1
|
Lu X, Yang S, Lu Q, Zhang Y, Cha Z, Huang W, Li T. Betulin ameliorates neuronal apoptosis and oxidative injury via DJ-1/Akt/Nrf2 signaling pathway after subarachnoid hemorrhage. CNS Neurosci Ther 2024; 30:e70019. [PMID: 39238115 PMCID: PMC11377304 DOI: 10.1111/cns.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/26/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024] Open
Abstract
AIMS We aimed to resolve the uncertainty as to whether betulin exerted neuroprotection on early brain injury (EBI) caused by subarachnoid hemorrhage (SAH), and to investigate the related molecular mechanisms. METHODS Bioinformatic analysis was performed to pre-study the differently expressed genes (DEGs) and the possible signaling pathways. Rat and cellular model of SAH were introduced in this study, and betulin, an activator of DJ-1 protein, was administered to reveal the effect. Gross assessment regarding mortality, neurofunctions, SAH grade, brain water content (BWC) along with multiple cellular and molecular studies in vivo or/and in vitro such as immunofluorescence (IF) staining, western blot (WB), reactive oxygen species (ROS) assay, and flow cytometry (FCM) were all conducted after SAH induction to verify the protective effect and the relevant mechanisms of DJ-1 in diverse levels. In addition, MK2206 (selective inhibitor of Akt) and iRNADj-1 (interfering RNA to Dj-1) were utilized to confirm the mechanisms of the effect. RESULTS The data from our study showed that DJ-1 protein was moderately expressed in neurons, microglia, and astrocytes; its level in brain tissue elevated and peaked at 24-72 h after SAH induction. Betulin could efficaciously induce the expression of DJ-1 which in turn activated Akt and Bcl-2, and anti-oxidative enzymes SOD2 and HO-1, functioning to reduce the activation of cleaved caspase-3 (c-Casp-3) and reactive oxygen species (ROS). The induced DJ-1 could upregulate the expression of Nrf2. However, Akt seemed no direct effect on elevating the expression of Nrf2. DJ-1 alone could as well activate Akt-independent antiapoptotic pathway via suppressing the activation of caspase-8 (Casp-8). CONCLUSIONS Betulin which was a potent agonist of DJ-1 had the ability to induce its expression in brain tissue. DJ-1 had neuroprotective effect on EBI through comprehensive mechanisms, including facilitating intrinsic and extrinsic antiapoptotic pathway, and reducing oxidative injury by upregulating the expression of redox proteins. Betulin as an inexpensive drug showed the potential for SAH treatment.
Collapse
Affiliation(s)
- Xiaoyang Lu
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Shu Yang
- Department of Neurology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qixiong Lu
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yuansheng Zhang
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zaihong Cha
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Wei Huang
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Tao Li
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
2
|
Yang G, Khan A, Liang W, Xiong Z, Stegbauer J. Aortic aneurysm: pathophysiology and therapeutic options. MedComm (Beijing) 2024; 5:e703. [PMID: 39247619 PMCID: PMC11380051 DOI: 10.1002/mco2.703] [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: 12/19/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Aortic aneurysm (AA) is an aortic disease with a high mortality rate, and other than surgery no effective preventive or therapeutic treatment have been developed. The renin-angiotensin system (RAS) is an important endocrine system that regulates vascular health. The ACE2/Ang-(1-7)/MasR axis can antagonize the adverse effects of the activation of the ACE/Ang II/AT1R axis on vascular dysfunction, atherosclerosis, and the development of aneurysms, thus providing an important therapeutic target for the prevention and treatment of AA. However, products targeting the Ang-(1-7)/MasR pathway still lack clinical validation. This review will outline the epidemiology of AA, including thoracic, abdominal, and thoracoabdominal AA, as well as current diagnostic and treatment strategies. Due to the highest incidence and most extensive research on abdominal AA (AAA), we will focus on AAA to explain the role of the RAS in its development, the protective function of Ang-(1-7)/MasR, and the mechanisms involved. We will also describe the roles of agonists and antagonists, suggest improvements in engineering and drug delivery, and provide evidence for Ang-(1-7)/MasR's clinical potential, discussing risks and solutions for clinical use. This study will enhance our understanding of AA and offer new possibilities and promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Guang Yang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Institute of Translational Medicine Shenzhen Second People's Hospital The First Affiliated Hospital of Shenzhen University Shenzhen China
- Department of Life Sciences Yuncheng University Yuncheng China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Abbas Khan
- Department of Nutrition and Health Promotion University of Home Economics Lahore Pakistan Lahore Pakistan
| | - Wei Liang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Zibo Xiong
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Johannes Stegbauer
- Department of Nephrology Medical Faculty University Hospital Düsseldorf Heinrich Heine University Düsseldorf Düsseldorf Germany
| |
Collapse
|
3
|
Pan H, Huang M, Zhu C, Lin S, He L, Shen R, Chen Y, Fang F, Qiu Y, Qin M, Bao P, Tan Y, Xu J, Ding J, Chen S. A novel compound alleviates oxidative stress via PKA/CREB1-mediated DJ-1 upregulation. J Neurochem 2024; 168:3034-3049. [PMID: 38994800 DOI: 10.1111/jnc.16161] [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: 06/10/2023] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024]
Abstract
Oxidative stress is one of the major culprits causing dopaminergic neuron loss in Parkinson's disease (PD). DJ-1 is a protein with multiple actions against oxidative stress, apoptosis, neuroinflammation, etc. DJ-1 expression is decreased in sporadic PD, therefore increasing DJ-1 expression might be beneficial in PD treatment. However, drugs known to upregulate DJ-1 are still lacking. In this study, we identified a novel DJ-1-elevating compound called ChemJ through luciferase assay-based high-throughput compound screening in SH-SY5Y cells and confirmed that ChemJ upregulated DJ-1 in SH-SY5Y cell line and primary cortical neurons. DJ-1 upregulation by ChemJ alleviated MPP+-induced oxidative stress. In exploring the underlying mechanisms, we found that the transcription factor CREB1 bound to DJ-1 promoter and positively regulated its expression under both unstressed and 1-methyl-4-phenylpyridinium-induced oxidative stress conditions and that ChemJ promoted DJ-1 expression via activating PKA/CREB1 pathway in SH-SY5Y cells. Our results demonstrated that ChemJ alleviated the MPP+-induced oxidative stress through a PKA/CREB1-mediated regulation of DJ-1 expression, thus offering a novel and promising avenue for PD treatment.
Collapse
Affiliation(s)
- Hong Pan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China
| | - Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxiang Zhu
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China
| | - Suzhen Lin
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruinan Shen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimeng Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Fang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinghui Qiu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiling Qin
- Institute of Neuroscience and State key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Puhua Bao
- Institute of Neuroscience and State key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yuyan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Xu
- Institute of Neuroscience and State key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China
| |
Collapse
|
4
|
Liu T, Li J, Sun L, Zhu C, Wei J. The role of ACE2 in RAS axis on microglia activation in Parkinson's disease. Neuroscience 2024; 553:128-144. [PMID: 38986737 DOI: 10.1016/j.neuroscience.2024.06.024] [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: 03/26/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The classic renin-angiotensin system (RAS) induces organ damage, while the ACE2/Ang-(1-7)/MasR axis opposes it. However, the role of ACE2 in the brain is unclear. We studied ACE2's role in the brain. METHOD We used male C57BL/6J (WT) mice, ACE2 knockout (KO) mice, and MPTP-induced mice. Behavioral tests confirmed successful modeling. We assessed the impact of ACE2 KO on the RAS axis and PD index, including ACE, ACE2, AT1, AT2, MasR, TH, α-syn, and Iba1. We investigated ACE2 and MasR's involvement in microglial activation via western blot and immunofluorescence. GSE10867 and GSE26532 datasets were used to analyze the effects of AT1 antagonists and in vitro PD models on microglia. RESULT Behavioral tests revealed that MPTP mice displayed motor deficits, depression, anxiety, and increased inflammatory markers in the SN and CPU, with reduced antioxidant capacity. ACE2 KO worsened these symptoms and exacerbated inflammation and oxidative stress. LPS-induced ACE2/MasR activation in BV2 cells demonstrated anti-inflammatory and neuroprotective effects by modulating microglial polarization. Antagonists inhibited microglial activation via inflammation and ROS processes. CONCLUSION The RAS axis regulates inflammation and oxidative stress to maintain CNS function, suggesting potential targets for neurologic disease treatment. Understanding microglial RAS activation can offer new therapeutic strategies.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Jingwen Li
- Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475000, China.
| | - Chaoyang Zhu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China.
| |
Collapse
|
5
|
Wang Y, Zhao M, Li J, Liu Y. Loganin exerts neuroprotective effect by inhibiting neuronal pyroptosis in rat with cerebral haemorrhage. Clin Exp Pharmacol Physiol 2024; 51:e13858. [PMID: 38636940 DOI: 10.1111/1440-1681.13858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/25/2024] [Accepted: 03/18/2024] [Indexed: 04/20/2024]
Abstract
Intracerebral haemorrhage (ICH) presents significant challenges in clinical management because of the high morbidity and mortality, necessitating novel therapeutic approaches. This study aimed to assess the neuroprotective effects of loganin in a rat ICH model. Sprague-Dawley rats were used, subjected to a collagenase-induced ICH model, followed by loganin treatment at doses of 2.5, 5 and 10 mg/kg. Neurological functions were evaluated using the modified neurological severity score (mNSS) and a rotarod test. Results indicated a significant improvement in neurological functions in loganin-treated groups, evident from the mNSS and rotarod tests, suggesting dose-dependent neuroprotection. Loganin also effectively reduced the blood-brain barrier (BBB) permeability and cerebral oedema. Additionally, it mitigated cellular pyroptosis, as shown by terminal deoxynucleotidyl transferase dUTP nick-end labelling staining and western blot analysis, which indicated reduced levels of pyroptosis markers in treated rats. Furthermore, loganin's regulatory effects on the adenosine A2A receptor and myosin light chain kinase pathways were observed, potentially underpinning its protective mechanism against ICH. The study concludes that loganin exhibits significant neuroprotective properties in a rat ICH model, highlighting its potential as a novel therapeutic strategy. Despite promising results, the study needs further research to determine loganin's therapeutic potential in human ICH patients. This research paves the way for further exploration into loganin's clinical applications, potentially revolutionizing treatment strategies for patients suffering from intracerebral haemorrhage.
Collapse
Affiliation(s)
- Yu Wang
- Center of Encephalopathy, Changchun University of Traditional Chinese Medicine Affiliated Third Clinical Hospital, Changchun, China
| | - Min Zhao
- Center of Encephalopathy, The Third Clinical Hospital Affiliated to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jing Li
- Center of Encephalopathy, Changchun University of Traditional Chinese Medicine Affiliated Third Clinical Hospital, Changchun, China
| | - Yue Liu
- Center of Encephalopathy, Changchun University of Traditional Chinese Medicine Affiliated Third Clinical Hospital, Changchun, China
| |
Collapse
|
6
|
Hong J, Li Y, Chen L, Han D, Li Y, Mi X, Liu K, Wang Q, Song Y, Liu T, Yang N, Liu Y, Li Z, Guo X. A53T α-synuclein mutation increases susceptibility to postoperative delayed neurocognitive recovery via hippocampal Ang-(1-7)/MasR axis. Biochem Pharmacol 2024; 224:116261. [PMID: 38705534 DOI: 10.1016/j.bcp.2024.116261] [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: 01/15/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Delayed neurocognitive recovery (dNCR) is a common complication in geriatric surgical patients. The impact of anesthesia and surgery on patients with neurodegenerative diseases, such as Parkinson's disease (PD) or prion disease, has not yet been reported. In this study, we aimed to determine the association between a pre-existing A53T genetic background, which involves a PD-related point mutation, and the development of postoperative dNCR. We observed that partial hepatectomy induced hippocampus-dependent cognitive deficits in 5-month-old A53T transgenic mice, a model of early-stage PD without cognitive deficits, unlike in age-matched wild-type (WT) mice. We respectively examined molecular changes at 6 h, 1 day, and 2 days after partial hepatectomy and observed that cognitive changes were accompanied by weakened angiotensin-(1-7)/Mas receptor [Ang-(1-7)/MasR] axis, increased alpha-synuclein (α-syn) expression and phosphorylation, decreased methylated protein phosphatase-2A (Me-PP2A), and prompted microglia M1 polarization and neuronal apoptosis in the hippocampus at 1 day after surgery. Nevertheless, no changes in blood-brain barrier (BBB) integrity or plasma α-syn levels in either A53T or WT mice. Furthermore, intranasal administration of selective MasR agonist AVE 0991, reversed the mentioned cognitive deficits in A53T mice, enhanced MasR expression, reduced α-syn accumulation and phosphorylation, and attenuated microglia activation and apoptotic response. Our findings suggest that individuals with the A53T genetic background may be more susceptible to developing postoperative dNCR. This susceptibility could be linked to central α-syn accumulation mediated by the weakened Ang-(1-7)/MasR/methyl-PP2A signaling pathway in the hippocampus following surgery, independent of plasma α-syn level and BBB.
Collapse
Affiliation(s)
- Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yitong Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Kaixi Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yanan Song
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yajie Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| |
Collapse
|
7
|
Hao J, Wang T, Cao C, Li X, Li H, Gao H, Li J, Shen H, Chen G. LPCAT3 exacerbates early brain injury and ferroptosis after subarachnoid hemorrhage in rats. Brain Res 2024; 1832:148864. [PMID: 38484924 DOI: 10.1016/j.brainres.2024.148864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
AIMS Lysophosphatidylcholine acyltransferase 3 (LPCAT3) is known to play a pivotal role in lipid metabolism, but its role in the early brain injury (EBI) following subarachnoid hemorrhage (SAH) remains unclear. This study provides insights into LPCAT3 expression alterations and functional implications in EBI following SAH. METHODS SAH models of adult male Sprague-Dawley (SD) rats were established by intravascular perforation. Lentivirus vectors were administered by intracerebroventricular injection (i.c.v.) to either induce LPCAT3 overexpression or knockdown 14 days before SAH induction. Western blot, immunofluorescence, Nissl staining, MDA detection, ROS detection, iron content detection, and short-term and long-term neurobehavioral tests were performed to investigate the effects of regulated-LPCAT3 after SAH. RESULTS LPCAT3 levels were found to be significantly elevated in SAH. Suppression of LPCAT3 expression via shRNA improved oxidative stress, reduced brain edema, alleviated behavioral and cognitive deficits following SAH and decreased neuronal death, while upregulating LPCAT3 expression showed opposing effects. CONCLUSION LPCAT3 is involved in SAH-induced EBI and associated with ferroptosis. Our findings provide a referential basis for potential therapeutic interventions aimed at alleviating EBI following SAH.
Collapse
Affiliation(s)
- Jiahui Hao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Tong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Intensive Care Unit, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China; Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Heng Gao
- Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China
| | - Jinquan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
8
|
Zhu K, Bi S, Zhu Z, Zhang W, Yang X, Li J, Liang G, Yu C, Pan P. Edaravone dexborneol attenuates oxidative stress in experimental subarachnoid hemorrhage via Keap1/Nrf2 signaling pathway. Front Pharmacol 2024; 15:1342226. [PMID: 38873422 PMCID: PMC11169797 DOI: 10.3389/fphar.2024.1342226] [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: 11/21/2023] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Background Subarachnoid hemorrhage (SAH) serves as a disease characterized by high incidence rate, which is exceedingly prevalent and severe. Presently, there is no unambiguous or efficacious intervention for the neurological impairment following SAH. Administering multi-targeted neuroprotective agents to reduce oxidative stress (OS) and neuroinflammation caused by early brain injury (EBI) has been demonstrated to improve neurological function and prognosis following SAH. Edaravone dexborneol (EDB), a novel multi targeted neuroprotective medication, combines four parts edaravone (EDA) with 1 part (+)-borneol in proportion. Clinical trials conducted in China have revealed during 2 days of acute ischemic stroke (AIS), early administration of EDB leads to improved therapeutic outcomes compared to treatment in EDA monotherapy. Currently, there is no clear evidence that EDB can effectively treat SAH, therefore, our study aims to investigate its potential therapeutic effects and mechanisms on EBI after SAH. Method We used the intravascular threading method to establish a mouse model of SAH to explore whether EDA and EDB could produce anti-OS and anti-apoptosis effects. Behavioral assessment of mice was conducted using the balance beam experiment and the modified Garcia scoring system. Neuronal damage due to OS and Keap1/Nrf2 signaling pathway were detected through techniques of immunofluorescence, Western blotting, spectrophotometry. The group of EDA and EDB were injected intraperitoneally for 72 h after SAH. Results The experiment results indicated that EDB lead to remarkably positive results by significantly enhancing neurological function, reducing blood-brain barrier (BBB) injury, and effectively inhibiting neuronal apoptosis after SAH. Further examination indicated EDB significantly reduced the expression of Keap1 and increased the expression of Nrf2, and it inhibited MDA, and enhanced SOD activity after SAH. These outcomes surpassed the effectiveness observed in EDA monotherapy. However, the application of ML385 reversed the anti-OS effects of EDB and EDA. Conclusion Our experimental findings indicated that EDB could activate Keap1/Nrf2 signaling pathway to reduce OS damage, thereby protecting neurological function and enhancing behavioral abilities after SAH. These outcomes could facilitate the creation of new approaches for the clinical management of SAH.
Collapse
Affiliation(s)
- Kunyuan Zhu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
- China Medical University, Shenyang, Liaoning, China
| | - Shijun Bi
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zechao Zhu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
- China Medical University, Shenyang, Liaoning, China
| | - Wenxu Zhang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
- China Medical University, Shenyang, Liaoning, China
| | - Xinyu Yang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
- China Medical University, Shenyang, Liaoning, China
| | - Jiashuo Li
- China Medical University, Shenyang, Liaoning, China
| | - Guobiao Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Chunyong Yu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Pengyu Pan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
9
|
Hu Q, Zhang R, Dong X, Yang D, Yu W, Du Q. Huperzine A ameliorates neurological deficits after spontaneous subarachnoid hemorrhage through endothelial cell pyroptosis inhibition. Acta Biochim Biophys Sin (Shanghai) 2024; 56:645-656. [PMID: 38529553 DOI: 10.3724/abbs.2024037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH) is a kind of hemorrhagic stroke which causes neurological deficits in survivors. Huperzine A has a neuroprotective effect, but its role in SAH is unclear. Therefore, we explore the effect of Huperzine A on neurological deficits induced by SAH and the related mechanism. In this study, Evans blue assay, TUNEL staining, immunofluorescence, western blot analysis, and ELISA are conducted. We find that Huperzine A can improve neurological deficits and inhibit the apoptosis of nerve cells in SAH rats. Huperzine A treatment can improve the upregulation of brain water content, damage of blood-brain barrier, fibrinogen and matrix metalloprotein 9 expressions and the downregulation of ZO-1 and occludin expressions induced by SAH. Huperzine A inhibit the expressions of proteins involved in pyroptosis in endothelial cells in SAH rats. The increase in MDA content and decrease in SOD activity in SAH rats can be partly reversed by Huperzine A. The ROS inducer H 2O 2 can induce pyroptosis and inhibit the expressions of ZO-1 and occludin in endothelial cells, which can be blocked by Huperzine A. In addition, the increase in the entry of p65 into the nucleus in endothelial cells can be partly reversed by Huperzine A. Huperzine A may delay the damage of blood-brain barrier in SAH rats by inhibiting oxidative stress-mediated pyroptosis and tight junction protein expression downregulation through the NF-κB pathway. Overall, Huperzine A may have clinical value for treating SAH.
Collapse
Affiliation(s)
- Qiang Hu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Rong Zhang
- Medical Examination Center, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
| | - Xiaoqiao Dong
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Dingbo Yang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Wenhua Yu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| |
Collapse
|
10
|
Deng X, Ren J, Chen K, Zhang J, Zhang Q, Zeng J, Li T, Tang Q, Lin J, Zhu J. Mas receptor activation facilitates innate hematoma resolution and neurological recovery after hemorrhagic stroke in mice. J Neuroinflammation 2024; 21:106. [PMID: 38658922 PMCID: PMC11041011 DOI: 10.1186/s12974-024-03105-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/18/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a devastating neurological disease causing severe sensorimotor dysfunction and cognitive decline, yet there is no effective treatment strategy to alleviate outcomes of these patients. The Mas axis-mediated neuroprotection is involved in the pathology of various neurological diseases, however, the role of the Mas receptor in the setting of ICH remains to be elucidated. METHODS C57BL/6 mice were used to establish the ICH model by injection of collagenase into mice striatum. The Mas receptor agonist AVE0991 was administered intranasally (0.9 mg/kg) after ICH. Using a combination of behavioral tests, Western blots, immunofluorescence staining, hematoma volume, brain edema, quantitative-PCR, TUNEL staining, Fluoro-Jade C staining, Nissl staining, and pharmacological methods, we examined the impact of intranasal application of AVE0991 on hematoma absorption and neurological outcomes following ICH and investigated the underlying mechanism. RESULTS Mas receptor was found to be significantly expressed in activated microglia/macrophages, and the peak expression of Mas receptor in microglia/macrophages was observed at approximately 3-5 days, followed by a subsequent decline. Activation of Mas by AVE0991 post-treatment promoted hematoma absorption, reduced brain edema, and improved both short- and long-term neurological functions in ICH mice. Moreover, AVE0991 treatment effectively attenuated neuronal apoptosis, inhibited neutrophil infiltration, and reduced the release of inflammatory cytokines in perihematomal areas after ICH. Mechanistically, AVE0991 post-treatment significantly promoted the transformation of microglia/macrophages towards an anti-inflammatory, phagocytic, and reparative phenotype, and this functional phenotypic transition of microglia/macrophages by Mas activation was abolished by both Mas inhibitor A779 and Nrf2 inhibitor ML385. Furthermore, hematoma clearance and neuroprotective effects of AVE0991 treatment were reversed after microglia depletion in ICH. CONCLUSIONS Mas activation can promote hematoma absorption, ameliorate neurological deficits, alleviate neuron apoptosis, reduced neuroinflammation, and regulate the function and phenotype of microglia/macrophages via Akt/Nrf2 signaling pathway after ICH. Thus, intranasal application of Mas agonist ACE0991 may provide promising strategy for clinical treatment of ICH patients.
Collapse
Affiliation(s)
- Xiangyang Deng
- Department of Neurosurgery, Wenzhou Municipal Key Laboratory of Neurodevelopmental Pathology and Physiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109, Xueyuan Road, Wenzhou, 325027, Zhejiang, China
- Department of Neurosurgery, Huashan Hospital, National Center for Neurological Disorders, National Key Lab. for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Lab. of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China
| | - Junwei Ren
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kezhu Chen
- Department of Neurosurgery, Huashan Hospital, National Center for Neurological Disorders, National Key Lab. for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Lab. of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China
| | - Jin Zhang
- The First Affiliated Hospital of the Naval Medical University, Shanghai, China
| | - Quan Zhang
- Department of Neurosurgery, Huashan Hospital, National Center for Neurological Disorders, National Key Lab. for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Lab. of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China
| | - Jun Zeng
- Department of Neurosurgery, Huashan Hospital, National Center for Neurological Disorders, National Key Lab. for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Lab. of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China
| | - Tianwen Li
- Department of Neurosurgery, Huashan Hospital, National Center for Neurological Disorders, National Key Lab. for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Lab. of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, National Center for Neurological Disorders, National Key Lab. for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Lab. of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China
| | - Jian Lin
- Department of Neurosurgery, Wenzhou Municipal Key Laboratory of Neurodevelopmental Pathology and Physiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109, Xueyuan Road, Wenzhou, 325027, Zhejiang, China.
| | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, National Center for Neurological Disorders, National Key Lab. for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Lab. of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China.
| |
Collapse
|
11
|
Wu Y, Xu Y, Sun J, Dai K, Wang Z, Zhang J. Inhibiting RIPK1-driven neuroinflammation and neuronal apoptosis mitigates brain injury following experimental subarachnoid hemorrhage. Exp Neurol 2024; 374:114705. [PMID: 38290652 DOI: 10.1016/j.expneurol.2024.114705] [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: 09/29/2023] [Revised: 01/14/2024] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
RIPK1, a receptor-interacting serine/threonine protein kinase, plays a crucial role in maintaining cellular and tissue homeostasis by integrating inflammatory responses and cell death signaling pathways including apoptosis and necroptosis, which have been implicated in diverse physiological and pathological processes. Suppression of RIPK1 activation is a promising strategy for restraining the pathological progression of many human diseases. Neuroinflammation and neuronal apoptosis are two pivotal factors in the pathogenesis of brain injury following subarachnoid hemorrhage (SAH). In this study, we established in vivo and in vitro models of SAH to investigate the activation of RIPK1 kinase in both microglia and neurons. We observed the correlation between RIPK1 kinase activity and microglia-mediated inflammation as well as neuronal apoptosis. We then investigated whether inhibition of RIPK1 could alleviate neuroinflammation and neuronal apoptosis following SAH, thereby reducing brain edema and ameliorating neurobehavioral deficits. Additionally, the underlying mechanisms were also explored. Our research findings revealed the activation of RIPK1 kinase in both microglia and neurons following SAH, as marked by the phosphorylation of RIPK1 at serine 166. The upregulation of p-RIPK1(S166) resulted in a significant augmentation of inflammatory cytokines and chemokines, including TNF-α, IL-6, IL-1α, CCL2, and CCL5, as well as neuronal apoptosis. The activation of RIPK1 in microglia and neurons following SAH could be effectively suppressed by administration of Nec-1 s, a specific inhibitor of RIPK1. Consequently, inhibition of RIPK1 resulted in a downregulation of inflammatory cytokines and chemokines and attenuation of neuronal apoptosis after SAH in vitro. Furthermore, the administration of Nec-1 s effectively mitigated neuroinflammation, neuronal apoptosis, brain edema, and neurobehavioral deficits in mice following SAH. Our findings suggest that inhibiting RIPK1 kinase represents a promising therapeutic strategy for mitigating brain injury after SAH by attenuating RIPK1-driven neuroinflammation and neuronal apoptosis.
Collapse
Affiliation(s)
- Yan Wu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yao Xu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingshan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kun Dai
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Jian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
12
|
Liu H, Guo D, Wang J, Zhang W, Zhu Z, Zhu K, Bi S, Pan P, Liang G. Aloe-emodin from Sanhua Decoction inhibits neuroinflammation by regulating microglia polarization after subarachnoid hemorrhage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117583. [PMID: 38122912 DOI: 10.1016/j.jep.2023.117583] [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: 04/09/2023] [Revised: 11/20/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Subarachnoid hemorrhage (SAH) triggers a cascade of events that lead to early brain injury (EBI), which contributes to poor outcomes and appears within 3 days after SAH initiation. EBI involves multiple process including neuronal death, blood-brain barrier (BBB) injury and inflammation response. Microglia are cluster of immune cells originating in the brain which respond to SAH by changing their states and releasing inflammatory molecules through various signaling pathways. M0, M1, M2 are three states of microglia represent resting state, promoting inflammation state, and anti-inflammation state respectively, which can be modulated by pharmacological strategies. AIM OF THE STUDY After identified potential active ingredients and targets of Sanhua Decoction (SHD) for SAH, we selected aloe-emodin (AE) as a potential ingredient modulating microglia activation states. MATERIALS AND METHODS Molecular mechanisms, targets and pathways of SHD were reveal by network pharmacology technique. The effects of AE on SAH were evaluated in vivo by assessing neurological deficits, neuronal apoptosis and BBB integrity in a mouse SAH model. Furthermore, BV-2 cells were used to examine the effects of AE on microglial polarization. The influence of AE on microglia transformation was measured by Iba-1, TNF-α, CD68, Arg-1 and CD206 staining. The signal pathways of neuronal apoptosis and microglia polarization was measured by Western blot. RESULTS Network pharmacology identified potential active ingredients and targets of SHD for SAH. And AE is one of the active ingredients. We also confirmed that AE via NF-κB and PKA/CREB pathway inhibited the microglia activation and promoted transformation from M1 phenotype to M2 at EBI stage after SAH. CONCLUSIONS AE, as one ingredient of SHD, can alleviate the inflammatory response and protecting neurons from SAH-induced injury. AE has potential value for treating SAH-induced nerve injury and is expected to be applied in clinical practice.
Collapse
Affiliation(s)
- Hui Liu
- Department of Clinical Medicine, College of Medicine, Lishui University, Lishui, China
| | - Dan Guo
- Department of First Outpatients, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiao Wang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Lishui University, Lishui, China
| | - Wenxu Zhang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zechao Zhu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Kunyuan Zhu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Shijun Bi
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Pengyu Pan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Guobiao Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
13
|
Yu J, Li J, Matei N, Wang W, Tang L, Pang J, Li X, Fang L, Tang J, Zhang JH, Yan M. Intranasal administration of recombinant prosaposin attenuates neuronal apoptosis through GPR37/PI3K/Akt/ASK1 pathway in MCAO rats. Exp Neurol 2024; 373:114656. [PMID: 38114054 PMCID: PMC10922973 DOI: 10.1016/j.expneurol.2023.114656] [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: 09/27/2023] [Revised: 11/19/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023]
Abstract
Studies have reported that Prosaposin (PSAP) is neuroprotective in cerebrovascular diseases. We hypothesized that PSAP would reduce infarct volume by attenuating neuronal apoptosis and promoting cell survival through G protein-coupled receptor 37(GPR37)/PI3K/Akt/ASK1 pathway in middle cerebral artery occlusion (MCAO) rats. Two hundred and thirty-five male and eighteen female Sprague-Dawley rats were used. Recombinant human PSAP (rPSAP) was administered intranasally 1 h (h) after reperfusion. PSAP small interfering ribonucleic acid (siRNA), GPR37 siRNA, and PI3K specific inhibitor LY294002 were administered intracerebroventricularly 48 h before MCAO. Infarct volume, neurological score, immunofluorescence staining, Western blot, Fluoro-Jade C (FJC) and TUNEL staining were examined. The expression of endogenous PSAP and GPR37 were increased after MCAO. Intranasal administration of rPSAP reduced brain infarction, neuronal apoptosis, and improved both short- and long-term neurological function. Knockdown of endogenous PSAP aggravated neurological deficits. Treatment with exogenous rPSAP increased PI3K expression, Akt and ASK1 phosphorylation, and Bcl-2 expression; phosphorylated-JNK and Bax levels were reduced along with the number of FJC and TUNEL positive neurons. GPR37 siRNA and LY294002 abolished the anti-apoptotic effect of rPSAP at 24 h after MCAO. In conclusion, rPSAP attenuated neuronal apoptosis and improved neurological function through GPR37/PI3K/Akt/ASK1 pathway after MCAO in rats. Therefore, further exploration of PSAP as a potential treatment option in ischemic stroke is warranted.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jinlan Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nathanael Matei
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA; Department of Ophthalmology, University of Southern California, Los Angeles, CA 90007, USA
| | - Wenna Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Lihui Tang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jinwei Pang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Xue Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Lili Fang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiping Tang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA.
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Fu W, Che X, Tan J, Cui S, Ma Y, Xu D, Long H, Yang X, Wen T, He Z. Rasd1 is involved in white matter injury through neuron-oligodendrocyte communication after subarachnoid hemorrhage. CNS Neurosci Ther 2024; 30:e14452. [PMID: 37735980 PMCID: PMC10916428 DOI: 10.1111/cns.14452] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/16/2023] [Accepted: 08/14/2023] [Indexed: 09/23/2023] Open
Abstract
AIMS Rasd1 has been reported to be correlated with neurotoxicity, metabolism, and rhythm, but its effect in case of subarachnoid hemorrhage (SAH) remained unclear. White matter injury (WMI) and ferroptosis participate in the early brain injury (EBI) after SAH. In this work, we have investigated whether Rasd1 can cause ferroptosis and contribute to SAH-induced WMI. METHODS Lentivirus for Rasd1 knockdown/overexpression was administrated by intracerebroventricular (i.c.v) injection at 7 days before SAH induction. SAH grade, brain water content, short- and long-term neurobehavior, Western blot, real-time PCR, ELISA, biochemical estimation, immunofluorescence, diffusion tensor imaging (DTI), and transmission electron microscopy (TEM) were systematically performed. Additionally, genipin, a selective uncoupling protein 2(UCP2) inhibitor, was used in primary neuron and oligodendrocyte co-cultures for further in vitro mechanistic studies. RESULTS Rasd1 knockdown has improved the neurobehavior, glia polarization, oxidative stress, neuroinflammation, ferroptosis, and demyelination. Conversely, Rasd1 overexpression aggravated these changes by elevating the levels of reactive oxygen species (ROS), inflammatory cytokines, MDA, free iron, and NCOA4, as well as contributing to the decrease of the levels of UCP2, GPX4, ferritin, and GSH mechanistically. According to the in vitro study, Rasd1 can induce oligodendrocyte ferroptosis through inhibiting UCP2, increasing reactive oxygen species (ROS), and activating NCOA4-mediated ferritinophagy. CONCLUSIONS It can be concluded that Rasd1 exerts a modulated role in oligodendrocytes ferroptosis in WMI following SAH.
Collapse
Affiliation(s)
- Wenqiao Fu
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xudong Che
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jiahe Tan
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Shizhen Cui
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yinrui Ma
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Daiqi Xu
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Haibo Long
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaolin Yang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Tangmin Wen
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhaohui He
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
15
|
Tang S, Lai N, Xu L. Neuronal pyroptosis mediated by STAT3 in early brain injury after subarachnoid hemorrhage. Brain Res 2024; 1822:148666. [PMID: 37949309 DOI: 10.1016/j.brainres.2023.148666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Neuroinflammation induced by early brain injury (EBI) seriously affects the prognosis of patients after subarachnoid hemorrhage (SAH). Pyroptosis can aggravate inflammatory injury by promoting the secretion of inflammatory cytokines. Meanwhile, STAT3 plays a critical role in the inflammatory response of EBI after SAH. However, whether it plays a pyroptotic role in SAH is mainly unknown. This study aimed to explore the mechanism of STAT3 in pyroptosis in EBI after SAH. C57BL/6J mice were used to establish the SAH model. Brain tissues were collected at different time points for q-RT-PCR and western blot to detect the expression level of STAT3. After intracerebroventricular injection of STAT3 inhibitor S3I-201, they were divided into sham, SAH, SAH + Vehicle, and SAH + S3I-201. Then, the SAH grade, cerebral edema content, blood-brain barrier (BBB) damage, and neurological scores of mice in each group were detected. qRT-PCR and western blot were used to detect related genes and proteins, and enzyme-linked immunosorbent assay (ELISA) was used to detect the expression levels of IL-18 and IL-1β. Immunofluorescence staining was used to observe the expression level of proteins. At the same time, S3I-201 was added to the primary neuron cells of the culture medium containing OxyHb to simulate the in vitro experiment, and the relevant indicators consistent with the in vivo experiment were detected. The expression of STAT3 was upregulated after SAH. Inhibition of STAT3 with S3I-201 attenuated neurological deficits, cerebral edema, and BBB damage after SAH. In addition, S3I-201 can also reduce the expression of pyroptosis-related inflammasomes such as GSDMD, NLRP3, Caspase 1, and AIM2 after SAH and the neurological damage caused by IL-18 and IL-1β. Further studies have shown that STAT3 regulates pyroptosis by promoting the nuclear translocation of NF-κB p65. Our finding demonstrated that STAT3 regulates neuronal pyroptosis in EBI after SAH. Inhibition of STAT3 may be a potential target to attenuate the damage that triggers neuroinflammation after SAH.
Collapse
Affiliation(s)
- Shengjie Tang
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Niansheng Lai
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Liang Xu
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University (The First People's Hospital of Chuzhou), Chuzhou, China.
| |
Collapse
|
16
|
Liao L, Wang H, Wei D, Yi M, Gu Y, Zhang M, Wang L. Exosomal microRNAs: implications in the pathogenesis and clinical applications of subarachnoid hemorrhage. Front Mol Neurosci 2023; 16:1300864. [PMID: 38143562 PMCID: PMC10748509 DOI: 10.3389/fnmol.2023.1300864] [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: 09/23/2023] [Accepted: 11/22/2023] [Indexed: 12/26/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a severe acute neurological disorder with a high fatality rate. Early brain injury (EBI) and cerebral vasospasm are two critical complications of SAH that significantly contribute to poor prognosis. Currently, surgical intervention and interventional therapy are the main treatment options for SAH, but their effectiveness is limited. Exosomes, which are a type of extracellular vesicles, play a crucial role in intercellular communication and have been extensively studied in the past decade due to their potential influence on disease progression, diagnosis, and treatment. As one of the most important components of exosomes, miRNA plays both direct and indirect roles in affecting disease progression. Previous research has found that exosomal miRNA is involved in the development of various diseases, such as tumors, chronic hepatitis, atherosclerosis, diabetes, and SAH. This review focuses on exploring the impact of exosomal miRNA on SAH, including its influence on neuronal apoptosis, inflammatory response, and immune activation following SAH. Furthermore, this review highlights the potential clinical applications of exosomal miRNA in the treatment of SAH. Although current research on this topic is limited and the clinical application of exosomal miRNA has inherent limitations, we aim to provide a concise summary of existing research progress and offer new insights for future research directions and trends in this field.
Collapse
Affiliation(s)
- Lishang Liao
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Haoran Wang
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Deli Wei
- Department of Neurosurgery, The People’s Hospital of Fushun County, Zigong, China
| | - Mingliang Yi
- Department of Neurosurgery, The People’s Hospital of Fushun County, Zigong, China
| | - Yingjiang Gu
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The People’s Hospital of Fushun County, Zigong, China
| | - Mingwei Zhang
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Li Wang
- Department of Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
17
|
Yang H, Ding C, Cheng M, Sheng Z, Chen L, Chen J, Wang Y. Perampanel attenuates oxidative stress and pyroptosis following subarachnoid hemorrhage via the SIRT3/FOXO3α pathway. Sci Rep 2023; 13:21320. [PMID: 38044382 PMCID: PMC10694148 DOI: 10.1038/s41598-023-48802-1] [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: 07/10/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) occurs most commonly after rupture of an aneurysm, resulting in high disability and mortality due to the absence of effective therapy. Its subsequent stage, early brain injury (EBI), promotes the sustainable development of injury in the brain and ultimately leads to poor prognosis. As a new antiepileptic drug, the effect of perampanel on EBI after SAH is unknown. Pyroptosis, a process of inflammatory programmed cell death, has been confirmed in most studies to play a substantial role in aggravating SAH-post EBI. Similarly, oxidative stress is closely involved in neuronal pyroptosis and the pathophysiological mechanism of SAH-post EBI, leading to a devastating outcome for SAH patients. Nonetheless, no studies have been conducted to determine whether perampanel reduces pyroptosis and oxidative stress in the context of SAH-induced EBI. Rat SAH model via endovascular perforation was constructed in this study, to assess the neuroprotective effect of perampanel on SAH-post EBI, and to clarify the possible molecular mechanism. By means of the neurological score, brain edema detection, FJB staining, immunofluorescence, WB, ELISA, and ROS assay, we found that perampanel can improve neuroscores and reduce brain edema and neuronal degeneration at 24 h after SAH; we also found that perampanel reduced oxidative stress, neuronal pyroptosis, and inhibition of the SIRT3-FOXO3α pathway at 24 h after SAH. When 3-TYP, an inhibitor of SIRT3, was administered, the effects of perampanel on the SIRT3-FOXO3a pathway, antioxidant stress, and neuronal pyroptosis were reversed. Taken together, our data indicate that perampanel attenuates oxidative stress and pyroptosis following subarachnoid hemorrhage via the SIRT3/FOXO3α pathway. This study highlights the application value of perampanel in subarachnoid hemorrhage and lays a foundation for clinical research and later transformation of perampanel in SAH.
Collapse
Affiliation(s)
- Hongqiao Yang
- Wuxi Clinical College of Anhui Medical University, Wuxi, China
- The Fifth Clinical College of Anhui Medical University, Hefei, China
| | - Changgeng Ding
- Wuxi Clinical College of Anhui Medical University, Wuxi, China
- The Fifth Clinical College of Anhui Medical University, Hefei, China
| | - Ming Cheng
- Wuxi Clinical College of Anhui Medical University, Wuxi, China
- The Fifth Clinical College of Anhui Medical University, Hefei, China
| | - Zhengwei Sheng
- Wuxi Clinical College of Anhui Medical University, Wuxi, China
- The Fifth Clinical College of Anhui Medical University, Hefei, China
| | - Lei Chen
- Wuxi Clinical College of Anhui Medical University, Wuxi, China
- The Fifth Clinical College of Anhui Medical University, Hefei, China
| | - Junhui Chen
- Wuxi Clinical College of Anhui Medical University, Wuxi, China.
- The Fifth Clinical College of Anhui Medical University, Hefei, China.
| | - Yuhai Wang
- Wuxi Clinical College of Anhui Medical University, Wuxi, China.
- The Fifth Clinical College of Anhui Medical University, Hefei, China.
| |
Collapse
|
18
|
Liang H, Tang LY, Ge HY, Chen MM, Lu SY, Zhang HX, Shen CL, Shen Y, Fei J, Wang ZG. Neuronal survival factor TAFA2 suppresses apoptosis through binding to ADGRL1 and activating cAMP/PKA/CREB/BCL2 signaling pathway. Life Sci 2023; 334:122241. [PMID: 37944639 DOI: 10.1016/j.lfs.2023.122241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
AIMS TAFA2, a cytokine specifically expressed in the central nervous system, plays a vital role in neuronal cell survival. TAFA2 deficiency has been correlated to various neurological disorders in mice and humans. However, the underlying mechanism remains elusive, especially its membrane-binding receptor through which TAFA2 functions. This study aimed to identify the specific binding receptor responsible for the anti-apoptotic effects of TAFA2. MAIN METHOD Co-immunoprecipitation (Co-IP) and quantitative mass spectrometry-based proteomic analysis were employed to identify potential TAFA2 binding proteins in V5 knockin mouse brain lysates. Subsequent validation involved in vitro and in vivo Co-IP and pull-down using specific antibodies. The functional analysis included evaluating the effects of ADGRL1 knockout, overexpression, and Lectin-like domain (Lec) deletion mutant on TAFA2's anti-apoptotic activity and analyzing the intracellular signaling pathways mediated by TAFA2 through ADGRL1. KEY FINDINGS Our study identified ADGRL1 as a potential receptor for TAFA2, which directly binds to TAFA2 through its lectin-like domain. Overexpression ADGRL1, but not ADGRL1ΔLec, induced apoptosis, which could be effectively suppressed by recombinant TAFA2 (rTAFA2). In ADGRL1-/- cells or re-introducing with ADGRL1ΔLec, responses to rTAFA2 in suppressing cell apoptosis were compromised. Increased cAMP, p-PKA, p-CREB, and BCL2 levels were also observed in response to rTAFA2 treatment, with these responses attenuated in ADGRL1-/- or ADGRL1ΔLec-expressing cells. SIGNIFICANCE Our results demonstrated that TAFA2 directly binds to the lectin-like domain of ADGRL1, activating cAMP/PKA/CREB/BCL2 signaling pathway, which is crucial in preventing cell death. These results implicate TAFA2 and its receptor ADGRL1 as potential therapeutic targets for neurological disorders.
Collapse
Affiliation(s)
- Hui Liang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ling Yun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hao Yang Ge
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ming Mei Chen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shun Yuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong Xin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chun Ling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Fei
- Tongji University, Shanghai 200092, China
| | - Zhu Gang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
19
|
Tian Q, Li Y, Feng S, Liu C, Guo Y, Wang G, Wei H, Chen Z, Gu L, Li M. Inhibition of CCR1 attenuates neuroinflammation via the JAK2/STAT3 signaling pathway after subarachnoid hemorrhage. Int Immunopharmacol 2023; 125:111106. [PMID: 37925951 DOI: 10.1016/j.intimp.2023.111106] [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: 08/23/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND AND PURPOSE Neuroinflammation is an important mechanism underlying brain injury caused by subarachnoid hemorrhage (SAH). C-C chemokine receptor type 1 (CCR1)-mediated inflammation is involved in the pathology of many central nervous system diseases. Herein, we investigated whether inhibition of CCR1 alleviated neuroinflammation after experimental SAH and aimed to elucidate the mechanisms of its potential protective effects. METHODS To analyze SAH transcriptome data R studio was used, and a mouse model of SAH was established using endovascular perforations. In this model, the selective CCR1 antagonist Met-RANTES (Met-R) and the CCR1 agonist recombinant CCL5 (rCCL5) were administered 1 h after SAH induction. To investigate the possible downstream mechanisms of CCR1, the JAK2 inhibitor AG490 and the JAK2 activator coumermycin A1 (C-A1) were administered 1 h after SAH induction. Furthermore, post-SAH evaluation, including SAH grading, neurological function tests, Western blot, the terminal deoxynucleotidyl transferase dUTP nick end labeling assay, and Fluoro-Jade B and fluorescent immunohistochemical staining were performed. Cerebrospinal fluid (CSF) samples were detected by ELISA. RESULTS CCL5 and CCR1 expression levels increased significantly following SAH. Met-R significantly improved neurological deficits in mice, decreased apoptosis and degeneration of ipsilateral cerebral cortex neurons, reduced infiltrating neutrophils, and promoted microglial activation after SAH induction. Furthermore, Met-R inhibited the expression of p-JAK2, p-STAT3, interleukin-1β, and tumor necrosis factor-α. However, the protective effects of Met-R were abolished by C-A1 treatment. Furthermore, rCCL5 injection aggravated neurological dysfunction and increased the expression of p-JAK2, p-STAT3, interleukin-1β, and tumor necrosis factor-α in SAH mice, all of which were reversed by the administration of AG490. Finally, the levels of CCL5 and CCR1 were elevate in the CSF of SAH patient and high level of CCL5 and CCR1 levels were associated with poor outcome. CONCLUSION The present results suggested that inhibition of CCR1 attenuates neuroinflammation after SAH via the JAK2/STAT3 signaling pathway, which may provide a new target for the treatment of SAH.
Collapse
Affiliation(s)
- Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Shi Feng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Chengli Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yujia Guo
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Guijun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Heng Wei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
20
|
Ma SJ, Li C, Yan C, Liu N, Jiang GY, Yang HR, Yan HC, Li JY, Liu HL, Gao C. Melatonin alleviates early brain injury by inhibiting the NRF2-mediated ferroptosis pathway after subarachnoid hemorrhage. Free Radic Biol Med 2023; 208:555-570. [PMID: 37717795 DOI: 10.1016/j.freeradbiomed.2023.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Ferroptosis is a novel form of cell death that plays a critical role in the pathological and physiological processes of early brain injury following subarachnoid hemorrhage. Melatonin, as the most potent endogenous antioxidant, has shown strong protective effects against pathological changes following subarachnoid hemorrhage, but its impact on ferroptosis induced by subarachnoid hemorrhage remains unexplored. In our study, we established a subarachnoid hemorrhage model in male SD rats. We found that subarachnoid hemorrhage induced changes in ferroptosis-related indicators such as lipid peroxidation and iron metabolism, while intraperitoneal injection of melatonin (40 mg/kg) effectively ameliorated these changes to a certain degree. Moreover, in a subset of rats with subarachnoid hemorrhage who received pre-treatment via intravenous injection of the melatonin receptor antagonist Luzindole (1 mg/kg) and 4P-PDOT (1 mg/kg), we found that the protective effect of melatonin against subarachnoid hemorrhage includes inhibition of lipid peroxidation and reduction of iron accumulation depended on melatonin receptor 1B (MT2). Furthermore, our study demonstrated that melatonin inhibited neuronal ferroptosis by activating the NRF2 signaling pathway, as evidenced by in vivo inhibition of NRF2. In summary, melatonin acts through MT2 and activates NRF2 and downstream genes such as HO-1/NQO1 to inhibit ferroptosis in subarachnoid hemorrhage-induced neuronal injury, thereby improving neurological function in rats. These results suggest that melatonin is a promising therapeutic target for subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Sheng-Ji Ma
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Chen Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Cong Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Nan Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Guang-You Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hong-Rui Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hao-Chen Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Ji-Yi Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Huai-Lei Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Cheng Gao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China.
| |
Collapse
|
21
|
Liu N, Lyu X, Zhang X, Zhang F, Chen Y, Li G. Astaxanthin attenuates cognitive deficits in Alzheimer's disease models by reducing oxidative stress via the SIRT1/PGC-1α signaling pathway. Cell Biosci 2023; 13:173. [PMID: 37710272 PMCID: PMC10503143 DOI: 10.1186/s13578-023-01129-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023] Open
Abstract
OBJECTIVE Oxidative stress plays a pivotal role in neurodegenerative diseases. Astaxanthin (AST) can play a neuroprotective role owing to its long-chain conjugated unsaturated double bond, which imparts potent antioxidant, anti-neuroinflammatory, and anti-apoptotic properties. However, the biological mechanisms underlying these effects remain unknown. Therefore, this study aimed to investigate and validate the protective effect of AST on neuronal senescence and apoptosis caused by oxidative stress induced by Aβ25-35 peptide, with the goal of preventing the onset of cognitive dysfunction. METHODS Alzheimer's disease models comprising ICR mice and PC12 cells were established using Aβ25-35. The Morris water maze test was used to assess mouse behavior. Nissl staining revealed morphological changes in the mouse hippocampal neurons. To elucidate the mechanism of action of AST, ICR mice and PC12 cells were treated with the silent information regulator 1 (SIRT1) inhibitor nicotinamide (NAM). Additionally, immunofluorescence, western blotting, and reverse transcription polymerase chain reaction were used to evaluate changes in the expression of Bcl-2 and Bax in the mouse hippocampus, and SIRT1/PGC-1α signaling pathway proteins were detected. Moreover, the oxidative stress markers in ICR mice and PC12 cells were evaluated. Further, CCK-8 assays, Annexin V/PI double staining, and β-galactosidase activity assays were performed in PC12 cells to evaluate the anti-senescence and apoptotic effects of AST. RESULTS In vivo experiments showed that Aβ25-35 impaired cognitive function, promoted morphological changes in hippocampal neurons, decreased Bcl-2 expression, increased Bax expression, decreased superoxide dismutase and GSH-px levels, and increased reactive oxygen species and malondialdehyde levels. Conversely, AST alleviated the impact of Aβ25-35 in mice, with reversed outcomes. NAM administration reduced SIRT1 and PGC-1α expression in the hippocampus. This decrease was accompanied by cognitive dysfunction and hippocampal neuron atrophy, which were also evident in the mice. Additionally, in vitro experiments showed that Aβ25-35 could promote oxidative stress and induce the senescence and apoptosis of PC12 cells. Nonetheless, AST treatment counteracted this effect by inhibiting oxidative stress and altering the state of PC12 cells. Notably, the Aβ + NAM group exhibited the most significant rates of senescence and apoptosis in PC12 cells following NAM treatment. CONCLUSION AST can improve cellular senescence and apoptosis mediated by oxidative stress via the SIRT1/PGC-1α signaling pathway and plays a vital role in inhibiting neuronal senescence and apoptosis and enhancing cognitive ability.
Collapse
Affiliation(s)
- Ning Liu
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Xiaohong Lyu
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
| | - Xianglin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Fan Zhang
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Yiming Chen
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Gang Li
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| |
Collapse
|
22
|
Li C, Lu P, Zhang L, He Y, Zhang L, Yang L, Zhang F, Kong X, Tao Q, Zhou J, Wu J, Peng T, Xie B, Jiang Y, Peng J. Apolipoprotein E Polymorphism Impacts White Matter Injury Through Microglial Phagocytosis After Experimental Subarachnoid Hemorrhage. Neuroscience 2023; 524:220-232. [PMID: 37290684 DOI: 10.1016/j.neuroscience.2023.05.020] [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: 01/26/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/10/2023]
Abstract
Apolipoprotein E (apoE, protein; APOE, gene), divided into three alleles of E2, E3 and E4 in humans, is associated with the progression of white matter lesion load. However, mechanism evidence has not been reported regarding the APOE genotype in early white matter injury (WMI) under subarachnoid hemorrhage (SAH) conditions. In the present study, we investigated the effects of APOE gene polymorphisms, by constructing microglial APOE3 and APOE4-specific overexpression, on WMI and underlying mechanisms of microglia phagocytosis in a mice model of SAH. A total of 167 male C57BL/6J mice (weight 22-26 g) were used. SAH and bleeding environment were induced by endovascular perforation in vivo and oxyHb in vitro, respectively. Multi-technology approaches, including immunohistochemistry, high throughput sequencing, gene editing for adeno-associated viruses, and several molecular biotechnologies were used to validate the effects of APOE polymorphisms on microglial phagocytosis and WMI after SAH. Our results revealed that APOE4 significantly aggravated the WMI and decreased neurobehavioral function by impairing microglial phagocytosis after SAH. Indicators negatively associated with microglial phagocytosis increased like CD16, CD86 and the ratio of CD16/CD206, while the indicators positively associated with microglial phagocytosis decreased like Arg-1 and CD206. The increased ROS and aggravating mitochondrial damage demonstrated that the damaging effects of APOE4 in SAH may be associated with microglial oxidative stress-dependent mitochondrial damage. Inhibiting mitochondrial oxidative stress by Mitoquinone (mitoQ) can enhance the phagocytic function of microglia. In conclusion, anti-oxidative stress and phagocytosis protection may serve as promising treatments in the management of SAH.
Collapse
Affiliation(s)
- Chaojie Li
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Peng Lu
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lihan Zhang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yijing He
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China
| | - Lifang Zhang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lei Yang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Fan Zhang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xi Kong
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China
| | - Qianke Tao
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jian Zhou
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jinpeng Wu
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tangming Peng
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Bingqing Xie
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China.
| | - Jianhua Peng
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
23
|
Huang D, Jia N, Pei C, Shen Z, Zhao S, Wang Y, Wu Y, Shi S, Li S, Wang Z. Rosavidin protects against PM2.5-induced lung toxicity via inhibition of NLRP3 inflammasome-mediated pyroptosis by activating the PI3K/AKT pathway. Biochem Pharmacol 2023; 213:115623. [PMID: 37244433 DOI: 10.1016/j.bcp.2023.115623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Fine particulate matter (PM2.5) contributes to adverse health effects through the promotion of inflammatory cytokine release. Rosavidin (Ro), a phenylpropanoid compound having multiple biological activities, is extracted from Rhodiola crenulata, a medicine and food homology plant. However, the protective role and mechanism of Ro in PM2.5-induced lung toxicity have not been previously studied. This study aimed to investigate the potential protective effect and mechanism of Ro in PM2.5-induced lung toxicity. A lung toxicity rat model was established through trachea drip of PM2.5 suspension after the different dose pretreatment of Ro (50 mg/kg and 100 mg/kg) to evaluate the effect of Ro on PM2.5 caused lung toxicity. The results showed that Ro attenuated the pathological changes, edema, and inflammation response in rats. The PI3K/AKT signaling pathway may be associated with the protective effect of Ro against pulmonary toxicity. Subsequently, we verified the role of PI3K/AKT in the PM2.5 exposure lung tissue. Moreover, expression levels of p-PI3K and p-AKT were lower, and those of NLRP3, ASC, cleaved caspase-1, cleaved IL-1β, and GSDMD-N were higher in PM2.5 group compared to those in control group. Whereas pre-administration of Ro reversed the expression trends of these proteins in lung tissue. Notably, those protective effects of Ro were not observed after pretreatment with a combination of Ro with nigericin or LY294002. These results indicate that Ro mitigates PM2.5-caused lung toxicity by inhibiting NLRP3 inflammasome-mediated pyroptosis through activation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Nan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Sijing Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yongcan Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shuiqin Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
24
|
Kong W, Peng Y, Ji C, Liu Z, Gao S, Zhang Y, Chen J, Li X, Bao M, Zhang Y, Jiang Q, Wang F, Li Z, Bian X, Ye J. Akt2 deficiency alleviates oxidative stress in the heart and liver via up-regulating SIRT6 during high-fat diet-induced obesity. Clin Sci (Lond) 2023; 137:823-841. [PMID: 37184210 DOI: 10.1042/cs20230433] [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: 05/05/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/16/2023]
Abstract
The present study aims to investigate the role of AKT2 in the pathogenesis of hepatic and cardiac lipotoxicity induced by lipid overload-induced obesity and identify its downstream targets. WT and Akt2 KO mice were fed either normal diet, or high-fat diet (HFD) to induce obesity model in vivo. Human hepatic cell line (L02 cells) and neonatal rat cardiomyocytes (NRCMs) were used as in vitro models. We observed that during HFD-induced obesity, Akt2 loss-of-function mitigated lipid accumulation and oxidative stress in the liver and heart tissue. Mechanistically, down-regulation of Akt2 promotes SIRT6 expression in L02 cells and NRCMs, the latter deacetylates SOD2, which promotes SOD2 activity and therefore alleviates oxidative stress-induced injury of hepatocytes and cardiomyocytes. Furthermore, we also proved that AKT2 inhibitor protects hepatocytes and cardiomyocytes from HFD-induced oxidative stress. Therefore, our work prove that AKT2 plays an important role in the regulation of obesity-induced lipid metabolic disorder in the liver and heart. Our study also indicates AKT2 inhibitor as a potential therapy for obesity-induced hepatic and cardiac injury.
Collapse
Affiliation(s)
- Weixian Kong
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Yue Peng
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Caoyu Ji
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Zekun Liu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Shuya Gao
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Yuexin Zhang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Jiawen Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Xie Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Mengmeng Bao
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Yubin Zhang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Qizhou Jiang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Fuqun Wang
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou 514031, China
| | - Zhe Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xiaohong Bian
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Junmei Ye
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| |
Collapse
|
25
|
Zhang Y, Chen Y, Yuan S, Yu Q, Fu J, Chen L, Liu J, He Y. Effect of gastrodin against cognitive impairment and neurodegeneration in APP/PS1 mice via regulating gut microbiota-gut-brain axis. Exp Brain Res 2023; 241:1661-1673. [PMID: 37199774 DOI: 10.1007/s00221-023-06632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Gastrodin (Gas) has exhibited protective activity in neurological disorders. Here, we investigated the neuroprotective effect and potential mechanisms of Gas against cognitive impairment via regulating gut microbiota. APPswe/PSEN1dE9 transgenic (APP/PS1) mice were treated intragastrically with Gas for 4 weeks, and then cognitive deficits, deposits of amyloid-β (Aβ) and phosphorylation of tau were analyzed. The expression levels of insulin-like growth factor-1 (IGF-1) pathway-related proteins, such as cAMP response element-binding protein (CREB), were detected. Meanwhile, gut microbiota composition was evaluated. Our results showed that Gas treatment significantly improved cognitive deficits and Aβ deposition in APP/PS1 mice. Moreover, Gas treatment increased the level of Bcl-2 and decreased level of Bax and ultimately inhibited neuronal apoptosis. Gas treatment markedly increased the expression levels of IGF-1 and CREB in APP/PS1 mice. Moreover, Gas treatment improved abnormal composition and structure of gut microbiota in APP/PS1 mice. These findings revealed that Gas actively participated in regulating the IGF-1 pathway to inhibit neuronal apoptosis via the gut-brain axis and that it can be considered a new therapeutic strategy against Alzheimer's disease.
Collapse
Affiliation(s)
- Yuhe Zhang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yan Chen
- Department of Neurology, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, 311899, Zhejiang, China
| | - Shushu Yuan
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Qingxia Yu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jianjiong Fu
- Department of Neurology, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, 311899, Zhejiang, China
| | - Luyun Chen
- Department of Neurology, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, 311899, Zhejiang, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Yuping He
- Department of Neurology, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, 311899, Zhejiang, China.
| |
Collapse
|
26
|
Fan L, Zhang S, Li X, Hu Z, Yang J, Zhang S, Zheng H, Su Y, Luo H, Liu X, Fan Y, Sun H, Zhang Z, Miao J, Song B, Xia Z, Shi C, Mao C, Xu Y. CHCHD2 p.Thr61Ile knock-in mice exhibit motor defects and neuropathological features of Parkinson's disease. Brain Pathol 2023; 33:e13124. [PMID: 36322611 PMCID: PMC10154378 DOI: 10.1111/bpa.13124] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/07/2022] [Indexed: 05/04/2023] Open
Abstract
The p.Thr61Ile (p.T61I) mutation in coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) was deemed a causative factor in Parkinson's disease (PD). However, the pathomechanism of the CHCHD2 p.T61I mutation in PD remains unclear. Few existing mouse models of CHCHD2-related PD completely reproduce the features of PD, and no transgenic or knock-in (KI) mouse models of CHCHD2 mutations have been reported. In the present study, we generated a novel CHCHD2 p.T61I KI mouse model, which exhibited accelerated mortality, progressive motor deficits, and dopaminergic (DA) neurons loss with age, accompanied by the accumulation and aggregation of α-synuclein and p-α-synuclein in the brains of the mutant mice. The mitochondria of mouse brains and induced pluripotent stem cells (iPSCs)-derived DA neurons carrying the CHCHD2 p.T61I mutation exhibited aberrant morphology and impaired function. Mechanistically, proteomic and RNA sequencing analysis revealed that p.T61I mutation induced mitochondrial dysfunction in aged mice likely through repressed insulin-degrading enzyme (IDE) expression, resulting in the degeneration of the nervous system. Overall, this CHCHD2 p.T61I KI mouse model recapitulated the crucial clinical and neuropathological aspects of patients with PD and provided a novel tool for understanding the pathogenic mechanism and therapeutic interventions of CHCHD2-related PD.
Collapse
Affiliation(s)
- Liyuan Fan
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Shuo Zhang
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Xinwei Li
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Zhengwei Hu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Jing Yang
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Shuyu Zhang
- Neuro‐Intensive Care UnitThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Huimin Zheng
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Yun Su
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Haiyang Luo
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Xinjing Liu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Yu Fan
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Huifang Sun
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Zhongxian Zhang
- Sino‐British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Jinxin Miao
- Sino‐British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Academy of Chinese Medicine ScienceHenan University of Chinese MedicineZhengzhouChina
| | - Bo Song
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Zongping Xia
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Clinical Systems Biology LaboratoriesZhengzhou UniversityZhengzhouChina
| | - Changhe Shi
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Chengyuan Mao
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Sino‐British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yuming Xu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| |
Collapse
|
27
|
Wu K, Chen L, Qiu Z, Zhao B, Hou J, Lei S, Jiang M, Xia Z. Protective Effect and Mechanism of Xbp1s Regulating HBP/O-GlcNAcylation through GFAT1 on Brain Injury after SAH. Biomedicines 2023; 11:biomedicines11051259. [PMID: 37238930 DOI: 10.3390/biomedicines11051259] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 05/28/2023] Open
Abstract
(1) SAH induces cellular stress and endoplasmic reticulum stress, activating the unfolded protein response (UPR) in nerve cells. IRE1 (inositol-requiring enzyme 1) is a protein that plays a critical role in cellular stress response. Its final product, Xbp1s, is essential for adapting to changes in the external environment. This process helps maintain proper cellular function in response to various stressors. O-GlcNAcylation, a means of protein modification, has been found to be involved in SAH pathophysiology. SAH can increase the acute O-GlcNAcylation level of nerve cells, which enhances the stress capacity of nerve cells. The GFAT1 enzyme regulates the level of O-GlcNAc modification in cells, which could be a potential target for neuroprotection in SAH. Investigating the IRE1/XBP1s/GFAT1 axis could offer a promising avenue for future research. (2) Methods: SAH was induced using a suture to perforate an artery in mice. HT22 cells with Xbp1 loss- and gain-of-function in neurons were generated. Thiamet-G was used to increase O-GlcNAcylation; (3) Results: Severe neuroinflammation caused by subarachnoid hemorrhage leads to extensive endoplasmic reticulum stress of nerve cells. Xbp1s, the final product of unfolded proteins induced by endoplasmic reticulum stress, can induce the expression of the hexosamine pathway rate limiting enzyme GFAT1, increase the level of O-GlcNAc modification of cells, and have a protective effect on neural cells; (4) Conclusions: The correlation between Xbp1s displayed by immunohistochemistry and O-GlcNAc modification suggests that the IRE1/XBP1 branch of unfolded protein reaction plays a key role in subarachnoid hemorrhage. IRE1/XBP1 branch is a new idea to regulate protein glycosylation modification, and provides a promising strategy for clinical perioperative prevention and treatment of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Kefan Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Lili Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Shaoqin Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Meng Jiang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| |
Collapse
|
28
|
Wang Y, Yang X, Cao Y, Li X, Xu R, Yan J, Guo Z, Sun S, Sun X, Wu Y. Electroacupuncture alleviates early brain injury via modulating microglia polarization and suppressing neuroinflammation in a rat model of subarachnoid hemorrhage. Heliyon 2023; 9:e14475. [PMID: 36967957 PMCID: PMC10036651 DOI: 10.1016/j.heliyon.2023.e14475] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Subarachnoid hemorrhage refers to an uncommon but severe subtype of stroke leading to high mortality and disability rates. Electroacupuncture, a traditional Chinese medical therapy combined with modern technology, shows evident curative effects on cerebral vascular diseases. This study attempts to investigate the possible treatment effects and mechanisms of EA on early brain injury after SAH. Data were gathered among sham group, SAH-induced group, and EA-treated group of male SD rats, concerning mortality rates, weight loss, rotarod latencies, cerebral blood flow, cell apoptosis, pro-inflammatory cytokines releasing, apoptotic protein level, microglia activation and related signal pathway. All results were collected 24-72 h after SAH induction. EA treatment demonstrated significant improvement on motor function 24 h after SAH without significant changes in mortality rate, weight loss, and cerebral blood flow. Another important finding was that EA regulated Bax and Bcl-2 imbalance and reduced cleaved casepase-3 caused by SAH. Additionally, levels of TNF-α, IL-1β, IL-6 were suppressed. The neuron apoptosis was suppressed by EA. The M1 polarization of activated microglia decreased while M2 polarized phenotype increased after EA treatment. Furthermore, pSTAT3-NOX2 signal axis, the M1 phenotype related activation pathway, was depressed after EA treatment. These findings suggested that EA improved motor deficits and ameliorated early brain injury after SAH probably via decreasing neuron apoptosis and anti-inflammation, which may involve modulation of microglia polarization. Taken together, EA may be a potential therapy for SAH treatment.
Collapse
|
29
|
Deng Y, Wang SY, Wang QG, Xu ZH, Peng Q, Chen SY, Zhu L, Zhang YD, Duan R. AVE 0991 Suppresses Astrocyte-Mediated Neuroinflammation of Alzheimer's Disease by Enhancing Autophagy. J Inflamm Res 2023; 16:391-406. [PMID: 36755969 PMCID: PMC9900155 DOI: 10.2147/jir.s392599] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
Purpose Our previous study has shown that AVE 0991, a nonpeptide analogue of Ang-(1-7), ameliorates cognitive decline and inhibits NLRP3 inflammasome of astrocytes in Alzheimer's disease model mice. Additionally, several studies have suggested that activation of autophagy appears to effectively inhibit the progression of neuroinflammation. However, it is unclear whether AVE 0991 can modulate astrocyte autophagy to suppress neuroinflammation in Alzheimer's disease. Materials and Methods APP/PS1 mice and Aβ-treated primary astrocytes were used as the research objects in vivo and in vitro, respectively. Water maze test was used to evaluate cognitive function of mice, Nissl staining and immunofluorescence staining was used to assess neuronal damage. ELISA kits were used to detect the levels of Ang-(1-7) and Aβ in the cortex, and qRT-PCR was used to detect the expression of cortical inflammation-related mediators. The expression of autophagy-related proteins in cortex were detected by Western blot. The upstream molecular responses involved in inflammation inhibition by AVE 0991 were validated by means of using the Mas1 antagonist and autophagy inhibitor. Results We found that 30 days of intraperitoneal administration of AVE 0991 improved. Aβ deposition, neuronal death, and cognitive deficits in APP/PS1 Alzheimer's disease model mice. Moreover, AVE 0991 treatment greatly suppressed astrocyte-mediated inflammation and up-regulated the expression of autophagy. Furthermore, the inhibitory effect of AVE 0991 on the expression of inflammatory factors was reversed by 3-MA, an autophagy inhibitor. Conclusion These findings suggest that regulation of autophagy is critical for inhibiting astrocyte neuroinflammatory responses and demonstrate a potential neuroprotective mechanism by which AVE 0991 could suppress neuroinflammatory responses by enhancing autophagy.
Collapse
Affiliation(s)
- Yang Deng
- Department of Neurology, Nanjing First Hospital, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Si-Yu Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Qing-Guang Wang
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, People’s Republic of China
| | - Zhao-Han Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Shuai-Yu Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Ying-Dong Zhang
- Department of Neurology, Nanjing First Hospital, China Pharmaceutical University, Nanjing, People’s Republic of China,Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China,Correspondence: Ying-Dong Zhang; Rui Duan, Department of Neurology, Nanjing First Hospital, China Pharmaceutical University, No.68, Changle Road, Nanjing, Jiangsu, People’s Republic of China, Email ;
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, China Pharmaceutical University, Nanjing, People’s Republic of China,Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
30
|
Hemoglobin Derived from Subarachnoid Hemorrhage-Induced Pyroptosis of Neural Stem Cells via ROS/NLRP3/GSDMD Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4383332. [PMID: 36703912 PMCID: PMC9871413 DOI: 10.1155/2023/4383332] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 01/17/2023]
Abstract
Accumulating evidence has demonstrated that neural stem cells (NSCs) have regenerative capacity after brain injuries, such as in aneurysmal subarachnoid hemorrhage (SAH). The reactive oxygen species (ROS)-induced NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome triggers inflammatory responses and pyroptosis of cells; however, whether ROS-induced neuroinflammation modulates the fate of endogenous NSCs after SAH remains largely unknown. In this study, the level of IL-1β was increased in the cerebrospinal fluid (CSF) of patients with SAH. In an endovascular perforation model of SAH in mice, the secretion of IL-1β increased to a peak at 24 h following SAH, and the expression of Caspase1 and NLRP3 was elevated in the hippocampus. Primary cultured NSCs were incubated with hemoglobin (Hb) to mimic SAH in vitro. The cell viability, LDH release, intracellular ROS levels, scanning electron microscopy (SEM), and the expression of NLRP3 and pyroptosis indicators (GSDMD, ASC, and Caspase-1) in NSCs after SAH were examined to investigate the process of pyroptosis. We found that pyroptotic death featuring cellular swelling, cell membrane pore formation and elevated IL-1β was increased in cultured primary NSCs after Hb treatment, as was the expression of NLRP3, ASC, Caspase-1, and GSDMD. In addition, we found that ROS-induced pyroptosis of NSCs by activating the NLRP3/GSDMD pathway. These findings suggest that pyroptosis of NSCs induced by Hb can impede neural regeneration after SAH.
Collapse
|
31
|
Duan J, Yuan W, Jiang J, Wang J, Yan X, Liu F, Liu A. ASK1 inhibitor NQDI‑1 decreases oxidative stress and neuroapoptosis via the ASK1/p38 and JNK signaling pathway in early brain injury after subarachnoid hemorrhage in rats. Mol Med Rep 2023; 27:47. [PMID: 36633130 PMCID: PMC9879074 DOI: 10.3892/mmr.2023.12934] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 11/30/2022] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress and neuroapoptosis are key pathological processes after subarachnoid hemorrhage (SAH). The present study evaluated the anti‑oxidation and anti‑apoptotic neuroprotective effects of the apoptosis signal‑regulating kinase 1 (ASK1) inhibitor ethyl‑2,7‑dioxo‑2,7‑dihydro‑3H‑naphtho(1,2,3‑de)quinoline‑1‑carboxylate (NQDI‑1) in early brain injury (EBI) following SAH in a rat model. A total of 191 rats were used and the SAH model was induced using monofilament perforation. Western blotting was subsequently used to detect the endogenous expression levels of proteins. Immunofluorescence was then used to confirm the nerve cellular localization of ASK1. Short‑term neurological function was assessed using the modified Garcia scores and the beam balance test 24 h after SAH, whereas long‑term neurological function was assessed using the rotarod test and the Morris water maze test. Apoptosis of neurons was assessed by TUNEL staining and oxidative stress was assessed by dihydroethidium staining 24 h after SAH. The protein expression levels of phosphorylated (p‑)ASK1 and ASK1 rose following SAH. NQDI‑1 was intracerebroventricularly injected 1 h after SAH and demonstrated significant improvements in both short and long‑term neurological function and significantly reduced oxidative stress and neuronal apoptosis. Injection of NQDI‑1 caused a significant decrease in protein expression levels of p‑ASK1, p‑p38, p‑JNK, 4 hydroxynonenal, and Bax and significantly increased the protein expression levels of heme oxygenase 1 and Bcl‑2. The use of the p38 inhibitor BMS‑582949 or the JNK inhibitor SP600125 led to significant decreases in the protein expression levels of p‑p38 or p‑JNK, respectively, and a significant reduction in oxidative stress and neuronal apoptosis; however, these inhibitors did not demonstrate an effect on p‑ASK1 or ASK1 protein expression levels. In conclusion, treatment with NQDI‑1 improved neurological function and decreased oxidative stress and neuronal apoptosis in EBI following SAH in rats, possibly via inhibition of ASK1 phosphorylation and the ASK1/p38 and JNK signaling pathway. NQDI‑1 may be considered a potential agent for the treatment of patients with SAH.
Collapse
Affiliation(s)
- Jiajia Duan
- Department of Neurosurgery, Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Wen Yuan
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Juan Jiang
- Department of Anatomy and Neurobiology, Xiangya Medicine School, Central South University, Changsha, Hunan 410000, P.R. China
| | - Jikai Wang
- Department of Neurosurgery, The Fifth Sun Yet-sen Hospital, Sun Yet-sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Xiangya Medicine School, Central South University, Changsha, Hunan 410000, P.R. China
| | - Fei Liu
- Department of Neurosurgery, The Fifth Sun Yet-sen Hospital, Sun Yet-sen University, Zhuhai, Guangdong 519000, P.R. China,Correspondence to: Professor Fei Liu, Department of Neurosurgery, The Fifth Sun Yet-sen Hospital, Sun Yet-sen University, 52 Meihuadong Road, Xiangzhou, Zhuhai, Guangdong 519000, P.R. China, E-mail:
| | - Aihua Liu
- Department of Neurosurgery, Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, P.R. China,Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China,Professor Aihua Liu, Beijing Neurosurgical Institute, Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai, Beijing 100070, P.R. China, E-mail:
| |
Collapse
|
32
|
Li MC, Tian Q, Liu S, Han SM, Zhang W, Qin XY, Chen JH, Liu CL, Guo YJ. The mechanism and relevant mediators associated with neuronal apoptosis and potential therapeutic targets in subarachnoid hemorrhage. Neural Regen Res 2023; 18:244-252. [PMID: 35900398 PMCID: PMC9396483 DOI: 10.4103/1673-5374.346542] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a dominant cause of death and disability worldwide. A sharp increase in intracranial pressure after SAH leads to a reduction in cerebral perfusion and insufficient blood supply for neurons, which subsequently promotes a series of pathophysiological responses leading to neuronal death. Many previous experimental studies have reported that excitotoxicity, mitochondrial death pathways, the release of free radicals, protein misfolding, apoptosis, necrosis, autophagy, and inflammation are involved solely or in combination in this disorder. Among them, irreversible neuronal apoptosis plays a key role in both short- and long-term prognoses after SAH. Neuronal apoptosis occurs through multiple pathways including extrinsic, mitochondrial, endoplasmic reticulum, p53 and oxidative stress. Meanwhile, a large number of blood contents enter the subarachnoid space after SAH, and the secondary metabolites, including oxygenated hemoglobin and heme, further aggravate the destruction of the blood-brain barrier and vasogenic and cytotoxic brain edema, causing early brain injury and delayed cerebral ischemia, and ultimately increasing neuronal apoptosis. Even there is no clear and effective therapeutic strategy for SAH thus far, but by understanding apoptosis, we might excavate new ideas and approaches, as targeting the upstream and downstream molecules of apoptosis-related pathways shows promise in the treatment of SAH. In this review, we summarize the existing evidence on molecules and related drugs or molecules involved in the apoptotic pathway after SAH, which provides a possible target or new strategy for the treatment of SAH.
Collapse
|
33
|
Wang X, Xu P, Liu Y, Wang Z, Lenahan C, Fang Y, Lu J, Zheng J, Wang K, Wang W, Zhou J, Chen S, Zhang J. New Insights of Early Brain Injury after Subarachnoid Hemorrhage: A Focus on the Caspase Family. Curr Neuropharmacol 2023; 21:392-408. [PMID: 35450528 PMCID: PMC10190145 DOI: 10.2174/1570159x20666220420115925] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/10/2022] [Accepted: 04/14/2022] [Indexed: 11/22/2022] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH), primarily caused by ruptured intracranial aneurysms, remains a prominent clinical challenge with a high rate of mortality and morbidity worldwide. Accumulating clinical trials aiming at the prevention of cerebral vasospasm (CVS) have failed to improve the clinical outcome of patients with SAH. Therefore, a growing number of studies have shifted focus to the pathophysiological changes that occur during the periods of early brain injury (EBI). New pharmacological agents aiming to alleviate EBI have become a promising direction to improve outcomes after SAH. Caspases belong to a family of cysteine proteases with diverse functions involved in maintaining metabolism, autophagy, tissue differentiation, regeneration, and neural development. Increasing evidence shows that caspases play a critical role in brain pathology after SAH. Therefore, caspase regulation could be a potential target for SAH treatment. Herein, we provide an overview pertaining to the current knowledge on the role of caspases in EBI after SAH, and we discuss the promising therapeutic value of caspase-related agents after SAH.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Penglei Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zefeng Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA
- Burrell College of Osteopathic Medicine, Las Cruces, New Mexico
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingwei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kaikai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingyi Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine (Huzhou Central Hospital), Huzhou, China
| | - Jianming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Ramanathan D, Huang L, Wilson T, Boling W. Molecular hydrogen therapy for neurological diseases: a review of current evidence. Med Gas Res 2022; 13:94-98. [PMID: 36571372 PMCID: PMC9979207 DOI: 10.4103/2045-9912.359677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Reactive oxygen species and other free radicals cause oxidative stress which is the underlying pathogenesis of cellular injury in various neurological diseases. Molecular hydrogen therapy with its unique biological property of selectively scavenging pathological free radicals has demonstrated therapeutic potential in innumerable animal studies and some clinical trials. These studies have implicated several cellular pathways affected by hydrogen therapy in explaining its anti-inflammatory and antioxidative effects. This article reviews relevant animal and clinical studies that demonstrate neuroprotective effects of hydrogen therapy in stroke, neurodegenerative diseases, neurotrauma, and global brain injury.
Collapse
Affiliation(s)
- Dinesh Ramanathan
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA,Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Taylor Wilson
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| | - Warren Boling
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA,Correspondence to: Warren Boling, E-mail:
| |
Collapse
|
35
|
Liu Y, Liu R, Huang L, Zuo G, Dai J, Gao L, Shi H, Fang Y, Lu Q, Okada T, Wang Z, Hu X, Lenahan C, Tang J, Xiao J, Zhang JH. Inhibition of Prostaglandin E2 Receptor EP3 Attenuates Oxidative Stress and Neuronal Apoptosis Partially by Modulating p38MAPK/FOXO3/Mul1/Mfn2 Pathway after Subarachnoid Hemorrhage in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7727616. [PMID: 36531208 PMCID: PMC9757947 DOI: 10.1155/2022/7727616] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/23/2022] [Accepted: 11/19/2022] [Indexed: 09/30/2023]
Abstract
Oxidative stress and neuronal apoptosis contribute to pathological processes of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Previous studies demonstrated that the inhibition of prostaglandin E2 receptor EP3 suppressed oxidative stress and apoptotic effects after Alzheimer's disease and intracerebral hemorrhage. This study is aimed at investigating the antioxidative stress and antiapoptotic effect of EP3 inhibition and the underlying mechanisms in a rat mode of SAH. A total of 263 Sprague-Dawley male rats were used. SAH was induced by endovascular perforation. Selective EP3 antagonist L798106 was administered intranasally at 1 h, 25 h, and 49 h after SAH induction. EP3 knockout CRISPR and FOXO3 activation CRISPR were administered intracerebroventricularly at 48 h prior to SAH, while selective EP3 agonist sulprostone was administered at 1 h prior to SAH. SAH grade, neurological deficits, western blots, immunofluorescence staining, Fluoro-Jade C staining, TUNEL staining, 8-OHdG staining, and Nissl staining were conducted after SAH. The expression of endogenous PGES2 increased and peaked at 12 h while the expression of EP1, EP2, EP3, EP4, and Mul1 increased and peaked at 24 h in the ipsilateral brain after SAH. EP3 was expressed mainly in neurons. The inhibition of EP3 with L798106 or EP3 KO CRISPR ameliorated the neurological impairments, brain tissue oxidative stress, and neuronal apoptosis after SAH. To examine potential downstream mediators of EP3, we examined the effect of the increased expression of activated FOXO3 following the administration of FOXO3 activation CRISPR. Mechanism studies demonstrated that L798106 treatment significantly decreased the expression of EP3, p-p38, p-FOXO3, Mul1, 4-HNE, Bax, and cleaved caspase-3 but upregulated the expression of Mfn2 and Bcl-2 in SAH rats. EP3 agonist sulprostone or FOXO3 activation CRISPR abolished the neuroprotective effects of L798106 and its regulation on expression of p38MAPK/FOXO3/Mul1/Mfn2 in the ipsilateral brain after SAH. In conclusion, the inhibition of EP3 by L798106 attenuated oxidative stress and neuronal apoptosis partly through p38MAPK/FOXO3/Mul1/Mfn2 pathway post-SAH in rats. EP3 may serve as a potential therapeutic target for SAH patients.
Collapse
Affiliation(s)
- Yu Liu
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Rui Liu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jiaxing Dai
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Ling Gao
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Hui Shi
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuanjian Fang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Qin Lu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Takeshi Okada
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Zhifei Wang
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China
| | - Xiao Hu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jie Xiao
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Emergency, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China
| | - John H. Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Neurosurgery and Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92350, USA
| |
Collapse
|
36
|
Lu J, Huang X, Deng A, Yao H, Wu G, Wang N, Gui H, Ren M, Guo S. miR-452-3p Targets HDAC3 to Inhibit p65 Deacetylation and Activate the NF-κB Signaling Pathway in Early Brain Injury after Subarachnoid Hemorrhage. Neurocrit Care 2022; 37:558-571. [PMID: 35641805 DOI: 10.1007/s12028-022-01509-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/05/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Subarachnoid hemorrhage (SAH) is a subtype of stroke, and early brain injury (EBI) is a contributor to its unfavorable outcome. microRNA (miRNA) is abundantly expressed in the brain and participates in brain injury. This study investigated the effect of miR-452-3p on EBI after SAH. METHODS The murine model of SAH was established. miR-452-3p expression was detected 48 h after the model establishment. Neurobehavioral function, blood-brain barrier permeability, brain water content, neuronal apoptosis, and inflammatory factors were evaluated. The cell model of SAH was induced by oxygen hemoglobin. Apoptosis rate, lactate dehydrogenase, and reactive oxygen species were detected. The targeting relationship between miR-452-3p and histone deacetylase 3 (HDAC3) was verified. The acetylation of p65 and the binding of HDAC3 to p65 were detected. The inhibitory protein of the nuclear factor κB pathway (IκBα) was detected. Suberoylanilide hydroxamic acid was injected into the SAH mice treated with miR-452-3p inhibitor. RESULTS SAH mice showed upregulated miR-452-3p expression; reduced the neurological score; increased blood-brain barrier permeability, brain water content, and neuronal apoptosis; elevated pro-inflammatory factors; and reduced anti-inflammatory factors. SAH increased the apoptosis rate, lactate dehydrogenase release, and reactive oxygen species levels in oxygen-hemoglobin-treated neuron cells. Inhibition of miR-452-3p reversed the above trends. miR-452-3p targeted HDAC3. SAH upregulated p65 acetylation. miR-452-3p inhibitor promoted the binding of HDAC3 to p65, decreased p65 acetylation, and upregulated IκBα. Suberoylanilide hydroxamic acid reversed the protective effect of miR-452-3p inhibitor on SAH mice and aggravated brain injury. CONCLUSIONS miR-452-3p targeted HDAC3 to inhibit the deacetylation of p65 and activate the nuclear factor κB pathway, thus aggravating EBI after SAH.
Collapse
Affiliation(s)
- Junti Lu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Xiaodong Huang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Aiping Deng
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Hong Yao
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Gao Wu
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Na Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Hui Gui
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Mojie Ren
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 People's South Road, Shiyan, 442000, Hubei, People's Republic of China
| | - Shiwen Guo
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
37
|
Xin Y, Chen J, Zhang H, Ostrowski RP, Liang Y, Zhao J, Xiang X, Liang F, Fu W, Huang H, Wu X, Su J, Deng J, He Z. Dexras1 Induces Dysdifferentiation of Oligodendrocytes and Myelin Injury by Inhibiting the cAMP-CREB Pathway after Subarachnoid Hemorrhage. Cells 2022; 11:cells11192976. [PMID: 36230939 PMCID: PMC9564295 DOI: 10.3390/cells11192976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
White matter damage (WMD), one of the research hotspots of subarachnoid hemorrhage (SAH), mainly manifests itself as myelin injury and oligodendrocyte differentiation disorder after SAH, although the specific mechanism remains unclear. Dexamethasone-induced Ras-related protein 1(Dexras1) has been reported to be involved in nervous system damage in autoimmune encephalitis and multiple sclerosis. However, whether Dexras1 participates in dysdifferentiation of oligodendrocytes and myelin injury after SAH has yet to be examined, which is the reason for creating the research content of this article. Here, intracerebroventricular lentiviral administration was used to modulate Dexras1 levels in order to determine its functional influence on neurological injury after SAH. Immunofluorescence, transmission electron microscopy, and Western blotting methods, were used to investigate the effects of Dexras1 on demyelination, glial cell activation, and differentiation of oligodendrocyte progenitor cells (OPCs) after SAH. Primary rat brain neurons were treated with oxyhemoglobin to verify the association between Dexras1 and cAMP-CREB. The results showed that Dexras1 levels were significantly increased upon in vivo SAH model, accompanied by OPC differentiation disturbances and myelin injury. Dexras1 overexpression significantly worsened OPC dysdifferentiation and myelin injury after SAH. In contrast, Dexras1 knockdown ameliorated myelin injury, OPC dysdifferentiation, and glial cell activation. Further research of the underlying mechanism discovered that the cAMP-CREB pathway was inhibited after Dexras1 overexpression in the in vitro model of SAH. This study is the first to confirm that Dexras1 induced oligodendrocyte dysdifferentiation and myelin injury after SAH by inhibiting the cAMP-CREB pathway. This present research may reveal novel therapeutic targets for the amelioration of brain injury and neurological dysfunction after SAH.
Collapse
Affiliation(s)
- Yuanjun Xin
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Jie Chen
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Hongxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Robert P. Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yidan Liang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Jun Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Xiang Xiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Fuming Liang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Wenqiao Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Hao Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Xintong Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Jun Su
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Jiewen Deng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Friendship Road, Chongqing 400016, China
- Correspondence: ; Tel.:+86-138-830-32981; Fax: +86-21-640-85875
| |
Collapse
|
38
|
Castillo G, Barrios-Arpi L, Ramos-Gonzalez M, Vidal P, Gonzales-Irribarren A, Ramos-Cevallos N, Rodríguez JL. Neurotoxicity associated with oxidative stress and inflammasome gene expression induced by allethrin in SH-SY5Y cells. Toxicol Ind Health 2022; 38:777-788. [PMID: 36074087 DOI: 10.1177/07482337221089585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pyrethroids, including allethrin, have largely been used as commercial insecticides. The toxicity of allethrin is little known, but it is assumed that, as occurs with other pyrethroids, it could cause alterations of the nervous system and pose both occupational and non-occupational health hazards. To evaluate the neurotoxicity of allethrin we used the MTT assay of SH-SY5Y neuroblastoma cells to determine cell viability. Dose-dependent reductions of cell viability served to compare the vehicle-group and the IC50 for allethrin, which was 49.19 μM. ROS production increased significantly at concentrations of 10-200 μM of allethrin, and NO levels were significantly increased by the effect of allethrin at a minimum concentration of 50 μM. Lipid peroxidation increased by the effect of allethrin at concentrations of 25, 50, 100, and 200 μM. Caspase 3/7 activity was induced by allethrin concentrations of 50, 100, and 200 μM. Here, we suggest that allethrin might affect the inflammasome complex (Caspase-1, NLRP3, and PYDC1) and apoptosis (Bax and Bcl-2) gene expression by mRNA fold change expression levels shown in Caspase-1 (2.46-fold), NLRP3 (1.57-fold), PYDC1 (1.48-fold), and Bax (2.1-fold). These results demonstrated that allethrin induced neurotoxicity effects on SH-SY5Y cells through activation of inflammasome pathways, cell death, and oxidative stress.
Collapse
Affiliation(s)
- Giovana Castillo
- Faculty of Pharmacy and Biochemistry, Research Institute Juan de Dios Guevara, 33209Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Luis Barrios-Arpi
- Animal Phisiology Laboratory, Faculty of Veterinary Medicine, 33209Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Mariella Ramos-Gonzalez
- Zootechnics and Animal Production Laboratory, Faculty of Veterinary Medicine, 33209Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Paola Vidal
- Animal Phisiology Laboratory, Faculty of Veterinary Medicine, 33209Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Alejandro Gonzales-Irribarren
- Pharmacology and Toxicology Laboratory, Faculty of Veterinary Medicine, 33209Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Norma Ramos-Cevallos
- Faculty of Pharmacy and Biochemistry, Research Institute Juan de Dios Guevara, 33209Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - José-Luis Rodríguez
- Pharmacology and Toxicology Laboratory, Faculty of Veterinary Medicine, 33209Universidad Nacional Mayor de San Marcos, Lima, Peru.,Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
39
|
Kutsche HS, Schreckenberg R, Schlüter KD. Uncoupling Proteins in Striated Muscle Tissue: Known Facts and Open Questions. Antioxid Redox Signal 2022; 37:324-335. [PMID: 35044239 DOI: 10.1089/ars.2021.0258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Uncoupling proteins (UCPs) are a family of proteins that allow proton leakage across the inner mitochondrial membrane. Although UCP1, also known as thermogenin, is well known and important for heat generation in brown adipose tissue, striated muscles express two distinct members of UCP, namely UCP2 and UCP3. Unlike UCP1, the main function of UCP2 and UCP3 does not appear to be heat production. Recent Advances: Interestingly, UCP2 is the main isoform expressed in cardiac tissues, whereas UCP3 is the dominant isoform in skeletal muscles. In the past years, researchers have started to investigate the regulation of UCP2 and UCP3 expression in striated muscles. Furthermore, concepts about the proposed functions of UCP2 and UCP3 in striated muscles are developed but are still a matter of debate. Critical Issues: Potential functions of UCP2 and UCP3 in striated muscles include a role in protection against mitochondria-dependent oxidative stress, as transporter for pyruvate, fatty acids, and protons into and out of the mitochondria, and in metabolic sensing. In this context, the different isoform expression of UCP2 and UCP3 in the skeletal and cardiac muscle may be related to different metabolic requirements of the two organs. Future Directions: The level of expression of UCP2 and UCP3 in striated muscles changes in different disease stages. This suggests that UCPs may become drug targets for therapy in the future. Antioxid. Redox Signal. 37, 324-335.
Collapse
Affiliation(s)
| | - Rolf Schreckenberg
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | | |
Collapse
|
40
|
Zhang Z, Zhang A, Liu Y, Hu X, Fang Y, Wang X, Luo Y, Lenahan C, Chen S. New Mechanisms and Targets of Subarachnoid Hemorrhage: A Focus on Mitochondria. Curr Neuropharmacol 2022; 20:1278-1296. [PMID: 34720082 PMCID: PMC9881073 DOI: 10.2174/1570159x19666211101103646] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH) accounts for 5-10% of all strokes and is a subtype of hemorrhagic stroke that places a heavy burden on health care. Despite great progress in surgical clipping and endovascular treatment for ruptured aneurysms, cerebral vasospasm (CVS) and delayed cerebral ischemia (DCI) threaten the long-term outcomes of patients with SAH. Moreover, there are limited drugs available to reduce the risk of DCI and adverse outcomes in SAH patients. New insight suggests that early brain injury (EBI), which occurs within 72 h after the onset of SAH, may lay the foundation for further DCI development and poor outcomes. The mechanisms of EBI mainly include excitotoxicity, oxidative stress, neuroinflammation, blood-brain barrier (BBB) destruction, and cellular death. Mitochondria are a double-membrane organelle, and they play an important role in energy production, cell growth, differentiation, apoptosis, and survival. Mitochondrial dysfunction, which can lead to mitochondrial membrane potential (Δψm) collapse, overproduction of reactive oxygen species (ROS), release of apoptogenic proteins, disorders of mitochondrial dynamics, and activation of mitochondria-related inflammation, is considered a novel mechanism of EBI related to DCI as well as post-SAH outcomes. In addition, mitophagy is activated after SAH. In this review, we discuss the latest perspectives on the role of mitochondria in EBI and DCI after SAH. We emphasize the potential of mitochondria as therapeutic targets and summarize the promising therapeutic strategies targeting mitochondria for SAH.
Collapse
Affiliation(s)
- Zeyu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,These authors contributed equally to this work.
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,These authors contributed equally to this work.
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,These authors contributed equally to this work.
| | - Xiaoming Hu
- Department of Neurosurgery, Taizhou Hospital, Taizhou, Zhejiang Province, China;
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
| | - Yujie Luo
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,Address correspondence to this author at the Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China; Tel: +86-571-87784815; Fax: +86-571-87784755; E-mail:
| |
Collapse
|
41
|
Xiao X, Sun S, Li Y, Cen X, Wu S, Lu A, Cai J, Zhao J, Li S. Geniposide attenuates early brain injury by inhibiting oxidative stress and neurocyte apoptosis after subarachnoid hemorrhage in rats. Mol Biol Rep 2022; 49:6303-6311. [PMID: 35474057 DOI: 10.1007/s11033-022-07438-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Oxidative stress and neurocyte apoptosis are crucial pathophysiological process in early brain injury (EBI) after subarachnoid hemorrhage (SAH). Geniposide (GNP) has been reported to exert neuroprotective effects by reducing oxidative injury and neurocyte apoptosis. However, the effect of GNP has not been clarified in EBI after SAH. The study was performed to evaluate the neuroprotective effects and mechanisms of GNP in EBI after SAH. METHODS AND RESULTS A total of 60 male Wistar rats were randomly divided into five groups. The prechiasmatic cistern SAH model was used in this study. SAH grade was evaluated using a grading system. Neurological function was evaluated using the Garcia scores. Brain edema was measured by the wet-dry method. Blood-brain barrier (BBB) permeability was measured by the extravasation of Evans Blue (EB). The neurocyte apoptosis was observed using TUNEL assay. The levels of malondialdehyde (MDA) and superoxide dismutase (SOD), as well as the expressions of nuclear factor erythroid 2-related factor 2 (Nrf2), hemeoxygenase-1 (HO-1), glutathione S-transferase (GST) and quinone oxidoreductase-1 (NQO-1) were performed. The results showed that GNP reduced brain edema, attenuated BBB permeability, inhibited neurocyte apoptosis and improved neurological function. Moreover, GNP also decreased the levels of ROS and MDA, elevated Nrf2 expression in the temporal cortex and up-regulated the expression of NQO-1, HO-1 and GST after SAH. CONCLUSIONS GNP could ameliorate oxidative stress and neurocyte apoptosis to exert neuroprotective effects by Nrf2 pathway.
Collapse
Affiliation(s)
- Xiaolan Xiao
- Department of Pediatrics, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Shuangxi Sun
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingbin Li
- Department of Neurosurgery, Hospital of Guangzhou University Mega Center, Guangdong Provincial Hospital of Chinese Medicine, 111 Dade Road, Guangzhou, 510120, Guangdong Province, China
| | - Xuecheng Cen
- Department of Neurosurgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Shibiao Wu
- Neurological Intensive Care Unit, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Aili Lu
- Neurological Intensive Care Unit, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jun Cai
- Department of Neurosurgery, Hospital of Guangzhou University Mega Center, Guangdong Provincial Hospital of Chinese Medicine, 111 Dade Road, Guangzhou, 510120, Guangdong Province, China
| | - Junjie Zhao
- Department of Neurosurgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Shaoxue Li
- Department of Neurosurgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
42
|
Zhou F, Wang Z, Xiong K, Zhang M, Wang Y, Wang M. Alantolactone reduced neuron injury via activating PI3K/Akt signaling pathway after subarachnoid hemorrhage in rats. PLoS One 2022; 17:e0270410. [PMID: 35749405 PMCID: PMC9231788 DOI: 10.1371/journal.pone.0270410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/09/2022] [Indexed: 12/23/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a common disease with high morbidity and mortality, which can cause pathological, physiological, and biological reactions. SAH causes a series of responses such as neuronal and cerebral cortex damage, which in turn leads to inflammation and apoptosis. Traditional Chinese medicine has a strong anti-inflammatory effect, such as Alantolactone (ATL). However, studies on ATL therapy for SAH have not been reported. We observed the neurological scores, brain water content, Evans blue (EB) extravasation, neuroinflammation, and apoptosis via performing an enzyme-linked immunosorbent assay (ELISA), western blotting, immunofluorescence staining, and other methods after SAH. In this study, we found that ATL treatment attenuated the neurologic deficits, inhibited neuronal apoptosis and inflammatory reaction, promoted polarization of microglia toward the M2 phenotype, and activated the PI3K/Akt signaling pathway. ATL can reduce the neurons and cerebral cortex damage of SAH rats through activating PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Neurosurgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of Neurosurgery, the Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Zhenzhi Wang
- Department of Chinese and Western Medicine, the Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Kang Xiong
- Department of Chinese and Western Medicine, the Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Meiling Zhang
- Department of Chinese and Western Medicine, the Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yuan Wang
- Combination of Acupuncture and Medicine Innovation Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Maode Wang
- Department of Neurosurgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- * E-mail:
| |
Collapse
|
43
|
Liu Y, Wang Z, Cao C, Xu Z, Lu J, Shen H, Li X, Li H, Wu J, Chen G. Aquaporin 4 Depolarization-Enhanced Transferrin Infiltration Leads to Neuronal Ferroptosis after Subarachnoid Hemorrhage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8808677. [PMID: 35761873 PMCID: PMC9233479 DOI: 10.1155/2022/8808677] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 12/19/2022]
Abstract
The infiltration of blood components into the brain parenchyma through the lymphoid system is an important cause of subarachnoid hemorrhage injury. AQP4, a water channel protein located at the astrocyte foot, has been reported to regulate blood-brain barrier integrity, and its polarization is disrupted after SAH. Neuronal ferroptosis is involved in subarachnoid hemorrhage- (SAH-) induced brain injury, but the inducing factors are not completely clear. Transferrin is one of the inducing factors of ferroptosis. This study is aimed at researching the role and mechanism of AQP4 in brain injury after subarachnoid hemorrhage in mice. An experimental mouse SAH model was established by endovascular perforation. An AAV vector encoding AQP4 with a GFAP-specific promoter was administered to mice to achieve specific overexpression of AQP4 in astrocytes. PI staining, Fer-1 intervention, and transmission electron microscopy were used to detect neuronal ferroptosis, and dextran (40 kD) leakage was used to detect BBB integrity. Western blot analysis of perfused brain tissue protein samples was used to detect transferrin infiltration. First, neuronal ferroptosis 24 h after SAH was observed by PI staining and Fer-1 intervention. Second, a significant increase in transferrin infiltration was found in the brain parenchyma 24 h after SAH modeling, while transferrin content was positively correlated with neuronal ferroptosis. Then, we observed that AQP4 overexpression effectively improved AQP depolarization and BBB injury induced by SAH and significantly reduced transferrin infiltration and neuronal ferroptosis after SAH. Finally, we found that AQP4 overexpression could effectively improve the neurobehavioral ability of SAH mice, and the neurobehavioral ability was negatively correlated with transferrin brain content. Taken together, these data indicate that overexpression of AQP4 in the mouse brain can effectively improve post-SAH neuronal ferroptosis and brain injury, at least partly by inhibiting transferrin infiltration into the brain parenchyma in the glymphatic system.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Chang Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Zhongmou Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| |
Collapse
|
44
|
Hashimoto M, Fujimoto M, Konno K, Lee ML, Yamada Y, Yamashita K, Toda C, Tomura M, Watanabe M, Inanami O, Kitamura H. Ubiquitin-Specific Protease 2 in the Ventromedial Hypothalamus Modifies Blood Glucose Levels by Controlling Sympathetic Nervous Activation. J Neurosci 2022; 42:4607-4618. [PMID: 35504726 PMCID: PMC9186793 DOI: 10.1523/jneurosci.2504-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/07/2022] [Accepted: 04/16/2022] [Indexed: 11/21/2022] Open
Abstract
Ubiquitin-specific protease 2 (USP2) participates in glucose metabolism in peripheral tissues such as the liver and skeletal muscles. However, the glucoregulatory role of USP2 in the CNS is not well known. In this study, we focus on USP2 in the ventromedial hypothalamus (VMH), which has dominant control over systemic glucose homeostasis. ISH, using a Usp2-specific probe, showed that Usp2 mRNA is present in VMH neurons, as well as other glucoregulatory nuclei, in the hypothalamus of male mice. Administration of a USP2-selective inhibitor ML364 (20 ng/head), into the VMH elicited a rapid increase in the circulating glucose level in male mice, suggesting USP2 has a suppressive role on glucose mobilization. ML364 treatment also increased serum norepinephrine concentration, whereas it negligibly affected serum levels of insulin and corticosterone. ML364 perturbated mitochondrial oxidative phosphorylation in neural SH-SY5Y cells and subsequently promoted the phosphorylation of AMP-activated protein kinase (AMPK). Consistent with these findings, hypothalamic ML364 treatment stimulated AMPKα phosphorylation in the VMH. Inhibition of hypothalamic AMPK prevented ML364 from increasing serum norepinephrine and blood glucose. Removal of ROS restored the ML364-evoked mitochondrial dysfunction in SH-SY5Y cells and impeded the ML364-induced hypothalamic AMPKα phosphorylation as well as prevented the elevation of serum norepinephrine and blood glucose levels in male mice. These results indicate hypothalamic USP2 attenuates perturbations in blood glucose levels by modifying the ROS-AMPK-sympathetic nerve axis.SIGNIFICANCE STATEMENT Under normal conditions (excluding hyperglycemia or hypoglycemia), blood glucose levels are maintained at a constant level. In this study, we used a mouse model to identify a hypothalamic protease controlling blood glucose levels. Pharmacological inhibition of USP2 in the VMH caused a deviation in blood glucose levels under a nonstressed condition, indicating that USP2 determines the set point of the blood glucose level. Modification of sympathetic nervous activity accounts for the USP2-mediated glucoregulation. Mechanistically, USP2 mitigates the accumulation of ROS in the VMH, resulting in attenuation of the phosphorylation of AMPK. Based on these findings, we uncovered a novel glucoregulatory axis consisting of hypothalamic USP2, ROS, AMPK, and the sympathetic nervous system.
Collapse
Affiliation(s)
- Mayuko Hashimoto
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 0698501, Japan
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 5848450, Japan
| | | | - Kohtarou Konno
- Department of Anatomy and Embryology, Graduate School of Medicine, Hokkaido University, Sapporo 0600808, Japan
| | - Ming-Liang Lee
- Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 0600808, Japan
| | - Yui Yamada
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 0698501, Japan
| | | | - Chitoku Toda
- Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 0600808, Japan
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 5848450, Japan
| | - Masahiko Watanabe
- Department of Anatomy and Embryology, Graduate School of Medicine, Hokkaido University, Sapporo 0600808, Japan
| | | | - Hiroshi Kitamura
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 0698501, Japan
| |
Collapse
|
45
|
Mi L, Min X, Chai Y, Zhang J, Chen X. NLRP1 Inflammasomes: A Potential Target for the Treatment of Several Types of Brain Injury. Front Immunol 2022; 13:863774. [PMID: 35707533 PMCID: PMC9189285 DOI: 10.3389/fimmu.2022.863774] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/28/2022] [Indexed: 12/28/2022] Open
Abstract
NOD-like receptor (NLR) family pyrin domain-containing 1 (NLRP1) is a member of the NLR family. The NLRP1 inflammasome consists of the NLRP1 protein, the adaptor protein apoptosis-associated speck-like protein containing a CARD domain, and the effector molecule pro-caspase-1. When stimulated, the inflammasome initiates the cleavage of pro-caspase-1 and converts it into its active form, caspase-1; then, caspase-1 facilitates the cleavage of the proinflammatory cytokines interleukin-1β and interleukin-18 into their active and secreted forms. In addition, caspase-1 also mediates the cleavage of gasdermin D, which leads to pyroptosis, an inflammatory form of cell death. Pathological events that damage the brain and result in neuropathological conditions can generally be described as brain injury. Neuroinflammation, especially that driven by NLRP1, plays a considerable role in the pathophysiology of brain injury, such as early brain injury (EBI) of subarachnoid hemorrhage, ischemic brain injury during stroke, and traumatic brain injury (TBI). In this article, a thorough overview of NLRP1 is presented, including its structure, mechanism of activation, and role in neuroinflammation. We also present recent studies on NLRP1 as a target for the treatment of EBI, ischemic brain injury, TBI, and other types of brain injury, thus highlighting the perspective of NLRP1 as an effective mediator of catastrophic brain injury.
Collapse
Affiliation(s)
- Liang Mi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xiaobin Min
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Baodi Clinical College, Tianjin Medical University, Tianjin, China
| | - Yan Chai
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, 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 Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, 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 Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
- *Correspondence: Xin Chen,
| |
Collapse
|
46
|
Dai S, Wei J, Zhang H, Luo P, Yang Y, Jiang X, Fei Z, Liang W, Jiang J, Li X. Intermittent fasting reduces neuroinflammation in intracerebral hemorrhage through the Sirt3/Nrf2/HO-1 pathway. J Neuroinflammation 2022; 19:122. [PMID: 35624490 PMCID: PMC9137193 DOI: 10.1186/s12974-022-02474-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/15/2022] [Indexed: 01/23/2023] Open
Abstract
Background Inflammation contributes to the poor prognosis of intracerebral hemorrhage (ICH). Intermittent fasting (IF) has been shown to be protective against inflammation in multiple pathogenic processes. In the present study, we aimed to investigated the beneficial effects of IF in attenuating neuroinflammation and neurological deficits in a mouse model of ICH and to investigate the underlying mechanism. Methods ICH was modeled by intrastriatal injection of autologous blood and IF was modeled by every-other-day feeding in male control mice (C57BL/6), mice with and microglia specific knockout Sirt3f/f;Cx3cr1-Cre (Sirt3 cKO), and Sirt3f/f (wild-type) mice. Brain tissues and arterial blood were harvested at 1, 3, 7 and 28 days after ICH for immunohistochemistry analysis of Iba-1, DARPP-32 and HO-1, morphological analysis by HE staining and inflammatory factor release tests by ELISA. Neurological functions were approached by corner test and cylinder test. Fluorescent double-labeled staining of Iba-1 with CD16, Arg1 or Sirt3 was used to provide direct image of co-expression of these molecules in microglia. TUNEL, cleaved caspase-3 and Nissl staining was performed to evaluate cellular injuries. Results IF alleviated neurological deficits in both acute and chronic phases after ICH. Morphologically, IF enhanced hematoma clearance, reduced brain edema in acute phase and attenuated striatum atrophy in chronic phase. In addition, IF decreased the numbers of TUNEL+ cells and increased Nissl+ neuron number at day 1, 3 and 7 after ICH. IF suppressed CD16+Iba-1+ microglia activation at day 3 after ICH and reduced inflammatory releases, such as IL-1β and TNF-α. The above effects of IF were attenuated by microglia Sirt3 deletion partly because of an inhibition of Nrf2/HO-1 signaling pathway. Interestingly, IF increased Iba-1+ microglia number at day 7 which mainly expressed Arg1 while decreased the proinflammatory factor levels. In mice with microglia-specific Sirt3 deletion, the effects of IF on Iba-1+ microglia activation and anti-inflammatory factor expressions were attenuated when compared with wild-type Sirt3f/f mice. Conclusions IF protects against ICH by suppressing the inflammatory responses via the Sirt3/Nrf2/HO-1 pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02474-2.
Collapse
Affiliation(s)
- Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China.,National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, 169 Changlexi Road, Xi'an, China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China.,Department of Health Service, Fourth Military Medical University, Xi'an, China
| | - Hongchen Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China
| | - Yuefan Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China
| | - Wenbin Liang
- University of Ottawa Heart Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jianli Jiang
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, 169 Changlexi Road, Xi'an, China.
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 127 Changlexi Road, Xi'an, China.
| |
Collapse
|
47
|
Inhibiting Microglia-Derived NLRP3 Alleviates Subependymal Edema and Cognitive Dysfunction in Posthemorrhagic Hydrocephalus after Intracerebral Hemorrhage via AMPK/Beclin-1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4177317. [PMID: 35620574 PMCID: PMC9129981 DOI: 10.1155/2022/4177317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 04/29/2022] [Indexed: 12/14/2022]
Abstract
For posthemorrhagic hydrocephalus (PHH) patients, whether occur subependymal edema indicates poor outcomes, partially manifested as cognitive impairment. In the brain, NLRP3 inflammasome mainly derived from microglia/macrophages is involved in proinflammatory and neurodeficits after hemorrhage, and autophagy is vital for neuronal homeostasis and functions. Accumulating evidence suggest that NLRP3 inflammasome and autophagy played an essential role after intracerebral hemorrhage (ICH). We aimed to dissect the mechanisms underlying subependymal edema formation and cognitive dysfunction. Here, based on the hydrocephalus secondary to ICH break into ventricular (ICH-IVH) in rats, this study investigated whether microglia/macrophage-derived NLRP3 induced subependymal edema formation and neuron apoptosis in subventricular zones (SVZ). In the acute phase of ICH-IVH, both the expression of NLRP3 inflammasome of microglia/macrophages and the autophagy of neurons were upregulated. The activated NLRP3 in microglia/macrophages promoted the release of IL-1beta to extracellular, which contributed to excessive autophagy, leading to neurons apoptosis both in vivo and in vitro through the AMPK/Beclin-1 pathway combined with transcriptomics. Administration of MCC950 (NLRP3 inflammasome specific inhibitor) after ICH-IVH significantly reduced edema formation and improved cognitive dysfunction. Thus, inhibiting NLRP3 activation in SVZ may be a promising therapeutic strategy for PHH patients that warrants further investigation.
Collapse
|
48
|
Wang Y, Xu J, You W, Shen H, Li X, Yu Z, Li H, Chen G. Roles of Rufy3 in experimental subarachnoid hemorrhage-induced early brain injury via accelerating neuronal axon repair and synaptic plasticity. Mol Brain 2022; 15:35. [PMID: 35461284 PMCID: PMC9034509 DOI: 10.1186/s13041-022-00919-6] [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: 12/12/2021] [Accepted: 04/06/2022] [Indexed: 11/25/2022] Open
Abstract
RUN and FYVE domain-containing 3 (Rufy3) is a well-known adapter protein of a small GTPase protein family and is bound to the activated Ras family protein to maintain neuronal polarity. However, in experimental subarachnoid hemorrhage (SAH), the role of Rufy3 has not been investigated. Consequently, we aimed to investigate the potential role of Rufy3 in an in vivo model of SAH-induced early brain injury (EBI). In addition, we investigated the relevant brain-protective mechanisms. Oxyhemoglobin (OxyHb) stimulation of cultured primary neurons simulated vitro SAH condition. The SAH rat model was induced by infusing autologous blood into the optic chiasma pool and treating the rats with lentivirus-negative control 1 (LV-NC1), lentivirus-Rufy3 shRNA (LV-shRNA), lentivirus-negative control 2 (LV-NC2), lentivirus-Rufy3 (LV-Rufy3), or 8-pCPT-2′-O-Me-cAMP (8p-CPT) (Rap1 agonist). In experiment one, we found that the protein level of Rufy3 decreased and neuronal axon injury in the injured neurons but was rectified by LV-Rufy3 treatment. In experiment two, mRNA and protein levels of Rufy3 were downregulated in brain tissue and reached the lowest level at 24 h after SAH. In addition, the expression of Myelin Basic Protein was downregulated and that of anti-hypophosphorylated neurofilament H (N52) was upregulated after SAH. In experiments three and four, Rufy3 overexpression (LV-Rufy3) increased the interactions between Rufy3 and Rap1, the level of Rap1-GTP, and the ratio of Rap1-GTP/total GTP. In addition, LV-Rufy3 treatment inhibited axon injury and accelerated axon repair by activating the Rap1/Arap3/Rho/Fascin signaling pathway accompanied by upregulated protein expression levels of ARAP3, Rho, Fascin, and Facin. LV-Rufy3 also enhanced synaptic plasticity by activating the Rap1/MEK/ERK/synapsin I signaling pathway accompanied by upregulated protein expression levels of ERK1, p-ERK1, MEK1, p-MEK1, synaspin I, and p-synaspin I. Moreover, LV-Rufy3 also alleviated brain damage indicators, including cortical neuronal cell apoptosis and degeneration, brain edema, and cognitive impairment after SAH. However, the downregulation of Rufy3 had the opposite effect and aggravated EBI induced by SAH. Notably, the combined application of LV-Rufy3 and 8p-CPT showed a significant synergistic effect on the aforementioned parameters. Our findings suggest that enhanced Rufy3 expression may reduce EBI by inhibiting axon injury and promoting neuronal axon repair and synaptic plasticity after SAH.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.,Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jianguo Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Wanchun You
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| |
Collapse
|
49
|
Chen Q, Cai Y, Zhu X, Wang J, Gao F, Yang M, Mao L, Zhang Z, Sun B. Edaravone Dexborneol Treatment Attenuates Neuronal Apoptosis and Improves Neurological Function by Suppressing 4-HNE-Associated Oxidative Stress After Subarachnoid Hemorrhage. Front Pharmacol 2022; 13:848529. [PMID: 35529450 PMCID: PMC9068884 DOI: 10.3389/fphar.2022.848529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Edaravone dexborneol is a novel neuroprotective drug that comprises edaravone and (+)-borneol in a 4:1 ratio. Phase II and III studies have demonstrated that Chinese patients treated with edaravone dexborneol within 48 h of AIS onset have better functional outcomes than those treated with edaravone alone. However, the effect of edaravone dexborneol on subarachnoid hemorrhage (SAH) has not yet been elucidated. This study aimed to investigate the therapeutic effects of edaravone dexborneol on SAH-induced brain injury and long-term behavioral deficits and to explore the possible mechanisms. The experimental rat SAH model was induced by an intraluminal puncture of the left middle cerebral artery (MCA). Edaravone dexborneol or edaravone at a clinical dose was infused into the tail vein for 3 days post-SAH surgery. Behavioral outcomes were assessed by a modified Garcia scoring system and rotarod, foot-fault, and corner tests. Immunofluorescence, Western blot, and ELISA methods were used to evaluate neuronal damage and oxidative stress. Our results showed that a post-SAH therapeutic regimen with edaravone dexborneol helped improve neurological function up to 21 days after SAH surgery and demonstrated a greater beneficial effect than edaravone alone, accompanied by an obvious inhibition of neuronal apoptosis in the CA1 hippocampus and basal cortex regions. Mechanistically, edaravone dexborneol not only suppressed the lipid peroxidation product malondialdehyde (MDA) but also improved the total antioxidant capability (TAC) 3 days after SAH. Notably, edaravone dexborneol treatment significantly inhibited the expression of another lipid peroxidation product, 4-hydroxynonenal (4-HNE), in the CA1 hippocampus and basal cortex, which are vital participants in the process of neuronal oxidative damage and death after SAH because of their acute cytotoxicity. Together, our results demonstrate that edaravone dexborneol confers neuroprotection and stabilizes long-term behavioral ability after SAH injury, possibly by suppressing 4-HNE-associated oxidative stress. These results may help develop new clinical strategies for SAH treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leilei Mao
- *Correspondence: Leilei Mao, ; Zongyong Zhang, ; Baoliang Sun,
| | - Zongyong Zhang
- *Correspondence: Leilei Mao, ; Zongyong Zhang, ; Baoliang Sun,
| | - Baoliang Sun
- *Correspondence: Leilei Mao, ; Zongyong Zhang, ; Baoliang Sun,
| |
Collapse
|
50
|
Solár P, Zamani A, Lakatosová K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids Barriers CNS 2022; 19:29. [PMID: 35410231 PMCID: PMC8996682 DOI: 10.1186/s12987-022-00312-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The response of the blood-brain barrier (BBB) following a stroke, including subarachnoid hemorrhage (SAH), has been studied extensively. The main components of this reaction are endothelial cells, pericytes, and astrocytes that affect microglia, neurons, and vascular smooth muscle cells. SAH induces alterations in individual BBB cells, leading to brain homeostasis disruption. Recent experiments have uncovered many pathophysiological cascades affecting the BBB following SAH. Targeting some of these pathways is important for restoring brain function following SAH. BBB injury occurs immediately after SAH and has long-lasting consequences, but most changes in the pathophysiological cascades occur in the first few days following SAH. These changes determine the development of early brain injury as well as delayed cerebral ischemia. SAH-induced neuroprotection also plays an important role and weakens the negative impact of SAH. Supporting some of these beneficial cascades while attenuating the major pathophysiological pathways might be decisive in inhibiting the negative impact of bleeding in the subarachnoid space. In this review, we attempt a comprehensive overview of the current knowledge on the molecular and cellular changes in the BBB following SAH and their possible modulation by various drugs and substances.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Klaudia Lakatosová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|