape the mitochondrial network and contribute to mitochondrial function and quality control. Mitochondrial fusion and division are integrated into diverse cellular functions and respond to changes in cell physiology. Imbalanced mitochondrial dynamics are associated with a range of diseases that are broadly characterized by impaired mitochondrial function and increased cell death. In various disease models, modulating mitochondrial fusion and division with either small molecules or genetic approaches has improved function. Although additional mechanistic understanding of mitochondrial fusion and division will be critical to inform further therapeutic approaches, mitochondrial dynamics represent a powerful therapeutic target in a wide range of human diseases.
Collapse
|
24
|
Mayer K, Sommer N, Hache K, Hecker A, Reiche S, Schneck E, Weissmann N, Seeger W, Hecker M. Resolvin E1 Improves Mitochondrial Function in Human Alveolar Epithelial Cells during Severe Inflammation. Lipids 2019; 54:53-65. [DOI: 10.1002/lipd.12119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 01/23/2023]
Affiliation(s)
- Konstantin Mayer
- Medical Clinic II, University of Giessen and Marburg Lung Center (UGMLC); Justus-Liebig-University; Klinikstr. 33, 35392 Giessen Germany
| | - Natascha Sommer
- Medical Clinic II, University of Giessen and Marburg Lung Center (UGMLC); Justus-Liebig-University; Klinikstr. 33, 35392 Giessen Germany
| | - Karl Hache
- Medical Clinic II, University of Giessen and Marburg Lung Center (UGMLC); Justus-Liebig-University; Klinikstr. 33, 35392 Giessen Germany
| | - Andreas Hecker
- Department of General and Thoracic Surgery; University Hospital of Giessen, Rudolf-Buchheim-Str. 7; 35392 Giessen Germany
| | - Sylvia Reiche
- Medical Clinic II, University of Giessen and Marburg Lung Center (UGMLC); Justus-Liebig-University; Klinikstr. 33, 35392 Giessen Germany
| | - Emmanuel Schneck
- Department of Anesthesiology and Intensive Care Medicine; University Hospital of Giessen, Rudolf-Buchheim-Str. 7; 35392 Giessen Germany
| | - Norbert Weissmann
- Medical Clinic II, University of Giessen and Marburg Lung Center (UGMLC); Justus-Liebig-University; Klinikstr. 33, 35392 Giessen Germany
| | - Werner Seeger
- Medical Clinic II, University of Giessen and Marburg Lung Center (UGMLC); Justus-Liebig-University; Klinikstr. 33, 35392 Giessen Germany
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodelling, Ludwigstr. 43; 61231 Bad Nauheim Germany
| | - Matthias Hecker
- Medical Clinic II, University of Giessen and Marburg Lung Center (UGMLC); Justus-Liebig-University; Klinikstr. 33, 35392 Giessen Germany
| |
Collapse
|
25
|
He Y, Gan X, Zhang L, Liu B, Zhu Z, Li T, Zhu J, Chen J, Yu H. CoCl 2 induces apoptosis via a ROS-dependent pathway and Drp1-mediated mitochondria fission in periodontal ligament stem cells. Am J Physiol Cell Physiol 2018; 315:C389-C397. [PMID: 29768044 DOI: 10.1152/ajpcell.00248.2017] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oxygen deficiency is associated with various oral diseases, including chronic periodontitis, age-related alveolar bone loss, and mechanical stress-linked cell injury from orthodontic appliances. Nevertheless, our understanding of the impact of hypoxia on periodontal tissues and its biochemical mechanism is still rudimentary. The purpose of this research was to elucidate the effects of hypoxia on the apoptosis of human periodontal ligament stem cells (PDLSCs) in vitro and the underlying mechanism. Herein, we showed that cobalt chloride (CoCl2) triggered cell dysfunction in human PDLSCs in a concentration-dependent manner and resulted in cell apoptosis and oxidative stress overproduction and accumulation in PDLSCs. In addition, CoCl2 promoted mitochondrial fission in PDLSCs. Importantly, CoCl2 increased the expression of dynamin-related protein 1 (Drp1), the major regulator in mitochondrial fission, in PDLSCs. Mitochondrial division inhibitor-1, pharmacological inhibition of Drp1, not only inhibited mitochondrial fission but also protected against CoCl2-induced PDLSC dysfunction, as shown by increased mitochondrial membrane potential, increased ATP level, reduced reactive oxygen species (ROS) level, and decreased apoptosis. Furthermore, N-acety-l-cysteine, a pharmacological inhibitor of ROS, also abolished CoCl2-induced expression of Drp1 and protected against CoCl2-induced PDLSC dysfunction, as shown by restored mitochondrial membrane potential, ATP level, inhibited mitochondrial fission, and decreased apoptosis. Collectively, our data provide new insights into the role of the ROS-Drp1-dependent mitochondrial pathway in CoCl2-induced apoptosis in PDLSCs, indicating that ROS and Drp1 are promising therapeutic targets for the treatment of CoCl2-induced PDLSC dysfunction.
Collapse
Affiliation(s)
- Yuting He
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China.,West China-Washington Mitochondria and Metabolism Center, Department of Anesthesiology, West China Hospital, Sichuan University , Chengdu , People's Republic of China
| | - Xueqi Gan
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China
| | - Ling Zhang
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China
| | - Beilei Liu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China
| | - Zhuoli Zhu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China
| | - Tao Li
- West China-Washington Mitochondria and Metabolism Center, Department of Anesthesiology, West China Hospital, Sichuan University , Chengdu , People's Republic of China
| | - Junfei Zhu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China
| | - Junsheng Chen
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China
| | - Haiyang Yu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , People's Republic of China
| |
Collapse
|
26
|
Ježek J, Cooper KF, Strich R. Reactive Oxygen Species and Mitochondrial Dynamics: The Yin and Yang of Mitochondrial Dysfunction and Cancer Progression. Antioxidants (Basel) 2018; 7:E13. [PMID: 29337889 PMCID: PMC5789323 DOI: 10.3390/antiox7010013] [Citation(s) in RCA: 311] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/02/2018] [Accepted: 01/09/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria are organelles with a highly dynamic ultrastructure maintained by a delicate equilibrium between its fission and fusion rates. Understanding the factors influencing this balance is important as perturbations to mitochondrial dynamics can result in pathological states. As a terminal site of nutrient oxidation for the cell, mitochondrial powerhouses harness energy in the form of ATP in a process driven by the electron transport chain. Contemporaneously, electrons translocated within the electron transport chain undergo spontaneous side reactions with oxygen, giving rise to superoxide and a variety of other downstream reactive oxygen species (ROS). Mitochondrially-derived ROS can mediate redox signaling or, in excess, cause cell injury and even cell death. Recent evidence suggests that mitochondrial ultrastructure is tightly coupled to ROS generation depending on the physiological status of the cell. Yet, the mechanism by which changes in mitochondrial shape modulate mitochondrial function and redox homeostasis is less clear. Aberrant mitochondrial morphology may lead to enhanced ROS formation, which, in turn, may deteriorate mitochondrial health and further exacerbate oxidative stress in a self-perpetuating vicious cycle. Here, we review the latest findings on the intricate relationship between mitochondrial dynamics and ROS production, focusing mainly on its role in malignant disease.
Collapse
Affiliation(s)
- Jan Ježek
- Department of Molecular Biology, Rowan University Graduate School of Biomedical Sciences, Stratford, NJ 08084, USA.
| | - Katrina F Cooper
- Department of Molecular Biology, Rowan University Graduate School of Biomedical Sciences, Stratford, NJ 08084, USA.
| | - Randy Strich
- Department of Molecular Biology, Rowan University Graduate School of Biomedical Sciences, Stratford, NJ 08084, USA.
| |
Collapse
|
27
|
Smith G, Gallo G. To mdivi-1 or not to mdivi-1: Is that the question? Dev Neurobiol 2017; 77:1260-1268. [PMID: 28842943 DOI: 10.1002/dneu.22519] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/17/2017] [Accepted: 08/22/2017] [Indexed: 12/12/2022]
Abstract
The fission/division and fusion of mitochondria are fundamental aspects of mitochondrial biology. The balance of fission and fusion sets the length of mitochondria in cells to serve their physiological requirements. The fission of mitochondria is markedly induced in many disease states and in response to cellular injury, resulting in the fragmentation of mitochondria into dysfunctional units. The mechanism that drives fission is dependent on the dynamin related protein 1 (Drp1) GTPase. mdivi-1 is a quinazolinone originally described as a selective inhibitor of Drp1, over other dynamin family members, and reported to inhibit mitochondrial fission. A recent study has challenged the activity of mdivi-1 as an inhibitor of Drp1. This study raises serious issues regarding the interpretation of data addressing the effects of mdivi-1 as reflective of the inhibition of Drp1 and thus fission. This commentary considers the evidence for and against mdivi-1 as an inhibitor of Drp1 and presents the following considerations; (1) the activity of mdivi-1 toward Drp1 GTPase activity requires further biochemical investigation, (2) as there is a large body of literature using mdivi-1 in vitro with effects as predicted for inhibition of Drp1 and mitochondrial fission, reviewed herein, the evidence is in favor of mdivi-1's originally described bioactivity, and (3) until the issue is resolved, experimental interpretations for the effects of mdivi-1 on inhibition of fission in cell and tissue experiments warrants stringent positive controls directly addressing the effects of mdivi-1 on fission. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1260-1268, 2017.
Collapse
Affiliation(s)
- George Smith
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, Pennsylvania, 19140
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, 3500 North Broad Street, Philadelphia, Pennsylvania, 19140
| |
Collapse
|