1
|
Liu M, Li B, Yin Z, Yin L, Luo Y, Zeng Q, Zhang D, Wu A, Chen L. Targeting mitochondrial dynamics: A promising approach for intracerebral hemorrhage therapy. Life Sci 2024; 361:123317. [PMID: 39674268 DOI: 10.1016/j.lfs.2024.123317] [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: 10/04/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a major global health issue with high mortality and disability rates. Following ICH, the hematoma exerts direct pressure on brain tissue, and blood entering the brain directly damages neurons and the blood-brain barrier. Subsequently, oxidative stress, inflammatory responses, apoptosis, brain edema, excitotoxicity, iron toxicity, and metabolic dysfunction around the hematoma further exacerbate brain tissue damage, leading to secondary brain injury (SBI). Mitochondria, essential for energy production and the regulation of oxidative stress, are damaged after ICH, resulting in impaired ATP production, excessive reactive oxygen species (ROS) generation, and disrupted calcium homeostasis, all of which contribute to SBI. Therefore, a central factor in SBI is mitochondrial dysfunction. Mitochondrial dynamics regulate the shape, size, distribution, and quantity of mitochondria through fusion and fission, both of which are crucial for maintaining their function. Fusion repairs damaged mitochondria and preserves their health, while fission helps mitochondria adapt to cellular stress and removes damaged mitochondria through mitophagy. When this balance is disrupted following ICH, mitochondrial dysfunction worsens, oxidative stress and metabolic failure are exacerbated, ultimately contributing to SBI. Targeting mitochondrial dynamics offers a promising therapeutic approach to restoring mitochondrial function, reducing cellular damage, and improving recovery. This review explores the latest research on modulating mitochondrial dynamics and highlights its potential to enhance outcomes in ICH patients.
Collapse
Affiliation(s)
- Mengnan Liu
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Binru Li
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China.
| | - Zhixue Yin
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Lu Yin
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Ye Luo
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Qi Zeng
- Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Dechou Zhang
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Department of Cardiology, The Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Luzhou 646000, Sichuan, China; Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Li Chen
- Department of Neurology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China.
| |
Collapse
|
2
|
Gu F, Wang Z, Ding H, Tao X, Zhang J, Dai K, Li X, Shen H, Li H, Chen Z, Wang Z. Microglial mitochondrial DNA release contributes to neuroinflammation after intracerebral hemorrhage through activating AIM2 inflammasome. Exp Neurol 2024; 382:114950. [PMID: 39278588 DOI: 10.1016/j.expneurol.2024.114950] [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: 07/01/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
Intracerebral hemorrhage (ICH) is a severe disease that often leads to disability and death. Neuroinflammatory response is a key causative factor of early secondary brain injury after ICH. AIM2 is a DNA-sensing protein that recognizes cytosolic double-stranded DNA and take a significant part in neuroinflammation. Mitochondrial DNA participates in the translation of proteins such as the respiratory chain in the mitochondria. Whether mtDNA is involved in forming AIM2 inflammasome after ICH remains unclear. We used mice to construct ICH model in vivo and we used BV2 microglial cells treated with oxyhemoglobin to simulate ICH in vitro. Following lentiviral transfection to overexpress AIM2 antagonist P202, a notable decrease was observed in the levels of AIM2 inflammasome-associated proteins, leading to a reduction in dead neurons surrounding the hematoma and an enhancement in long-term and short-term behavior of neurological deficits. We further explored whether mtDNA took part in the AIM2 activation after ICH. The cytosolic mtDNA level was down-regulated by the mitochondrial division protector Mdivi-1 and up-regulated by transfection of mtDNA into cytoplasm. We found the expression level of AIM2 inflammasome-related proteins and inflammatory cytokines release were regulated by the cytosolic mtDNA level. In conclusion, after ICH, the mtDNA content in the cytoplasm of microglia around the hematoma rises, causing AIM2 inflammation leading to neuronal apoptosis, which leads to neurological deficits in mice. On the other hand, P202 was able to block inflammatory vesicle activation and improve neurological function by preventing the interaction between AIM2 protein and mitochondrial DNA.
Collapse
Affiliation(s)
- Feng Gu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haojie Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xinyu Tao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Kun Dai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
3
|
Fedoruk A, Shadyro O, Edimecheva I, Fedoruk D, Khrutskin V, Kirkovsky L, Sorokin V, Talkachova H. Free radical fragmentation and oxidation in the polar part of lysophospholipids: Results of the study of blood serum of healthy donors and patients with acute surgical pathology. Redox Biol 2024; 76:103309. [PMID: 39178730 PMCID: PMC11388268 DOI: 10.1016/j.redox.2024.103309] [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: 07/12/2024] [Revised: 08/03/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024] Open
Abstract
The interaction of reactive oxygen species with cell membrane lipids is usually considered in the context of lipid peroxidation in the nonpolar component of the membrane. In this work, for the first time, data were obtained indicating that damage to human cell membranes can occur in the polar part of lysophospholipids at the interface with the aqueous environment due to free radical fragmentation (FRF) processes. FRF products, namely 1-hexadecanoyloxyacetone (PAc) and 1-octadecanoyloxyacetone (SAc), were identified in human serum, and a GC-MS method was developed to quantify PAc and SAc. The content of FRF products in serum samples of 52 healthy donors was found to be in the range of 1.98-4.75 μmol/L. A linear regression equation, CPAc&SAc (μmol/L) = 0.51 + 0.064 × years, was derived to describe the relationship between age and content of FRF products. In 70 patients with acute surgical pathology in comparison with the control group of healthy donors, two distinct clusters with different concentration levels of FRF products were revealed. The first cluster: groups of 43 patients with various localized inflammatory-destructive lesions of hollow organ walls and bacterial translocation (septic inflammation) of abdominal cavity organs. These patients showed a 1.5-1.9-fold (p = 0.012) decrease in the total concentration of PAc and SAc in serum. In the second cluster: groups of 27 patients with ischemia-reperfusion tissue damage (aseptic inflammation), - a statistically significant increase in the concentration of FRF products was observed: in 2.2-4.0 times (p = 0.0001). The obtained data allow us to further understand the role of free-radical processes in the damage of lipid molecules. FRF products can potentially be used as markers of the degree of free-radical damage of hydroxyl containing phospholipids.
Collapse
Affiliation(s)
- Alexey Fedoruk
- Department of Hepatology and Minimally Invasive Surgery, Minsk Scientific and Practical Center for Surgery, Transplantology and Hematology, 220087, Semashko st., 8, Minsk, Belarus; Department of Transplantology, Institute of Advanced Studies and retraining of Healthcare Personnel in Educational Institutions «Belarusian State Medical University», 220083, Dzerzhinsky Ave., 83, Minsk, Belarus.
| | - Oleg Shadyro
- Laboratory of Chemistry of Free Radical Processes, Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, Leningradskaya st., 14, Minsk, Belarus; Department of Radiation Chemistry and Chemical Pharmaceutical Technologies, Belarusian State University, 220030, Nezavisimosti av., 4, Minsk, Belarus
| | - Irina Edimecheva
- Laboratory of Chemistry of Free Radical Processes, Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, Leningradskaya st., 14, Minsk, Belarus.
| | - Dmitry Fedoruk
- Department of Hepatology and Minimally Invasive Surgery, Minsk Scientific and Practical Center for Surgery, Transplantology and Hematology, 220087, Semashko st., 8, Minsk, Belarus.
| | - Valery Khrutskin
- Laboratory of Chemistry of Free Radical Processes, Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, Leningradskaya st., 14, Minsk, Belarus; Department of Radiation Chemistry and Chemical Pharmaceutical Technologies, Belarusian State University, 220030, Nezavisimosti av., 4, Minsk, Belarus.
| | - Leanid Kirkovsky
- Department of Hepatology and Minimally Invasive Surgery, Minsk Scientific and Practical Center for Surgery, Transplantology and Hematology, 220087, Semashko st., 8, Minsk, Belarus.
| | - Viktor Sorokin
- Laboratory of Chemistry of Free Radical Processes, Research Institute for Physical Chemical Problems of the Belarusian State University, 220006, Leningradskaya st., 14, Minsk, Belarus; Department of Radiation Chemistry and Chemical Pharmaceutical Technologies, Belarusian State University, 220030, Nezavisimosti av., 4, Minsk, Belarus.
| | - Halina Talkachova
- Department of Hepatology and Minimally Invasive Surgery, Minsk Scientific and Practical Center for Surgery, Transplantology and Hematology, 220087, Semashko st., 8, Minsk, Belarus.
| |
Collapse
|
4
|
Thapak P, Gomez-Pinilla F. The bioenergetics of traumatic brain injury and its long-term impact for brain plasticity and function. Pharmacol Res 2024; 208:107389. [PMID: 39243913 DOI: 10.1016/j.phrs.2024.107389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Mitochondria provide the energy to keep cells alive and functioning and they have the capacity to influence highly complex molecular events. Mitochondria are essential to maintain cellular energy homeostasis that determines the course of neurological disorders, including traumatic brain injury (TBI). Various aspects of mitochondria metabolism such as autophagy can have long-term consequences for brain function and plasticity. In turn, mitochondria bioenergetics can impinge on molecular events associated with epigenetic modifications of DNA, which can extend cellular memory for a long time. Mitochondrial dysfunction leads to pathological manifestations such as oxidative stress, inflammation, and calcium imbalance that threaten brain plasticity and function. Hence, targeting mitochondrial function may have great potential to lessen the outcomes of TBI.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
5
|
Chen CC, Ke CH, Wu CH, Lee HF, Chao Y, Tsai MC, Shyue SK, Chen SF. Transient receptor potential vanilloid 1 inhibition reduces brain damage by suppressing neuronal apoptosis after intracerebral hemorrhage. Brain Pathol 2024; 34:e13244. [PMID: 38308041 PMCID: PMC11328348 DOI: 10.1111/bpa.13244] [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: 11/17/2023] [Accepted: 01/19/2024] [Indexed: 02/04/2024] Open
Abstract
Intracerebral hemorrhage (ICH) induces a complex sequence of apoptotic cascades and inflammatory responses, leading to neurological impairment. Transient receptor potential vanilloid 1 (TRPV1), a nonselective cation channel with high calcium permeability, has been implicated in neuronal apoptosis and inflammatory responses. This study used a mouse ICH model and neuronal cultures to examine whether TRPV1 activation exacerbates brain damage and neurological deficits by promoting neuronal apoptosis and neuroinflammation. ICH was induced by injecting collagenase in both wild-type (WT) C57BL/6 mice and TRPV1-/- mice. Capsaicin (CAP; a TRPV1 agonist) or capsazepine (a TRPV1 antagonist) was administered by intracerebroventricular injection 30 min before ICH induction in WT mice. The effects of genetic deletion or pharmacological inhibition of TRPV1 using CAP or capsazepine on motor deficits, histological damage, apoptotic responses, blood-brain barrier (BBB) permeability, and neuroinflammatory reactions were explored. The antiapoptotic mechanisms and calcium influx induced by TRPV1 inactivation were investigated in cultured hemin-stimulated neurons. TRPV1 expression was upregulated in the hemorrhagic brain, and TRPV1 was expressed in neurons, microglia, and astrocytes after ICH. Genetic deletion of TRPV1 significantly attenuated motor deficits and brain atrophy for up to 28 days. Deletion of TRPV1 also reduced brain damage, neurodegeneration, microglial activation, cytokine expression, and cell apoptosis at 1 day post-ICH. Similarly, the administration of CAP ameliorated brain damage, neurodegeneration, brain edema, BBB permeability, and cytokine expression at 1 day post-ICH. In primary neuronal cultures, pharmacological inactivation of TRPV1 by CAP attenuated neuronal vulnerability to hemin-induced injury, suppressed apoptosis, and preserved mitochondrial integrity in vitro. Mechanistically, CAP reduced hemin-stimulated calcium influx and prevented the phosphorylation of CaMKII in cultured neurons, which was associated with reduced activation of P38 and c-Jun NH2-terminal kinase mitogen-activated protein kinase signaling. Our results suggest that TRPV1 inhibition may be a potential therapy for ICH by suppressing mitochondria-related neuronal apoptosis.
Collapse
Affiliation(s)
- Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Taoyuan, Taiwan, Republic of China
| | - Chia-Hua Ke
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Chun-Hu Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Hung-Fu Lee
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, Republic of China
| | - Yuan Chao
- Department of Medical Education, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
6
|
Wu F, Liang T, Liu Y, Sun Y, Wang B. Hydrogen mitigates brain injury by prompting NEDD4-CX43- mediated mitophagy in traumatic brain injury. Exp Neurol 2024; 379:114876. [PMID: 38942265 DOI: 10.1016/j.expneurol.2024.114876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Hydrogen (H2) has emerged as a potential therapeutic intervention for traumatic brain injury (TBI). However, the precise mechanism underlying H2's neuroprotective effects in TBI remain incompletely understood. METHODS TBI mouse model was induced using the controlled cortical impact (CCI) method, and a cell model was established by exposing astrocytes to lipopolysaccharide (LPS). Cell viability was detected by CCK-8 kits. Cell apoptosis was measured by flow cytometry. ELISA was used to detect cytokine quantification. Protein and gene expression was detected by western blot and RT-PCR analysis. Co-immunoprecipitation (CO-IP) were employed for protein-protein interactions. Morris water maze test and rotarod test were applied for TBI mice. RESULTS H2 treatment effectively inhibited the LPS-induced cell injury and cell apoptosis in astrocytes. NEDD4 expression was increased following H2 treatment coupled with enhanced mitophagy in LPS-treated astrocytes. Overexpression of NEDD4 and down-regulation of connexin 43 (CX43) mirrored the protective effects of H2 treatment in LPS-exposed astrocytes. NEDD4 interacts CX43 to regulates the ubiquitinated degradation of CX43. While overexpression of CX43 reversed the protective effects of H2 treatment in LPS-exposed astrocytes. In addition, H2 treatment significantly alleviated brain injury in TBI mouse model. CONCLUSION H2 promoted NEDD4-CX43 mediated mitophagy to protect brain injury induced by TBI, highlighting a novel pathway underlying the therapeutic effects of H2 in TBI.
Collapse
Affiliation(s)
- Fan Wu
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing 100095, PR China
| | - Tao Liang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Yang Liu
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing 100095, PR China
| | - Yongxing Sun
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing 100095, PR China.
| | - Baoguo Wang
- Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing 100095, PR China.
| |
Collapse
|
7
|
Zhu H, Zhang H, Zhao XJ, Zhang L, Liu X, Zhang ZY, Ren YZ, Feng Y. Tetramerization of PKM2 Alleviates Traumatic Brain Injury by Ameliorating Mitochondrial Damage in Microglia. J Neuroimmune Pharmacol 2024; 19:48. [PMID: 39196455 DOI: 10.1007/s11481-024-10138-6] [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: 08/20/2023] [Accepted: 06/27/2024] [Indexed: 08/29/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Microglial activation and neuroinflammation are key cellular events that determine the outcome of TBI, especially neuronal and cognitive function. Studies have suggested that the metabolic characteristics of microglia dictate their inflammatory response. The pyruvate kinase isoform M2 (PKM2), a key glycolytic enzyme, is involved in the regulation of various cellular metabolic processes, including mitochondrial metabolism. This suggests that PKM2 may also participate in the regulation of microglial activation during TBI. Therefore, the present study aimed to evaluate the role of PKM2 in regulating microglial activation and neuroinflammation and its effects on cognitive function following TBI. A controlled cortical impact (CCI) mouse model and inflammation-induced primary mouse microglial cells in vitro were used to investigate the potential effects of PKM2 inhibition and regulation. PKM2 was significantly increased during the acute and subacute phases of TBI and was predominantly detected in microglia rather than in neurons. Our results demonstrate that shikonin and TEPP-46 can inhibit microglial inflammation, improving mitochondria, improving mouse behavior, reducing brain defect volume, and alleviating pathological changes after TBI. There is a difference in the intervention of shikonin and TEPP-46 on PKM2. Shikonin directly inhibits General PKM2; TEPP-46 can promote the expression of PKM2 tetramer. In vitro experiments, TEPP-46 can promote the expression of PKM2 tetramer, enhance the interaction between PKM2 and MFN2, improve mitochondria, alleviate neuroinflammation. General inhibition and tetramerization activation of PKM2 attenuated cognitive function caused by TBI, whereas PKM2 tetramerization exhibited a better treatment effect. Our experiments demonstrated the non-metabolic role of PKM2 in the regulation of microglial activation following TBI. Both shikonin and TEPP-46 can inhibit pro-inflammatory factors, but only TEPP-46 can promote PKM2 tetramerization and upregulate the release of anti-inflammatory factors from microglia.
Collapse
Affiliation(s)
- Haiyan Zhu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Huiwen Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Xiao-Jing Zhao
- Department of Pathology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, China
| | - Lingyuan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Xue Liu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Zhi-Yuan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Yi-Zhi Ren
- Department of Clinical Genetics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China, 262 North Zhongshan Road.
| | - Yong Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210009, China, Baiziting 42.
| |
Collapse
|
8
|
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
|
9
|
Li Q, Wang B, Yang J, Wang Y, Duan F, Luo M, Zhao C, Wei W, Wang L, Liu S. Preliminary Analysis of Aging-Related Genes in Intracerebral Hemorrhage by Integration of Bulk and Single-Cell RNA Sequencing Technology. Int J Gen Med 2024; 17:2719-2740. [PMID: 38883702 PMCID: PMC11180471 DOI: 10.2147/ijgm.s457480] [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: 02/23/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
Background Aging is recognized as the key risk for intracerebral hemorrhage (ICH). The detailed mechanisms of aging in ICH warrant exploration. This study aimed to identify potential aging-related genes associated with ICH. Methods ICH-specific aging-related genes were determined by the intersection of differentially expressed genes (DEGs) between perihematomal tissues and corresponding contralateral parts of four patients with ICH (GSE24265) and 349 aging-related genes obtained from the Aging Atlas database. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) analyses were performed to identify the potential biological functions and pathways in which these ICH-specific aging-related genes may be involved. Then, PPI network was established to identify the hub genes of ICH-specific aging-related genes. Meanwhile, miRNA-mRNA and transcription factor (TF)-mRNA regulatory networks were constructed to further explore the ICH-specific aging-related genes regulation. The relationship between these hub genes and immune infiltration was also further explored. Additional single-cell RNA-seq analysis (scRNA-seq, GSE167593) was used to locate the hub genes in different cell types. Besides, expression levels of the hub genes were validated using clinical samples from our institute and another GEO dataset (GSE206971). Results This study identified 24 ICH-specific aging-related genes, including 22 up-regulated and 2 down-regulated genes. The results of GO and KEGG suggested that the ICH-specific aging-related genes mainly enriched in immunity and inflammation-related pathways, suggesting that aging may affect the ich pathogenesis by regulating inflammatory and immune-related pathways. Conclusion Our study revealed 24 ICH-specific aging-related genes and their functions highly pertinent to ICH pathogenesis, providing new insights into the impact of aging on ICH.
Collapse
Affiliation(s)
- Qianfeng Li
- Department of Neurosurgery, Wuhan No.1 Hospital, Wuhan, People's Republic of China
| | - Bo Wang
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Jun Yang
- Huanggang Central Hospital of Yangtze University, Huanggang, People's Republic of China
| | - Yuan Wang
- Department of Neurosurgery, Wuhan No.1 Hospital, Wuhan, People's Republic of China
| | - Faliang Duan
- Department of Neurosurgery, Wuhan No.1 Hospital, Wuhan, People's Republic of China
| | - Ming Luo
- Department of Neurosurgery, Wuhan No.1 Hospital, Wuhan, People's Republic of China
| | - Chungang Zhao
- Jilin Jianda Modern Agricultural Research Institute, Changchun, People's Republic of China
| | - Wei Wei
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Lei Wang
- Huanggang Central Hospital of Yangtze University, Huanggang, People's Republic of China
| | - Sha Liu
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Department of General Practice, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
10
|
Li Q, Ni H, Rui Q, Ding J, Kong X, Kan X, Gao R, Shen H. Armcx1 Reduces Neurological Damage Via a Mitochondrial Transport Pathway Involving Miro1 After Traumatic Brain Injury. Neuroscience 2024; 545:111-124. [PMID: 38492796 DOI: 10.1016/j.neuroscience.2024.03.009] [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: 07/11/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Armcx1 is a member of the ARMadillo repeat-Containing protein on the X chromosome (ARMCX) family, which is recognized to have evolutionary conserved roles in regulating mitochondrial transport and dynamics. Previous research has shown that Armcx1 is expressed at higher levels in mice after axotomy and in adult retinal ganglion cells after crush injury, and this protein increases neuronal survival and axonal regeneration. However, its role in traumatic brain injury (TBI) is unclear. Therefore, the aim of this study was to assess the expression of Armcx1 after TBI and to explore possible related mechanisms by which Armcx1 is involved in TBI. We used C57BL/6 male mice to model TBI and evaluated the role of Armcx1 in TBI by transfecting mice with Armcx1 small interfering RNA (siRNA) to inhibit Armcx1 expression 24 h before TBI modeling. Western blotting, immunofluorescence, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining, Nissl staining, transmission electron microscopy, adenosine triphosphate (ATP) level measurement, neuronal apoptosis analysis, neurological function scoring and the Morris water maze were performed. The results demonstrated that Armcx1 protein expression was elevated after TBI and that the Armcx1 protein was localized in neurons and astroglial cells in cortical tissue surrounding the injury site. In addition, inhibition of Armcx1 expression further led to impaired mitochondrial transport, abnormal morphology, reduced ATP levels, aggravation of neuronal apoptosis and neurological dysfunction, and decrease Miro1 expression. In conclusion, our findings indicate that Armcx1 may exert neuroprotective effects by ameliorating neurological injury after TBI through a mitochondrial transport pathway involving Miro1.
Collapse
Affiliation(s)
- Qiuying Li
- Department of Geriatrics, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou 215600, China
| | - Haibo Ni
- Department of Neurosurgery, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou 215600, China; Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qin Rui
- Department of Laboratory, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou 215600, China
| | - Jiasheng Ding
- Department of Neurosurgery, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou 215600, China
| | - Xianghu Kong
- Department of Burn and Plastic Surgery, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou 215600, China
| | - Xugang Kan
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Rong Gao
- Department of Neurosurgery, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou 215600, China.
| | - Hongbo Shen
- Department of Geriatrics, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou 215600, China.
| |
Collapse
|
11
|
Allen J, Dames SS, Foldi CJ, Shultz SR. Psychedelics for acquired brain injury: a review of molecular mechanisms and therapeutic potential. Mol Psychiatry 2024; 29:671-685. [PMID: 38177350 DOI: 10.1038/s41380-023-02360-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Acquired brain injury (ABI), such as traumatic brain injury and stroke, is a leading cause of disability worldwide, resulting in debilitating acute and chronic symptoms, as well as an increased risk of developing neurological and neurodegenerative disorders. These symptoms can stem from various neurophysiological insults, including neuroinflammation, oxidative stress, imbalances in neurotransmission, and impaired neuroplasticity. Despite advancements in medical technology and treatment interventions, managing ABI remains a significant challenge. Emerging evidence suggests that psychedelics may rapidly improve neurobehavioral outcomes in patients with various disorders that share physiological similarities with ABI. However, research specifically focussed on psychedelics for ABI is limited. This narrative literature review explores the neurochemical properties of psychedelics as a therapeutic intervention for ABI, with a focus on serotonin receptors, sigma-1 receptors, and neurotrophic signalling associated with neuroprotection, neuroplasticity, and neuroinflammation. The promotion of neuronal growth, cell survival, and anti-inflammatory properties exhibited by psychedelics strongly supports their potential benefit in managing ABI. Further research and translational efforts are required to elucidate their therapeutic mechanisms of action and to evaluate their effectiveness in treating the acute and chronic phases of ABI.
Collapse
Affiliation(s)
- Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Shannon S Dames
- Psychedelic-Assisted Therapy Post-Graduate Program, Health Sciences and Human Services, Vancouver Island University, Nanaimo, BC, Canada
| | - Claire J Foldi
- Department of Physiology, Monash University, Clayton, VIC, Australia
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Centre for Trauma and Mental Health Research, Health Sciences and Human Services, Vancouver Island University, Nanaimo, BC, Canada.
| |
Collapse
|
12
|
Kureshi S, Mendizabal M, Francis J, Djalilian HR. Conservative Management of Acute Sports-Related Concussions: A Narrative Review. Healthcare (Basel) 2024; 12:289. [PMID: 38338173 PMCID: PMC10855441 DOI: 10.3390/healthcare12030289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
This review explores the application of the conservative management model for pain to sports-related concussions (SRCs), framing concussions as a distinct form of pain syndrome with a pathophysiological foundation in central sensitization. Drawing parallels with proven pain management models, we underscore the significance of a proactive approach to concussion management. Recognizing concussions as a pain syndrome allows for the tailoring of interventions in alignment with conservative principles. This review first covers the epidemiology and controversies surrounding prolonged concussion recovery and persistent post-concussion symptoms (PPCS). Next, the pathophysiology of concussions is presented within the central sensitization framework, emphasizing the need for early intervention to mitigate the neuroplastic changes that lead to heightened pain sensitivity. Five components of the central sensitization process specific to concussion injuries are highlighted as targets for conservative interventions in the acute period: peripheral sensitization, cerebral metabolic dysfunction, neuroinflammation, glymphatic system dysfunction, and pain catastrophizing. These proactive interventions are emphasized as pivotal in accelerating concussion recovery and reducing the risk of prolonged symptoms and PPCS, in line with the philosophy of conservative management.
Collapse
Affiliation(s)
- Sohaib Kureshi
- Neurosurgical Medical Clinic, San Diego, CA 92111, USA
- TBI Virtual, San Diego, CA 92111, USA
| | | | | | - Hamid R. Djalilian
- TBI Virtual, San Diego, CA 92111, USA
- Departments of Otolaryngology, Neurological Surgery, and Biomedical Engineering, University of California, Irvine, CA 92697, USA
| |
Collapse
|
13
|
Wang R, Wang J, Zhang Z, Ma B, Sun S, Gao L, Gao G. FGF21 alleviates endothelial mitochondrial damage and prevents BBB from disruption after intracranial hemorrhage through a mechanism involving SIRT6. Mol Med 2023; 29:165. [PMID: 38049769 PMCID: PMC10696847 DOI: 10.1186/s10020-023-00755-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: 06/04/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Disruption of the BBB is a harmful event after intracranial hemorrhage (ICH), and this disruption contributes to a series of secondary injuries. We hypothesized that FGF21 may have protective effects after intracranial hemorrhage (ICH) and investigated possible underlying molecular mechanisms. METHODS Blood samples of ICH patients were collected to determine the relationship between the serum level of FGF21 and the [Formula: see text]GCS%. Wild-type mice, SIRT6flox/flox mice, endothelial-specific SIRT6-homozygous-knockout mice (eSIRT6-/- mice) and cultured human brain microvascular endothelial cells (HCMECs) were used to determine the protective effects of FGF21 on the BBB. RESULTS We obtained original clinical evidence from patient data identifying a positive correlation between the serum level of FGF21 and [Formula: see text]GCS%. In mice, we found that FGF21 treatment is capable of alleviating BBB damage, mitigating brain edema, reducing lesion volume and improving neurofunction after ICH. In vitro, after oxyhemoglobin injury, we further explored the protective effects of FGF21 on endothelial cells (ECs), which are a significant component of the BBB. Mitochondria play crucial roles during various types of stress reactions. FGF21 significantly improved mitochondrial biology and function in ECs, as evidenced by alleviated mitochondrial morphology damage, reduced ROS accumulation, and restored ATP production. Moreover, we found that the crucial regulatory mitochondrial factor deacylase sirtuin 6 (SIRT6) played an irreplaceable role in the effects of FGF21. Using endothelial-specific SIRT6-knockout mice, we found that SIRT6 deficiency largely diminished these neuroprotective effects of FGF21. Then, we revealed that FGF21 might promote the expression of SIRT6 via the AMPK-Foxo3a pathway. CONCLUSIONS We provide the first evidence that FGF21 is capable of protecting the BBB after ICH by improving SIRT6-mediated mitochondrial homeostasis.
Collapse
Affiliation(s)
- Runfeng Wang
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Jin Wang
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Zhiguo Zhang
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Bo Ma
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Shukai Sun
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
14
|
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
|
15
|
Wang J, Yang S, Li H, Shen H, Lu X, Li X, Chen G. Downregulation of mitochondrial calcium uptake family 3 attenuates secondary brain injury after intracerebral hemorrhage in rats. Exp Neurol 2023; 361:114302. [PMID: 36549422 DOI: 10.1016/j.expneurol.2022.114302] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Intracerebral hemorrhage (ICH) is one type of stroke with a high incidence and mortality. Mitochondria provide energy for various life processes and regulate calcium-mediated signaling pathways by taking up calcium ions from cytoplasm. Mitochondrial calcium uptake family 3 (MICU3) is a tissue-specific enhancer of mitochondrial calcium uptake. The effects and mechanisms of MICU3 in ICH are unknown. In this study, we aimed to explore the role of MICU3 in ICH in rats and neuronal models. First, we constructed ICH model both in vivo and in vitro and observed increased expression of MICU3. Then lentivirus was transduced to knock down MICU3. We observed that knockdown of MICU3 significantly reduced mitochondrial uptake of calcium in primary neurons. Moreover, the downregulation of MICU3 attenuated cell apoptosis and decreased the accumulation of reactive oxygen species (ROS). Recovery of neurobehavioral and cognitive function also benefited from downregulation of MICU3. The findings demonstrated that MICU3 played an important role in cell apoptosis, oxidative stress, and maintenance of mitochondrial structure and function, and promoted rehabilitation of neurobehavior. In conclusion, MICU3 is expected to be a molecular marker and a potential therapeutic target for ICH.
Collapse
Affiliation(s)
- Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Siyuan Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Xiaocheng Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou 215006, China
| |
Collapse
|
16
|
Mitophagy and Traumatic Brain Injury: Regulatory Mechanisms and Therapeutic Potentials. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:1649842. [PMID: 36846712 PMCID: PMC9957633 DOI: 10.1155/2023/1649842] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/25/2022] [Accepted: 01/21/2023] [Indexed: 02/19/2023]
Abstract
Traumatic brain injury (TBI), a kind of external trauma-induced brain function alteration, has posed a financial burden on the public health system. TBI pathogenesis involves a complicated set of events, including primary and secondary injuries that can cause mitochondrial damage. Mitophagy, a process in which defective mitochondria are specifically degraded, segregates and degrades defective mitochondria allowing a healthier mitochondrial network. Mitophagy ensures that mitochondria remain healthy during TBI, determining whether neurons live or die. Mitophagy acts as a critical regulator in maintaining neuronal survival and healthy. This review will discuss the TBI pathophysiology and the consequences of the damage it causes to mitochondria. This review article will explore the mitophagy process, its key factors, and pathways and reveal the role of mitophagy in TBI. Mitophagy will be further recognized as a therapeutic approach in TBI. This review will offer new insights into mitophagy's role in TBI progression.
Collapse
|
17
|
Hu H, Chen X, Zhao K, Zheng W, Gao C. Recent Advances in Biomaterials-Based Therapies for Alleviation and Regeneration of Traumatic Brain Injury. Macromol Biosci 2023; 23:e2200577. [PMID: 36758541 DOI: 10.1002/mabi.202200577] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Indexed: 02/11/2023]
Abstract
Traumatic brain injury (TBI), a major public health problem accompanied with numerous complications, usually leads to serve disability and huge financial burden. The adverse and unfavorable pathological environment triggers a series of secondary injuries, resulting in serious loss of nerve function and huge obstacle of endogenous nerve regeneration. With the advances in adaptive tissue regeneration biomaterials, regulation of detrimental microenvironment to reduce the secondary injury and to promote the neurogenesis becomes possible. The adaptive biomaterials could respond and regulate biochemical, cellular, and physiological events in the secondary injury, including excitotoxicity, oxidative stress, and neuroinflammation, to rebuild circumstances suitable for regeneration. In this review, the development of pathology after TBI is discussed, followed by the introduction of adaptive biomaterials based on various pathological characteristics. The adaptive biomaterials carried with neurotrophic factors and stem cells for TBI treatment are then summarized. Finally, the current drawbacks and future perspective of biomaterials for TBI treatment are suggested.
Collapse
Affiliation(s)
- Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiping Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Kefei Zhao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.,Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| |
Collapse
|
18
|
Stevens AR, Hadis M, Milward M, Ahmed Z, Belli A, Palin W, Davies DJ. Photobiomodulation in Acute Traumatic Brain Injury: A Systematic Review and Meta-Analysis. J Neurotrauma 2023; 40:210-227. [PMID: 35698294 DOI: 10.1089/neu.2022.0140] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Photobiomodulation (PBM) is a therapeutic modality that has gained increasing interest in neuroscience applications, including acute traumatic brain injury (TBI). Its proposed mechanisms for therapeutic effect when delivered to the injured brain include antiapoptotic and anti-inflammatory effects. This systematic review summarizes the available evidence for the value of PBM in improving outcomes in acute TBI and presents a meta-analysis of the pre-clinical evidence for neurological severity score (NSS) and lesion size in animal models of TBI. A systematic review of the literature was performed, with searches and data extraction performed independently in duplicate by two authors. Eighteen published articles were identified for inclusion: seventeen pre-clinical studies of in vivo animal models and one clinical study in human patients. The available human study supports safety and feasibility of PBM in acute moderate TBI. For pre-clinical studies, meta-analysis for NSS and lesion size were found to favor intervention versus control. Subgroup analysis based on PBM parameter variables for these outcomes was performed. Favorable parameters were identified as: wavelengths in the region of 665 nm and 810 nm; time to first administration of PBM ≤4 h; total number of daily treatments ≤3. No differences were identified between pulsed and continuous wave modes or energy delivery. Mechanistic substudies within included in vivo studies are presented and were found to support hypotheses of antiapoptotic, anti-inflammatory, and pro-proliferative effects, and a modulation of cellular metabolism. This systematic review provides substantial meta-analysis evidence of the benefits of PBM on functional and histological outcomes of TBI in in vivo mammalian models. Study design and PBM parameters should be closely considered for future human clinical studies.
Collapse
Affiliation(s)
- Andrew Robert Stevens
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - Mohammed Hadis
- Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - Michael Milward
- Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - Zubair Ahmed
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Antonio Belli
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - William Palin
- Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - David James Davies
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom.,Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
19
|
Smith AN, Shaughness M, Collier S, Hopkins D, Byrnes KR. Therapeutic targeting of microglia mediated oxidative stress after neurotrauma. Front Med (Lausanne) 2022; 9:1034692. [PMID: 36405593 PMCID: PMC9671221 DOI: 10.3389/fmed.2022.1034692] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 10/06/2023] Open
Abstract
Inflammation is a primary component of the central nervous system injury response. Traumatic brain and spinal cord injury are characterized by a pronounced microglial response to damage, including alterations in microglial morphology and increased production of reactive oxygen species (ROS). The acute activity of microglia may be beneficial to recovery, but continued inflammation and ROS production is deleterious to the health and function of other cells. Microglial nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), mitochondria, and changes in iron levels are three of the most common sources of ROS. All three play a significant role in post-traumatic brain and spinal cord injury ROS production and the resultant oxidative stress. This review will evaluate the current state of therapeutics used to target these avenues of microglia-mediated oxidative stress after injury and suggest avenues for future research.
Collapse
Affiliation(s)
- Austin N. Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Michael Shaughness
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Sean Collier
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Deanna Hopkins
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
20
|
Dave KM, Dobbins DX, Farinelli MN, Sullivan A, Milosevic J, Stolz DB, Kim J, Zheng S, Manickam DS. Engineering Extracellular Vesicles to Modulate Their Innate Mitochondrial Load. Cell Mol Bioeng 2022; 15:367-389. [PMID: 36444353 PMCID: PMC9700543 DOI: 10.1007/s12195-022-00738-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Extracellular vesicles (EVs) are promising carriers for the delivery of biotherapeutic cargo such as RNA and proteins. We have previously demonstrated that the innate EV mitochondria in microvesicles (MVs), but not exosomes (EXOs) can be transferred to recipient BECs and mouse brain slice neurons. Here, we sought to determine if the innate EV mitochondrial load can be further increased via increasing mitochondrial biogenesis in the donor cells. We hypothesized that mitochondria-enriched EVs ("mito-EVs") may increase the recipient BEC ATP levels to a greater extent than naïve MVs. Methods We treated NIH/3T3, a fibroblast cell line and hCMEC/D3, a human brain endothelial cell (BEC) line using resveratrol to activate peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), the central mediator of mitochondrial biogenesis. Naïve EVs and mito-EVs isolated from the non-activated and activated donor cells were characterized using transmission electron microscopy, dynamic light scattering and nanoparticle tracking analysis. The effect of mito-EVs on resulting ATP levels in the recipient BECs were determined using Cell Titer Glo ATP assay. The uptake of Mitotracker Red-stained EVs into recipient BECs and their colocalization with recipient BEC mitochondria were studied using flow cytometry and fluorescence microscopy. Results Resveratrol treatment increased PGC-1α expression in the donor cells. Mito-MVs but not mito-EXOs showed increased expression of mitochondrial markers ATP5A and TOMM20 compared to naïve MVs. TEM images showed that a greater number of mito-MVs contained mitochondria compared to naïve MVs. Mito-MVs but not mito-EXOs showed a larger particle diameter compared to their naïve EV counterparts from the non-activated cells suggesting increased mitochondria incorporation. Mito-EVs were generated at higher particle concentrations compared to naïve EVs from non-activated cells. Mito-EVs increased the cellular ATP levels and transferred their mitochondrial load into the recipient BECs. Mito-MV mitochondria also colocalized with recipient BEC mitochondria. Conclusions Our results suggest that the pharmacological modulation of mitochondrial biogenesis in the donor cells can change the mitochondrial load in the secreted MVs. Outcomes of physicochemical characterization studies and biological assays confirmed the superior effects of mito-MVs compared to naïve MVs-suggesting their potential to improve mitochondrial function in neurovascular and neurodegenerative diseases. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00738-8.
Collapse
Affiliation(s)
- Kandarp M. Dave
- Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, 453 Mellon Hall, Pittsburgh, PA USA
| | - Duncan X. Dobbins
- Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, 453 Mellon Hall, Pittsburgh, PA USA
| | - Maura N. Farinelli
- Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, 453 Mellon Hall, Pittsburgh, PA USA
- Department of Biochemistry and Molecular Biology, Gettysburg College, Gettysburg, PA USA
| | - Abigail Sullivan
- Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, 453 Mellon Hall, Pittsburgh, PA USA
- Psychological and Brain Sciences, Villanova University, Villanova, PA USA
| | - Jadranka Milosevic
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA USA
- Captis Diagnostics Inc, Pittsburgh, PA USA
| | - Donna B. Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA USA
| | - Jeongyun Kim
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA USA
| | - Siyang Zheng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA USA
| | - Devika S. Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, 453 Mellon Hall, Pittsburgh, PA USA
| |
Collapse
|
21
|
Unraveling axonal mechanisms of traumatic brain injury. Acta Neuropathol Commun 2022; 10:140. [PMID: 36131329 PMCID: PMC9494812 DOI: 10.1186/s40478-022-01414-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Axonal swellings (AS) are one of the neuropathological hallmark of axonal injury in several disorders from trauma to neurodegeneration. Current evidence proposes a role of perturbed Ca2+ homeostasis in AS formation, involving impaired axonal transport and focal distension of the axons. Mechanisms of AS formation, in particular moments following injury, however, remain unknown. Here we show that AS form independently from intra-axonal Ca2+ changes, which are required primarily for the persistence of AS in time. We further show that the majority of axonal proteins undergoing de/phosphorylation immediately following injury belong to the cytoskeleton. This correlates with an increase in the distance of the actin/spectrin periodic rings and with microtubule tracks remodeling within AS. Observed cytoskeletal rearrangements support axonal transport without major interruptions. Our results demonstrate that the earliest axonal response to injury consists in physiological adaptations of axonal structure to preserve function rather than in immediate pathological events signaling axonal destruction.
Collapse
|
22
|
The Role of Hydrogen Sulfide Targeting Autophagy in the Pathological Processes of the Nervous System. Metabolites 2022; 12:metabo12090879. [PMID: 36144282 PMCID: PMC9502065 DOI: 10.3390/metabo12090879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Autophagy is an important cellular process, involving the transportation of cytoplasmic contents in the double membrane vesicles to lysosomes for degradation. Autophagy disorder contributes to many diseases, such as immune dysfunction, cancers and nervous system diseases. Hydrogen sulfide (H2S) is a volatile and toxic gas with a rotten egg odor. For a long time, it was considered as an environmental pollution gas. In recent years, H2S is regarded as the third most important gas signal molecule after NO and CO. H2S has a variety of biological functions and can play an important role in a variety of physiological and pathological processes. Increasingly more evidences show that H2S can regulate autophagy to play a protective role in the nervous system, but the mechanism is not fully understood. In this review, we summarize the recent literatures on the role of H2S in the pathological process of the nervous system by regulating autophagy, and analyze the mechanism in detail, hoping to provide the reference for future related research.
Collapse
|
23
|
Understanding Acquired Brain Injury: A Review. Biomedicines 2022; 10:biomedicines10092167. [PMID: 36140268 PMCID: PMC9496189 DOI: 10.3390/biomedicines10092167] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/02/2022] [Accepted: 08/26/2022] [Indexed: 01/19/2023] Open
Abstract
Any type of brain injury that transpires post-birth is referred to as Acquired Brain Injury (ABI). In general, ABI does not result from congenital disorders, degenerative diseases, or by brain trauma at birth. Although the human brain is protected from the external world by layers of tissues and bone, floating in nutrient-rich cerebrospinal fluid (CSF); it remains susceptible to harm and impairment. Brain damage resulting from ABI leads to changes in the normal neuronal tissue activity and/or structure in one or multiple areas of the brain, which can often affect normal brain functions. Impairment sustained from an ABI can last anywhere from days to a lifetime depending on the severity of the injury; however, many patients face trouble integrating themselves back into the community due to possible psychological and physiological outcomes. In this review, we discuss ABI pathologies, their types, and cellular mechanisms and summarize the therapeutic approaches for a better understanding of the subject and to create awareness among the public.
Collapse
|
24
|
Hamdan AME, Mohammedsaleh ZM, Aboelnour A, Elkannishy SM. Preclinical study for the ameliorating effect of l-ascorbic acid for the oxidative stress of chronic administration of organic nitrates on myocardial tissue in high sucrose/fat rat model. Saudi Pharm J 2022; 30:1405-1417. [PMID: 36387332 PMCID: PMC9649357 DOI: 10.1016/j.jsps.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
Background The therapeutic activity of Glyceryl trinitrate (GTN) is mainly regulated by liberating nitric oxide (NO) and reactive nitrogen species (RNS). During this biotransformation, oxidative stress and lipid peroxidation inside the red blood cells (RBCs) occur. Hemoglobin tightly binds to NO forming methemoglobin altering the erythrocytic antioxidant defense system. Aim The principal objective of our research is to show the ameliorating effect of l-ascorbic acid for the deleterious effects of chronic administration of nitrovasodilator drugs used in cardiovascular diseases such as oxidative stresses and tolerance. Method We studied some biochemical parameters for the oxidative stress using groups of high sucrose/fat (HSF) diet Wistar male rats chronically orally administered different concentrations of Isosorbide-5-mononitrate (ISMN) 0.3 mg/kg, 0.6 mg/kg and 1.2 mg/kg. Afterwards, we evaluated the role of l-ascorbic acid against these biochemical changes in cardiac tissues. Results Chronic treatment with organic nitrates caused elevated serum levels of lipid peroxidation, hemoglobin derivatives as methemoglobin and carboxyhemoglobin, rate of hemoglobin autoxidation, the cellular levels of the pro-inflammatory cytokines marker (NF-κB) and apoptosis markers (caspase-3) in the myocardium muscles in a dose-dependent manner. Meanwhile, such exposure caused a decline in the enzymatic effect of SOD, GSH and CAT accompanied by a decrease in the level of mitochondrial oxidative stress marker (nrf2) in the myocardium muscles and a decrease in the serum iron and total iron-binding capacity (TIBC) in a dose-dependent manner. Concomitant treatment with l-ascorbic acid significantly diminished these changes for all examined parameters. Conclusion Chronic administration of organic nitrates leads to the alteration of the level of oxidative stress factors in the myocardium tissue due to the generation of reactive oxygen species. Using l-ascorbic acid can effectively ameliorate such intoxication to overcome nitrate tolerance.
Collapse
|
25
|
Li Y, Liu H, Tian C, An N, Song K, Wei Y, Sun Y, Xing Y, Gao Y. Targeting the multifaceted roles of mitochondria in intracerebral hemorrhage and therapeutic prospects. Biomed Pharmacother 2022; 148:112749. [PMID: 35219118 DOI: 10.1016/j.biopha.2022.112749] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/19/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a severe, life-threatening subtype of stoke that constitutes a crucial health and socioeconomic problem worldwide. However, the current clinical treatment can only reduce the mortality of patients to a certain extent, but cannot ameliorate neurological dysfunction and has a high recurrence rate. Increasing evidence has demonstrated that mitochondrial dysfunction occurs in the early stages of brain injury and participates in all stages of secondary brain injury (SBI) after ICH. As the energy source of cells, various pathobiological processes that lead to SBI closely interact with the mitochondria, such as oxidative stress, calcium overload, and neuronal injury. In this review, we discussed the structure and function of mitochondria and the abnormal morphological changes after ICH. In addition, we discussed recent research on the involvement of mitochondrial dynamics in the pathological process of SBI after ICH and introduced the pathological variations and related molecular mechanisms of mitochondrial dysfunction in the occurrence of brain injury. Finally, we summarized the latest progress in mitochondrion-targeted agents for ICH, which provides a direction for the development of emerging therapeutic strategies targeting the mitochondria after ICH.
Collapse
Affiliation(s)
- Yuanyuan Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing 100700, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haoqi Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Chao Tian
- Beijing University of Chinese Medicine, Beijing 100029, China; China-Japan Friendship Hospital, Beijing 100029, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ke Song
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yufei Wei
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Guangxi 530000, China
| | - Yikun Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yanwei Xing
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|