1
|
Zhou H, Wu C, Jin Y, Wu O, Chen L, Guo Z, Wang X, Chen Q, Kwan KYH, Li YM, Xia D, Chen T, Wu A. Role of oxidative stress in mitochondrial dysfunction and their implications in intervertebral disc degeneration: Mechanisms and therapeutic strategies. J Orthop Translat 2024; 49:181-206. [PMID: 39483126 PMCID: PMC11526088 DOI: 10.1016/j.jot.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/03/2024] [Accepted: 08/22/2024] [Indexed: 11/03/2024] Open
Abstract
Background Intervertebral disc degeneration (IVDD) is widely recognized as one of the leading causes of low back pain. Intervertebral disc cells are the main components of the intervertebral disc (IVD), and their functions include synthesizing and secreting collagen and proteoglycans to maintain the structural and functional stability of the IVD. In addition, IVD cells are involved in several physiological processes. They help maintain nutrient metabolism balance in the IVD. They also have antioxidant and anti-inflammatory effects. Because of these roles, IVD cells are crucial in IVDD. When IVD cells are subjected to oxidative stress, mitochondria may become damaged, affecting normal cell function and accelerating degenerative changes. Mitochondria are the energy source of the cell and regulate important intracellular processes. As a key site for redox reactions, excessive oxidative stress and reactive oxygen species can damage mitochondria, leading to inflammation, DNA damage, and apoptosis, thus accelerating disc degeneration. Aim of review Describes the core knowledge of IVDD and oxidative stress. Comprehensively examines the complex relationship and potential mechanistic pathways between oxidative stress, mitochondrial dysfunction and IVDD. Highlights potential therapeutic targets and frontier therapeutic concepts. Draws researchers' attention and discussion on the future research of all three. Key scientific concepts of review Origin, development and consequences of IVDD, molecular mechanisms of oxidative stress acting on mitochondria, mechanisms of oxidative stress damage to IVD cells, therapeutic potential of targeting mitochondria to alleviate oxidative stress in IVDD. The translational potential of this article Targeted therapeutic strategies for oxidative stress and mitochondrial dysfunction are particularly critical in the treatment of IVDD. Using antioxidants and specific mitochondrial therapeutic agents can help reduce symptoms and pain. This approach is expected to significantly improve the quality of life for patients. Individualized therapeutic approaches, on the other hand, are based on an in-depth assessment of the patient's degree of oxidative stress and mitochondrial functional status to develop a targeted treatment plan for more precise and effective IVDD management. Additionally, we suggest preventive measures like customized lifestyle changes and medications. These are based on understanding how IVDD develops. The aim is to slow down the disease and reduce the chances of it coming back. Actively promoting clinical trials and evaluating the safety and efficacy of new therapies helps translate cutting-edge treatment concepts into clinical practice. These measures not only improve patient outcomes and quality of life but also reduce the consumption of healthcare resources and the socio-economic burden, thus having a positive impact on the advancement of the IVDD treatment field.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, 315010, China
| | - Chenyu Wu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, 315010, China
| | - Yuxin Jin
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Ouqiang Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Zhenyu Guo
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Xinzhou Wang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| | - Qizhu Chen
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200082, China
| | - Kenny Yat Hong Kwan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 5/F Professorial Block, Queen Mary Hospital, 102 Pokfulam Road, Pokfulam, China
| | - Yan Michael Li
- Minimally Invasive Brain and Spine Institute, Upstate Medical University 475 Irving Ave, #402 Syracuse, NY, 13210, USA
| | - Dongdong Xia
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, 315010, China
| | - Tao Chen
- Department of Orthopaedics, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital, Tongji University School of Medicine, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China
| |
Collapse
|
2
|
Huang Y, Huang Y, Cai Z, Ferrari MW, Li C, Zhang T, Lyu G, Wang Z. MiR-21-3p inhibitor exerts myocardial protective effects by altering macrophage polarization state and reducing excessive mitophagy. Commun Biol 2024; 7:1371. [PMID: 39438580 PMCID: PMC11496525 DOI: 10.1038/s42003-024-07050-3] [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: 03/12/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Chronic heart failure (CHF) is closely associated with inflammation and mitochondrial dysfunction in cardiomyocytes. This study attempts to investigate the effects of microRNA-21-3p (miR-21-3p) on macrophage polarization and mitophagy in CHF. Here we found miR-21-3p was upregulated in CHF and negatively correlated with carnitine palmitoyl transferase 1A (CPT1A). L-palmitoyl carnitine (L-PC) exacerbated isoproterenol (ISO)-induced myocardial structural disruption and fibrosis in rats, which was exacerbated by miR-21-3p. Mechanistically, miR-21-3p accelerated M1 macrophage polarization. Both miR-21-3p inhibitor and CPT1A overexpression suppressed mitophagy. The inhibition of CPT1A on mitophagy was reversed by miR-21-3p. MiR-21-3p targeted CPT1A mRNA and co-localized with CPT1A protein in cardiomyocytes. In the co-culture system of M1 macrophages and H9c2 cells, miR-21-3p mimics in H9c2 cells promoted M1 polarization, whereas miR-21-3p inhibitor reduced M1 phenotype. M1 macrophages exacerbated H9c2 cell damage. These findings support the potential therapeutic targeting of miR-21-3p to regulate inflammation and mitophagy by inducing CPT1A in CHF.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yalin Huang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhaoling Cai
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Markus W Ferrari
- Department of Internal Medicine 1, Helios-HSK Clinics, Wiesbaden, Germany
| | - Chengyi Li
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Tianzhang Zhang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Guorong Lyu
- Department of Ultrasound Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Collaborative Innovation Center for Maternal and Infant Health Service Application Technology of Education Ministry, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Zhenhua Wang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| |
Collapse
|
3
|
Chang H, Zhang W, Xu L, Li Z, Lin C, Shen Y, Zhang G, Mao L, Ma C, Liu N, Lu H. Copper aggravated synaptic damage after traumatic brain injury by downregulating BNIP3-mediated mitophagy. Autophagy 2024:1-17. [PMID: 39415457 DOI: 10.1080/15548627.2024.2409613] [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: 07/12/2023] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
Synaptic damage is a crucial pathological process in traumatic brain injury. However, the mechanisms driving this process remain poorly understood. In this report, we demonstrate that the accumulation of damaged mitochondria, resulting from impaired mitphagy, plays a significant role in causing synaptic damage. Moreover, copper induced downregulation of BNIP3 is a key player in regulating mitophagy. DMSA alleviates synaptic damage and mitochondrial dysfunction by promoting urinary excretion of copper. Mechanistically, we find that copper downregulate BNIP3 by increasing the nuclear translocation of NFKB, which is triggered by TRIM25-mediated ubiquitination-dependent degradation of NFKBIA. Our study underscores the importance of copper accumulation in the regulation of BNIP3-mediated mitophagy and suggests that therapeutic targeting of the copper-TRIM25-NFKB-BNIP3 axis holds promise to attenuate synaptic damage after traumatic brain injury.
Collapse
Affiliation(s)
- Hanxiao Chang
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Weiwei Zhang
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Xu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Zheng Li
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Chao Lin
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Yuqi Shen
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Guangjian Zhang
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Lei Mao
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Chencheng Ma
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Ning Liu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Hua Lu
- Department of Neurosurgery, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Li L, Yuan R, Wu M, Yin X, Zhang M, Chen Z. Progress in the regulatory mechanism of mitophagy in chronic cerebral ischemic neuronal injury. Exp Neurol 2024; 383:115003. [PMID: 39419436 DOI: 10.1016/j.expneurol.2024.115003] [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: 08/17/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
Chronic cerebral ischemia (CCI) is a common clinical syndrome that can impact various cerebrovascular diseases. Its pathological mechanism of injury involves energy imbalance, oxidative stress, inflammatory response, and many other processes. Neuronal damage occurs in a complex and multifaceted manner. This article provides a detailed discussion of the activation and inhibition mechanisms of mitophagy under cerebral ischemia and considers the advantages and disadvantages of mitophagy in the recovery process of ischemic brain injury. Finally, we address the future direction of research on neuronal injury and the regulatory mechanisms of mitophagy in chronic cerebral ischemia. Future studies should focus on drug intervention at specific regulatory points and the cross-regulation of related signaling pathways to comprehensively deepen understanding of the mechanisms of neuronal injury in chronic cerebral ischemia. Promising interventions could potentially improve the treatment and outcomes of chronic cerebral ischemia.
Collapse
Affiliation(s)
- Lihong Li
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Rui Yuan
- Department of Otolaryngology, Head and Neck Surgery, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Moxin Wu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Manqing Zhang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang 332005, Jiangxi, China.
| | - Zhiying Chen
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang 332005, Jiangxi, China.
| |
Collapse
|
5
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [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] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
Yin J, Xu X, Guo Y, Sun C, Yang Y, Liu H, Yu P, Wu T, Song X. Repair and regeneration: ferroptosis in the process of remodeling and fibrosis in impaired organs. Cell Death Discov 2024; 10:424. [PMID: 39358326 PMCID: PMC11447141 DOI: 10.1038/s41420-024-02181-2] [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: 04/02/2024] [Revised: 09/01/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
As common clinical-pathological processes, wound healing and tissue remodelling following injury or stimulation are essential topics in medical research. Promoting the effective healing of prolonged wounds, improving tissue repair and regeneration, and preventing fibrosis are important and challenging issues in clinical practice. Ferroptosis, which is characterized by iron overload and lipid peroxidation, is a nontraditional form of regulated cell death. Emerging evidence indicates that dysregulated metabolic pathways and impaired iron homeostasis play important roles in various healing and regeneration processes via ferroptosis. Thus, we review the intrinsic mechanisms of tissue repair and remodeling via ferroptosis in different organs and systems under various conditions, including the inflammatory response in skin wounds, remodeling of joints and cartilage, and fibrosis in multiple organs. Additionally, we summarize the common underlying mechanisms, key molecules, and targeted drugs for ferroptosis in repair and regeneration. Finally, we discuss the potential of therapeutic agents, small molecules, and novel materials emerging for targeting ferroptosis to promote wound healing and tissue repair and attenuate fibrosis.
Collapse
Affiliation(s)
- Jiali Yin
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xinjun Xu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Ying Guo
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Caiyu Sun
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yujuan Yang
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Huifang Liu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Second Clinical Medicine College, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Pengyi Yu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Tong Wu
- Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China.
| | - Xicheng Song
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China.
| |
Collapse
|
7
|
Zhang Y, Hu K, Shang Z, Yang X, Cao L. Ferroptosis: Regulatory mechanisms and potential targets for bone metabolism: A review. Medicine (Baltimore) 2024; 103:e39158. [PMID: 39331895 PMCID: PMC11441915 DOI: 10.1097/md.0000000000039158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2024] Open
Abstract
Bone homeostasis is a homeostasis process constructed by osteoblast bone formation and osteoclast bone resorption. Bone homeostasis imbalance and dysfunction are the basis for the development of various orthopedic diseases such as osteoporosis, osteoarthritis, and steroid-induced avascular necrosis of femoral head. Previous studies have demonstrated that ferroptosis can induce lipid peroxidation through the generation of reactive oxygen species, activate a number of signaling pathways, and participate in the regulation of osteoblast bone formation and osteoclast bone resorption, resulting in bone homeostasis imbalance, which is an important factor in the pathogenesis of many orthopedic diseases, but the mechanism of ferroptosis is still unknown. In recent years, it has been found that, in addition to iron metabolism and intracellular antioxidant system imbalance, organelle dysfunction is also a key factor affecting ferroptosis. This paper takes this as the starting point, reviews the latest literature reports at home and abroad, elaborates the pathogenesis and regulatory pathways of ferroptosis and the relationship between ferroptosis and various organelles, and summarizes the mechanism by which ferroptosis mediates bone homeostasis imbalance, with the aim of providing new directions for the research related to ferroptosis and new ideas for the prevention and treatment of bone and joint diseases.
Collapse
Affiliation(s)
- Yongjie Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kangyi Hu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhengya Shang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaorui Yang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linzhong Cao
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
- The Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
8
|
Poznyak AV, Orekhov NA, Churov AV, Starodubtseva IA, Beloyartsev DF, Kovyanova TI, Sukhorukov VN, Orekhov AN. Mitochondrial Dysfunction in Systemic Lupus Erythematosus: Insights and Therapeutic Potential. Diseases 2024; 12:226. [PMID: 39329895 PMCID: PMC11430897 DOI: 10.3390/diseases12090226] [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: 08/06/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by the presence of various serum autoantibodies and multi-system effects, predominantly affecting young female patients. The pathogenesis of SLE involves a combination of genetic factors, environmental triggers, and pathogen invasions that disrupt immune cell activation, leading to the release of autoantibodies and chronic inflammation. Mitochondria, as the primary cellular powerhouses, play a crucial role in SLE development through their control of energy generation, reactive oxygen species (ROS) production, and cellular apoptotic pathways. Dysregulation of mitochondrial structure and function can contribute to the immune dysregulation, oxidative stress, and inflammation seen in SLE. Recent research has highlighted the impact of mitochondrial dysfunction on various immune cells involved in SLE pathogenesis, such as T-lymphocytes, B-lymphocytes, neutrophils, and plasmacytoid dendritic cells. Mitochondrial dysfunction in these immune cells leads to increased ROS production, disrupted mitophagy, and alterations in energy metabolism, contributing to immune dysregulation and inflammation. Moreover, genetic variations in mitochondrial DNA (mtDNA) and abnormalities in mitochondrial dynamics have been linked to the pathogenesis of SLE, exacerbating oxidative stress and immune abnormalities. Targeting mitochondrial function has emerged as a promising therapeutic approach for SLE. Drugs such as sirolimus, N-acetylcysteine, coenzyme Q10, and metformin have shown potential in restoring mitochondrial homeostasis, reducing oxidative stress, and modulating immune responses in SLE. These agents have demonstrated efficacy in preclinical models and clinical studies by improving disease activity, reducing autoantibody titers, and ameliorating organ damage in SLE patients. In conclusion, this review underscores the critical role of mitochondria in the pathogenesis of SLE and the potential of targeting mitochondrial dysfunction as a novel therapeutic strategy for improving outcomes in SLE patients. Further investigation into the mechanisms underlying mitochondrial involvement in SLE and the development of targeted mitochondrial therapies hold promise for advancing SLE treatment and enhancing patient care.
Collapse
Affiliation(s)
- Anastasia V Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Nikolay A Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Alexey V Churov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Russian Gerontology Clinical Research Centre, Institute on Aging Research, Pirogov Russian National Research Medical University, Russian Federation, 16 1st Leonova Street, 129226 Moscow, Russia
| | - Irina A Starodubtseva
- Department of Polyclinic Therapy, N.N. Burdenko Voronezh State Medical University, 10 Studencheskaya Street, 394036 Voronezh, Russia
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, 117997 Moscow, Russia
| | - Tatiana I Kovyanova
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Vasily N Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| |
Collapse
|
9
|
Xing Y, Huang B, Cui Z, Zhang Q, Ma H. Dioscin improves fatty liver hemorrhagic syndrome by promoting ERα-AMPK mediated mitophagy in laying hens. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156056. [PMID: 39342780 DOI: 10.1016/j.phymed.2024.156056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/06/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Mitochondria play a crucial role in upholding metabolic homeostasis. Mitochondrial damage closely associated with the pathogenesis of fatty liver hemorrhagic syndrome (FLHS), while mitophagy being among the most effective methods for eliminating the damaged mitochondria. Dioscin, a natural extract, can activate autophagy; however, its effects on FLHS regarding mitophagy regulation remain unelucidated. PURPOSE We explored the impact of dioscin on FLHS induced by a high-energy and low-protein (HELP) diet in laying hens, mainly focused the protective effects of dioscin on mitochondrial injury. METHOD To investigate the impact of dioscin on fatty liver syndrome in laying hens, we first induced the condition by feeding them a high-energy and low-protein diet. Then, we assessed lipid metabolism-related markers using oil red staining and a commercial detection kit. In addition, the role of dioscin on fatty liver syndrome in laying hens was confirmed by assessing the activation of hepatocyte fat deposition and hepatocyte apoptosis; and the mechanism of dioscin in FLHS was investigated through LMH cell experiment in vitro. Furthermore, CETSA and molecular docking were conducted for additional confirmation. RESULT The results showed that dioscin alleviated mitochondrial damage, relieved the excessive deposition of hepatic lipid droplets and oxidative stress induced by HELP diet in laying hens. Furthermore, dioscin regulated the mitophagy by activating the estrogen receptor α (ERα)/adenosine 5'-monophosphate-activated protein kinase (AMPK) signaling pathway, thus mitigating mitochondria injury and apoptosis in hepatocytes. In addition, we found that dioscin promoted the translocation of nuclear transcription factor into nucleus by activating ERα-AMPK signaling, facilitating autophagic flux in the liver of laying hens and LMH cells. Furthermore, cells pretreated with the lysosomal acidification inhibitor bafilomycin A1 blocked the inhibitory effect of dioscin on the apoptosis induced by palmitic acid (PA)-stimulation in LMH cells, suggesting that dioscin reduces PA-induced apoptosis by activating mitophagy. Moreover, dioscin-induced lysosomal acidification and mitochondrial biogenesis were reversed in PA-induced LMH cells pretreated with ERα-specific inhibitor methylpiperidino pyrazole. CONCLUSION This study firstly demonstrated that dioscin alleviates fatty liver syndrome induced by HELP diet in laying hens. The findings from this study illustrated that dioscin plays a significant role in reducing mitochondrial damage and apoptosis, and these beneficial effects mainly achieve through promotion of ERα-AMPK signaling, which mediates autophagy within the liver of laying hens fed a HELP-diets. These findings provide a theoretical basis for considering dioscin as a possible treatment option for mitigating FLHS in egg-laying hens.
Collapse
Affiliation(s)
- Yuxiao Xing
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Benzeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ziyi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Quanwei Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
10
|
Yu S, Zhu W, Yu L. The role of rapamycin in the PINK1/Parkin signaling pathway in mitophagy in podocytes. Open Life Sci 2024; 19:20220958. [PMID: 39290494 PMCID: PMC11406223 DOI: 10.1515/biol-2022-0958] [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: 05/18/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
This study aimed to clarify the role of rapamycin in the PINK1/Parkin signaling pathway in mitophagy in podocytes and the role of voltage-dependent anion channel 1 (VDAC1) in the PINK1/Parkin signaling pathway in mouse glomerular podocytes. For this purpose, podocytes were cultured with rapamycin and observed using microscopy. The apoptosis rate of podocytes was detected by flow cytometry. Changes in the mitochondrial membrane potential were measured. The autophagy-related proteins VDAC1, PINK1, Parkin, and LC3 were detected, and mitochondrial autophagosomes were observed via transmission electron microscopy. In the present study, we demonstrated that the number of podocytes treated with rapamycin was significantly reduced. Compared with those in the control group, the apoptosis rate of podocytes and the degree of mitochondrial membrane potential depolarization were significantly higher. We also found the expression levels of VDAC1, PINK1, Parkin, and LC3 were significantly increased. In the rapamycin-treated group, the numbers of swollen mitochondria and mitochondrial autophagosomes were significantly higher. Finally, we showed that rapamycin can upregulate the expression of VDAC1, PINK1, Parkin, and LC3 in glomerular podocytes, which is correlated with mitophagy. VDAC1 is involved in mitophagy and is related to the PINK1/Parkin signaling pathway, serving as an indicator of mitophagy in podocytes.
Collapse
Affiliation(s)
- Shengyou Yu
- Department of Pediatrics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong Province, China
- Department of Pediatrics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P. R. China
| | - Weixue Zhu
- Department of Pediatrics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong Province, China
| | - Li Yu
- Department of Pediatrics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong Province, China
- Department of Pediatrics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, P. R. China
| |
Collapse
|
11
|
Meng X, Mao H, Wan M, Lu L, Chen Z, Zhang L. Mitochondrial homeostasis in odontoblast: Physiology, pathogenesis and targeting strategies. Life Sci 2024; 352:122797. [PMID: 38917871 DOI: 10.1016/j.lfs.2024.122797] [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: 02/17/2024] [Revised: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024]
Abstract
Caries and pulpitis remain a major global disease burden and affect the quality of life of patients. Odontoblasts are key players in the progression of caries and pulpitis, not only secreting and mineralizing to form dentin, but also acting as a wall of defense to initiate immune defenses. Mitochondrion is an information processor for numerous cellular activities, and dysregulation of mitochondrion homeostasis not only affects cellular metabolism but also triggers a wide range of diseases. Elucidating mitochondrial homeostasis in odontoblasts can help deepen scholars' understanding of odontoblast-associated diseases. Articles on mitochondrial homeostasis in odontoblasts were evaluated for information pertinent to include in this narrative review. This narrative review focused on understanding the complex interplay between mitochondrial homeostasis in odontoblasts under physiological and pathological conditions. Furthermore, mitochondria-centered therapeutic strategies (including mitochondrial base editing, targeting platforms, and mitochondrial transplantation) were emphasized by resolving key genes that regulate mitochondrial function. Mitochondria are involved in odontoblast differentiation and function, and act as mitochondrial danger-associated molecular patterns (mtDAMPs) to mediate odontoblast pathological progression. Novel mitochondria-centered therapeutic strategies are particularly attractive as emerging therapeutic approaches for the maintenance of mitochondrial homeostasis. It is expected to probe key events of odontoblast differentiation and advance the clinical resolution of dentin formation and mineralization disorders and odontoblast-related diseases.
Collapse
Affiliation(s)
- Xiang Meng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Hanqing Mao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Minting Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Linxin Lu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Endodontics, School and Hospital of Stomatology, Wuhan University, HongShan District, LuoYu Road No. 237, Wuhan 430079, China.
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Endodontics, School and Hospital of Stomatology, Wuhan University, HongShan District, LuoYu Road No. 237, Wuhan 430079, China.
| |
Collapse
|
12
|
Sun B, Ding P, Song Y, Zhou J, Chen X, Peng C, Liu S. FDX1 downregulation activates mitophagy and the PI3K/AKT signaling pathway to promote hepatocellular carcinoma progression by inducing ROS production. Redox Biol 2024; 75:103302. [PMID: 39128228 PMCID: PMC11366913 DOI: 10.1016/j.redox.2024.103302] [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] [Revised: 07/16/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Mitochondrial dysfunction and metabolic reprogramming can lead to the development and progression of hepatocellular carcinoma (HCC). Ferredoxin 1 (FDX1) is a small mitochondrial protein and recent studies have shown that FDX1 plays an important role in tumor cuproptosis, but its role in HCC is still elusive. In this study, we aim to investigate the expression and novel functions of FDX1 in HCC. METHODS FDX1 expression was first analyzed in publicly available datasets and verified by immunohistochemistry, qRT-PCR and Western blot. In vitro and in vivo experiments were applied to explore the functions of FDX1. Non-targeted metabolomics and RNA-sequencing were used to determine molecular mechanism. mRFP-GFP-LC3 lentivirus transfection, Mito-Tracker Red and Lyso-Tracker Green staining, transmission electron microscopy, flow cytometry, JC-1 staining, etc. were used to analyze mitophagy or ROS levels. Hydrodynamic tail vein injection (HTVi) and patient-derived organoid (PDO) models were used to analyze effect of FDX1 overexpression. RESULTS FDX1 expression is significantly downregulated in HCC tissues. FDX1 downregulation promotes HCC cell proliferation, invasion in vitro and growth, metastasis in vivo. In addition, FDX1 affects metabolism of HCC cells and is associated with autophagy. We then confirmed that FDX1 deficiency increases ROS levels, activates mitophagy and the PI3K/AKT signaling pathway in HCC cells. Interestingly, scavenging ROS attenuates the tumor-promoting role and mitophagy of FDX1 downregulation. The results of HTVi and PDO models both find that FDX1 elevation significantly inhibits HCC progression. Moreover, low FDX1 expression is associated with shorter survival and is an independent risk factor for prognosis in HCC patients. CONCLUSIONS Our research had investigated novel functions of FDX1 in HCC. Downregulation of FDX1 contributes to metabolic reprogramming and leads to ROS-mediated activation of mitophagy and the PI3K/AKT signaling pathway. FDX1 is a potential prognostic biomarker and increasing FDX1 expression may be a potential therapeutic approach to inhibit HCC progression.
Collapse
Affiliation(s)
- Bo Sun
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China; Hunan Engineering Research Center of Digital Hepatobiliary Medicine, Changsha, 410005, China; Hunan Key Laboratory for the Prevention and Treatment of Biliary Tract Diseases, Changsha, 410005, China
| | - Peng Ding
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Yinghui Song
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Jia Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Xu Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Chuang Peng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China; Hunan Key Laboratory for the Prevention and Treatment of Biliary Tract Diseases, Changsha, 410005, China.
| | - Sulai Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China; Hunan Engineering Research Center of Digital Hepatobiliary Medicine, Changsha, 410005, China; Hunan Key Laboratory for the Prevention and Treatment of Biliary Tract Diseases, Changsha, 410005, China.
| |
Collapse
|
13
|
Liu JY, Liu JX, Li R, Zhang ZQ, Zhang XH, Xing SJ, Sui BD, Jin F, Ma B, Zheng CX. AMPK, a hub for the microenvironmental regulation of bone homeostasis and diseases. J Cell Physiol 2024:e31393. [PMID: 39210747 DOI: 10.1002/jcp.31393] [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: 03/19/2024] [Revised: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
AMP-activated protein kinase (AMPK), a crucial regulatory kinase, monitors energy levels, conserving ATP and boosting synthesis in low-nutrition, low-energy states. Its sensitivity links microenvironmental changes to cellular responses. As the primary support structure and endocrine organ, the maintenance, and repair of bones are closely associated with the microenvironment. While a series of studies have explored the effects of specific microenvironments on bone, there is lack of angles to comprehensively evaluate the interactions between microenvironment and bone cells, especially for bone marrow mesenchymal stem cells (BMMSCs) which mediate the differentiation of osteogenic lineage. It is noteworthy that accumulating evidence has indicated that AMPK may serve as a hub between BMMSCs and microenvironment factors, thus providing a new perspective for us to understand the biology and pathophysiology of stem cells and bone. In this review, we emphasize AMPK's pivotal role in bone microenvironment modulation via ATP, inflammation, reactive oxygen species (ROS), calcium, and glucose, particularly in BMMSCs. We further explore the use of AMPK-activating drugs in the context of osteoarthritis and osteoporosis. Moreover, building upon the foundation of AMPK, we elucidate a viewpoint that facilitates a comprehensive understanding of the dynamic relationship between the microenvironment and bone homeostasis, offering valuable insights for prospective investigations into stem cell biology and the treatment of bone diseases.
Collapse
Affiliation(s)
- Jin-Yu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Jie-Xi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Rang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Zi-Qi Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xiao-Hui Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Shu-Juan Xing
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
- College of Life Science, Northwest University, Xi'an, China
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Fang Jin
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Bo Ma
- State Key Laboratory of National Security Specially Needed Medicines, Academy of Military Medical Sciences, Beijing, China
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
14
|
Li J, Wang T, Hou X, Li Y, Zhang J, Bai W, Qian H, Sun Z. Extracellular vesicles: opening up a new perspective for the diagnosis and treatment of mitochondrial dysfunction. J Nanobiotechnology 2024; 22:487. [PMID: 39143493 PMCID: PMC11323404 DOI: 10.1186/s12951-024-02750-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
Mitochondria are crucial organelles responsible for energy generation in eukaryotic cells. Oxidative stress, calcium disorders, and mitochondrial DNA abnormalities can all cause mitochondrial dysfunction. It is now well documented that mitochondrial dysfunction significantly contributes to the pathogenesis of numerous illnesses. Hence, it is vital to investigate innovative treatment methods targeting mitochondrial dysfunction. Extracellular vesicles (EVs) are cell-derived nanovesicles that serve as intercellular messengers and are classified into small EVs (sEVs, < 200 nm) and large EVs (lEVs, > 200 nm) based on their sizes. It is worth noting that certain subtypes of EVs are rich in mitochondrial components (even structurally intact mitochondria) and possess the ability to transfer them or other contents including proteins and nucleic acids to recipient cells to modulate their mitochondrial function. Specifically, EVs can modulate target cell mitochondrial homeostasis as well as mitochondria-controlled apoptosis and ROS generation by delivering relevant substances. In addition, the artificial modification of EVs as delivery carriers for therapeutic goods targeting mitochondria is also a current research hotspot. In this article, we will focus on the ability of EVs to modulate the mitochondrial function of target cells, aiming to offer novel perspectives on therapeutic approaches for diverse conditions linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jiali Li
- Department of Gerontology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Tangrong Wang
- Department of Gerontology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaomei Hou
- The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, 450000, China
| | - Yu Li
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jiaxin Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Wenhuan Bai
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Zixuan Sun
- Department of Gerontology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
15
|
Toledano JM, Puche-Juarez M, Galvez-Navas JM, Moreno-Fernandez J, Diaz-Castro J, Ochoa JJ. Pregnancy Disorders: A Potential Role for Mitochondrial Altered Homeostasis. Antioxidants (Basel) 2024; 13:979. [PMID: 39199225 PMCID: PMC11351112 DOI: 10.3390/antiox13080979] [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: 06/14/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 09/01/2024] Open
Abstract
Pregnancy is a complex and challenging process associated with physiological changes whose objective is to adapt the maternal organism to the increasing energetic requirements due to embryo and fetal development. A failed adaptation to these demands may lead to pregnancy complications that threaten the health of both mothers and their offspring. Since mitochondria are the main organelle responsible for energy generation in the form of ATP, the adequate state of these organelles seems crucial for proper pregnancy development and healthy pregnancy outcomes. The homeostasis of these organelles depends on several aspects, including their content, biogenesis, energy production, oxidative stress, dynamics, and signaling functions, such as apoptosis, which can be modified in relation to diseases during pregnancy. The etiology of pregnancy disorders like preeclampsia, fetal growth restriction, and gestational diabetes mellitus is not yet well understood. Nevertheless, insufficient placental perfusion and oxygen transfer are characteristic of many of them, being associated with alterations in the previously cited different aspects of mitochondrial homeostasis. Therefore, and due to the capacity of these multifactorial organelles to respond to physiological and pathophysiological stimuli, it is of great importance to gather the currently available scientific information regarding the relationship between main pregnancy complications and mitochondrial alterations. According to this, the present review is intended to show clear insight into the possible implications of mitochondria in these disorders, thus providing relevant information for further investigation in relation to the investigation and management of pregnancy diseases.
Collapse
Affiliation(s)
- Juan M. Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - María Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Jose Maria Galvez-Navas
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain;
- Cáncer Registry of Granada, Andalusian School of Public Health, Cuesta del Observatorio 4, Campus Universitario de Cartuja, 18011 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| |
Collapse
|
16
|
Pan B, Li Y, Han H, Zhang L, Hu X, Pan Y, Peng Z. FoxG1/BNIP3 axis promotes mitophagy and blunts cisplatin resistance in osteosarcoma. Cancer Sci 2024; 115:2565-2577. [PMID: 38932521 PMCID: PMC11309937 DOI: 10.1111/cas.16242] [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: 03/13/2024] [Revised: 05/19/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Cisplatin (CDDP) is a commonly used chemotherapeutic for osteosarcoma (OS) patients, and drug resistance remains as a major hurdle to undermine the treatment outcome. Here, we investigated the potential involvement of FoxG1 and BNIP3 in CDDP resistance of OS cells. FoxG1 and BNIP3 expression levels were detected in the CDDP-sensitive and CDDP-resistant OS tumors and cell lines. Mitophagy was observed through transmission electron microscope analysis. The sensitivity to CDDP in OS cells upon FoxG1 overexpression was examined in cell and animal models. We found that FoxG1 and BNIP3 showed significant downregulation in the CDDP-resistant OS tumor samples and cell lines. CDDP-resistant OS tumor specimens and cells displayed impaired mitophagy. FoxG1 overexpression promoted BNIP3 expression, enhanced mitophagy in CDDP-resistant OS cells, and resensitized the resistant cells to CDDP treatment in vitro and in vivo. Our data highlighted the role of the FoxG1/BNIP3 axis in regulating mitophagy and dictating CDDP resistance in OS cells, suggesting targeting FoxG1/BNIP3-dependent mitophagy as a potential strategy to overcome CDDP resistance in OS.
Collapse
Affiliation(s)
- Baolong Pan
- Health Examination CenterSixth Affiliated Hospital of Kunming Medical UniversityYuxiYunnanChina
| | - Yan Li
- Department of NeuroendocrineYuxi Children's HospitalYuxiYunnanChina
| | - Huiyun Han
- Department of PharmacyKunming Children's HospitalKunmingYunnanChina
| | - Lu Zhang
- Department of Asset ManagementThird Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Xuemei Hu
- Health Examination CenterSixth Affiliated Hospital of Kunming Medical UniversityYuxiYunnanChina
| | - Yanyu Pan
- College of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Zhuohui Peng
- Second Department of OrthopedicsThird Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
17
|
Chen Y, Huang J, Li Y, Chen Y, Gong Z, Xu M, Ma Y, Hu D, Peng X, Xu G, Cai S, Liu L, Zhao W, Zhao H. Bongkrekic acid alleviates airway inflammation via breaking the mPTP/mtDAMPs/RAGE feedback loop in a steroid-insensitive asthma model. Biomed Pharmacother 2024; 177:117111. [PMID: 39013220 DOI: 10.1016/j.biopha.2024.117111] [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: 04/11/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/18/2024] Open
Abstract
Mitochondrial dysfunction is critical in the pathogenesis of asthma. Mitochondrial permeability transition pore (mPTP) regulates the release of mitochondrial damage-associated molecular patterns (mtDAMPs) to maintain mitochondrial homeostasis. Bongkrekic acid (BKA) is a highly selective inhibitor of mPTP opening, participates the progression of various diseases. This research investigated the exact roles of BKA and mPTP in the pathogenesis of asthma and elucidated its underlying mechanisms. In the present study, cytochrome c, one of the mtDAMPs, levels were elevated in asthmatic patients, and associated to airway inflammation and airway obstruction. BKA, the inhibitor of mPTP markedly reversed TDI-induced airway hyperresponsiveness, airway inflammation, and mitochondrial dysfunction. Pretreatment with mitochondrial precipitation, to simulate the release of mtDAMPs, further increased TDI-induced airway inflammation and the expression of RAGE in mice. Administration of the inhibitor of RAGE, FPS-ZM1, alleviated the airway inflammation, the abnormal open of mPTP and mitochondrial dysfunction induced by mtDAMPs and TDI. Furthermore, stimulation with different mtDAMPs activated RAGE signaling in human bronchial epithelial cells. Accordingly, our study indicated that mPTP was important and BKA was efficient in alleviating inflammation in TDI-induced asthma. A positive feedback loop involving mPTP, mtDAMPs and RAGE was present in TDI-induced asthma, indicating that mPTP might serve as a potential therapeutic target for asthma.
Collapse
Affiliation(s)
- Ying Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junwen Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuemao Li
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yaoxin Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhaoqian Gong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Maosheng Xu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanyan Ma
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dapeng Hu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xianru Peng
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guilin Xu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Laiyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenqu Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Haijin Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
18
|
Yang J, Zhao H, Qu S. Phytochemicals targeting mitophagy: Therapeutic opportunities and prospects for treating Alzheimer's disease. Biomed Pharmacother 2024; 177:117144. [PMID: 39004063 DOI: 10.1016/j.biopha.2024.117144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder and the leading cause of age-related cognitive decline. Recent studies have established a close relationship between mitophagy and the pathogenesis of AD. Various phytochemicals have shown promising therapeutic effects in mitigating the onset and progression of AD. This review offers a comprehensive overview of the typical features of mitophagy and the underlying mechanisms leading to its occurrence in AD, highlighting its significance in the disease's pathogenesis and progression. Additionally, we examine the therapeutic mechanisms of synthetic drugs that induce mitophagy in AD. Finally, we summarize recent advances in research on phytochemicals that regulate mitophagy in the treatment of AD, potentially guiding the development of new anti-AD drugs.
Collapse
Affiliation(s)
- Jing Yang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China.
| | - He Zhao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China.
| | - Shengtao Qu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China.
| |
Collapse
|
19
|
Yu Y, Jiang Y, Glandorff C, Sun M. Exploring the mystery of tumor metabolism: Warburg effect and mitochondrial metabolism fighting side by side. Cell Signal 2024; 120:111239. [PMID: 38815642 DOI: 10.1016/j.cellsig.2024.111239] [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: 05/06/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The metabolic reconfiguration of tumor cells constitutes a pivotal aspect of tumor proliferation and advancement. This study delves into two primary facets of tumor metabolism: the Warburg effect and mitochondrial metabolism, elucidating their contributions to tumor dominance. The Warburg effect facilitates efficient energy acquisition by tumor cells through aerobic glycolysis and lactic acid fermentation, offering metabolic advantages conducive to growth and proliferation. Simultaneously, mitochondrial metabolism, serving as the linchpin of sustained tumor vitality, orchestrates the tricarboxylic acid cycle and electron transport chain, furnishing a steadfast and dependable wellspring of biosynthesis for tumor cells. Regarding targeted therapy, this discourse examines extant strategies targeting tumor glycolysis and mitochondrial metabolism, underscoring their potential efficacy in modulating tumor metabolism while envisaging future research trajectories and treatment paradigms in the realm of tumor metabolism. By means of a thorough exploration of tumor metabolism, this study aspires to furnish crucial insights into the regulation of tumor metabolic processes, thereby furnishing valuable guidance for the development of novel therapeutic modalities. This comprehensive deliberation is poised to catalyze advancements in tumor metabolism research and offer novel perspectives and pathways for the formulation of cancer treatment strategies in the times ahead.
Collapse
Affiliation(s)
- Yongxin Yu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yulang Jiang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Christian Glandorff
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; University Clinic of Hamburg at the HanseMerkur Center of TCM, Hamburg, Germany
| | - Mingyu Sun
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
20
|
Forte M, D'Ambrosio L, Schiattarella GG, Salerno N, Perrone MA, Loffredo FS, Bertero E, Pilichou K, Manno G, Valenti V, Spadafora L, Bernardi M, Simeone B, Sarto G, Frati G, Perrino C, Sciarretta S. Mitophagy modulation for the treatment of cardiovascular diseases. Eur J Clin Invest 2024; 54:e14199. [PMID: 38530070 DOI: 10.1111/eci.14199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Defects of mitophagy, the selective form of autophagy for mitochondria, are commonly observed in several cardiovascular diseases and represent the main cause of mitochondrial dysfunction. For this reason, mitophagy has emerged as a novel and potential therapeutic target. METHODS In this review, we discuss current evidence about the biological significance of mitophagy in relevant preclinical models of cardiac and vascular diseases, such as heart failure, ischemia/reperfusion injury, metabolic cardiomyopathy and atherosclerosis. RESULTS Multiple studies have shown that cardiac and vascular mitophagy is an adaptive mechanism in response to stress, contributing to cardiovascular homeostasis. Mitophagy defects lead to cell death, ultimately impairing cardiac and vascular function, whereas restoration of mitophagy by specific compounds delays disease progression. CONCLUSIONS Despite previous efforts, the molecular mechanisms underlying mitophagy activation in response to stress are not fully characterized. A comprehensive understanding of different forms of mitophagy active in the cardiovascular system is extremely important for the development of new drugs targeting this process. Human studies evaluating mitophagy abnormalities in patients at high cardiovascular risk also represent a future challenge.
Collapse
Affiliation(s)
| | - Luca D'Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nadia Salerno
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francesco S Loffredo
- Division of Cardiology, Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino-Italian IRCCS Cardiology Network, Genoa, Italy
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Valenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- ICOT Istituto Marco Pasquali, Latina, Italy
| | | | - Marco Bernardi
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | | | | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
21
|
Dai C, Zeng X, Zhang X, Liu Z, Cheng S. Machine learning-based integration develops a mitophagy-related lncRNA signature for predicting the progression of prostate cancer: a bioinformatic analysis. Discov Oncol 2024; 15:316. [PMID: 39073679 PMCID: PMC11286916 DOI: 10.1007/s12672-024-01189-5] [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: 05/26/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
Prostate cancer remains a complex and challenging disease, necessitating innovative approaches for prognosis and therapeutic guidance. This study integrates machine learning techniques to develop a novel mitophagy-related long non-coding RNA (lncRNA) signature for predicting the progression of prostate cancer. Leveraging the TCGA-PRAD dataset, we identify a set of four key lncRNAs and formulate a riskscore, revealing its potential as a prognostic indicator. Subsequent analyses unravel the intricate connections between riskscore, immune cell infiltration, mutational landscapes, and treatment outcomes. Notably, the pan-cancer exploration of YEATS2-AS1 highlights its pervasive impact, demonstrating elevated expression across various malignancies. Furthermore, drug sensitivity predictions based on riskscore guide personalized chemotherapy strategies, with drugs like Carmustine and Entinostat showing distinct suitability for high and low-risk group patients. Regression analysis exposes significant correlations between the mitophagy-related lncRNAs, riskscore, and key mitophagy-related genes. Molecular docking analyses reveal promising interactions between Cyclophosphamide and proteins encoded by these genes, suggesting potential therapeutic avenues. This comprehensive study not only introduces a robust prognostic tool but also provides valuable insights into the molecular intricacies and potential therapeutic interventions in prostate cancer, paving the way for more personalized and effective clinical approaches.
Collapse
Affiliation(s)
- Caixia Dai
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xiangju Zeng
- Department of Outpatient, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xiuhong Zhang
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Ziqi Liu
- Department of Acupuncture and Moxibustion, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shunhua Cheng
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
22
|
Sun Y, Li G, Zhang H, Xie M. Knockdown of CPSF4 Inhibits Bladder Cancer Cell Growth by Upregulating NRF1. Biochem Genet 2024:10.1007/s10528-024-10891-6. [PMID: 39039322 DOI: 10.1007/s10528-024-10891-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 03/15/2024] [Indexed: 07/24/2024]
Abstract
Increasing studies have shown that nuclear respiratory factor 1 (NRF1) deficiency frequently occurs in many human diseases, and its activation can protect neurons and other cells from degenerative diseases and malignant tumors. However, how NRF1 is regulated in bladder cancer remains unknown. Our research aims to reveal the role of leavage and polyadenylation-specific factor 4 (CPSF4) on the growth inhibition effect of bladder cancer and clarify its relationship with NRF1. Here, cell proliferation assay, transwell migration assay and multicellular tumor spheroids (MCTS) formation assay in the bladder cancer cell lines were carried out to measure tumor cell growth. Western bolt assay was carried out to identify the relationship between NRF1 and CPSF4. Also, subcutaneous xenograft tumors in nude mice were established to further validate the inhibition effect of CPSF4 on bladder tumor and the regulation on NRF1. The results in vitro showed that knockdown of CPSF4 strongly reduced the proliferation and migration, and inhibited MCTS formation in 5637 and HT1376 cell lines, while an additional knockdown of increased NRF1 induced by CPSF4 knockdown partially abolished these effects. The results in vivo showed that knockdown of CPSF4 strongly reduced the volume and weight of subcutaneous tumor, and decreased the expression of Ki-67 in tumor tissue, while NRF1 knockdown partially reversed these effects induced by CPSF4 knockdown. Western bolt assay demonstrated that CPSF4 could negatively regulate NRF1. Our results indicated that knock-down of CPSF4 inhibited bladder cancer cell growth by upregulating NRF1, which might provide evidence of CPSF4 as a therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Yixiang Sun
- Department of Urology, Yantai Yuhuangding Hospital, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Guanglei Li
- Department of Urology, Yantai Yeda Hospital, No. 23-1, Huanghe Road, Economic and Technological Development Area, Yantai, 264000, Shandong, China
| | - Hanlin Zhang
- Department of Urology, Yantai Muping District Hospital of Traditional Chinese Medicine, No. 505, Government Street, Muping District, Yantai, 264000, Shandong, China
| | - Mao Xie
- Department of Urology, Yantai Yuhuangding Hospital, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China.
| |
Collapse
|
23
|
Zhong FF, Wei B, Bao GX, Lou YP, Wei ME, Wang XY, Xiao X, Tian JJ. FABP3 Induces Mitochondrial Autophagy to Promote Neuronal Cell Apoptosis in Brain Ischemia-Reperfusion Injury. Neurotox Res 2024; 42:35. [PMID: 39008165 DOI: 10.1007/s12640-024-00712-4] [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: 01/15/2024] [Revised: 05/30/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
This study elucidates the molecular mechanisms by which FABP3 regulates neuronal apoptosis via mitochondrial autophagy in the context of cerebral ischemia-reperfusion (I/R). Employing a transient mouse model of middle cerebral artery occlusion (MCAO) established using the filament method, brain tissue samples were procured from I/R mice. High-throughput transcriptome sequencing on the Illumina CN500 platform was performed to identify differentially expressed mRNAs. Critical genes were selected by intersecting I/R-related genes from the GeneCards database with the differentially expressed mRNAs. The in vivo mechanism was explored by infecting I/R mice with lentivirus. Brain tissue injury, infarct volume ratio in the ischemic penumbra, neurologic deficits, behavioral abilities, neuronal apoptosis, apoptotic factors, inflammatory factors, and lipid peroxidation markers were assessed using H&E staining, TTC staining, Longa scoring, rotation experiments, immunofluorescence staining, and Western blot. For in vitro validation, an OGD/R model was established using primary neuron cells. Cell viability, apoptosis rate, mitochondrial oxidative stress, morphology, autophagosome formation, membrane potential, LC3 protein levels, and colocalization of autophagosomes and mitochondria were evaluated using MTT assay, LDH release assay, flow cytometry, ROS/MDA/GSH-Px measurement, transmission electron microscopy, MitoTracker staining, JC-1 method, Western blot, and immunofluorescence staining. FABP3 was identified as a critical gene in I/R through integrated transcriptome sequencing and bioinformatics analysis. In vivo experiments revealed that FABP3 silencing mitigated brain tissue damage, reduced infarct volume ratio, improved neurologic deficits, restored behavioral abilities, and attenuated neuronal apoptosis, inflammation, and mitochondrial oxidative stress in I/R mice. In vitro experiments demonstrated that FABP3 silencing restored OGD/R cell viability, reduced neuronal apoptosis, and decreased mitochondrial oxidative stress. Moreover, FABP3 induced mitochondrial autophagy through ROS, which was inhibited by the free radical scavenger NAC. Blocking mitochondrial autophagy with sh-ATG5 lentivirus confirmed that FABP3 induces mitochondrial dysfunction and neuronal apoptosis by activating mitochondrial autophagy. In conclusion, FABP3 activates mitochondrial autophagy through ROS, leading to mitochondrial dysfunction and neuronal apoptosis, thereby promoting cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Fang-Fang Zhong
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China.
| | - Bo Wei
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Guo-Xiang Bao
- Department of Clinical Laboratory Center, Shaoxing People's Hospital, Shaoxing, China
| | - Yi-Ping Lou
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Ming-Er Wei
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Xin-Yue Wang
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Xiao Xiao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Jin-Jin Tian
- School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
24
|
Wang S, Liu J, Zhou L, Xu H, Zhang D, Zhang X, Wang Q, Zhou Q. Research progresses on mitochondrial-targeted biomaterials for bone defect repair. Regen Biomater 2024; 11:rbae082. [PMID: 39055307 PMCID: PMC11272180 DOI: 10.1093/rb/rbae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/23/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
In recent years, the regulation of the cell microenvironment has opened up new avenues for bone defect repair. Researchers have developed novel biomaterials to influence the behavior of osteoblasts and immune cells by regulating the microenvironment, aiming to achieve efficient bone repair. Mitochondria, as crucial organelles involved in energy conversion, biosynthesis and signal transduction, play a vital role in maintaining bone integrity. Dysfunction of mitochondria can have detrimental effects on the transformation of the immune microenvironment and the differentiation of stem cells, thereby hindering bone tissue regeneration. Consequently, targeted therapy strategies focusing on mitochondria have emerged. This approach offers a wide range of applications and reliable therapeutic effects, thereby providing a new treatment option for complex and refractory bone defect diseases. In recent studies, more biomaterials have been used to restore mitochondrial function and promote positive cell differentiation. The main directions are mitochondrial energy metabolism, mitochondrial biogenesis and mitochondrial quality control. In this review, we investigated the biomaterials used for mitochondria-targeted treatment of bone defect repair in recent years from the perspective of progress and strategies. We also summarized the micro-molecular mechanisms affected by them. Through discussions on energy metabolism, oxidative stress regulation and autophagy regulation, we emphasized the opportunities and challenges faced by mitochondria-targeted biomaterials, providing vital clues for developing a new generation of bone repair materials.
Collapse
Affiliation(s)
- Shuze Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Jialin Liu
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Linxi Zhou
- Department of Orthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- National Center for Stomatology, Shanghai 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Hao Xu
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Dan Zhang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Xing Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Qing Zhou
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| |
Collapse
|
25
|
Li Y, Wei X, Xiao R, Chen Y, Xiong T, Fang ZM, Huo B, Guo X, Luo H, Wu X, Liu L, Zhu XH, Hu Q, Jiang DS, Yi X. SMYD2-Methylated PPARγ Facilitates Hypoxia-Induced Pulmonary Hypertension by Activating Mitophagy. Circ Res 2024; 135:93-109. [PMID: 38770649 DOI: 10.1161/circresaha.124.323698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Hyperproliferation of pulmonary arterial smooth muscle cells (PASMCs) and consequent pulmonary vascular remodeling are the crucial pathological features of pulmonary hypertension (PH). Protein methylation has been shown to be critically involved in PASMC proliferation and PH, but the underlying mechanism remains largely unknown. METHODS PH animal models were generated by treating mice/rats with chronic hypoxia for 4 weeks. SMYD2-vTg mice (vascular smooth muscle cell-specific suppressor of variegation, enhancer of zeste, trithorax and myeloid Nervy DEAF-1 (deformed epidural auto-regulatory factor-1) domain-containing protein 2 transgenic) or wild-type rats and mice treated with LLY-507 (3-cyano-5-{2-[4-[2-(3-methylindol-1-yl)ethyl]piperazin-1-yl]-phenyl}-N-[(3-pyrrolidin-1-yl)propyl]benzamide) were used to investigate the function of SMYD2 (suppressor of variegation, enhancer of zeste, trithorax and myeloid Nervy DEAF-1 domain-containing protein 2) on PH development in vivo. Primary cultured rat PASMCs with SMYD2 knockdown or overexpression were used to explore the effects of SMYD2 on proliferation and to decipher the underlying mechanism. RESULTS We demonstrated that the expression of the lysine methyltransferase SMYD2 was upregulated in the smooth muscle cells of pulmonary arteries from patients with PH and hypoxia-exposed rats/mice and in the cytoplasm of hypoxia-induced rat PASMCs. More importantly, targeted inhibition of SMYD2 by LLY-507 significantly attenuated hypoxia-induced pulmonary vascular remodeling and PH development in both male and female rats in vivo and reduced rat PASMC hyperproliferation in vitro. In contrast, SMYD2-vTg mice exhibited more severe PH phenotypes and related pathological changes than nontransgenic mice after 4 weeks of chronic hypoxia treatment. Furthermore, SMYD2 overexpression promoted, while SMYD2 knockdown suppressed, the proliferation of rat PASMCs by affecting the cell cycle checkpoint between S and G2 phases. Mechanistically, we revealed that SMYD2 directly interacted with and monomethylated PPARγ (peroxisome proliferator-activated receptor gamma) to inhibit the nuclear translocation and transcriptional activity of PPARγ, which further promoted mitophagy to facilitate PASMC proliferation and PH development. Furthermore, rosiglitazone, a PPARγ agonist, largely abolished the detrimental effects of SMYD2 overexpression on PASMC proliferation and PH. CONCLUSIONS Our results demonstrated that SMYD2 monomethylates nonhistone PPARγ and inhibits its nuclear translocation and activation to accelerate PASMC proliferation and PH by triggering mitophagy, indicating that targeting SMYD2 or activating PPARγ are potential strategies for the prevention of PH.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Rats
- Cell Proliferation
- Cells, Cultured
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/complications
- Hypoxia/metabolism
- Methylation
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitophagy
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- PPAR gamma/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- Rats, Sprague-Dawley
- Vascular Remodeling
Collapse
Affiliation(s)
- Yi Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
- Key Laboratory of Organ Transplantation, Ministry of Education (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- NHC Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
| | - Rui Xiao
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan (R.X., Q.H.)
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China (R.X., Q.H.)
| | - Yongjie Chen
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, China (Y.C.)
| | - Tianxin Xiong
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Ze-Min Fang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Bo Huo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Xian Guo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Hanshen Luo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Xingliang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, China (X. Wu, L.L., X.Y.)
| | - Liyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China (X. Wu, L.L., X.Y.)
| | - Xue-Hai Zhu
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
| | - Qinghua Hu
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan (R.X., Q.H.)
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China (R.X., Q.H.)
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.L., X. Wei, T.X., Z.-M.F., B.H., X.G., H.L., X.-H.Z., D.-S.J.)
- Key Laboratory of Organ Transplantation, Ministry of Education (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- NHC Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation (X. Wei, D.-S.J.), Chinese Academy of Medical Sciences, Wuhan, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, China (X. Wu, L.L., X.Y.)
| |
Collapse
|
26
|
Wu K, Shieh JS, Qin L, Guo JJ. Mitochondrial mechanisms in the pathogenesis of chronic inflammatory musculoskeletal disorders. Cell Biosci 2024; 14:76. [PMID: 38849951 PMCID: PMC11162051 DOI: 10.1186/s13578-024-01259-9] [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: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Chronic inflammatory musculoskeletal disorders characterized by prolonged muscle inflammation, resulting in enduring pain and diminished functionality, pose significant challenges for the patients. Emerging scientific evidence points to mitochondrial malfunction as a pivotal factor contributing to these ailments. Mitochondria play a critical role in powering skeletal muscle activity, but in the context of persistent inflammation, disruptions in their quantity, configuration, and performance have been well-documented. Various disturbances, encompassing alterations in mitochondrial dynamics (such as fission and fusion), calcium regulation, oxidative stress, biogenesis, and the process of mitophagy, are believed to play a central role in the progression of these disorders. Additionally, unfolded protein responses and the accumulation of fatty acids within muscle cells may adversely affect the internal milieu, impairing the equilibrium of mitochondrial functioning. The structural discrepancies between different mitochondrial subsets namely, intramyofibrillar and subsarcolemmal mitochondria likely impact their metabolic capabilities and susceptibility to inflammatory influences. The release of signals from damaged mitochondria is known to incite inflammatory responses. Intriguingly, migrasomes and extracellular vesicles serve as vehicles for intercellular transfer of mitochondria, aiding in the removal of impaired mitochondria and regulation of inflammation. Viral infections have been implicated in inducing stress on mitochondria. Prolonged dysfunction of these vital organelles sustains oxidative harm, metabolic irregularities, and heightened cytokine release, impeding the body's ability to repair tissues. This review provides a comprehensive analysis of advancements in understanding changes in the intracellular environment, mitochondrial architecture and distribution, biogenesis, dynamics, autophagy, oxidative stress, cytokines associated with mitochondria, vesicular structures, and associated membranes in the context of chronic inflammatory musculoskeletal disorders. Strategies targeting key elements regulating mitochondrial quality exhibit promise in the restoration of mitochondrial function, alleviation of inflammation, and enhancement of overall outcomes.
Collapse
Affiliation(s)
- Kailun Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of Soochow University/Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People's Republic of China
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Ju-Sheng Shieh
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 11490, Taiwan
| | - Ling Qin
- Musculoskeletal Research Laboratory of the Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Jiong Jiong Guo
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.
- MOE China-Europe Sports Medicine Belt and Road Joint Laboratory, Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Kong L, Li S, Fu Y, Cai Q, Du X, Liang J, Ma T. Mitophagy in relation to chronic inflammation/ROS in aging. Mol Cell Biochem 2024:10.1007/s11010-024-05042-9. [PMID: 38834837 DOI: 10.1007/s11010-024-05042-9] [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: 04/07/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Various assaults on mitochondria occur during the human aging process, contributing to mitochondrial dysfunction. This mitochondrial dysfunction is intricately connected with aging and diseases associated with it. In vivo, the accumulation of defective mitochondria can precipitate inflammatory and oxidative stress, thereby accelerating aging. Mitophagy, an essential selective autophagy process, plays a crucial role in managing mitochondrial quality control and homeostasis. It is a highly specialized mechanism that systematically removes damaged or impaired mitochondria from cells, ensuring their optimal functioning and survival. By engaging in mitophagy, cells are able to maintain a balanced and stable environment, free from the potentially harmful effects of dysfunctional mitochondria. An ever-growing body of research highlights the significance of mitophagy in both aging and age-related diseases. Nonetheless, the association between mitophagy and inflammation or oxidative stress induced by mitochondrial dysfunction remains ambiguous. We review the fundamental mechanisms of mitophagy in this paper, delve into its relationship with age-related stress, and propose suggestions for future research directions.
Collapse
Affiliation(s)
- Liang Kong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Shuhao Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Yu Fu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Qinyun Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Xinyun Du
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
28
|
Halfon M, Tankeu AT, Ribi C. Mitochondrial Dysfunction in Systemic Lupus Erythematosus with a Focus on Lupus Nephritis. Int J Mol Sci 2024; 25:6162. [PMID: 38892349 PMCID: PMC11173067 DOI: 10.3390/ijms25116162] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease affecting mostly women of child-bearing age. Immune dysfunction in SLE results from disrupted apoptosis which lead to an unregulated interferon (IFN) stimulation and the production of autoantibodies, leading to immune complex formation, complement activation, and organ damage. Lupus nephritis (LN) is a common and severe complication of SLE, impacting approximately 30% to 40% of SLE patients. Recent studies have demonstrated an alteration in mitochondrial homeostasis in SLE patients. Mitochondrial dysfunction contributes significantly to SLE pathogenesis by enhancing type 1 IFN production through various pathways involving neutrophils, platelets, and T cells. Defective mitophagy, the process of clearing damaged mitochondria, exacerbates this cycle, leading to increased immune dysregulation. In this review, we aim to detail the physiopathological link between mitochondrial dysfunction and disease activity in SLE. Additionally, we will explore the potential role of mitochondria as biomarkers and therapeutic targets in SLE, with a specific focus on LN. In LN, mitochondrial abnormalities are observed in renal cells, correlating with disease progression and renal fibrosis. Studies exploring cell-free mitochondrial DNA as a biomarker in SLE and LN have shown promising but preliminary results, necessitating further validation and standardization. Therapeutically targeting mitochondrial dysfunction in SLE, using drugs like metformin or mTOR inhibitors, shows potential in modulating immune responses and improving clinical outcomes. The interplay between mitochondria, immune dysregulation, and renal involvement in SLE and LN underscores the need for comprehensive research and innovative therapeutic strategies. Understanding mitochondrial dynamics and their impact on immune responses offers promising avenues for developing personalized treatments and non-invasive biomarkers, ultimately improving outcomes for LN patients.
Collapse
Affiliation(s)
- Matthieu Halfon
- Transplantation Center, Lausanne University Hospital, Rue du Bugnon 44, CH-1010 Lausanne, Switzerland;
| | - Aurel T. Tankeu
- Transplantation Center, Lausanne University Hospital, Rue du Bugnon 44, CH-1010 Lausanne, Switzerland;
| | - Camillo Ribi
- Division of Immunology and Allergy, Lausanne University Hospital, CH-1010 Lausanne, Switzerland;
| |
Collapse
|
29
|
Kou L, Wang Y, Li J, Zou W, Jin Z, Yin S, Chi X, Sun Y, Wu J, Wang T, Xia Y. Mitochondria-lysosome-extracellular vesicles axis and nanotheranostics in neurodegenerative diseases. Exp Neurol 2024; 376:114757. [PMID: 38508481 DOI: 10.1016/j.expneurol.2024.114757] [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: 12/21/2023] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
The intricate functional interactions between mitochondria and lysosomes play a pivotal role in maintaining cellular homeostasis and proper cellular functions. This dynamic interplay involves the exchange of molecules and signaling, impacting cellular metabolism, mitophagy, organellar dynamics, and cellular responses to stress. Dysregulation of these processes has been implicated in various neurodegenerative diseases. Additionally, mitochondrial-lysosomal crosstalk regulates the exosome release in neurons and glial cells. Under stress conditions, neurons and glial cells exhibit mitochondrial dysfunction and a fragmented network, which further leads to lysosomal dysfunction, thereby inhibiting autophagic flux and enhancing exosome release. This comprehensive review synthesizes current knowledge on mitochondrial regulation of cell death, organelle dynamics, and vesicle trafficking, emphasizing their significant contributions to neurodegenerative diseases. Furthermore, we explore the emerging field of nanomedicine in the management of neurodegenerative diseases. The review provides readers with an insightful overview of nano strategies that are currently advancing the mitochondrial-lysosome-extracellular vesicle axis as a therapeutic approach for mitigating neurodegenerative diseases.
Collapse
Affiliation(s)
- Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
30
|
Sedraoui S, Leduc-Gaudet JP, Mayaki D, Moamer A, Huck L, Gouspillou G, Petrof BJ, Hussain S. Lack of compensatory mitophagy in skeletal muscles during sepsis. J Physiol 2024; 602:2823-2838. [PMID: 38748778 DOI: 10.1113/jp286216] [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: 01/02/2024] [Accepted: 05/02/2024] [Indexed: 06/15/2024] Open
Abstract
Skeletal muscle dysfunction is a major problem in critically ill patients suffering from sepsis. This condition is associated with mitochondrial dysfunction and increased autophagy in skeletal muscles. Autophagy is a proteolytic mechanism involved in eliminating dysfunctional cellular components, including mitochondria. The latter process, referred to as mitophagy, is essential for maintaining mitochondrial quality and skeletal muscle health. Recently, a fluorescent reporter system called mito-QC (i.e. mitochondrial quality control) was developed to specifically quantify mitophagy levels. In the present study, we used mito-QC transgenic mice and confocal microscopy to morphologically monitor mitophagy levels during sepsis. To induce sepsis, Mito-QC mice received Escherichia coli lipopolysaccharide (10 mg kg-1 i.p.) or phosphate-buffered saline and skeletal muscles (hindlimb and diaphragm) were excised 48 h later. In control groups, there was a negative correlation between the basal mitophagy level and overall muscle mitochondrial content. Sepsis increased general autophagy in both limb muscles and diaphragm but had no effect on mitophagy levels. Sepsis was associated with a downregulation of certain mitophagy receptors (Fundc1, Bcl2L13, Fkbp8 and Phbb2). The present study suggests that general autophagy and mitophagy can be dissociated from one another, and that the characteristic accumulation of damaged mitochondria in skeletal muscles under the condition of sepsis may reflect a failure of adequate compensatory mitophagy. KEY POINTS: There was a negative correlation between the basal level of skeletal muscle mitophagy and the mitochondrial content of individual muscles. Mitophagy levels in limb muscles and the diaphragm were unaffected by lipopolysaccharide (LPS)-induced sepsis. With the exception of BNIP3 in sepsis, LPS administration induced either no change or a downregulation of mitophagy receptors in skeletal muscles.
Collapse
Affiliation(s)
- Sami Sedraoui
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Jean-Philippe Leduc-Gaudet
- Department of Medical Biology, Faculty of Health Sciences, Université du Québec à Trois-Rivieres, Trois-Rivieres, QC, Canada
| | - Dominique Mayaki
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Alaa Moamer
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Laurent Huck
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Gilles Gouspillou
- Département des Sciences de l'Activité Physique, Faculté des Sciences, Université du Québec à Montréal, Montréal, QC, Canada
| | - Basil J Petrof
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Sabah Hussain
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
- Department of Critical Care Medicine, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
31
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
32
|
Lv Y, Yu Z, Zhang P, Zhang X, Li H, Liang T, Guo Y, Cheng L, Peng F. The structure and function of FUN14 domain-containing protein 1 and its contribution to cardioprotection by mediating mitophagy. Front Pharmacol 2024; 15:1389953. [PMID: 38828457 PMCID: PMC11140143 DOI: 10.3389/fphar.2024.1389953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Cardiovascular disease (CVD) is a serious public health risk, and prevention and treatment efforts are urgently needed. Effective preventive and therapeutic programs for cardiovascular disease are still lacking, as the causes of CVD are varied and may be the result of a multifactorial combination. Mitophagy is a form of cell-selective autophagy, and there is increasing evidence that mitophagy is involved in cardioprotective processes. Recently, many studies have shown that FUN14 domain-containing protein 1 (FUNDC1) levels and phosphorylation status are highly associated with many diseases, including heart disease. Here, we review the structure and functions of FUNDC1 and the path-ways of its mediated mitophagy, and show that mitophagy can be effectively activated by dephosphorylation of Ser13 and Tyr18 sites, phosphorylation of Ser17 site and ubiquitination of Lys119 site in FUNDC1. By effectively activating or inhibiting excessive mitophagy, the quality of mitochondria can be effectively controlled. The main reason is that, on the one hand, improper clearance of mitochondria and accumulation of damaged mitochondria are avoided, and on the other hand, excessive mitophagy causing apoptosis is avoided, both serving to protect the heart. In addition, we explore the possible mechanisms by which FUNDC1-mediated mitophagy is involved in exercise preconditioning (EP) for cardioprotection. Finally, we also point out unresolved issues in FUNDC1 and its mediated mitophagy and give directions where further research may be needed.
Collapse
Affiliation(s)
- Yuhu Lv
- College of Physical Education, Guangdong University of Education, Guangzhou, China
- Research Center for Adolescent Sports and Health Promotion of Guangdong Province, Guangzhou, China
| | - Zhengze Yu
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Peiwen Zhang
- College of Nursing and Rehabilitation, Xi an FanYi University, Xi’an, China
| | - Xiqian Zhang
- College of Physical Education, Guangdong University of Education, Guangzhou, China
- Research Center for Adolescent Sports and Health Promotion of Guangdong Province, Guangzhou, China
| | - Huarui Li
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Ting Liang
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Yanju Guo
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Lin Cheng
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Fenglin Peng
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| |
Collapse
|
33
|
Zhang P, Zhu L, Pan X. A comprehensive analysis of the oncogenic and prognostic role of TBC1Ds in human hepatocellular carcinoma. PeerJ 2024; 12:e17362. [PMID: 38766486 PMCID: PMC11100476 DOI: 10.7717/peerj.17362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
Backgrounds TBC1D family members (TBC1Ds) are a group of proteins that contain the Tre2-Bub2-Cdc16 (TBC) domain. Recent studies have shown that TBC1Ds are involved in tumor growth, but no analysis has been done of expression patterns and prognostic values of TBC1Ds in hepatocellular carcinoma (HCC). Methods The expression levels of TBC1Ds were evaluated in HCC using the TIMER, UALCN and Protein Atlas databases. The correlation between the mRNA levels of TBC1Ds and the prognosis of patients with HCC in the GEPIA database was then analyzed. An enrichment analysis then revealed genes that potentially interact with TBC1Ds. The correlation between levels of TBC1Ds and tumor-infiltrating immune cells (TIICs) in HCC were studied using the TIMER 2.0 database. Finally, a series of in vitro assays verified the role of TBC1Ds in HCC progression. Results This study revealed the upregulated expression of TBC1Ds in HCC and the strong positive correlation between the mRNA levels of TBC1Ds and poor prognosis of patients with HCC. The functions of TBC1Ds were mainly related to autophagy and the AMPK pathway. There was also a significant correlation between level of TBC1Ds and tumor-infiltrating immune cells (TIICs) in HCC. The promoting role of TBC1Ds in HCC progression was verified in vitro assays. Conclusion The results of this analysis indicate that TBC1Ds may serve as new biomarkers for early diagnosis and treatment of HCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- Prognosis
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Autophagy/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Cell Line, Tumor
Collapse
Affiliation(s)
- Pei Zhang
- The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Zhu
- The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaodong Pan
- The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
34
|
Li J, Ma Y, Qiu T, Wang J, Zhang J, Sun X, Jiang L, Li Q, Yao X. Autophagy-dependent lysosomal calcium overload and the ATP5B-regulated lysosomes-mitochondria calcium transmission induce liver insulin resistance under perfluorooctane sulfonate exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116318. [PMID: 38626609 DOI: 10.1016/j.ecoenv.2024.116318] [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: 09/26/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/18/2024]
Abstract
Perfluorooctane sulfonate (PFOS), an officially listed persistent organic pollutant, is a widely distributed perfluoroalkyl substance. Epidemiological studies have shown that PFOS is intimately linked to the occurrence of insulin resistance (IR). However, the detailed mechanism remains obscure. In previous studies, we found that mitochondrial calcium overload was concerned with hepatic IR induced by PFOS. In this study, we found that PFOS exposure noticeably raised lysosomal calcium in L-02 hepatocytes from 0.5 h. In the PFOS-cultured L-02 cells, inhibiting autophagy alleviated lysosomal calcium overload. Inhibition of mitochondrial calcium uptake aggravated the accumulation of lysosomal calcium, while inhibition of lysosomal calcium outflowing reversed PFOS-induced mitochondrial calcium overload and IR. Transient receptor potential mucolipin 1 (TRPML1), the calcium output channel of lysosomes, interacted with voltage-dependent anion channel 1 (VDAC1), the calcium intake channel of mitochondria, in the PFOS-cultured cells. Moreover, we found that ATP synthase F1 subunit beta (ATP5B) interacted with TRPML1 and VDAC1 in the L-02 cells and the liver of mice under PFOS exposure. Inhibiting ATP5B expression or restraining the ATP5B on the plasma membrane reduced the interplay between TRPML1 and VDAC1, reversed the mitochondrial calcium overload and deteriorated the lysosomal calcium accumulation in the PFOS-cultured cells. Our research unveils the molecular regulation of the calcium crosstalk between lysosomes and mitochondria, and explains PFOS-induced IR in the context of activated autophagy.
Collapse
Affiliation(s)
- Jixun Li
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Yu Ma
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Tianming Qiu
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Jianyu Wang
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Jingyuan Zhang
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Xiance Sun
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Liping Jiang
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Qiujuan Li
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China
| | - Xiaofeng Yao
- Occupation and Environment Health Department, Dalian Medical University, 9 West Lvshun South Road, Dalian, China.
| |
Collapse
|
35
|
Wu Q, Wang Z, Chen S, She X, Zhu S, Li P, Liu L, Zhao C, Li K, Liu A, Huang C, Chen Y, Hu F, Wang G, Hu J. USP26 promotes colorectal cancer tumorigenesis by restraining PRKN-mediated mitophagy. Oncogene 2024; 43:1581-1593. [PMID: 38565942 DOI: 10.1038/s41388-024-03009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Deubiquitinating enzymes (DUBs) are promising targets for cancer therapy because of their pivotal roles in various physiological and pathological processes. Among these, ubiquitin-specific peptidase 26 (USP26) is a protease with crucial regulatory functions. Our study sheds light on the upregulation of USP26 in colorectal cancer (CRC), in which its increased expression correlates with an unfavorable prognosis. Herein, we evidenced the role of USP26 in promoting CRC tumorigenesis in a parkin RBR E3 ubiquitin-protein ligase (PRKN) protein-dependent manner. Our investigation revealed that USP26 directly interacted with PRKN protein, facilitating its deubiquitination, and subsequently reducing its activity. Additionally, we identified the K129 site on PRKN as a specific target for USP26-mediated deubiquitination. Our research highlights that a K-to-R mutation at the site on PRKN diminishes its potential for activation and ability to mediate mitophagy. In summary, our findings underscore the significance of USP26-mediated deubiquitination in restraining the activation of the PRKN-mediated mitophagy pathway, ultimately driving CRC tumorigenesis. This study not only elucidated the multifaceted role of USP26 in CRC but also introduced a promising avenue for therapeutic exploration through the development of small molecule inhibitors targeting USP26. This strategy holds promise as a novel therapeutic approach for CRC.
Collapse
Affiliation(s)
- Qi Wu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihong Wang
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Siqi Chen
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaowei She
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shengyu Zhu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pengcheng Li
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lang Liu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chongchong Zhao
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, 150001, China
| | - Kangdi Li
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Anyi Liu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Changsheng Huang
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaqi Chen
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fuqing Hu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guihua Wang
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Junbo Hu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
36
|
Dong Z, Yang B, Jia M, Yang C, Wang S, Mu H, Wang J. DDIT3/CHOP promotes LPS/ATP-induced pyroptosis in osteoblasts via mitophagy inhibition. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119712. [PMID: 38521466 DOI: 10.1016/j.bbamcr.2024.119712] [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: 09/20/2023] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Inflammatory environments can trigger endoplasmic reticulum (ER) stress and lead to pyroptosis in various tissues and cells, including liver, brain, and immune cells. As a key factor of ER stress, DNA damage-inducible transcript 3 (DDIT3)/CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) is upregulated in osteoblasts during inflammatory stimulation. DDIT3/CHOP may therefore regulate osteoblast pyroptosis in inflammatory conditions. During this investigation, we found that lipopolysaccharides (LPS)/adenosine 5'-triphosphate (ATP) stimulation in vitro induced osteoblasts to undergo pyroptosis, and the expression of DDIT3/CHOP was increased during this process. The overexpression of DDIT3/CHOP further promoted osteoblast pyroptosis as evidenced by the increased expression of the inflammasome NLR family pyrin domain containing 3 (NLRP3) and ratios of caspase-1 p20/caspase-1 and cleaved gasdermin D (GSDMD)/GSDMD. To explore the specific mechanism of this effect, we found through fluorescence imaging and Western blot analysis that LPS/ATP stimulation promoted PTEN-induced kinase 1 (PINK1)/E3 ubiquitin-protein ligase parkin (Parkin)-mediated mitophagy in osteoblasts, and this alteration was suppressed by the DDIT3/CHOP overexpression, resulting in increased ratio of pyroptosis compared with the control groups. The impact of DDIT3/CHOP on pyroptosis in osteoblasts was reversed by the application of carbonyl cyanide 3-chlorophenylhydrazone (CCCP), a specific mitophagy agonist. Therefore, our data demonstrated that DDIT3/CHOP promotes osteoblast pyroptosis by inhibiting PINK1/Parkin-mediated mitophagy in an inflammatory environment.
Collapse
Affiliation(s)
- Zhipeng Dong
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Beining Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Meie Jia
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Chang Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Shuo Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Hailin Mu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Jiawei Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China.
| |
Collapse
|
37
|
Ying X, Zheng X, Zhang X, Yin Y, Wang X. Kynurenine in IDO1 high cancer cell-derived extracellular vesicles promotes angiogenesis by inducing endothelial mitophagy in ovarian cancer. J Transl Med 2024; 22:267. [PMID: 38468343 PMCID: PMC10929174 DOI: 10.1186/s12967-024-05054-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Mitophagy, a prominent cellular homeostasis process, has been implicated in modulating endothelial cell function. Emerging evidence suggests that extracellular vesicles (EVs) participate in intercellular communication, which could modulate tumor angiogenesis, a hallmark of ovarian cancer (OC) progression. However, the underlying mechanisms through how EVs regulate endothelial mitophagy associated with tumor angiogenesis during OC development remain obscure. METHODS The effect of cancer cell-derived EVs on endothelial mitophagy and its correlation with tumor angiogenesis and OC development were explored by in vitro and in vivo experiments. Multi-omics integration analysis was employed to identify potential regulatory mechanisms of cancer cell-derived EVs on endothelial mitophagy, which is involved in tumor angiogenesis associated with OC development. These insights were then further corroborated through additional experiments. An orthotopic OC mouse model was constructed to assess the antiangiogenic and therapeutic potential of the Indoleamine 2,3 dioxygenase-1 (IDO1) inhibitor. RESULTS Cancer cell-derived EVs promoted tumor angiogenesis via the activation of endothelial mitophagy, contributing to the growth and metastasis of OC. The aberrantly high expression of IDO1 mediated abnormal tryptophan metabolism in cancer cells and promoted the secretion of L-kynurenine (L-kyn)-enriched EVs, with associated high levels of L-kyn in EVs isolated from both the tumor tissues and patient plasma in OC. EVs derived from IDO1high ovarian cancer cells elevated nicotinamide adenine dinucleotide (NAD +) levels in endothelial cells via delivering L-kyn. Besides, IDO1high ovarian cancer cell-derived EVs upregulated sirt3 expression in endothelial cells by increasing acetylation modification. These findings are crucial for promoting endothelial mitophagy correlated with tumor angiogenesis. Notably, both endothelial mitophagy and tumor angiogenesis could be suppressed by the IDO1 inhibitor in the orthotopic OC mouse model. CONCLUSIONS Together, our findings unveil a mechanism of mitophagy in OC angiogenesis and indicate the clinical relevance of EV enriched L-kyn as a potential biomarker for tumorigenesis and progression. Additionally, IDO1 inhibitors might become an alternative option for OC adjuvant therapy.
Collapse
Affiliation(s)
- Xiang Ying
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xiaoqian Zhang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Rd, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
38
|
Chen Z, Li S, Liu M, Yin M, Chen J, Li Y, Li Q, Zhou Y, Xia Y, Chen A, Lu D, Li C, Chen Y, Qian J, Ge J. Nicorandil alleviates cardiac microvascular ferroptosis in diabetic cardiomyopathy: Role of the mitochondria-localized AMPK-Parkin-ACSL4 signaling pathway. Pharmacol Res 2024; 200:107057. [PMID: 38218357 DOI: 10.1016/j.phrs.2024.107057] [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: 10/03/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
Mitochondria-associated ferroptosis exacerbates cardiac microvascular dysfunction in diabetic cardiomyopathy (DCM). Nicorandil, an ATP-sensitive K+ channel opener, protects against endothelial dysfunction, mitochondrial dysfunction, and DCM; however, its effects on ferroptosis and mitophagy remain unexplored. The present study aimed to assess the beneficial effects of nicorandil against endothelial ferroptosis in DCM and the underlying mechanisms. Cardiac microvascular perfusion was assessed using a lectin perfusion assay, while mitophagy was assessed via mt-Keima transfection and transmission electron microscopy. Ferroptosis was examined using mRNA sequencing, fluorescence staining, and western blotting. The mitochondrial localization of Parkin, ACSL4, and AMPK was determined via immunofluorescence staining. Following long-term diabetes, nicorandil treatment improved cardiac function and remodeling by alleviating cardiac microvascular injuries, as evidenced by the improved microvascular perfusion and structural integrity. mRNA-sequencing and biochemical analyses showed that ferroptosis occurred and Pink1/Parkin-dependent mitophagy was suppressed in cardiac microvascular endothelial cells after diabetes. Nicorandil treatment suppressed mitochondria-associated ferroptosis by promoting the Pink1/Parkin-dependent mitophagy. Moreover, nicorandil treatment increased the phosphorylation level of AMPKα1 and promoted its mitochondrial translocation, which further inhibited the mitochondrial translocation of ACSL4 via mitophagy and ultimately suppressed mitochondria-associated ferroptosis. Importantly, overexpression of mitochondria-localized AMPKα1 (mitoAα1) shared similar benefits with nicorandil on mitophagy, ferroptosis and cardiovascular protection against diabetic injury. In conclusion, the present study demonstrated the therapeutic effects of nicorandil against cardiac microvascular ferroptosis in DCM and revealed that the mitochondria-localized AMPK-Parkin-ACSL4 signaling pathway mediates mitochondria-associated ferroptosis and the development of cardiac microvascular dysfunction.
Collapse
Affiliation(s)
- Zhangwei Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Muyin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ming Yin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jinxiang Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Youran Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Qiyu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yan Xia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ao Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Danbo Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Chenguang Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Yuqiong Chen
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
39
|
Wang C, Zhuo JJ, Li WQ, Zhou ML, Cheng KJ. Role of autophagy and mitophagy of group 2 innate lymphoid cells in allergic and local allergic rhinitis. World Allergy Organ J 2024; 17:100852. [PMID: 38298830 PMCID: PMC10827603 DOI: 10.1016/j.waojou.2023.100852] [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: 08/04/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 02/02/2024] Open
Abstract
Background Roles of ILC2s in allergic rhinitis (AR) and local allergic rhinitis (LAR) are unclear. In this study, we are determined to find the levels of autophagy and mitophagy of ILC2s in allergic nasal inflammation. Methods ELISA was used to detect type 2 inflammatory cytokines. Hematoxylin and eosin (H&E) staining were used to compare the eosinophil (EOS) infiltration of nasal tissue specimens. Flow cytometry was used to detect the levels of ILC2s and Th2 cells. Immunohistochemistry (IHC) and Western blot (WB) were used to detect the levels of Beclin1, LC3, p62, PINK1, Parkin, FUNDC1, and BNIP3 in nasal mucosa. The levels of autophagy related proteins and mitophagy related proteins of the ILC2s were detected by WB. The number of autophagosomes of ILC2s was observed by transmission electron microscopy. The co-localization levels of GFP-LC3 and Mito tracker in ILC2s were observed by confocal microscopy using immunofluorescence. Results We found that the level of type 2 inflammation in AR and LAR mice was significantly increased. The levels of autophagy and mitophagy of AR and LAR mice in nasal mucosa and ILC2s were both increased. Conclusions ILC2s may be associated with the occurrence and development of nasal allergic inflammation. The abnormal increase of autophagy and mitophagy levels in the nose may be associated with the incidence of AR and LAR. Abnormal autophagy and mitophagy levels of ILC2s cells may be one of the causes of allergic nasal inflammation.
Collapse
Affiliation(s)
- Chen Wang
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jin-Jing Zhuo
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wen-Qian Li
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Min-Li Zhou
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ke-Jia Cheng
- Department of Otolaryngology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
40
|
Chen Z, Chen J, Li Y, Wang B, Lu Y, Jian J, Tang J, Cai J. Functional properties of ATPIF1 in the orange-spotted grouper (Epinephelus coioides) in response to viral infection. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109329. [PMID: 38154763 DOI: 10.1016/j.fsi.2023.109329] [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: 11/14/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
ATP synthase inhibitory factor 1 (ATPIF1) can activate mitochondrial autophagic pathway and mediates immune response by regulating ATP synthase activity. However, the role of fish ATPIF1 on viral infection is still unknown. In this study, we identified an ATPIF1 homolog (Ec-ATPIF1) from orange-spotted grouper (Epinephelus coioides). Ec-ATPIF1 is mainly expressed in the kidney and liver. The expression of Ec-ATPIF1 was significantly up-regulated after RGNNV stimulation in vitro. Further experiments showed that overexpression of Ec-ATPIF1 inhibited the expression of viral genes (CP and RdRp) and intracellular ATP synthesis. Ec-ATPIF1 overexpression also promoted the expression of mitophagy related genes (PINK1, Parkin, BNIP3, NIX, FUNDC1, LC3), inflammation-related factors (IL-1β, IL-6, IL-8, IL-10, TNF-α, TLR2) and interferon pathway factors (IRF1, IRF3, IRF7, MX1, ISG15, ISG56, MDA5, TRIF). While the knockdown of Ec-ATPIF1 exhibited the opposite effects on the expression of viral genes and immune-related factors above. These data suggest that Ec-ATPIF1 can impact viral infection by regulating mitophagy, ATP synthesis, the expression of inflammatory factors and interferon pathway factors. These findings will be beneficial to better explore the immune regulatory mechanisms of fish respond to viral infection.
Collapse
Affiliation(s)
- Zhaofeng Chen
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Junxi Chen
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Yi Li
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Bei Wang
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Yishan Lu
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Jichang Jian
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Jufen Tang
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China
| | - Jia Cai
- College of Fisheries, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, PR China; Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Academy of Sciences, Nanning, 530007, PR China.
| |
Collapse
|
41
|
Nagy-Grócz G, Spekker E, Vécsei L. Kynurenines, Neuronal Excitotoxicity, and Mitochondrial Oxidative Stress: Role of the Intestinal Flora. Int J Mol Sci 2024; 25:1698. [PMID: 38338981 PMCID: PMC10855176 DOI: 10.3390/ijms25031698] [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: 01/04/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
The intestinal flora has been the focus of numerous investigations recently, with inquiries not just into the gastrointestinal aspects but also the pathomechanism of other diseases such as nervous system disorders and mitochondrial diseases. Mitochondrial disorders are the most common type of inheritable metabolic illness caused by mutations of mitochondrial and nuclear DNA. Despite the intensive research, its diagnosis is usually difficult, and unfortunately, treating it challenges physicians. Metabolites of the kynurenine pathway are linked to many disorders, such as depression, schizophrenia, migraine, and also diseases associated with impaired mitochondrial function. The kynurenine pathway includes many substances, for instance kynurenic acid and quinolinic acid. In this review, we would like to show a possible link between the metabolites of the kynurenine pathway and mitochondrial stress in the context of intestinal flora. Furthermore, we summarize the possible markers of and future therapeutic options for the kynurenine pathway in excitotoxicity and mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Gábor Nagy-Grócz
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary;
- Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| | | | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary;
- HUN-REN-SZTE Neuroscience Research Group, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
42
|
Wang J, Zheng F, Wang D, Yang Q. Regulation of ULK1 by WTAP/IGF2BP3 axis enhances mitophagy and progression in epithelial ovarian cancer. Cell Death Dis 2024; 15:97. [PMID: 38286802 PMCID: PMC10824720 DOI: 10.1038/s41419-024-06477-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/31/2024]
Abstract
There is a pressing need for innovative therapeutic strategies for patients with epithelial ovarian cancer (EOC). Previous studies have shown that UNC-51-like kinase 1 (ULK1), a serine/threonine kinase, is crucial in regulating cellular autophagy and mitophagy across various tumor types. However, the clinical implications, biological functions, and potential mechanisms of ULK1 in EOC remain poorly understood. This study demonstrates that ULK1 expression is upregulated in EOC tissue samples and EOC cell lines, with increased ULK1 expression correlating with poor prognosis. Functionally, overexpressed ULK1 enhances the proliferation and migration abilities of EOC cells both in vitro and in vivo. Mechanistically, ULK1 was identified as an m6A target of WTAP. WTAP-mediated m6A modification of ULK1 enhanced its mRNA stability in an IGF2BP3-dependent manner, leading to elevated ULK1 expression and enhanced mitophagy in EOC. In summary, our research reveals that the WTAP/IGF2BP3-ULK1 axis significantly influences protective mitophagy in EOC, contributing to its progression. Therefore, the regulatory mechanisms and biological function of ULK1 identify it as a potential molecular target for therapeutic intervention in EOC.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Fei Zheng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Dandan Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
43
|
Zheng Z, Zhao X, Yuan B, Jiang S, Yan R, Dong X, Yao Q, Liang H. Soy isoflavones induces mitophagy to inhibit the progression of osteosarcoma by blocking the AKT/mTOR signaling pathway. Mol Med 2024; 30:5. [PMID: 38191316 PMCID: PMC10775635 DOI: 10.1186/s10020-024-00778-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: 08/08/2023] [Accepted: 01/02/2024] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Soy isoflavones (SI) is a natural bioactive substance exhibiting beneficial effects on human health. This study aims to elucidate the therapeutic potential of SI in the treatment of osteosarcoma (OS) and to investigate the underlying mechanisms, particularly focusing on mitophagy. METHODS The effects of SI on the proliferation, apoptosis, migration, and invasion of U2OS cells were analyzed. Mitophagy was assessed through multiple parameters: mitochondrial autophagosomes, mitochondrial membrane potential, autophagy-related proteins, reactive oxygen species (ROS), and oxygen consumption rate (OCR). Protein levels related to apoptosis, autophagy, and the AKT/mTOR pathway were analyzed using western blot. The therapeutic efficacy of SI was further identified using a mouse tumor xenograft model. Cell apoptosis and proliferation in tumor xenografts were detected by TUNEL staining and immunohistochemistry (IHC), respectively. RESULTS SI dose-dependently suppressed the viability, colony formation, migration, and invasion of U2OS cells, and enhanced the apoptosis. SI also dose-dependently induced mitophagy in OS cells, evidenced by an increase in autophagosomes and ROS levels, a decrease in mitochondrial membrane potential and OCR, and concomitant changes in autophagy-related proteins. Mdivi-1, an inhibitor of mitophagy, reversed the anti-tumor effects of SI on U2OS cells. In addition, SI blocked the AKT/mTOR pathway in U2OS cells. SC-79, an AKT agonist, reversed the effect of SI on inducing mitophagy. Moreover, SI also promoted cell apoptosis and mitophagy in tumor xenografts in vivo. CONCLUSIONS SI induces mitophagy in OS cells by blocking the AKT/mTOR pathway, contributing to the inhibition of OS.
Collapse
Affiliation(s)
- Ziang Zheng
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Xinghan Zhao
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Bo Yuan
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Shan Jiang
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Rushan Yan
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Xiaowei Dong
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Qijun Yao
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Haidong Liang
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China.
| |
Collapse
|
44
|
Xu J, Bian L, You D, Li Z, Wang T, Li Y, Ren X, He Y. PDGF-BB accelerates TSCC via fibroblast lactates limiting miR-26a-5p and boosting mitophagy. Cancer Cell Int 2024; 24:5. [PMID: 38169376 PMCID: PMC10763357 DOI: 10.1186/s12935-023-03172-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
The tumor microenvironment and cancer-associated fibroblasts (CAFs) play crucial roles in tumor development, and their metabolic coupling remains unclear. Clinical data showed a positive correlation between PDGF-BB, CAFs, and glycolysis in the tumor microenvironment of oral tongue squamous cell carcinoma patients. In vitro, CAFs are derived from hOMF cells treated with PDGF-BB, which induces their formation and promotes aerobic glycolysis. Mitophagy increased the PDGF-BB-induced formation of CAF phenotypes and aerobic glycolysis, while autophagy inhibition blocked PDGF-BB-induced effects. Downregulation of miR-26a-5p was observed in CAFs; upregulation of miR-26a-5p inhibited the expression of mitophagy-related proteins ULKI, Parkin, PINK1, and LC3 and aerobic glycolysis in PDGF-BB-induced CAFs. PDGF-BB-induced CAFs promoted tumor cell proliferation, invasion, metastasis, NF-κB signaling pathway activation, and PDGF-BB secretion. Thus, PDGF-BB is associated with lactate-induced CAF formation and glucose metabolism reprogramming. These findings indicate potential therapeutic targets in oral tongue squamous cell carcinoma.
Collapse
Affiliation(s)
- Jianguo Xu
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Li Bian
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650106, China
| | - Dingyun You
- School of Public Health, Kunming Medical University, Kunming, 650500, China
| | - Ziliang Li
- Department of oral Implantology, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
| | - Tingting Wang
- Department of Stomatology, The First People's Hospital of Yunnan Province, Kunming, 650032, China
| | - Yiting Li
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
| | - Xiaobin Ren
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, 1088 Haiyuan Central Road, Kunming, Yunnan, 650106, China.
| | - Yongwen He
- Department of Dental Research, Kunming Medical University School and Hospital of Stomatology, 1088 Haiyuan Central Road, Kunming, Yunnan, 650106, China.
- Qujing Medical College, Qujing, 655011, China.
| |
Collapse
|
45
|
Gao A, Wang M, Tang X, Shi G, Hou K, Fang J, Zhou L, Zhou H, Jiang W, Li Y, Ouyang F. NDP52 SUMOylation contributes to low-dose X-rays-induced cardiac hypertrophy through PINK1/Parkin-mediated mitophagy via MUL1/SUMO2 signalling. J Cell Physiol 2024; 239:79-96. [PMID: 37942585 DOI: 10.1002/jcp.31145] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023]
Abstract
Radiation-induced heart damage caused by low-dose X-rays has a significant impact on tumour patients' prognosis, with cardiac hypertrophy being the most severe noncarcinogenic adverse effect. Our previous study demonstrated that mitophagy activation promoted cardiac hypertrophy, but the underlying mechanisms remained unclear. In the present study, PARL-IN-1 enhanced excessive hypertrophy of cardiomyocytes and exacerbated mitochondrial damage. Isobaric tags for relative and absolute quantification-based quantitative proteomics identified NDP52 as a crucial target mediating cardiac hypertrophy induced by low-dose X-rays. SUMOylation proteomics revealed that the SUMO E3 ligase MUL1 facilitated NDP52 SUMOylation through SUMO2. Co-IP coupled with LC-MS/MS identified a critical lysine residue at position 262 of NDP52 as the key site for SUMO2-mediated SUMOylation of NDP52. The point mutation plasmid NDP52K262R inhibited mitophagy under MUL1 overexpression, as evidenced by inhibition of LC3 interaction with NDP52, PINK1 and LAMP2A. A mitochondrial dissociation study revealed that NDP52K262R inhibited PINK1 targeting to endosomes early endosomal marker (EEA1), late/lysosome endosomal marker (LAMP2A) and recycling endosomal marker (RAB11), and laser confocal microscopy confirmed that NDP52K262R impaired the recruitment of mitochondria to the autophagic pathway through EEA1/RAB11 and ATG3, ATG5, ATG16L1 and STX17, but did not affect mitochondrial delivery to lysosomes via LAMP2A for degradation. In conclusion, our findings suggest that MUL1-mediated SUMOylation of NDP52 plays a crucial role in regulating mitophagy in the context of low-dose X-ray-induced cardiac hypertrophy. Two hundred sixty-second lysine of NDP52 is identified as a key SUMOylation site for low-dose X-ray promoting mitophagy activation and cardiac hypertrophy. Collectively, this study provides novel implications for the development of therapeutic strategies aimed at preventing the progression of cardiac hypertrophy induced by low-dose X-rays.
Collapse
Affiliation(s)
- Anbo Gao
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Mengjie Wang
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Gangqing Shi
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Kai Hou
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Jinren Fang
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Linlin Zhou
- Hengyang Medical School, Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Hong Zhou
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weimin Jiang
- Department of Cardiology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Fan Ouyang
- Department of Cardiology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| |
Collapse
|
46
|
Yao BF, Luo XJ, Peng J. A review for the correlation between optic atrophy 1-dependent mitochondrial fusion and cardiovascular disorders. Int J Biol Macromol 2024; 254:127910. [PMID: 37939779 DOI: 10.1016/j.ijbiomac.2023.127910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/19/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Mitochondrial dynamics homeostasis is sustained by continuous and balanced fission and fusion, which are determinants of morphology, abundance, biogenesis and mitophagy of mitochondria. Optic atrophy 1 (OPA1), as the only inner mitochondrial membrane fusion protein, plays a key role in stabilizing mitochondrial dynamics. The disturbance of mitochondrial dynamics contributes to the pathophysiological progress of cardiovascular disorders, which are the main cause of death worldwide in recent decades and result in tremendous social burden. In this review, we describe the latest findings regarding OPA1 and its role in mitochondrial fusion. We summarize the post-translational modifications (PTMs) for OPA1 and its regulatory role in mitochondrial dynamics. Then the diverse cell fates caused by OPA1 expression during cardiovascular disorders are discussed. Moreover, cardiovascular disorders (such as heart failure, myocardial ischemia/reperfusion injury, cardiomyopathy and cardiac hypertrophy) relevant to OPA1-dependent mitochondrial dynamics imbalance have been detailed. Finally, we highlight the potential that targeting OPA1 to impact mitochondrial fusion may be used as a novel strategy against cardiovascular disorders.
Collapse
Affiliation(s)
- Bi-Feng Yao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| |
Collapse
|
47
|
Yang Y, Jiang Y, Qian D, Wang Z, Xiao L. Prevention and treatment of osteoporosis with natural products: Regulatory mechanism based on cell ferroptosis. J Orthop Surg Res 2023; 18:951. [PMID: 38082321 PMCID: PMC10712195 DOI: 10.1186/s13018-023-04448-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
CONTEXT With the development of society, the number of patients with osteoporosis is increasing. The prevention and control of osteoporosis has become a serious and urgent issue. With the continuous progress of biomedical research, ferroptosis has attracted increased attention. However, the pathophysiology and mechanisms of ferroptosis and osteoporosis still need further study. Natural products are widely used in East Asian countries for osteoporosis prevention and treatment. OBJECTIVE In this paper, we will discuss the basic mechanisms of ferroptosis, the relationship between ferroptosis and osteoclasts and osteoblasts, and in vitro and in vivo studies of natural products to prevent osteoporosis by interfering with ferroptosis. METHODS This article takes ferroptosis, natural products, osteoporosis, osteoblasts and osteoclast as key words. Retrieve literature from 2012 to 2023 indexed in databases such as PubMed Central, PubMed, Web of Science, Scopus and ISI. RESULTS Ferroptosis has many regulatory mechanisms, including the system XC -/GSH/GPX4, p62/Keap1/Nrf2, FSP1/NAD (P) H/CoQ10, P53/SAT1/ALOX15 axes etc. Interestingly, we found that natural products, such as Artemisinin, Biochanin A and Quercetin, can play a role in treating osteoporosis by promoting ferroptosis of osteoclast and inhibiting ferroptosis of osteoblasts. CONCLUSIONS Natural products have great potential to regulate OBs and OCs by mediating ferroptosis to prevent and treat osteoporosis, and it is worthwhile to explore and discover more natural products that can prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Yunshang Yang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
| | - Yifan Jiang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
| | - Daoyi Qian
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China
| | - Zhirong Wang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
| | - Long Xiao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
- Department of Orthopedics, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang, 215600, Jiangsu, China.
| |
Collapse
|
48
|
Giansanti M, Theinert T, Boeing SK, Haas D, Schlegel PG, Vacca P, Nazio F, Caruana I. Exploiting autophagy balance in T and NK cells as a new strategy to implement adoptive cell therapies. Mol Cancer 2023; 22:201. [PMID: 38071322 PMCID: PMC10709869 DOI: 10.1186/s12943-023-01893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Autophagy is an essential cellular homeostasis pathway initiated by multiple stimuli ranging from nutrient deprivation to viral infection, playing a key role in human health and disease. At present, a growing number of evidence suggests a role of autophagy as a primitive innate immune form of defense for eukaryotic cells, interacting with components of innate immune signaling pathways and regulating thymic selection, antigen presentation, cytokine production and T/NK cell homeostasis. In cancer, autophagy is intimately involved in the immunological control of tumor progression and response to therapy. However, very little is known about the role and impact of autophagy in T and NK cells, the main players in the active fight against infections and tumors. Important questions are emerging: what role does autophagy play on T/NK cells? Could its modulation lead to any advantages? Could specific targeting of autophagy on tumor cells (blocking) and T/NK cells (activation) be a new intervention strategy? In this review, we debate preclinical studies that have identified autophagy as a key regulator of immune responses by modulating the functions of different immune cells and discuss the redundancy or diversity among the subpopulations of both T and NK cells in physiologic context and in cancer.
Collapse
Affiliation(s)
- Manuela Giansanti
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Tobias Theinert
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Sarah Katharina Boeing
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Dorothee Haas
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paola Vacca
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Francesca Nazio
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy.
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Ignazio Caruana
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
49
|
Girigoswami K, Pallavi P, Girigoswami A. Intricate subcellular journey of nanoparticles to the enigmatic domains of endoplasmic reticulum. Drug Deliv 2023; 30:2284684. [PMID: 37990530 PMCID: PMC10987057 DOI: 10.1080/10717544.2023.2284684] [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: 08/24/2023] [Accepted: 11/05/2023] [Indexed: 11/23/2023] Open
Abstract
It is evident that site-specific systemic drug delivery can reduce side effects, systemic toxicity, and minimal dosage requirements predominantly by delivering drugs to particular pathological sites, cells, and even subcellular structures. The endoplasmic reticulum (ER) and associated cell organelles play a vital role in several essential cellular functions and activities, such as the synthesis of lipids, steroids, membrane-associated proteins along with intracellular transport, signaling of Ca2+, and specific response to stress. Therefore, the dysfunction of ER is correlated with numerous diseases where cancer, neurodegenerative disorders, diabetes mellitus, hepatic disorder, etc., are very common. To achieve satisfactory therapeutic results in certain diseases, it is essential to engineer delivery systems that can effectively enter the cells and target ER. Nanoparticles are highly biocompatible, contain a variety of cargos or payloads, and can be modified in a pliable manner to achieve therapeutic effectiveness at the subcellular level when delivered to specific organelles. Passive targeting drug delivery vehicles, or active targeting drug delivery systems, reduce the nonselective accumulation of drugs while reducing side effects by modifying them with small molecular compounds, antibodies, polypeptides, or isolated bio-membranes. The targeting of ER and closely associated organelles in cells using nanoparticles, however, is still unsymmetrically understood. Therefore, here we summarized the pathophysiological prospect of ER stress, involvement of ER and mitochondrial response, disease related to ER dysfunctions, essential therapeutics, and nanoenabled modulation of their delivery to optimize therapy.
Collapse
Affiliation(s)
- Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| | - Pragya Pallavi
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| |
Collapse
|
50
|
Guo B, Song H, Fan J, Wang B, Chen L, Hu Q, Yin Y. The NR2B-targeted intervention alleviates the neuronal injuries at the sub-acute stage of cerebral ischemia: an exploration of stage-dependent strategy against ischemic insults. Exp Brain Res 2023; 241:2735-2750. [PMID: 37845379 DOI: 10.1007/s00221-023-06717-3] [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: 03/28/2023] [Accepted: 09/30/2023] [Indexed: 10/18/2023]
Abstract
Stroke is reported to be the second leading cause of death worldwide, among which ischemic stroke has fourfold greater incidence than intracerebral hemorrhage. Excitotoxicity induced by NMDAR plays a central role in ischemic stroke-induced neuronal death. However, intervention targeted NMDARs against ischemic stroke has failed, which may result from the complex composition of NMDARs and the dynamic changes of their subunits. In this current study, the levels of NR1, NR2A and NR2B subunits of NMDARs were observed upon different time points during the reperfusion after 1 h ischemia with the western blot assay. It was found that the changes of NR1 subunit were only detected after ischemia 1 h/reperfusion 1 day (1 d). While, the changes of NR2A and NR2B subunits may last to ischemia 1 h/reperfusion 7 day(7 d), indicating that NR2subunits may be a potential target for ischemia-reperfusion injuries at the sub-acute stage of ischemic stroke. Simultaneously, mitochondrial injuries in neurons were investigated with transmission electron microscopy (TEM), and mitochondrial dysfunction was evaluated with mitochondrial membrane proteins oxidative respiratory chain complex and OCR. When the antagonist of NMDARs was used before ischemic exposure, the neuronal mitochondrial dysfunction was alleviated, suggesting that these aberrant deviations of NMDARs from basal levels led to mitochondrial dysfunction. Furthermore, when the antagonist of NR2B was administrated intracerebroventricularly at the sub-acute cerebral ischemia, the volume of cerebral infarct region was decreased and the neural functions were improved. To sum up, the ratio of NR2B-containing NMDARs is vital for mitochondrial homeostasis and then neuronal survival. NR2B-targeted intervention should be chosen at the sub-acute stage of cerebral ischemia.
Collapse
Affiliation(s)
- Bei Guo
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Huimeng Song
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Jiahui Fan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Bin Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Lingyi Chen
- John Bapst Memorial High School, Bangor, CA, USA
| | - Qiandai Hu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Yanling Yin
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China.
| |
Collapse
|