1
|
Xu S, Jia J, Mao R, Cao X, Xu Y. Mitophagy in acute central nervous system injuries: regulatory mechanisms and therapeutic potentials. Neural Regen Res 2025; 20:2437-2453. [PMID: 39248161 DOI: 10.4103/nrr.nrr-d-24-00432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/22/2024] [Indexed: 09/10/2024] Open
Abstract
Acute central nervous system injuries, including ischemic stroke, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury, are a major global health challenge. Identifying optimal therapies and improving the long-term neurological functions of patients with acute central nervous system injuries are urgent priorities. Mitochondria are susceptible to damage after acute central nervous system injury, and this leads to the release of toxic levels of reactive oxygen species, which induce cell death. Mitophagy, a selective form of autophagy, is crucial in eliminating redundant or damaged mitochondria during these events. Recent evidence has highlighted the significant role of mitophagy in acute central nervous system injuries. In this review, we provide a comprehensive overview of the process, classification, and related mechanisms of mitophagy. We also highlight the recent developments in research into the role of mitophagy in various acute central nervous system injuries and drug therapies that regulate mitophagy. In the final section of this review, we emphasize the potential for treating these disorders by focusing on mitophagy and suggest future research paths in this area.
Collapse
Affiliation(s)
- Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Jiangsu University, Nanjing, Jiangsu Province, China
| | - Junqiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, Jiangsu Province, China
| | - Rui Mao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Jiangsu University, Nanjing, Jiangsu Province, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu Province, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu Province, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Jiangsu University, Nanjing, Jiangsu Province, China
- Department of Neurology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, Jiangsu Province, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu Province, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
- Nanjing Neurology Medical Center, Nanjing, Jiangsu Province, China
| |
Collapse
|
2
|
Shang J, Wen Y, Zhang X, Huang G, Chen W, Wang B, Wu K, Xiang Q, Liu X. Naoxintong capsule accelerates mitophagy in cerebral ischemia-reperfusion injury via TP53/PINK1/PRKN pathway based on network pharmacology analysis and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118721. [PMID: 39173723 DOI: 10.1016/j.jep.2024.118721] [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: 07/02/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The incidence and mortality of cerebrovascular diseases are increasing year by year. Cerebral ischemia-reperfusion injury (CIRI) is common in patients with ischemic stroke. Naoxintong (NXT) is composed of a variety of Chinese medicines and has the ability to treat CIRI. AIM OF THE STUDY The aim of this study is to investigate whether NXT regulates mitophagy in CIRI based on network pharmacology analysis and experimental validation. MATERIALS AND METHODS Oxygen and glucose deprivation/re-oxygenation (OGD/R, 2/22 h) model of PC12 cells and transient middle cerebral artery occlusion (tMCAO, 2/22 h) model of rats were established. Pharmacodynamic indicators include neurological deficit score, 2,3,5-triphenyte-trazoliumchloride (TTC) staining, hematoxylin-eosin (HE) staining and cell viability. Network pharmacology was used to predict pharmacological mechanisms. Pharmacological mechanism indexes include transmission electron microscopy (TEM), drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), immunohistochemistry (IHC), western blot (WB) and immunofluorescence (IF). Kevetrin (an agonists of p53) and pifithrin-α (an inhibitor of p53) used to detect the key role of p53 in mitophagy of NXT. RESULTS NXT (1% serum containing NXT and 110 mg/kg) improved the damage of OGD/R PC12 cells and tMCAO rats, and this protective effect was related to the anti-oxidation and ability to promote mitophagy of NXT. NXT and pifithrin-α increased the expression of promoting-mitophagy targets (PINK1, PRKN and LC3B) and inhibited the expression of inhibiting-mitophagy targets (p52) via restraining p53, and finally accelerated mitophagy caused by CIRI. CONCLUSION This study demonstrates that NXT promotes mitophagy in CIRI through restraining p53 and promoting PINK1/PRKN in vivo and in vitro.
Collapse
Affiliation(s)
- Jinfeng Shang
- Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yinlian Wen
- Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xiaolu Zhang
- Beijing University of Chinese Medicine, Beijing 102488, China.
| | | | - Wenbin Chen
- Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Bohong Wang
- Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Kai Wu
- Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Quan Xiang
- Gansu University of Chinese Medicine, Gansu 730101, China.
| | - Xin Liu
- Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
3
|
Wu W, Wei Z, Wu Z, Chen J, Liu J, Chen M, Yuan J, Zheng Z, Zhao Z, Lin Q, Liu N, Chen H. Exercise training alleviates neuronal apoptosis and re-establishes mitochondrial quality control after cerebral ischemia by increasing SIRT3 expression. Cell Biol Toxicol 2024; 41:10. [PMID: 39707047 DOI: 10.1007/s10565-024-09957-3] [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: 06/26/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Existing evidence indicates that exercise training can enhance neural function by regulating mitochondrial quality control (MQC), which can be impaired by cerebral ischemia, and that sirtuin-3 (SIRT3), a protein localized in mitochondria, is crucial in maintaining mitochondrial functions. However, the relationship among exercise training, SIRT3, and MQC after cerebral ischemia remains obscure. This study attempted to elucidate the relationship among exercise training, SIRT3 and MQC after cerebral ischemia in rats. Male adult SD rats received tMCAO after the transfection of adeno-associated virus encoding either sirtuin-3 (AAV-SIRT3) or SIRT3 knockdown (AAV-sh-SIRT3) into the ipsilateral striata and cortex. Subsequently, the animals were randomly selected for exercise training. The index changes were measured by transmission electron microscopy, Western blot analysis, nuclear magnetic resonance imaging, TUNEL staining, and immunofluorescence staining, etc. The results revealed that after cerebral ischemia, exercise training increased SIRT3 expression, significantly improved neural function, alleviated infarct volume and neuronal apoptosis, maintained the mitochondrial structural integrity, and re-established MQC. The latter promoted mitochondrial biogenesis, balanced mitochondrial fission/fusion, and enhanced mitophagy. These favorable benefits were reversed after SIRT3 interference. In addition, a cellular OGD/R model showed that the increased SIRT3 expression alleviates neuronal apoptosis and re-establishes mitochondrial quality control by activating the β-catenin pathway. These findings suggest that exercise training may optimize mitochondrial quality control by increasing the expression of SIRT3, thereby improving neural functions after cerebral ischemia, which illuminates the mechanism underlying the exercise training-conferred neural benefits and indicates SIRT3 as a therapeutic strategy for brain ischemia.
Collapse
Affiliation(s)
- Wenwen Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zengyu Wei
- Emergency Department, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhiyun Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Jianmin Chen
- Department of Rehabilitation Medicine, The First Afliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ji Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Jinjin Yuan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zhijian Zheng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zijun Zhao
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Qiang Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Luo X, Zhang S, Wang L, Li J. Pathological roles of mitochondrial dysfunction in endothelial cells during the cerebral no-reflow phenomenon: A review. Medicine (Baltimore) 2024; 103:e40951. [PMID: 39705421 DOI: 10.1097/md.0000000000040951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2024] Open
Abstract
Emergency intravascular interventional therapy is the most effective approach to rapidly restore blood flow and manage occlusion of major blood vessels during the initial phase of acute ischemic stroke. Nevertheless, several patients continue to experience ineffective reperfusion or cerebral no-reflow phenomenon, that is, hypoperfusion of cerebral blood supply after treatment. This is primarily attributed to downstream microcirculation disturbance. As integral components of the cerebral microvascular structure, endothelial cells (ECs) attach importance to regulating microcirculatory blood flow. Unlike neurons and microglia, ECs harbor a relatively low abundance of mitochondria, acting as key sensors of environmental and cellular stress in regulating the viability, structural integrity, and function of ECs rather than generating energy. Mitochondria dysfunction including increased mitochondrial reactive oxygen species levels and disturbed mitochondrial dynamics causes endothelial injury, further causing microcirculation disturbance involved in the cerebral no-reflow phenomenon. Therefore, this review aims to discuss the role of mitochondrial changes in regulating the role of ECs and cerebral microcirculation blood flow during I/R injury. The outcomes of the review will provide promising potential therapeutic targets for future prevention and effective improvement of the cerebral no-reflow phenomenon.
Collapse
Affiliation(s)
- Xia Luo
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shaotao Zhang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Longbing Wang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinglun Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Wang J, Wang D. Mitophagy in gynecological malignancies: roles, advances, and therapeutic potential. Cell Death Discov 2024; 10:488. [PMID: 39639053 PMCID: PMC11621523 DOI: 10.1038/s41420-024-02259-x] [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: 10/10/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Mitophagy is a process in which impaired or dysfunctional mitochondria are selectively eliminated through the autophagy mechanism to maintain mitochondrial quality control and cellular homeostasis. Based on specific target signals, several mitophagy processes have been identified. Defects in mitophagy are associated with various pathological conditions, including neurodegenerative disorders, cardiovascular diseases, metabolic diseases, and cancer. Mitophagy has been shown to play a critical role in the pathogenesis of gynecological malignancies and the development of drug resistance. In this review, we have summarized and discussed the role and recent advances in understanding the therapeutic potential of mitophagy in the development of gynecological malignancies. Therefore, the valuable insights provided in this review may serve as a basis for further studies that contribute to the development of novel treatment strategies and improved patient outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- 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.
| |
Collapse
|
6
|
Xi ZC, Ren HG, Ai L, Wang Y, Liu MF, Qiu YF, Feng JL, Fu W, Bi QQ, Wang F, Xu HX. Ginsenoside Rg1 mitigates cerebral ischaemia/reperfusion injury in mice by inhibiting autophagy through activation of mTOR signalling. Acta Pharmacol Sin 2024; 45:2474-2486. [PMID: 38937576 PMCID: PMC11579309 DOI: 10.1038/s41401-024-01334-4] [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: 01/25/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
Reperfusion injury, which is distinct from ischaemic injury, occurs when blood flow is restored in previously ischaemic brain tissue, further compromising neurons and other cells and worsening the injury. There is currently a lack of pharmaceutical agents and therapeutic interventions that specifically mitigate cerebral ischaemia/reperfusion (I/R) injury. Ginsenoside Rg1 (Rg1), a protopanaxatriol-type saponin isolated from Panax ginseng C. A. Meyer, has been found to protect against cerebral I/R injury, but its intricate protective mechanisms remain to be elucidated. Numerous studies have shown that autophagy plays a crucial role in protecting brain tissue during the I/R process and is emerging as a promising therapeutic strategy for effective treatment. In this study, we investigated whether Rg1 protected against I/R damage in vitro and in vivo by regulating autophagy. Both MCAO and OGD/R models were established. SK-N-AS and SH-SY5Y cells were subjected to OGD followed by reperfusion with Rg1 (4-32 μM). MCAO mice were injected with Rg1 (30 mg·kg-1·d-1. i.p.) for 3 days before and on the day of surgery. Rg1 treatment significantly mitigated ischaemia/reperfusion injury both in vitro and in vivo. Furthermore, we demonstrated that the induction of autophagy contributed to I/R injury, which was effectively inhibited by Rg1 in both in vitro and in vivo models of cerebral I/R injury. Rg1 inhibited autophagy through multiple steps, including impeding autophagy initiation, inducing lysosomal dysfunction and inhibiting cathepsin enzyme activities. We revealed that mTOR activation was pivotal in mediating the inhibitory effect of Rg1 on autophagy. Treatment with Torin-1, an autophagy inducer and mTOR-specific inhibitor, significantly reversed the impact of Rg1 on autophagy, decreasing its protective efficacy against I/R injury both in vitro and in vivo. In conclusion, our results suggest that Rg1 may serve as a promising drug candidate against cerebral I/R injury by inhibiting autophagy through activation of mTOR signalling.
Collapse
Affiliation(s)
- Zhi-Chao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China
| | - Han-Gui Ren
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China
| | - Lin Ai
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Yuan Wang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Meng-Fan Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China
| | - Yu-Fei Qiu
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Ji-Ling Feng
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Wang Fu
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Qian-Qian Bi
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Feng Wang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Hong-Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China.
| |
Collapse
|
7
|
Zhang W, Guo C, Li Y, Wang H, Wang H, Wang Y, Wu T, Wang H, Cheng G, Man J, Chen S, Fu S, Yang L. Mitophagy mediated by HIF-1α/FUNDC1 signaling in tubular cells protects against renal ischemia/reperfusion injury. Ren Fail 2024; 46:2332492. [PMID: 38584135 PMCID: PMC11000611 DOI: 10.1080/0886022x.2024.2332492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/14/2024] [Indexed: 04/09/2024] Open
Abstract
Acute kidney injury (AKI) is associated with a high mortality rate. Pathologically, renal ischemia/reperfusion injury (RIRI) is one of the primary causes of AKI, and hypoxia-inducible factor (HIF)-1α may play a defensive role in RIRI. This study assessed the role of hypoxia-inducible factor 1α (HIF-1α)-mediated mitophagy in protection against RIRI in vitro and in vivo. The human tubular cell line HK-2 was used to assess hypoxia/reoxygenation (H/R)-induced mitophagy through different in vitro assays, including western blotting, immunofluorescence staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), and reactive oxygen species (ROS) measurement. Additionally, a rat RIRI model was established for evaluation by renal histopathology, renal Doppler ultrasound, and transmission electron microscopy to confirm the in vitro data. The selective HIF-1α inhibitor LW6 reduced H/R-induced mitophagy but increased H/R-induced apoptosis and ROS production. Moreover, H/R treatment enhanced expression of the FUN14 domain-containing 1 (FUNDC1) protein. Additionally, FUNDC1 overexpression reversed the effects of LW6 on the altered expression of light chain 3 (LC3) BII and voltage-dependent anion channels as well as blocked the effects of HIF-1α inhibition in cells. Pretreatment of the rat RIRI model with roxadustat, a novel oral HIF-1α inhibitor, led to decreased renal injury and apoptosis in vivo. In conclusion, the HIF-1α/FUNDC1 signaling pathway mediates H/R-promoted renal tubular cell mitophagy, whereas inhibition of this signaling pathway protects cells from mitophagy, thus aggravating apoptosis, and ROS production. Accordingly, roxadustat may protect against RIRI-related AKI.
Collapse
Affiliation(s)
- Wenjun Zhang
- Department of Nephrology, Lanzhou University Affiliated Second Hospital, Lanzhou, China
- Gansu Provicne Clinical Research Center for Kidney Diseases, Lanzhou, China
| | - Chao Guo
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yi Li
- Department of Anesthesiology, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Hao Wang
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Huabing Wang
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Yingying Wang
- Department of Nephrology, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Tingting Wu
- Department of Functional Examination in Children, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Huinan Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Gang Cheng
- The Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jiangwei Man
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Siyu Chen
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Shengjun Fu
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Li Yang
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
- Gansu Provicne Clinical Research Center for Urology, Lanzhou, China
| |
Collapse
|
8
|
Jia Z, Li H, Xu K, Li R, Yang S, Chen L, Zhang Q, Li S, Sun X. MAM-mediated mitophagy and endoplasmic reticulum stress: the hidden regulators of ischemic stroke. Front Cell Neurosci 2024; 18:1470144. [PMID: 39640236 PMCID: PMC11617170 DOI: 10.3389/fncel.2024.1470144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemic stroke (IS) is the predominant subtype of stroke and a leading contributor to global mortality. The mitochondrial-associated endoplasmic reticulum membrane (MAM) is a specialized region that facilitates communication between the endoplasmic reticulum and mitochondria, and has been extensively investigated in the context of neurodegenerative diseases. Nevertheless, its precise involvement in IS remains elusive. This literature review elucidates the intricate involvement of MAM in mitophagy and endoplasmic reticulum stress during IS. PINK1, FUNDC1, Beclin1, and Mfn2 are highly concentrated in the MAM and play a crucial role in regulating mitochondrial autophagy. GRP78, IRE1, PERK, and Sig-1R participate in the unfolded protein response (UPR) within the MAM, regulating endoplasmic reticulum stress during IS. Hence, the diverse molecules on MAM operate independently and interact with each other, collectively contributing to the pathogenesis of IS as the covert orchestrator.
Collapse
Affiliation(s)
- Ziyi Jia
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongtao Li
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ke Xu
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ruobing Li
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyu Yang
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Long Chen
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qianwen Zhang
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shulin Li
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaowei Sun
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
9
|
Xie P, Xia M, Long T, Guo D, Cao W, Sun P, Yu W. GIV/Girdin Modulation of Microglial Activation in Ischemic Stroke: Impact of FTO-Mediated m6A Modification. Mol Neurobiol 2024:10.1007/s12035-024-04604-8. [PMID: 39560901 DOI: 10.1007/s12035-024-04604-8] [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: 03/06/2024] [Revised: 09/12/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024]
Abstract
Ischemic stroke (IS) is one of the most common causes of death in the world. The lack of effective pharmacological treatments for IS was primarily due to a lack of understanding of its pathogenesis. Gα-Interacting vesicle-associated protein (GIV/Girdin) is a multi-modular signal transducer and guanine nucleotide exchange factor that controls important signaling downstream of multiple receptors. The purpose of this study was to investigate the role of GIV in IS. In the present study, we found that GIV is highly expressed in the central nervous system (CNS). GIV protein level was decreased, while GIV transcript level was increased in the middle cerebral artery occlusion reperfusion (MCAO/R) mice model. Additionally, GIV was insensitive lipopolysaccharide (LPS) exposure. Interestingly, we found that GIV overexpression dramatically restrained microglial activation, inflammatory response, and M1 polarization in BV-2 microglia induced by oxygen-glucose deprivation and reoxygenation (OGD/R). On the contrary, GIV knockdown had the opposite impact. Mechanistically, we found that GIV activated the Wnt/β-catenin signaling pathway by interacting with DVL2 (disheveled segment polarity protein 2). Notably, m6A demethylase fat mass and obesity-associated protein (FTO) decreased the N6-methyladenosine (m6A) modification-mediated increase of GIV expression and attenuated the inflammatory response in BV-2 stimulated by OGD/R. Taken together, our results demonstrate that GIV inhibited the inflammatory response via activating the Wnt/β-catenin signaling pathway which expression regulated in an FTO-mediated m6A modification in IS. These results broaden our understanding of the role of the FTO-GIV axis in IS development.
Collapse
Affiliation(s)
- Peng Xie
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Mingyan Xia
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tingting Long
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Dongfen Guo
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wenpeng Cao
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ping Sun
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang, Guiyang, China
| | - Wenfeng Yu
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
10
|
Lian CY, Li HJ, Xia WH, Li Y, Zhou XL, Yang DB, Wan XM, Wang L. Insufficient FUNDC1-dependent mitophagy due to early environmental cadmium exposure triggers mitochondrial redox imbalance to aggravate diet-induced lipotoxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 361:124724. [PMID: 39142430 DOI: 10.1016/j.envpol.2024.124724] [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: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Cadmium (Cd) is a toxic contaminant widely spread in natural and industrial environments. Adolescent exposure to Cd increases risk for obesity-related morbidity in young adults including type 2 diabetes and metabolic dysfunction-associated steatotic liver disease (MASLD). Despite this recognition, the direct impact of adolescent Cd exposure on the progression of MASLD later in life, and the mechanisms underlying these effects, remain unclear. Here, adolescent rats received control diet or diets containing 2 mg Cd2+/kg feed for 4 weeks, and then HFD containing 15% lard or control diet in young adult rats was selected for 6 weeks to clarify this issue. Data firstly showed that HFD-fed rats in young adulthood due to adolescent Cd exposure exhibited more severe MASLD, evidenced by increased liver damage, disordered serum and hepatic lipid levels, and activated NLRP3 inflammasome. Hepatic transcriptome analysis revealed the potential effects of mitochondrial dysfunction in aggravated MASLD due to Cd exposure. Verification data further confirmed that mitochondrial structure and function were targeted and disrupted during this process, shown by broken mitochondrial ridges, decreased mitochondrial membrane potential, imbalanced mitochondrial dynamic, insufficient ATP concentration, and enhanced mitochondrial ROS generation. However, mitophagy is inactively involved in clearance of damaged mitochondria induced by early Cd in HFD condition due to inhibited mitophagy receptor FUNDC1. In contrast, FUNDC1-dependent mitophagy activation prevents lipotoxicity aggravated by early Cd via suppressing mitochondrial ROS generation. Collectively, our data show that insufficient FUNDC1-dependent mitophagy can drive the transition from HFD-induced MASLD to MASH, and accordingly, these findings will provide a better understanding of potential mechanism of diet-induced metabolic diseases in the context of early environmental Cd exposure.
Collapse
Affiliation(s)
- Cai-Yu Lian
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province, 271017, China
| | - Hui-Jia Li
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province, 271017, China
| | - Wei-Hao Xia
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province, 271017, China
| | - Yue Li
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province, 271017, China
| | - Xue-Lei Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, China
| | - Du-Bao Yang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province, 271017, China
| | - Xue-Mei Wan
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, China
| | - Lin Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province, 271017, China.
| |
Collapse
|
11
|
Wang N, Chen J, Dang Y, Zhao X, Tibenda JJ, Li N, Zhu Y, Wang X, Zhao Q, Sun L. Research progress of traditional Chinese medicine in the treatment of ischemic stroke by regulating mitochondrial dysfunction. Life Sci 2024; 357:123045. [PMID: 39251017 DOI: 10.1016/j.lfs.2024.123045] [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: 06/13/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Ischemic stroke (IS) is a severe cerebrovascular disease with increasing incidence and mortality rates in recent years. The pathogenesis of IS is highly complex, with mitochondrial dysfunction playing a critical role in its onset and progression. Thus, preserving mitochondrial function is a pivotal aspect of treating ischemic brain injury. In response, there has been growing interest among scholars in the regulation of mitochondrial function through traditional Chinese medicine (TCM), including herb-derived compounds, individual herbs, and herbal prescriptions. This article reviews recent research on the mechanisms of mitochondrial dysfunction in IS and explores the potential of TCM in treating this condition by targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Niuniu Wang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Jun Chen
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanning Dang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Xinlin Zhao
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Jonnea Japhet Tibenda
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Nuan Li
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Yafei Zhu
- School of Nursing, Ningxia Medical University, Yinchuan, China
| | - Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Qipeng Zhao
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China.
| | - Lei Sun
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
12
|
Liu Y, Liu Q, Shang H, Li J, Chai H, Wang K, Guo Z, Luo T, Liu S, Liu Y, Wang X, Zhang H, Wu C, Song SJ, Yang J. Potential application of natural compounds in ischaemic stroke: Focusing on the mechanisms underlying "lysosomocentric" dysfunction of the autophagy-lysosomal pathway. Pharmacol Ther 2024; 263:108721. [PMID: 39284368 DOI: 10.1016/j.pharmthera.2024.108721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/06/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Ischaemic stroke (IS) is the second leading cause of death and a major cause of disability worldwide. Currently, the clinical management of IS still depends on restoring blood flow via pharmacological thrombolysis or mechanical thrombectomy, with accompanying disadvantages of narrow therapeutic time window and risk of haemorrhagic transformation. Thus, novel pathophysiological mechanisms and targeted therapeutic candidates are urgently needed. The autophagy-lysosomal pathway (ALP), as a dynamic cellular lysosome-based degradative process, has been comprehensively studied in recent decades, including its upstream regulatory mechanisms and its role in mediating neuronal fate after IS. Importantly, increasing evidence has shown that IS can lead to lysosomal dysfunction, such as lysosomal membrane permeabilization, impaired lysosomal acidity, lysosomal storage disorder, and dysfunctional lysosomal ion homeostasis, which are involved in the IS-mediated defects in ALP function. There is tightly regulated crosstalk between transcription factor EB (TFEB), mammalian target of rapamycin (mTOR) and lysosomal function, but their relationship remains to be systematically summarized. Notably, a growing body of evidence emphasizes the benefits of naturally derived compounds in the treatment of IS via modulation of ALP function. However, little is known about the roles of natural compounds as modulators of lysosomes in the treatment of IS. Therefore, in this context, we provide an overview of the current understanding of the mechanisms underlying IS-mediated ALP dysfunction, from a lysosomal perspective. We also provide an update on the effect of natural compounds on IS, according to their chemical structural types, in different experimental stroke models, cerebral regions and cell types, with a primary focus on lysosomes and autophagy initiation. This review aims to highlight the therapeutic potential of natural compounds that target lysosomal and ALP function for IS treatment.
Collapse
Affiliation(s)
- Yueyang Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hanxiao Shang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jichong Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - He Chai
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kaixuan Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenkun Guo
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Tianyu Luo
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shiqi Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yan Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuemei Wang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hangyi Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Chunfu Wu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Jingyu Yang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
13
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
14
|
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
|
15
|
Wang S, Li X, Wang T, Sun Z, Feng E, Jin Y. Overexpression of USP35 enhances the protective effect of hUC-MSCs and their extracellular vesicles in oxygen-glucose deprivation/reperfusion-induced SH-SY5Y cells via stabilizing FUNDC1. Commun Biol 2024; 7:1330. [PMID: 39406943 PMCID: PMC11480199 DOI: 10.1038/s42003-024-07024-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemia-reperfusion (IR) injury is associated with neurological disorders such as stroke. The therapeutic potential of human umbilical cord mesenchymal stem cells (hUC-MSCs) and their secreted extracellular vesicles (EVs) in alleviating IR injury across various cell types including neuronal cells has been documented. However, the underlying mechanisms through which hUC-MSCs and hUC-MSC-EVs protect neuronal cells from IR-triggered damage are not well understood. In this study, we co-cultured SH-SY5Y neuroblastoma cells with hUC-MSCs or hUC-MSC-EVs and subjected them to oxygen-glucose deprivation/reperfusion (OGD/R) treatment. Our findings indicate that both hUC-MSCs and hUC-MSC-EVs significantly improved viability, reduced apoptosis, promoted autophagy of OGD/R-induced SH-SY5Y cells, and decreased mitochondrial reactive oxygen species levels within them. Furthermore, the neuroprotective effect of hUC-MSCs and hUC-MSC-EVs in OGD/R-induced SH-SY5Y cells was enhanced by overexpressing USP35, a deubiquitinase. Mechanistically, USP35 interacted with and stabilized FUNDC1, a positive regulator of mitochondrial metabolism. Knockdown of FUNDC1 in USP35-overexpressing hUC-MSCs and their secreted EVs eliminated the augmented neuroprotective function induced by excess USP35. In conclusion, these findings underscore the crucial role of USP35 in enhancing the neuroprotective function of hUC-MSCs and their secreted EVs, achieved through the stabilization of FUNDC1 in OGD/R-induced SH-SY5Y cells.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Xigong Li
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Tianjiao Wang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310051, People's Republic of China
| | - Zeyu Sun
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Erwei Feng
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Yongming Jin
- Department of Orthopedics Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, People's Republic of China.
| |
Collapse
|
16
|
Lin J, Chen X, Du Y, Li J, Guo T, Luo S. Mitophagy in Cell Death Regulation: Insights into Mechanisms and Disease Implications. Biomolecules 2024; 14:1270. [PMID: 39456203 PMCID: PMC11506020 DOI: 10.3390/biom14101270] [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: 06/18/2024] [Revised: 08/15/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Mitophagy, a selective form of autophagy, plays a crucial role in maintaining optimal mitochondrial populations, normal function, and intracellular homeostasis by monitoring and removing damaged or excess mitochondria. Furthermore, mitophagy promotes mitochondrial degradation via the lysosomal pathway, and not only eliminates damaged mitochondria but also regulates programmed cell death-associated genes, thus preventing cell death. The interaction between mitophagy and various forms of cell death has recently gained increasing attention in relation to the pathogenesis of clinical diseases, such as cancers and osteoarthritis, neurodegenerative, cardiovascular, and renal diseases. However, despite the abundant literature on this subject, there is a lack of understanding regarding the interaction between mitophagy and cell death. In this review, we discuss the main pathways of mitophagy, those related to cell death mechanisms (including apoptosis, ferroptosis, and pyroptosis), and the relationship between mitophagy and cell death uncovered in recent years. Our study offers potential directions for therapeutic intervention and disease diagnosis, and contributes to understanding the molecular mechanism of mitophagy.
Collapse
Affiliation(s)
| | | | | | | | | | - Sai Luo
- The 1st Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin 150000, China; (J.L.); (X.C.); (Y.D.); (J.L.); (T.G.)
| |
Collapse
|
17
|
Zhu Y, Zhang J, Deng Q, Chen X. Mitophagy-associated programmed neuronal death and neuroinflammation. Front Immunol 2024; 15:1460286. [PMID: 39416788 PMCID: PMC11479883 DOI: 10.3389/fimmu.2024.1460286] [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: 07/05/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Mitochondria are crucial organelles that play a central role in cellular metabolism and programmed cell death in eukaryotic cells. Mitochondrial autophagy (mitophagy) is a selective process where damaged mitochondria are encapsulated and degraded through autophagic mechanisms, ensuring the maintenance of both mitochondrial and cellular homeostasis. Excessive programmed cell death in neurons can result in functional impairments following cerebral ischemia and trauma, as well as in chronic neurodegenerative diseases, leading to irreversible declines in motor and cognitive functions. Neuroinflammation, an inflammatory response of the central nervous system to factors disrupting homeostasis, is a common feature across various neurological events, including ischemic, infectious, traumatic, and neurodegenerative conditions. Emerging research suggests that regulating autophagy may offer a promising therapeutic avenue for treating certain neurological diseases. Furthermore, existing literature indicates that various small molecule autophagy regulators have been tested in animal models and are linked to neurological disease outcomes. This review explores the role of mitophagy in programmed neuronal death and its connection to neuroinflammation.
Collapse
Affiliation(s)
- Yanlin Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Quanjun Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
18
|
Peng Y, Jia J, Zhang M, Ma W, Cui Y, Yu M. Transcription Factor TFAP2B Exerts Neuroprotective Effects Targeting BNIP3-Mediated Mitophagy in Ischemia/Reperfusion Injury. Mol Neurobiol 2024; 61:7319-7334. [PMID: 38381297 DOI: 10.1007/s12035-024-04004-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: 08/02/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) leads to malignant brain edema, blood-brain barrier destruction, and neuronal apoptosis. N6-methyladenosine (m6A) RNA modification in CIRI was still limited explored. In this study, MeRIP- and RNA-sequencing were performed of middle cerebral artery occlusion and reperfusion (MCAO/R) rats to find novel potential molecular targets. Transcription factor TFAP2B stood out of which its m6A abundance decreased associated with a marked reduction of its mRNA based on cojoint interactive bioinformatics analysis of the MeRIP- and RNA-sequencing data. It was suggested TFAP2B could have a role in CIRI. Functionally, overexpression of TFAP2B in cultured primary neurons could effectively improve the cell survival and pro-survival autophagy in parallel with reduced cell apoptosis during OGD/R in vitro. Through the RNA-sequencing of TFAP2B overexpressed primary neurons and subsequent validation experiments, it was found that mitophagy receptor BNIP3 was one of the important targets of TFAP2B in OGD/R neurons through which TFAP2B could bind to its promoter region for transcriptional activation of BNIP3, thereby enhancing BNIP3-mediated mitophagy to protect against OGD/R injury of neurons. Lastly, TFAP2B was demonstrated to alleviate the MCAO/R damage to a certain extent in vivo. Although it failed to confirm TFAP2B dysregulation was m6A dependent in current research, this is the first research of TFAP2B in CIRI field with important guiding significance.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Middle Road, Changsha, Hunan, 410011, People's Republic of China
| | - Jiaoying Jia
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Middle Road, Changsha, Hunan, 410011, People's Republic of China
| | - Mingming Zhang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Middle Road, Changsha, Hunan, 410011, People's Republic of China
| | - Wenjia Ma
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Middle Road, Changsha, Hunan, 410011, People's Republic of China
| | - Yan Cui
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Middle Road, Changsha, Hunan, 410011, People's Republic of China
| | - Mengqiang Yu
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Middle Road, Changsha, Hunan, 410011, People's Republic of China.
| |
Collapse
|
19
|
Burton JC, Royer F, Grimsey NJ. Spatiotemporal control of kinases and the biomolecular tools to trace activity. J Biol Chem 2024; 300:107846. [PMID: 39362469 PMCID: PMC11550616 DOI: 10.1016/j.jbc.2024.107846] [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: 07/02/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
The delicate balance of cell physiology is implicitly tied to the expression and activation of proteins. Post-translational modifications offer a tool to dynamically switch protein activity on and off to orchestrate a wide range of protein-protein interactions to tune signal transduction during cellular homeostasis and pathological responses. There is a growing acknowledgment that subcellular locations of kinases define the spatial network of potential scaffolds, adaptors, and substrates. These highly ordered and localized biomolecular microdomains confer a spatially distinct bias in the outcomes of kinase activity. Furthermore, they may hold essential clues to the underlying mechanisms that promote disease. Developing tools to dissect the spatiotemporal activation of kinases is critical to reveal these mechanisms and promote the development of spatially targeted kinase inhibitors. Here, we discuss the spatial regulation of kinases, the tools used to detect their activity, and their potential impact on human health.
Collapse
Affiliation(s)
- Jeremy C Burton
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, Athens, Georgia, USA
| | - Fredejah Royer
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, Athens, Georgia, USA
| | - Neil J Grimsey
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, Athens, Georgia, USA.
| |
Collapse
|
20
|
Liu F, Chen Y, Huang K. Electro-acupuncture Suppresses Ferroptosis to Alleviate Cerebral Ischemia-Reperfusion Injury Through KAT3B-Mediated Succinylation of ACSL4. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05063-6. [PMID: 39340629 DOI: 10.1007/s12010-024-05063-6] [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] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Electro-acupuncture (EA) is identified as an effective therapeutic method for cerebral ischemia/reperfusion injury (CIRI), which is a combination of Chinese traditional acupuncture and modern electro-therapy. However, the downstream molecular mechanisms of EA in CIRI process remains largely unknown. The purpose of the present study is to unveil the therapeutic effect of EA on CIRI rat and its regulatory mechanisms. At first, we constructed middle cerebral artery occlusion (MCAO) rat models and then treated them with EA to observe the pathological changes. The results indicated that EA decreased the infarct volume (43.81 ± 3.34 vs 15.96 ± 2.22) and the neurological scores (3.33 ± 0.52 vs 1.67 ± 0.52) and suppressed the apoptosis in MCAO model rats. For ferroptosis analysis, EA decreased the Fe2 + (0.08 ± 0.01 vs 0.06 ± 0.01), MDA (36.61 ± 4.29 vs 21.72 ± 2.79), and LPS (5.25 ± 0.69 vs 2.89 ± 0.42) contents and increased the GSH (4.94 ± 1.04 vs 11.69 ± 1.88) content in MCAO model rats. We next detected whether succinylation mediated EA-treated I/R injury. According to immunoprecipitation and western blot analysis, EA treatment could lower both levels of succinylation and KAT3B in MCAO rats. Moreover, mechanism experiments unveiled that KAT3B promoted the succinylation of the ferroptosis-related protein ACSL4 at K661 site and thus stabilizing ACSL4. Finally, EA-treated MCAO rats were further injected with KAT3B expression vector. The results showed that KAT3B overexpression increased the infarct volume (31.44 ± 3.92 vs 7.94 ± 2.84) and the neurological scores (2.67 ± 0.51 vs 1.33 ± 0.51) and promoted the apoptosis in EA treated MCAO model rats. For ferroptosis analysis, KAT3B overexpression increased the Fe2 + (0.08 ± 0.01 vs 0.05 ± 0.01), MDA (29.24 ± 4.30 vs 22.06 ± 1.89), and LPO (5.07 ± 0.45 vs 2.88 ± 0.49) contents and decreased the GSH (7.86 ± 1.09 vs 11.06 ± 1.76) content in EA treated MCAO model rats. Collectively, our study demonstrates that EA plays a therapeutic role in CIRI through suppressing KAT3B-induced stabilization of ACSL4 to inhibit ferroptosis. These findings contribute to our understanding of the molecular mechanisms underlying the neuroprotective effects of EA and open new avenues for the development of innovative therapeutic strategies for CIRI.
Collapse
Affiliation(s)
- Fang Liu
- Department of Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Chen
- Department of Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Kangbai Huang
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, China.
| |
Collapse
|
21
|
Wu Y, Xu Y, Deng H, Sun J, Li X, Tang J. Poricoic acid a ameliorates high glucose-induced podocyte injury by regulating the AMPKα/FUNDC1 pathway. Mol Biol Rep 2024; 51:1003. [PMID: 39305364 DOI: 10.1007/s11033-024-09921-8] [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: 05/23/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Poricoic acid A (PAA), a major triterpenoid component of Poria cocos with anti-tumor, anti-fibrotic, anti-inflammatory, and immune-regulating activities, has been shown to induce podocyte autophagy in diabetic kidney disease (DKD) by downregulating FUN14 domain containing 1 (FUNDC1). This study aimed to identify the role of adenosine monophosphate-activated protein kinase alpha (AMPKα) in PAA-mediated phosphorylation of FUNDC1 in podocyte injury occurring in the pathogenesis of DKD. METHODS AND RESULTS A cellular model of renal podocyte injury was established by culturing MPC5 cells under high-glucose (HG) conditions. MPC5 cells were subjected to transfection with small interfering RNA (siRNA) targeting AMPKα or siRNA targeting FUNDC1, an AMPKα activator, or PAA. PAA treatment induced the phosphorylation of AMPKα in HG-cultured podocytes. AMPKα activation was implicated in the inhibitory effect of PAA on FUNDC phosphorylation in HG-cultured podocytes. Treatment targeting the AMPKα activator also significantly augmented proliferation, migration, mitochondrial membrane potential, and autophagy levels, while reducing apoptosis levels, inhibiting oxidative stress, and suppressing the release of proinflammatory factors in HG-cultured MPC5 cells. In contrast, insufficient expression of AMPKα reversed the effects of PAA on the proliferation, migration, and apoptosis of podocytes and further exacerbated the reduction of phosphorylated FUNDC1 expression in podocytes under HG conditions. CONCLUSIONS AMPKα is involved in the regulation of FUNDC1 phosphorylation by PAA in HG-induced podocyte injury. Furthermore, the AMPKα/FUNDC1 pathway plays a crucial regulatory role in HG-induced podocyte injury. These findings support AMPKα, FUNDC1, and the AMPKα/FUNDC1 pathway as targets for PAA intervention.
Collapse
Affiliation(s)
- Yuwen Wu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, No.167 Donghu Road, Wuhan, 430071, Hubei, China.
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, No.167 Donghu Road, Wuhan, 430071, Hubei, China
| | - Haohua Deng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, No.167 Donghu Road, Wuhan, 430071, Hubei, China
| | - Jiazhong Sun
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, No.167 Donghu Road, Wuhan, 430071, Hubei, China
| | - Xin Li
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, No.167 Donghu Road, Wuhan, 430071, Hubei, China
| | - Jun Tang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, No.167 Donghu Road, Wuhan, 430071, Hubei, China
| |
Collapse
|
22
|
Jiang XL, Zhang ZB, Feng CX, Lin CJ, Yang H, Tan LL, Ding X, Xu LX, Li G, Pan T, Qin ZH, Sun B, Feng X, Li M. PHLDA1 contributes to hypoxic ischemic brain injury in neonatal rats via inhibiting FUNDC1-mediated mitophagy. Acta Pharmacol Sin 2024; 45:1809-1820. [PMID: 38750074 PMCID: PMC11336168 DOI: 10.1038/s41401-024-01292-x] [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: 01/22/2024] [Accepted: 04/14/2024] [Indexed: 08/22/2024] Open
Abstract
Hypoxia-ischemia (HI) is one of the main causes of neonatal brain injury. Mitophagy has been implicated in the degradation of damaged mitochondria and cell survival following neonatal brain HI injury. Pleckstrin homology-like domain family A member 1 (PHLDA1) plays vital roles in the progression of various disorders including the regulation of oxidative stress, the immune responses and apoptosis. In the present study we investigated the role of PHLDA1 in HI-induced neuronal injury and further explored the mechanisms underlying PHLDA1-regulated mitophagy in vivo and in vitro. HI model was established in newborn rats by ligation of the left common carotid artery plus exposure to an oxygen-deficient chamber with 8% O2 and 92% N2. In vitro studies were conducted in primary hippocampal neurons subjected to oxygen and glucose deprivation/-reoxygenation (OGD/R). We showed that the expression of PHLDA1 was significantly upregulated in the hippocampus of HI newborn rats and in OGD/R-treated primary neurons. Knockdown of PHLDA1 in neonatal rats via lentiviral vector not only significantly ameliorated HI-induced hippocampal neuronal injury but also markedly improved long-term cognitive function outcomes, whereas overexpression of PHLDA1 in neonatal rats via lentiviral vector aggravated these outcomes. PHLDA1 knockdown in primary neurons significantly reversed the reduction of cell viability and increase in intracellular reactive oxygen species (ROS) levels, and attenuated OGD-induced mitochondrial dysfunction, whereas overexpression of PHLDA1 decreased these parameters. In OGD/R-treated primary hippocampal neurons, we revealed that PHLDA1 knockdown enhanced mitophagy by activating FUNDC1, which was abolished by FUNDC1 knockdown or pretreatment with mitophagy inhibitor Mdivi-1 (25 μM). Notably, pretreatment with Mdivi-1 or the knockdown of FUNDC1 not only increased brain infarct volume, but also abolished the neuroprotective effect of PHLDA1 knockdown in HI newborn rats. Together, these results demonstrate that PHLDA1 contributes to neonatal HI-induced brain injury via inhibition of FUNDC1-mediated neuronal mitophagy.
Collapse
Affiliation(s)
- Xiao-Lu Jiang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Zu-Bin Zhang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsycho Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Chen-Xi Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Chen-Jie Lin
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Hui Yang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Lan-Lan Tan
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xin Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Li-Xiao Xu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Gen Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Tao Pan
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Zheng-Hong Qin
- Institute of Health Technology, Global Institute of Software Technology, Qingshan Road, Suzhou Science & Technology Tower, Hi-Tech Area, Suzhou, 215163, China
| | - Bin Sun
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xing Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
23
|
Tang T, Hu LB, Ding C, Zhang Z, Wang N, Wang T, Zhou H, Xia S, Fan L, Fu XJ, Yan F, Zhang X, Chen G, Li J. Src inhibition rescues FUNDC1-mediated neuronal mitophagy in ischaemic stroke. Stroke Vasc Neurol 2024; 9:367-379. [PMID: 37793899 PMCID: PMC11420917 DOI: 10.1136/svn-2023-002606] [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: 05/19/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Ischaemic stroke triggers neuronal mitophagy, while the involvement of mitophagy receptors in ischaemia/reperfusion (I/R) injury-induced neuronal mitophagy remain not fully elucidated. Here, we aimed to investigate the involvement of mitophagy receptor FUN14 domain-containing 1 (FUNDC1) and its modulation in neuronal mitophagy induced by I/R injury. METHODS Wild-type and FUNDC1 knockout mice were generated to establish models of neuronal I/R injury, including transient middle cerebral artery occlusion (tMCAO) in vivo and oxygen glucose deprivation/reperfusion in vitro. Stroke outcomes of mice with two genotypes were assessed. Neuronal mitophagy was analysed both in vivo and in vitro. Activities of FUNDC1 and its regulator Src were evaluated. The impact of Src on FUNDC1-mediated mitophagy was assessed through administration of Src antagonist PP1. RESULTS To our surprise, FUNDC1 knockout mice subjected to tMCAO showed stroke outcomes comparable to those of their wild-type littermates. Although neuronal mitophagy could be activated by I/R injury, FUNDC1 deletion did not disrupt neuronal mitophagy. Transient activation of FUNDC1, represented by dephosphorylation of Tyr18, was detected in the early stages (within 3 hours) of neuronal I/R injury; however, phosphorylated Tyr18 reappeared and even surpassed baseline levels in later stages (after 6 hours), accompanied by a decrease in FUNDC1-light chain 3 interactions. Spontaneous inactivation of FUNDC1 was associated with Src activation, represented by phosphorylation of Tyr416, which changed in parallel with the level of phosphorylated FUNDC1 (Tyr18) during neuronal I/R injury. Finally, FUNDC1-mediated mitophagy in neurons under I/R conditions can be rescued by pharmacological inhibition of Src. CONCLUSIONS FUNDC1 is inactivated by Src during the later stage (after 6 hours) of neuronal I/R injury, and rescue of FUNDC1-mediated mitophagy may serve as a potential therapeutic strategy for treating ischaemic stroke.
Collapse
Affiliation(s)
- Tianchi Tang
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Li-Bin Hu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Chao Ding
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhihua Zhang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ning Wang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tingting Wang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hang Zhou
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Siqi Xia
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Linfeng Fan
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Xiong-Jie Fu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Feng Yan
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Xiangnan Zhang
- Zhejiang University Department of Pharmacology, Hangzhou, Zhejiang, China
| | - Gao Chen
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jianru Li
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Xu L, Mi Y, Meng Q, Liu Y, Wang Y, Zhang Y, Yang Y, Chen G, Liu Y, Hou Y. A quinolinyl resveratrol derivative alleviates acute ischemic stroke injury by promoting mitophagy for neuroprotection via targeting CK2α'. Int Immunopharmacol 2024; 137:112524. [PMID: 38909494 DOI: 10.1016/j.intimp.2024.112524] [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: 05/02/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
Ischemic stroke (IS) is a serious threat to human health. The naturally derived small molecule (E)-5-(2-(quinolin-4-yl) ethenyl) benzene-1,3-diol (RV01) is a quinolinyl analog of resveratrol with great potential in the treatment of IS. The aim of this study was to investigate the potential mechanisms and targets for the protective effect of the RV01 on IS. The mouse middle cerebral artery occlusion and reperfusion (MCAO/R) and oxygen-glucose deprivation and reperfusion (OGD/R) models were employed to evaluate the effects of RV01 on ischemic injury and neuroprotection. RV01 was found to significantly increase the survival of SH-SY5Y cells and prevent OGD/R-induced apoptosis in SH-SY5Y cells. Furthermore, RV01 reduced oxidative stress and mitochondrial damage by promoting mitophagy in OGD/R-exposed SH-SY5Y cells. Knockdown of CK2α' abolished the RV01-mediated promotion on mitophagy and alleviation on mitochondrial damage as well as neuronal injury after OGD/R. These results were further confirmed by molecular docking, drug affinity responsive target stability and cellular thermal shift assay analysis. Importantly, in vivo study showed that treatment with the CK2α' inhibitor CX-4945 abolished the RV01-mediated alleviation of cerebral infarct volume, brain edema, cerebral blood flow and neurological deficit in MCAO/R mice. These data suggest that RV01 effectively reduces damage caused by acute ischemic stroke by promoting mitophagy through its interaction with CK2α'. These findings offer valuable insights into the underlying mechanisms through which RV01 exerts its therapeutic effects on IS.
Collapse
Affiliation(s)
- Libin Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yan Mi
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Qingqi Meng
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yeshu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yongping Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Ying Zhang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yuxin Yang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yueyang Liu
- Shenyang Key Laboratory of Vascular Biology, Science and Research Center, Department of Pharmacology, Shenyang Medical College, Shenyang, China.
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China.
| |
Collapse
|
25
|
Lyu Y, Meng Z, Hu Y, Jiang B, Yang J, Chen Y, Zhou J, Li M, Wang H. Mechanisms of mitophagy and oxidative stress in cerebral ischemia-reperfusion, vascular dementia, and Alzheimer's disease. Front Mol Neurosci 2024; 17:1394932. [PMID: 39169952 PMCID: PMC11335644 DOI: 10.3389/fnmol.2024.1394932] [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: 03/02/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Neurological diseases have consistently represented a significant challenge in both clinical treatment and scientific research. As research has progressed, the significance of mitochondria in the pathogenesis and progression of neurological diseases has become increasingly prominent. Mitochondria serve not only as a source of energy, but also as regulators of cellular growth and death. Both oxidative stress and mitophagy are intimately associated with mitochondria, and there is mounting evidence that mitophagy and oxidative stress exert a pivotal regulatory influence on the pathogenesis of neurological diseases. In recent years, there has been a notable rise in the prevalence of cerebral ischemia/reperfusion injury (CI/RI), vascular dementia (VaD), and Alzheimer's disease (AD), which collectively represent a significant public health concern. Reduced levels of mitophagy have been observed in CI/RI, VaD and AD. The improvement of associated pathology has been demonstrated through the increase of mitophagy levels. CI/RI results in cerebral tissue ischemia and hypoxia, which causes oxidative stress, disruption of the blood-brain barrier (BBB) and damage to the cerebral vasculature. The BBB disruption and cerebral vascular injury may induce or exacerbate VaD to some extent. In addition, inadequate cerebral perfusion due to vascular injury or altered function may exacerbate the accumulation of amyloid β (Aβ) thereby contributing to or exacerbating AD pathology. Intravenous tissue plasminogen activator (tPA; alteplase) and endovascular thrombectomy are effective treatments for stroke. However, there is a narrow window of opportunity for the administration of tPA and thrombectomy, which results in a markedly elevated incidence of disability among patients with CI/RI. It is regrettable that there are currently no there are still no specific drugs for VaD and AD. Despite the availability of the U.S. Food and Drug Administration (FDA)-approved clinical first-line drugs for AD, including memantine, donepezil hydrochloride, and galantamine, these agents do not fundamentally block the pathological process of AD. In this paper, we undertake a review of the mechanisms of mitophagy and oxidative stress in neurological disorders, a summary of the clinical trials conducted in recent years, and a proposal for a new strategy for targeted treatment of neurological disorders based on both mitophagy and oxidative stress.
Collapse
Affiliation(s)
- Yujie Lyu
- Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, China
- Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, China
| | - Zhipeng Meng
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Yunyun Hu
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Bing Jiang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Jiao Yang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Yiqin Chen
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Jun Zhou
- Xichang Hospital of Traditional Chinese Medicine, Xichang, China
| | - Mingcheng Li
- Qujing 69 Hospital, China RongTong Medical Healthcare Group Co. Ltd, Qujing, China
| | - Huping Wang
- Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, China
- Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, China
| |
Collapse
|
26
|
Zhao L, Chen Y, Li H, Ding X, Li J. Deciphering the neuroprotective mechanisms of RACK1 in cerebral ischemia-reperfusion injury: Pioneering insights into mitochondrial autophagy and the PINK1/Parkin axis. CNS Neurosci Ther 2024; 30:e14836. [PMID: 39097918 PMCID: PMC11298203 DOI: 10.1111/cns.14836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 05/23/2024] [Accepted: 06/16/2024] [Indexed: 08/06/2024] Open
Abstract
INTRODUCTION Cerebral ischemia-reperfusion injury (CIRI) is a common and debilitating complication of cerebrovascular diseases such as stroke, characterized by mitochondrial dysfunction and cell apoptosis. Unraveling the molecular mechanisms behind these processes is essential for developing effective CIRI treatments. This study investigates the role of RACK1 (receptor for activated C kinase 1) in CIRI and its impact on mitochondrial autophagy. METHODS We utilized high-throughput transcriptome sequencing and weighted gene co-expression network analysis (WGCNA) to identify core genes associated with CIRI. In vitro experiments used human neuroblastoma SK-N-SH cells subjected to oxygen and glucose deprivation (OGD) to simulate ischemia, followed by reperfusion (OGD/R). RACK1 knockout cells were created using CRISPR/Cas9 technology, and cell viability, apoptosis, and mitochondrial function were assessed. In vivo experiments involved middle cerebral artery occlusion/reperfusion (MCAO/R) surgery in rats, evaluating neurological function and cell apoptosis. RESULTS Our findings revealed that RACK1 expression increases during CIRI and is protective by regulating mitochondrial autophagy through the PINK1/Parkin pathway. In vitro, RACK1 knockout exacerbated cell apoptosis, while overexpression of RACK1 reversed this process, enhancing mitochondrial function. In vivo, RACK1 overexpression reduced cerebral infarct volume and improved neurological deficits. The regulatory role of RACK1 depended on the PINK1/Parkin pathway, with RACK1 knockout inhibiting PINK1 and Parkin expression, while RACK1 overexpression restored them. CONCLUSION This study demonstrates that RACK1 safeguards against neural damage in CIRI by promoting mitochondrial autophagy through the PINK1/Parkin pathway. These findings offer crucial insights into the regulation of mitochondrial autophagy and cell apoptosis by RACK1, providing a promising foundation for future CIRI treatments.
Collapse
Affiliation(s)
- Lanqing Zhao
- Department of Sleep Medicine CenterThe Shengjing Affiliated Hospital, China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Yu Chen
- Department of Otorhinolaryngology Head and NeckShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Hongxi Li
- Department of Pain ManagementShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Xiaoxu Ding
- Department of Otorhinolaryngology Head and NeckShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Jinwei Li
- Department of Neurology/Stroke CenterThe First Affiliated Hospital of China Medical University, China Medical UniversityShenyangLiaoningPeople's Republic of China
| |
Collapse
|
27
|
Han Q, Yu Y, Liu X, Guo Y, Shi J, Xue Y, Li Y. The Role of Endothelial Cell Mitophagy in Age-Related Cardiovascular Diseases. Aging Dis 2024:AD.2024.0788. [PMID: 39122456 DOI: 10.14336/ad.2024.0788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Aging is a major risk factor for cardiovascular diseases (CVD), and mitochondrial autophagy impairment is considered a significant physiological change associated with aging. Endothelial cells play a crucial role in maintaining vascular homeostasis and function, participating in various physiological processes such as regulating vascular tone, coagulation, angiogenesis, and inflammatory responses. As aging progresses, mitochondrial autophagy impairment in endothelial cells worsens, leading to the development of numerous cardiovascular diseases. Therefore, regulating mitochondrial autophagy in endothelial cells is vital for preventing and treating age-related cardiovascular diseases. However, there is currently a lack of systematic reviews in this area. To address this gap, we have written this review to provide new research and therapeutic strategies for managing aging and age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Quancheng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiujuan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yonghong Guo
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
28
|
Ma L, Li H, Xu H, Liu D. The potential roles of PKM2 in cerebrovascular diseases. Int Immunopharmacol 2024; 139:112675. [PMID: 39024754 DOI: 10.1016/j.intimp.2024.112675] [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: 05/29/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Pyruvate kinase M2 (PKM2), a key enzyme involved in glycolysis,plays an important role in regulating cell metabolism and growth under different physiological conditions. PKM2 has been intensively investigated in multiple cancer diseases. Recent years, many studies have found its pivotal role in cerebrovascular diseases (CeVDs), the disturbances in intracranial blood circulation. CeVDs has been confirmed to be closely associated with oxidative stress (OS), mitochondrial dynamics, systemic inflammation, and local neuroinflammation in the brain. It has further been revealed that PKM2 exerts various biological functions in the regulation of energy supply, OS, inflammatory responses, and mitochondrial dysfunction. The roles of PKM2 are closely related to its different isoforms, expression levels in subcellular localization, and post-translational modifications. Therefore, summarizing the roles of PKM2 in CeVDs will help further understanding the molecular mechanisms of CeVDs. In this review, we illustrate the characteristics of PKM2, the regulated PKM2 expression, and the biological roles of PKM2 in CeVDs.
Collapse
Affiliation(s)
- Ling Ma
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Huatao Li
- Department of Stroke Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Hu Xu
- Department of Stroke Center, Shandong Second Medical University, Weifang, Shandong 261000, China
| | - Dianwei Liu
- Department of Stroke Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Department of Neurosurgery, XuanWu Hospital Capital Medical University Jinan Branch, Jinan, Shandong 250100, China.
| |
Collapse
|
29
|
Guo X, Zhang Z, Gu J, Ke P, Liu J, Meng Y, Zheng W, Que W, Fan R, Luo J, Xiao F. FUDNC1-dependent mitophagy ameliorate motor neuron death in an amyotrophic lateral sclerosis mouse model. Neurobiol Dis 2024; 197:106534. [PMID: 38759931 DOI: 10.1016/j.nbd.2024.106534] [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: 01/11/2024] [Revised: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is one of the most common neurodegenerative diseases, yet effective treatment is lacking. Moreover, the underlying pathomechanisms of ALS remain unclear, with impaired mitophagy function being increasingly recognized as a contributing factor. FUN14 domain-containing protein 1 (FUNDC1) is an autophagy receptor localized to the outer mitochondrial membrane and a mitochondrial membrane protein that mediates mitophagy and therefore considered as important factor in neurodegenerative diseases. However, its specific role in ALS is not yet clear. Therefore, this study aimed to investigate the regulatory role of FUNDC1 in ALS and determine its regulatory mechanisms. ALS transgenic mice were obtained and maintained under standard conditions. Cell lines were generated by stable transfection with hSOD1G93A or control vectors. Mice received intrathecal injections of AAV9 vectors expressing FUNDC1 or EGFP. Motor function was assessed through behavioral tests, and histological and immunostaining analyses were performed. Colocalization analysis was conducted in transfected cells, and protein expression was evaluated via western blotting. We first observed that FUNDC1 was significantly downregulated in the spinal cord tissues of SOD1G93A mice. FUNDC1 overexpression considerably improved locomotor activity and prolonged survival time in SOD1G93A mice. Mechanistically, reduced expression of FUNDC1 resulted in decreased mitophagy, as indicated by decreased recruitment through LC3 in SOD1G93A mice and cellular models. Consequently, this led to increased mitochondrial accumulation and cell apoptosis, exacerbating the ALS phenotype. Furthermore, we identified transcription factor FOXD3 as an essential upstream factor of FUNDC1, resulting in reduced transcription of FUNDC1 in ALS lesions. This study suggests a novel strategy of targeting FUNDC1-mediated mitophagy for developing therapeutic interventions to mitigate disease progression and improve outcomes for ALS patients.
Collapse
Affiliation(s)
- Xia Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China; Department of Neurology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Zhuo Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Juan Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China; Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - PingYang Ke
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Jing Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Yuan Meng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Wei Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - WenJun Que
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Rui Fan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Jing Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China.
| | - Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China.
| |
Collapse
|
30
|
Chen Y, Zhang Y, Wu Q, Chen J, Deng Y. The neuroprotective effect of Chinese herbal medicine for cerebral ischemia reperfusion injury through regulating mitophagy. Front Pharmacol 2024; 15:1378358. [PMID: 38895624 PMCID: PMC11183336 DOI: 10.3389/fphar.2024.1378358] [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: 01/29/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
The incidence of ischemic stroke has been increasing annually with an unfavorable prognosis. Cerebral ischemia reperfusion injury can exacerbate nerve damage. Effective mitochondrial quality control including mitochondrial fission, fusion and autophagy, is crucial for maintaining cellular homeostasis. Several studies have revealed the critical role of mitophagy in Cerebral ischemia reperfusion injury. Cerebral ischemia and hypoxia induce mitophagy, and mitophagy exhibits positive and negative effects in cerebral ischemia reperfusion injury. Studies have shown that Chinese herbal medicine can alleviate Cerebral ischemia reperfusion injury and serve as a neuroprotective agent by inhibiting or promoting mitophagy-mediated pathways. This review focuses on the mitochondrial dynamics and mitophagy-related pathways, as well as the role of mitophagy in ischemia reperfusion injury. Additionally, it discusses the therapeutic potential and benefits of Chinese herbal monomers and decoctions in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yanling Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yanan Zhang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qin Wu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province Key Laboratory of Cerebrovascular Disease Prevention and Treatment of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yihui Deng
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
31
|
Geng Y, Qiu L, Cheng Y, Li J, Ma Y, Zhao C, Cai Y, Zhang X, Chen J, Pan Y, Wang K, Yao X, Guo D, Wu J. Alleviating Recombinant Tissue Plasminogen Activator-induced Hemorrhagic Transformation in Ischemic Stroke via Targeted Delivery of a Ferroptosis Inhibitor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309517. [PMID: 38647405 PMCID: PMC11199968 DOI: 10.1002/advs.202309517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/02/2024] [Indexed: 04/25/2024]
Abstract
Intravenous thrombolysis with recombinant tissue plasminogen activator (rtPA) is the primary treatment for ischemic stroke. However, rtPA treatment can substantially increase blood-brain barrier (BBB) permeability and susceptibility to hemorrhagic transformation. Herein, the mechanism underlying the side effects of rtPA treatment is investigated and demonstrated that ferroptosis plays an important role. The ferroptosis inhibitor, liproxstatin-1 (Lip) is proposed to alleviate the side effects. A well-designed macrocyclic carrier, glucose-modified azocalix[4]arene (GluAC4A), is prepared to deliver Lip to the ischemic site. GluAC4A bound tightly to Lip and markedly improved its solubility. Glucose, modified at the upper rim of GluAC4A, imparts BBB targeting to the drug delivery system owing to the presence of glucose transporter 1 on the BBB surface. The responsiveness of GluAC4A to hypoxia due to the presence of azo groups enabled the targeted release of Lip at the ischemic site. GluAC4A successfully improved drug accumulation in the brain, and Lip@GluAC4A significantly reduced ferroptosis, BBB leakage, and neurological deficits induced by rtPA in vivo. These findings deepen the understanding of the side effects of rtPA treatment and provide a novel strategy for their effective mitigation, which is of great significance for the treatment and prognosis of patients with ischemic stroke.
Collapse
Affiliation(s)
- Yan‐Qin Geng
- School of MedicineNankai UniversityTianjin300071China
- Tianjin Huanhu HospitalTianjin300350China
| | - Li‐Na Qiu
- Department of NeurologyTianjin Huanhu HospitalTianjin300350China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases Tianjin Neurosurgical InstituteTianjin Huanhu HospitalTianjin300350China
| | - Yuan‐Qiu Cheng
- College of ChemistryState Key Laboratory of Elemento‐Organic ChemistryKey Laboratory of Functional Polymer Materials (Ministry of Education)Frontiers Science Center for New Organic MatterCollaborative Innovation Center of Chemical Science and Engineering (Tianjin)Nankai UniversityTianjin300071China
| | - Juan‐Juan Li
- College of Chemistry and Environmental ScienceKey Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education)Key Laboratory of Chemical Biology of Hebei ProvinceHebei UniversityBaoding071002China
| | - Yi‐Lin Ma
- Clinical College of NeurologyNeurosurgery and NeurorehabilitationTianjin Medical UniversityTianjin300071China
| | - Cheng‐Cheng Zhao
- Clinical College of NeurologyNeurosurgery and NeurorehabilitationTianjin Medical UniversityTianjin300071China
| | - Ying Cai
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases Tianjin Neurosurgical InstituteTianjin Huanhu HospitalTianjin300350China
| | - Xue‐Bin Zhang
- Department of PathologyTianjin Huanhu HospitalTianjin300350China
| | - Jieli Chen
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases Tianjin Neurosurgical InstituteTianjin Huanhu HospitalTianjin300350China
| | - Yu‐Chen Pan
- College of ChemistryState Key Laboratory of Elemento‐Organic ChemistryKey Laboratory of Functional Polymer Materials (Ministry of Education)Frontiers Science Center for New Organic MatterCollaborative Innovation Center of Chemical Science and Engineering (Tianjin)Nankai UniversityTianjin300071China
| | - Ke‐Rang Wang
- College of Chemistry and Environmental ScienceKey Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education)Key Laboratory of Chemical Biology of Hebei ProvinceHebei UniversityBaoding071002China
| | - Xiu‐Hua Yao
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases Tianjin Neurosurgical InstituteTianjin Huanhu HospitalTianjin300350China
| | - Dong‐Sheng Guo
- College of ChemistryState Key Laboratory of Elemento‐Organic ChemistryKey Laboratory of Functional Polymer Materials (Ministry of Education)Frontiers Science Center for New Organic MatterCollaborative Innovation Center of Chemical Science and Engineering (Tianjin)Nankai UniversityTianjin300071China
- Xinjiang Key Laboratory of Novel Functional Materials ChemistryCollege of Chemistry and Environmental SciencesKashi UniversityKashi844000China
| | - Jia‐Ling Wu
- School of MedicineNankai UniversityTianjin300071China
- Tianjin Huanhu HospitalTianjin300350China
- Department of NeurologyTianjin Huanhu HospitalTianjin300350China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases Tianjin Neurosurgical InstituteTianjin Huanhu HospitalTianjin300350China
| |
Collapse
|
32
|
Wu J, Yang Y, Lin D, Wang Z, Ma J. SIRT3 and RORα are two prospective targets against mitophagy during simulated ischemia/reperfusion injury in H9c2 cells. Heliyon 2024; 10:e30568. [PMID: 38784556 PMCID: PMC11112282 DOI: 10.1016/j.heliyon.2024.e30568] [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: 01/31/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Autophagy during myocardial ischemia/reperfusion (MI/R) exacerbates cardiomyocyte injury. Melatonin (Mel) alleviates myocardial damage by regulating mitochondrial function and mitophagy, but the role of mitophagy in melatonin-induced cardioprotection remains unclear. This study aimed to explore the roles of sirtuin3 (SIRT3) and retinoid-related orphan nuclear receptor-α (RORα) in mitophagy during simulated ischemia reperfusion (SIR) in H9c2 cells. Our data showed that mitophagy was excessively activated after SIR injury, which was consistent with reduced cell survival, enhanced oxidative responses and mitochondrial dysfunction in H9c2 myocytes. Melatonin greatly enhanced cell viability, reduced oxidative stress and improved mitochondrial function. The effects of melatonin protection were involved in excessive mitophagy inhibition, as demonstrated by the reduced levels of mitophagy-linked proteins, including Parkin, Beclin1, NIX and BNIP3, and the LC3 II/LC3 I ratio and elevations in p62. Additionally, the decreases in SIRT3 and RORα in H9c2 myocytes after SIR were reversed by melatonin, and the above effects of melatonin were eliminated by small interfering RNA (siRNA)-mediated knockdown of SIRT3 and RORα. In brief, SIRT3 and RORα are two prospective targets in the cardioprotection of melatonin against mitophagy during SIR in H9c2 myocytes.
Collapse
Affiliation(s)
- Jinjing Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Yanli Yang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Zhaoqi Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| |
Collapse
|
33
|
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
|
34
|
Wang Y, Yang J. ER-organelle contacts: A signaling hub for neurological diseases. Pharmacol Res 2024; 203:107149. [PMID: 38518830 DOI: 10.1016/j.phrs.2024.107149] [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: 11/28/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Neuronal health is closely linked to the homeostasis of intracellular organelles, and organelle dysfunction affects the pathological progression of neurological diseases. In contrast to isolated cellular compartments, a growing number of studies have found that organelles are largely interdependent structures capable of communicating through membrane contact sites (MCSs). MCSs have been identified as key pathways mediating inter-organelle communication crosstalk in neurons, and their alterations have been linked to neurological disease pathology. The endoplasmic reticulum (ER) is a membrane-bound organelle capable of forming an extensive network of pools and tubules with important physiological functions within neurons. There are multiple MCSs between the ER and other organelles and the plasma membrane (PM), which regulate a variety of cellular processes. In this review, we focus on ER-organelle MCSs and their role in a variety of neurological diseases. We compared the biological effects between different tethering proteins and the effects of their respective disease counterparts. We also discuss how altered ER-organelle contacts may affect disease pathogenesis. Therefore, understanding the molecular mechanisms of ER-organelle MCSs in neuronal homeostasis will lay the foundation for the development of new therapies targeting ER-organelle contacts.
Collapse
Affiliation(s)
- Yunli Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Jinghua Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
35
|
He Y, He T, Li H, Chen W, Zhong B, Wu Y, Chen R, Hu Y, Ma H, Wu B, Hu W, Han Z. Deciphering mitochondrial dysfunction: Pathophysiological mechanisms in vascular cognitive impairment. Biomed Pharmacother 2024; 174:116428. [PMID: 38599056 DOI: 10.1016/j.biopha.2024.116428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Vascular cognitive impairment (VCI) encompasses a range of cognitive deficits arising from vascular pathology. The pathophysiological mechanisms underlying VCI remain incompletely understood; however, chronic cerebral hypoperfusion (CCH) is widely acknowledged as a principal pathological contributor. Mitochondria, crucial for cellular energy production and intracellular signaling, can lead to numerous neurological impairments when dysfunctional. Recent evidence indicates that mitochondrial dysfunction-marked by oxidative stress, disturbed calcium homeostasis, compromised mitophagy, and anomalies in mitochondrial dynamics-plays a pivotal role in VCI pathogenesis. This review offers a detailed examination of the latest insights into mitochondrial dysfunction within the VCI context, focusing on both the origins and consequences of compromised mitochondrial health. It aims to lay a robust scientific groundwork for guiding the development and refinement of mitochondrial-targeted interventions for VCI.
Collapse
Affiliation(s)
- Yuyao He
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Tiantian He
- Sichuan Academy of Chinese Medicine Sciences, China
| | - Hongpei Li
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wei Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Biying Zhong
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yue Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Runming Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yuli Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Huaping Ma
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Bin Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wenyue Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Zhenyun Han
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
36
|
Wu P, Xiao Y, Qing L, Mi Y, Tang J, Cao Z, Huang C. Emodin activates autophagy to suppress oxidative stress and pyroptosis via mTOR-ULK1 signaling pathway and promotes multi-territory perforator flap survival. Biochem Biophys Res Commun 2024; 704:149688. [PMID: 38387327 DOI: 10.1016/j.bbrc.2024.149688] [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: 01/09/2024] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Multi-territory perforator flap reconstruction has been proven effective in treating large skin and soft tissue defects in clinical settings. However, in view of that the multi-territory perforator flap is prone to partial postoperative necrosis, increasing its survival is the key to the success of reconstruction. In this study, we aimed to clarify the effect of emodin on multi-territory perforator flap survival. METHODS Flap survival was assessed by viability area analysis, infrared laser imaging detector, HE staining, immunohistochemistry, and angiography. Western blotting, immunofluorescence assays, and real-time fluorescent quantitative PCR were performed to detect the indicators of oxidative stress, pyroptosis and autophagy. RESULTS After emodin treatment, the multi-territory perforator flap showed a significantly increased survival rate, which was shown to be closely related to the inhibition of oxidative stress and pyroptosis and enhanced autophagy. Meanwhile, the use of autophagy inhibitor 3 MA was found to reverse the inhibitory effects of emodin on oxidative stress and pyroptosis and weaken the improving effect of emodin on flap survival, suggesting that autophagy plays a critical role in emodin-treated flaps. Interestingly, our mechanistic investigations revealed that the positive effect of emodin on multi-territory perforator flap was attributed to the mTOR-ULK1 signaling pathway activation. CONCLUSIONS Emodin can inhibit oxidative stress and pyroptosis by activating autophagy via the mTOR-ULK1 pathway, thereby improving the multi-territory perforator flap survival.
Collapse
Affiliation(s)
- Panfeng Wu
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Xiao
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liming Qing
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yanan Mi
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Juyu Tang
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zheming Cao
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Chengxiong Huang
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
37
|
Zhang LC, Li N, Chen JL, Sun J, Xu M, Liu WQ, Zuo ZF, Shi LL, Wang TH, Luo XY. Molecular network mechanism in cerebral ischemia-reperfusion rats treated with human urine stem cells. Heliyon 2024; 10:e27508. [PMID: 38560254 PMCID: PMC10979071 DOI: 10.1016/j.heliyon.2024.e27508] [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: 04/21/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Objective To explore the effect of human urine-derived stem cells (husc) in improving the neurological function of rats with cerebral ischemia-reperfusion (CIR), and report new molecular network by bioinformatics, combined with experiment validation. Methods After CIR model was established, and husc were transplanted into the lateral ventricle of rats,neurological severe score (NSS) andgene network analysis were performed. Firstly, we input the keywords "Cerebral reperfusion" and "human urine stem cells" into Genecard database and merged data with findings from PubMed so as to get their targets genes, and downloaded them to make Venny intersection plot. Then, Gene ontology (GO) analysis, kyoto encyclopedia of genes and genomes (KEGG) pathway analysis and protein-protein interaction (PPI) were performed to construct molecular network of core genes. Lastly, the expressional level of core genes was validated via quantitative real-time polymerase chain reaction (qRT-PCR), and localized by immunofluorescence. Results Compared with the Sham group, the neurological function of CIR rats was significantly improved after the injection of husc into the lateral ventricle; at 14 days, P = 0.028, which was statistically significant. There were 258 overlapping genes between CIR and husc, and integrated with 252 genes screened from PubMed and CNKI. GO enrichment analysis were mainly involved neutrophil degranulation, neutrophil activation in immune response and platelet positive regulation of degranulation, Hemostasis, blood coagulation, coagulation, etc. KEGG pathway analysis was mainly involved in complement and coagulation cascades, ECM-receptor. Hub genes screened by Cytoscape consist ofCD44, ACTB, FN1, ITGB1, PLG, CASP3, ALB, HSP90AA1, EGF, GAPDH. Lastly, qRT-PCR results showed statistic significance (P < 0.05) in ALB, CD44 and EGF before and after treatment, and EGF immunostaining was localized in neuron of cortex. Conclusion husc transplantation showed a positive effect in improving neural function of CIR rats, and underlying mechanism is involved in CD44, ALB, and EGF network.
Collapse
Affiliation(s)
- Lang-Chun Zhang
- Department of Neurosurgery, Xiang Ya Hospital of Central South University, Changsha, 410078, China
- Animal Canter Department of Anatomy, Kunming Medical University, Kunming, 650500, China
| | - Na Li
- Animal Canter Department of Anatomy, Kunming Medical University, Kunming, 650500, China
| | - Ji-Lin Chen
- Animal Canter Department of Anatomy, Kunming Medical University, Kunming, 650500, China
| | - Jie Sun
- Animal Canter Department of Anatomy, Kunming Medical University, Kunming, 650500, China
| | - Min Xu
- Animal Canter Department of Anatomy, Kunming Medical University, Kunming, 650500, China
| | - Wen-Qiang Liu
- College of Basic Medicine, Jinzhou Medical University, Jinzhou, 121000, China
| | - Zhong-Fu Zuo
- Department of Anatomy, Jinzhou Medical University, Jinzhou, China
| | - Lan-Lan Shi
- Department of Neurosurgery, Xiang Ya Hospital of Central South University, Changsha, 410078, China
| | - Ting-Hua Wang
- Department of Neurosurgery, Xiang Ya Hospital of Central South University, Changsha, 410078, China
| | - Xiang-Yin Luo
- Department of Neurosurgery, Xiang Ya Hospital of Central South University, Changsha, 410078, China
| |
Collapse
|
38
|
Prunotto P, Marie P, Lebouvier L, Hommet Y, Vivien D, Ali C. The Janus face of endogenous neuronal tPA: promoting self-protection and worsening the death of neighboring neurons. Cell Death Dis 2024; 15:261. [PMID: 38609369 PMCID: PMC11014960 DOI: 10.1038/s41419-024-06655-0] [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: 11/22/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
Recombinant tissue-type plasminogen activator (r-tPA/Actilyse) stands as the prevailing pharmacological solution for treating ischemic stroke patients, of whom because their endogenous circulating tPA alone is not sufficient to rescue reperfusion and to promote favorable outcome. Beyond the tPA contributed by circulating endothelial cells and hepatocytes, neurons also express tPA, sparking debates regarding its impact on neuronal fate ranging from pro-survival to neurotoxic properties. In order to investigate the role of neuronal tPA during brain injuries, we developed models leading to its conditional deletion in neurons, employing AAV9-pPlat-GFP and AAV9-pPlat-Cre-GFP along with tPA floxed mice. These models were subjected to N-methyl-D-aspartate (NMDA)-induced excitotoxicity or thromboembolic ischemic stroke in mice. Initially, we established that our AAV9 constructs selectively transduce neurons, bypassing other brain cell types. Subsequently, we demonstrated that tPA-expressing neurons exhibit greater resistance against NMDA-induced excitotoxicity compared to tPA negative neurons. The targeted removal of tPA in neurons heightened the susceptibility of these neurons to cell death and prevented a paracrine neurotoxic effect on tPA non-expressing neurons. Under ischemic conditions, the self-neuroprotective influence of tPA encompassed both excitatory (GFP+/Tbr1+) and inhibitory (GFP+/GABA+) neurons. Our data indicate that endogenous neuronal tPA is a protective or deleterious factor against neuronal death in an excitotoxic/ischemic context, depending on whether it acts as an autocrine or a paracrine mediator.
Collapse
Affiliation(s)
- Paul Prunotto
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Pauline Marie
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Laurent Lebouvier
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
- Department of clinical research, CHU de Caen Normandie, Caen, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France.
| |
Collapse
|
39
|
Ali NH, Al-Kuraishy HM, Al-Gareeb AI, Alnaaim SA, Alexiou A, Papadakis M, Saad HM, Batiha GES. The probable role of tissue plasminogen activator/neuroserpin axis in Alzheimer's disease: a new perspective. Acta Neurol Belg 2024; 124:377-388. [PMID: 37917293 DOI: 10.1007/s13760-023-02403-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia associated with amyloid beta (Aβ) deposition. Dysfunction of the neuronal clearance pathway promotes the accumulation of Aβ. The plasminogen-activating system (PAS) is controlled by various enzymes like tissue plasminogen activators (tPA). Neuronal tPA enhances the conversion of plasminogen to plasmin, which cleaves Aβ; this function is controlled by many inhibitors of PAS, including a plasminogen-activating inhibitor (PAI-1) and neuroserpin. Therefore, the objective of the present narrative review was to explore the potential role of tPA/neuroserpin in the pathogenesis of AD. PAI-1 activity is increased in AD, which is involved in accumulating Aβ. Progressive increase of Aβ level during AD neuropathology is correlated with the over-production of PAI-1 with subsequent reduction of plasmin and tPA activities. Reducing plasmin and tPA activities promote Aβ by reducing Aβ clearance. Neuroserpin plays a critical role in the pathogenesis of AD as it regulates the expression and accumulation of Aβ. Higher expression of neuroserpin inhibits the neuroprotective tPA and the generation of plasmin with subsequent reduction in the clearance of Aβ. These observations raise conflicting evidence on whether neuroserpin is neuroprotective or involved in AD progression. Thus, neuroserpin over-expression with subsequent reduction of tPA may propagate AD neuropathology.
Collapse
Affiliation(s)
- Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, PO Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, PO Box 14132, Baghdad, Iraq
| | - Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744, Matrouh, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
40
|
Gelen V, Özkanlar S, Kara A, Yeşildağ A. Citrate-coated silver nanoparticles loaded with agomelatine provide neuronal therapy in acute cerebral ischemia/reperfusion of rats by inhibiting the oxidative stress, endoplasmic reticulum stress, and P2X7 receptor-mediated inflammasome. ENVIRONMENTAL TOXICOLOGY 2024; 39:1531-1543. [PMID: 38009636 DOI: 10.1002/tox.24021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
Cerebral ischemia and reperfusion are related to various situations like injuries after various traumas, oxidative stress, increased calcium ion, capillary hypoperfusion, microvascular hyperpermeability, leukocyte infiltration, and blood-brain barrier disruption. An antidepressant Agomelatine which is a melatonin receptor (MT1/MT2) agonist and serotonin receptor (5-HT2C) antagonist has been reported by studies to have antioxidant and anti-inflammatory effects. In our study, we aimed to detect the effects of citrate-coated silver nanoparticle-loaded agomelatine application on neurodegeneration, endoplasmic reticulum stress, autophagic and apoptotic cell death, inflammation, and P2X7R expression in the cerebral ischemia-reperfusion model to facilitate the passage of blood-brain barrier. Forty two Sprague-Dawley rats in total were divided into six equal groups (n:7) and applications were performed. Acute cerebral injury in the ischemia-reperfusion model was created 2 h after internal carotid artery ligation in rats and then at the 2nd hour of reperfusion citrate-coated silver nanoparticles loaded with Agomelatine were applied. Twenty four hours later, neurologic analysis on animals in experimental groups was performed, animals were decapitated and GSH, GPx, SOD, CAT, MDA, IL-1β, and TNF-α parameters were examined after taking blood and the cerebral tissue samples. As a result, it was determined that ischemia-reperfusion caused endoplasmic reticulum stress in the cerebral tissues and thus caused cellular injury.
Collapse
Affiliation(s)
- Volkan Gelen
- Department of Physiology, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey
| | - Seçkin Özkanlar
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Adem Kara
- Department of Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
| | - Ali Yeşildağ
- Department of Bioengineering, Faculty of Engineering and Architecture, Kafkas University, Kars, Turkey
| |
Collapse
|
41
|
Li YY, Qin ZH, Sheng R. The Multiple Roles of Autophagy in Neural Function and Diseases. Neurosci Bull 2024; 40:363-382. [PMID: 37856037 PMCID: PMC10912456 DOI: 10.1007/s12264-023-01120-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/11/2023] [Indexed: 10/20/2023] Open
Abstract
Autophagy involves the sequestration and delivery of cytoplasmic materials to lysosomes, where proteins, lipids, and organelles are degraded and recycled. According to the way the cytoplasmic components are engulfed, autophagy can be divided into macroautophagy, microautophagy, and chaperone-mediated autophagy. Recently, many studies have found that autophagy plays an important role in neurological diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, neuronal excitotoxicity, and cerebral ischemia. Autophagy maintains cell homeostasis in the nervous system via degradation of misfolded proteins, elimination of damaged organelles, and regulation of apoptosis and inflammation. AMPK-mTOR, Beclin 1, TP53, endoplasmic reticulum stress, and other signal pathways are involved in the regulation of autophagy and can be used as potential therapeutic targets for neurological diseases. Here, we discuss the role, functions, and signal pathways of autophagy in neurological diseases, which will shed light on the pathogenic mechanisms of neurological diseases and suggest novel targets for therapies.
Collapse
Affiliation(s)
- Yan-Yan Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
42
|
Gao L, Peng L, Wang J, Zhang JH, Xia Y. Mitochondrial stress: a key role of neuroinflammation in stroke. J Neuroinflammation 2024; 21:44. [PMID: 38321473 PMCID: PMC10845693 DOI: 10.1186/s12974-024-03033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/27/2024] [Indexed: 02/08/2024] Open
Abstract
Stroke is a clinical syndrome characterized by an acute, focal neurological deficit, primarily caused by the occlusion or rupture of cerebral blood vessels. In stroke, neuroinflammation emerges as a pivotal event contributing to neuronal cell death. The occurrence and progression of neuroinflammation entail intricate processes, prominently featuring mitochondrial dysfunction and adaptive responses. Mitochondria, a double membrane-bound organelle are recognized as the "energy workshop" of the body. Brain is particularly vulnerable to mitochondrial disturbances due to its high energy demands from mitochondria-related energy production. The interplay between mitochondria and neuroinflammation plays a significant role in the pathogenesis of stroke. The biological and pathological consequences resulting from mitochondrial stress have substantial implications for cerebral function. Mitochondrial stress serves as an adaptive mechanism aimed at mitigating the stress induced by the import of misfolded proteins, which occurs in response to stroke. This adaptive response involves a reduction in misfolded protein accumulation and overall protein synthesis. The influence of mitochondrial stress on the pathological state of stroke is underscored by its capacity to interact with neuroinflammation. The impact of mitochondrial stress on neuroinflammation varies according to its severity. Moderate mitochondrial stress can bolster cellular adaptive defenses, enabling cells to better withstand detrimental stressors. In contrast, sustained and excessive mitochondrial stress detrimentally affects cellular and tissue integrity. The relationship between neuroinflammation and mitochondrial stress depends on the degree of mitochondrial stress present. Understanding its role in stroke pathogenesis is instrumental in excavating the novel treatment of stroke. This review aims to provide the evaluation of the cross-talk between mitochondrial stress and neuroinflammation within the context of stroke. We aim to reveal how mitochondrial stress affects neuroinflammation environment in stroke.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Li Peng
- Department of Ophthalmology, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
| | - Jian Wang
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA.
- Department of Neurosurgery and Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA.
| | - Ying Xia
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China.
| |
Collapse
|
43
|
Huang Q, Yu X, Fu P, Wu M, Yin X, Chen Z, Zhang M. Mechanisms and therapeutic targets of mitophagy after intracerebral hemorrhage. Heliyon 2024; 10:e23941. [PMID: 38192843 PMCID: PMC10772251 DOI: 10.1016/j.heliyon.2023.e23941] [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: 06/25/2023] [Revised: 11/03/2023] [Accepted: 12/15/2023] [Indexed: 01/10/2024] Open
Abstract
Mitochondria are dynamic organelles responsible for cellular energy production. In addition to regulating energy homeostasis, mitochondria are responsible for calcium homeostasis, clearance of damaged organelles, signaling, and cell survival in the context of injury and pathology. In stroke, the mechanisms underlying brain injury secondary to intracerebral hemorrhage are complex and involve cellular hypoxia, oxidative stress, inflammatory responses, and apoptosis. Recent studies have shown that mitochondrial damage and autophagy are essential for neuronal metabolism and functional recovery after intracerebral hemorrhage, and are closely related to inflammatory responses, oxidative stress, apoptosis, and other pathological processes. Because hypoxia and inflammatory responses can cause secondary damage after intracerebral hemorrhage, the restoration of mitochondrial function and timely clearance of damaged mitochondria have neuroprotective effects. Based on studies on mitochondrial autophagy (mitophagy), cellular inflammation, apoptosis, ferroptosis, the BNIP3 autophagy gene, pharmacological and other regulatory approaches, and normobaric oxygen (NBO) therapy, this article further explores the neuroprotective role of mitophagy after intracerebral hemorrhage.
Collapse
Affiliation(s)
- Qinghua Huang
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332000, China
| | - Xiaoqin Yu
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Peijie Fu
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
- Jiujiang Clinical Precision Medicine Research Center, 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
| | - Zhiying Chen
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332000, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi, 332000, China
| |
Collapse
|
44
|
Chen D, Zhou L, Chen G, Lin T, Lin J, Zhao X, Li W, Guo S, Wu R, Wang Z, Liu W. FUNDC1-induced mitophagy protects spinal cord neurons against ischemic injury. Cell Death Discov 2024; 10:4. [PMID: 38177127 PMCID: PMC10766648 DOI: 10.1038/s41420-023-01780-9] [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: 03/28/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024] Open
Abstract
Local ischemia and hypoxia are the most important pathological processes in the early phase of secondary spinal cord injury (SCI), in which mitochondria are the main target of ischemic injury. Mitochondrial autophagy, also known as mitophagy, acts as a selective autophagy that specifically identifies and degrades damaged mitochondria, thereby reducing mitochondria-dependent apoptosis. Accumulating evidence shows that the mitophagy receptor, FUN14 domain-containing 1 (FUNDC1), plays an important role in ischemic injury, but the role of FUNDC1 in SCI has not been reported. In this study, we aimed to investigate whether FUNDC1 can enhance mitophagy and inhibit neuronal apoptosis in the early stage of SCI. In a rat SCI model, we found that FUNDC1 overexpression enhanced neuronal autophagy and decreased neuronal apoptosis in the early stage of injury, thereby reducing spinal cord damage. In vitro studies showed that the neuroprotective effects of FUNDC1 were achieved by inhibiting mitochondria-dependent apoptosis and improving mitochondrial function. In addition, FUNDC1 enhanced mitophagy. The protective effects of FUNDC1 against apoptosis and mitochondrial dysfunction were reversed by 3-methyladenine (3-MA), an autophagy inhibitor. Taken together, our results confirm that FUNDC1 can protect against neuronal loss after SCI by inducing mitophagy, inhibiting mitochondria-dependent apoptosis, and improving mitochondrial function.
Collapse
Affiliation(s)
- Dehui Chen
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Linquan Zhou
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Gang Chen
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Taotao Lin
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Jiemin Lin
- School of Health, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Xin Zhao
- School of Health, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Wenwen Li
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Shengyu Guo
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Rongcan Wu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Zhenyu Wang
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| | - Wenge Liu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
45
|
Fang G, Wen X, Jiang Z, Du X, Liu R, Zhang C, Huang G, Liao W, Zhang Z. FUNDC1/PFKP-mediated mitophagy induced by KD025 ameliorates cartilage degeneration in osteoarthritis. Mol Ther 2023; 31:3594-3612. [PMID: 37838829 PMCID: PMC10727975 DOI: 10.1016/j.ymthe.2023.10.016] [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: 05/01/2023] [Revised: 08/12/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, but no disease-modifying drugs have been approved for OA treatment. Mitophagy participates in mitochondrial homeostasis regulation by selectively clearing dysfunctional mitochondria, which might contribute to cartilage degeneration in OA. Here, we provide evidence of impaired mitophagy in OA chondrocytes, which exacerbates chondrocyte degeneration. Among the several classic mitophagy-regulating pathways and receptors, we found that FUNDC1 plays a key role in preserving chondrocyte homeostasis by inducing mitophagy. FUNDC1 knockdown in vitro and knockout in vivo decreased mitophagy and exacerbated mitochondrial dysfunction, exacerbating chondrocyte degeneration and OA progression. FUNDC1 overexpression via intra-articular injection of adeno-associated virus alleviated cartilage degeneration in OA. Mechanistically, our study demonstrated that PFKP interacts with and dephosphorylates FUNDC1 to induce mitophagy in chondrocytes. Further analysis identified KD025 as a candidate drug for restoring chondrocyte mitophagy by increasing the FUNDC1-PFKP interaction and thus alleviating cartilage degeneration in mice with DMM-induced OA. Our study highlights the role of the FUNDC1-PFKP interaction in chondrocyte homeostasis via mitophagy induction and identifies KD025 as a promising agent for treating OA by increasing chondrocyte mitophagy.
Collapse
Affiliation(s)
- Guibin Fang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China; Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xingzhao Wen
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China; Department of Medicine, Solna, Karolinska Institutet, and Centre for Molecular Medicine, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Zongrui Jiang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China
| | - Xue Du
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China
| | - Ruonan Liu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China
| | - Chengyun Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China
| | - Guiwu Huang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China
| | - Weiming Liao
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China
| | - Zhiqi Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
46
|
Zhu X, An X, Chen M, Guo D, Xie P, Wang B, Huang Z, Yu W. Seipin overexpression attenuates cerebral ischemia-reperfusion injury via preventing apoptosis and autophagy. Brain Behav 2023; 13:e3195. [PMID: 37897134 PMCID: PMC10726895 DOI: 10.1002/brb3.3195] [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: 03/05/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Ischemic cerebrovascular disease (ICVD) is one of three fatal diseases in humans, along with heart disease and malignant tumors. Cerebral ischemia/reperfusion injury (CI/RI) is the primary cause of ICVD. Recently, seipin was reported to be crucial for lipid droplet formation, hepatic steatosis, and axonal atrophy. However, the function and mechanism of seipin in CI/RI has not been explicitly stated. METHODS Oxygen-glucose deprivation/reoxygenation (OGD/R) hippocampal neuron cell line (HT-22) and middle cerebral artery occlusion (MCAO) in rats were established. The levels of apoptosis- and autophagy-related proteins and seipin were confirmed by western blot. Cell proliferation was assessed with CCK-8, and ischemic infarction and pathological structure were monitored by TTC and H&E staining, and tissue apoptosis was assessed through TUNEL assay. RESULTS The proliferative activity was decreased, and apoptosis and autophagy pathways could also be induced in the OGD/R HT-22 cells. Seipin overexpression accelerated viability and inhibited apoptosis and autophagy in the OGD/R HT-22 cells. Moreover, the data revealed that seipin overexpression could also attenuate cerebral infarction, apoptosis. Autophagy pathways could be repressed by seipin in the MCAO rats. CONCLUSION Seipin has a protective role against CI/RI in rats, and its mechanism might be relevant to the suppression of apoptosis and autophagy, suggesting that seipin might be a latent target for CI/RI.
Collapse
Affiliation(s)
- Xiaoxi Zhu
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
- Cell engineering LaboratoryAffiliated Hospital of Zunyi Medical UniversityZunyi CityChina
| | - Xiaoqiong An
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Ming Chen
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Dongfen Guo
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Peng Xie
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Bi Wang
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Zhi Huang
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Wenfeng Yu
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| |
Collapse
|
47
|
Yang X, Zhang Y, Luo JX, Zhu T, Ran Z, Mu BR, Lu MH. Targeting mitophagy for neurological disorders treatment: advances in drugs and non-drug approaches. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3503-3528. [PMID: 37535076 DOI: 10.1007/s00210-023-02636-w] [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: 04/16/2023] [Accepted: 07/18/2023] [Indexed: 08/04/2023]
Abstract
Mitochondria serve as a vital energy source for nerve cells. The mitochondrial network also acts as a defense mechanism against external stressors that can threaten the stability of the nervous system. However, excessive accumulation of damaged mitochondria can lead to neuronal death. Mitophagy is an essential pathway in the mitochondrial quality control system and can protect neurons by selectively removing damaged mitochondria. In most neurological disorders, dysfunctional mitochondria are a common feature, and drugs that target mitophagy can improve symptoms. Here, we reviewed the role of mitophagy in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, stroke, and traumatic brain injuries. We also summarized drug and non-drug approaches to promote mitophagy and described their therapeutic role in neurological disorders in order to provide valuable insight into the potential therapeutic agents available for neurological disease treatment. However, most studies on mitophagy regulation are based on preclinical research using cell and animal models, which may not accurately reflect the effects in humans. This poses a challenge to the clinical application of drugs targeting mitophagy. Additionally, these drugs may carry the risk of intolerable side effects and toxicity. Future research should focus on the development of safer and more targeted drugs for mitophagy.
Collapse
Affiliation(s)
- Xiong Yang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yu Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jia-Xin Luo
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Zhu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhao Ran
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ben-Rong Mu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Mei-Hong Lu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
48
|
Tao H, Li L, Dong L, Chen H, Shan X, Zhuge L, Lou H. Growth differentiation factor 7 pretreatment enhances the therapeutic capacity of bone marrow-derived mesenchymal stromal cells against cerebral ischemia-reperfusion injury. Chem Biol Interact 2023; 386:110779. [PMID: 37879595 DOI: 10.1016/j.cbi.2023.110779] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/13/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) transplantation is a promising therapeutic strategy for cerebral ischemia/reperfusion (I/R) injury; however, the clinical outcome is barely satisfactory and demands further improvement. The present study aimed to investigate whether preconditioning of BMSCs by recombinant human growth differentiation factor 7 (rhGDF7) could enhance its therapeutic capacity against cerebral I/R injury. Mouse BMSCs and primary neurons were co-cultured and exposed to oxygen glucose deprivation/reperfusion (OGD/R) stimulation. To investigate the role of exosomal microRNA-369-3p (miR-369-3p), inhibitors, RNAi and the miR-369-3p antagomir were used. Meanwhile, mice were intravenously injected with rhGDF7-preconditioned BMSCs and then received cerebral I/R surgery. Markers of inflammation, oxidative stress and neural damage were evaluated. To inhibit AMP-activated protein kinase (AMPK), compound C was used in vivo and in vitro. Compared with cell-free transwell or vehicle-preconditioned BMSCs, rhGDF7-preconditioned BMSCs significantly prevented OGD/R-induced inflammation, oxidative stress and neural damage in vitro. Meanwhile, rhGDF7-preconditioned BMSCs could prevent I/R-induced cerebral inflammation and oxidative stress in vivo. Mechanistically, rhGDF7 preconditioning significantly increased exosomal miR-369-3p expression in BMSCs and then transferred exosomal miR-369-3p to primary neurons, where it bound to phosphodiesterase 4 D (Pde4d) 3'-UTR and downregulated PDE4D expression, thereby preventing I/R-induced inflammation, oxidative stress and neural damage through activating AMPK pathway. Our study identify GDF7 pretreatment as a promising adjuvant reagent to improve the therapeutic potency of BMSCs for cerebral I/R injury and ischemic stroke.
Collapse
Affiliation(s)
- Hongmiao Tao
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China
| | - Lin Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Lihua Dong
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China
| | - Haohao Chen
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China
| | - Xiaoyun Shan
- Department of Clinical Laboratory, Jinhua Municipal Central Hospital, Jinhua, 321000, Zhejiang, China
| | - Lujie Zhuge
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Hongqiang Lou
- Medical College, Jinhua Polytechnic, Jinhua, 321017, Zhejiang, China.
| |
Collapse
|
49
|
She R, Ge J, Mei Z. New Wine in an Old Bottle: tPA for Ischemic Stroke Management. Transl Stroke Res 2023:10.1007/s12975-023-01209-6. [PMID: 37921975 DOI: 10.1007/s12975-023-01209-6] [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: 10/12/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2023]
Abstract
As the only clinical thrombolytic drug approved by the FDA, tissue-type plasminogen activator (tPA) is the good standard acute treatment against ischemic stroke (IS) during the super-early stage. tPA forms the active principle of alteplase, a recombinant tissue-type plasminogen activator (rtPA), which is well known for its intravascular thrombolytic activity. However, the multifaceted functions of tPA in the central nervous system (CNS) hold untapped potential. Currently, increasing studies have explored the neuroprotective function of tPA in neurological diseases, particularly in acute ischemic stroke (AIS). A series of studies have indicated that tPA has anti-excitotoxic, neurotrophic, and anti-apoptotic effects on neurons; it is also involved in neuronal plasticity, axonal regeneration, and cerebral inflammatory processes, but how to deeply understand the underlying mechanism and take maximum advantage of tPA seems to be urgent. Therefore, more work is needed to illuminate how tPA performs with more diverse functions after stroke onset. In this comment, we focus on possible hypotheses about why and how tPA promotes ischemic neuronal survival in a comprehensive view. The text provides a holistic picture of the functions of tPA and enlists the considerations for the future, which might attract more attention toward the therapeutic potential of tPA in AIS.
Collapse
Affiliation(s)
- Ruining She
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, China.
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- Third-Grade Pharmacological Laboratory On Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei, China.
| |
Collapse
|
50
|
Li Z, Xing J. Contribution and therapeutic value of mitophagy in cerebral ischemia-reperfusion injury after cardiac arrest. Biomed Pharmacother 2023; 167:115492. [PMID: 37716121 DOI: 10.1016/j.biopha.2023.115492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Cardiopulmonary resuscitation and related life support technologies have improved substantially in recent years; however, mortality and disability rates from cardiac arrest (CA) remain high and are closely associated with the high incidence of cerebral ischemia-reperfusion injury (CIRI), which is explained by a "double-hit" model (i.e., resulting from both ischemia and reperfusion). Mitochondria are important power plants in the cell and participate in various biochemical processes, such as cell differentiation and signaling in eukaryotes. Various mitochondrial processes, including energy metabolism, calcium homeostasis, free radical production, and apoptosis, are involved in several important stages of the progression and development of CIRI. Mitophagy is a key mechanism of the endogenous removal of damaged mitochondria to maintain organelle function and is a critical target for CIRI treatment after CA. Mitophagy also plays an essential role in attenuating ischemia-reperfusion in other organs, particularly during post-cardiac arrest myocardial dysfunction. Regulation of mitophagy may influence necroptosis (a programmed cell death pathway), which is the main endpoint of organ ischemia-reperfusion injury. In this review, we summarize the main signaling pathways related to mitophagy and their associated regulatory proteins. New therapeutic methods and drugs targeting mitophagy in ischemia-reperfusion animal models are also discussed. In-depth studies of the mechanisms underlying the regulation of mitophagy will enhance our understanding of the damage and repair processes in CIRI after CA, thereby contributing to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|