1
|
Martínez‐Torres AM, Ramírez‐Celis C, Morán J. Cannabinoid Receptors Reduced Early Brain Damage by Regulating NOX-2 and the NLRP3 Inflammasome in an Animal Model of Intracerebral Hemorrhage. CNS Neurosci Ther 2025; 31:e70385. [PMID: 40245261 PMCID: PMC12005396 DOI: 10.1111/cns.70385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a leading cause of death and disability worldwide. Following the initial mechanical injury caused by hematoma expansion, a secondary injury occurs, characterized by the production of reactive oxygen species (ROS) generated by NOX-2 and neuroinflammation, which is exacerbated by the upregulation of the NLRP3 inflammasome. These conditions collectively aggravate brain damage. The endocannabinoid system (ECS), through the activation of the cannabinoid receptors, has demonstrated neuroprotective properties in various models of brain injury. However, the role of the ECS during ICH remains poorly understood, particularly regarding the action of the CB1 receptor in the activation of NOX-2 and the inflammasome. The present study investigates the neuroprotective effects of the cannabinoid receptor agonist WIN55,212-2 in an ICH animal model, specifically examining the roles of NLRP3 and NOX-2. METHODS Male C57BL/6 mice were subjected to ICH through an intracerebral injection of collagenase, followed by intraperitoneal administration of WIN55,212-2 and/or MCC950, a selective NLRP3 inhibitor. Various outcome measures were employed, including assessments of motor activity, hematoma volume, brain water content, and blood-brain barrier (BBB) permeability, which was evaluated using Evans blue assay. Additionally, the activity of NOX and the protein levels of crucial markers such as CB1, gp91phox, NLRP3, AQP4, and caspase-1 were measured via western blot analysis. RESULT The findings demonstrate that ICH induced a significant brain lesion characterized by hematoma formation, edema, BBB disruption, and subsequent motor impairments in the affected mice. Notably, these detrimental effects were markedly reduced in animals treated with WIN55,212-2. The study also revealed an activation of both NOX-2 and NLRP3 in response to ICH, which was reduced by cannabinoid receptor activation. Furthermore, the pharmacological inhibition of NLRP3 using MCC950 also led to a reduction in hematoma size, edema, and motor impairment secondary to ICH. CONCLUSIONS These results support a neuroprotective role of the cannabinoid receptor activation during ICH and suggest the involvement of NOX-2 and NLRP3.
Collapse
Affiliation(s)
- Ari Misael Martínez‐Torres
- División de Neurociencias, Instituto de Fisiología CelularUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMéxico
| | - Crisalde Ramírez‐Celis
- División de Neurociencias, Instituto de Fisiología CelularUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMéxico
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología CelularUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMéxico
| |
Collapse
|
2
|
Cai Y, Huang G, Ren M, Chai Y, Fu Y, Yan T, Zhu L. Exploring the Kidney-Brain Crosstalk: Biomarkers for Early Detection of Kidney Injury-Related Alzheimer's Disease. J Inflamm Res 2025; 18:827-846. [PMID: 39845024 PMCID: PMC11752830 DOI: 10.2147/jir.s499343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025] Open
Abstract
Background The phenomenon of "kidney-brain crosstalk" has stimulated scholarly inquiry into the correlations between kidney injury (KI) and Alzheimer's disease (AD). Nonetheless, the precise interactions and shared mechanisms between KI and AD have yet to be fully investigated. The primary goal of this study was to investigate the link between KI and AD, with a specific focus on identifying diagnostic biomarkers for KI-related AD. Methods The first step of the present study was to use Mendelian randomization (MR) analysis to investigate the link between KI and AD, followed by verification of in vivo and in vitro experiments. Subsequently, bioinformatics and machine learning techniques were used to identify biomarkers for KI-associated ferroptosis-related genes (FRGs) in AD, which were validated in following experiments. Moreover, the relationship between hub biomarkers and immune infiltration was assessed using CIBERSORT, and the potential drugs or small molecules associated with the core biomarkers were identified via the DGIdb database. Results MR analysis showed that KI may be a risk factor for AD. Experiments showed that the combination of D-galactose and aluminum chloride was found to induce both KI and AD, with ferroptosis emerging as a bridge to facilitate crosstalk between KI and AD. Besides, we identified EGFR and RELA have significant diagnostic value. These biomarkers are associated with NK_cells_resting and B_cells_memory and could be targeted for intervention in KI-related AD by treating gefitinib and plumbagin. Conclusion Our study elucidates that ferroptosis may be an important pathway for kidney-brain crosstalk. Notably, gefitinib and plumbagin may be therapeutic candidates for intervening in KI-associated AD by targeting EGFR and RELA.
Collapse
Affiliation(s)
- Yawen Cai
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Guiqin Huang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Menghui Ren
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Yuhui Chai
- Department of Pharmacy, Shanghai Changhai Hospital, Second Military University, Shanghai, 200433, China
| | - Yu Fu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Tianhua Yan
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Lingpeng Zhu
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| |
Collapse
|
3
|
Fang S, Huang W, Qu X, Chai W. The mitochondria as a potential therapeutic target in cerebral I/R injury. Front Neurosci 2025; 18:1500647. [PMID: 39844858 PMCID: PMC11752919 DOI: 10.3389/fnins.2024.1500647] [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: 09/23/2024] [Accepted: 12/04/2024] [Indexed: 01/24/2025] Open
Abstract
Ischemic stroke is a major cause of mortality and disability worldwide. Among patients with ischemic stroke, the primary treatment goal is to reduce acute cerebral ischemic injury and limit the infarct size in a timely manner by ensuring effective cerebral reperfusion through the administration of either intravenous thrombolysis or endovascular therapy. However, reperfusion can induce neuronal death, known as cerebral reperfusion injury, for which effective therapies are lacking. Accumulating data supports a paradigm whereby cerebral ischemia/reperfusion (I/R) injury is coupled with impaired mitochondrial function, contributing to the pathogenesis of ischemic stroke. Herein, we review recent evidence demonstrating a heterogeneous mitochondrial response following cerebral I/R injury, placing a specific focus on mitochondrial protein modifications, reactive oxygen species, calcium (Ca2+), inflammation, and quality control under experimental conditions using animal models.
Collapse
Affiliation(s)
- Susu Fang
- The Second Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Wenzhou Huang
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, Jiangxi, China
| | - Xinhui Qu
- The Second Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Wen Chai
- Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
4
|
Akif A, My Nguyen TT, Liu L, Xu X, Kulkarni A, Jiang J, Zhang Y, Hao J. Targeting NLRP3 signaling with a novel sulfonylurea compound for the treatment of vascular cognitive impairment and dementia. RESEARCH SQUARE 2024:rs.3.rs-5611378. [PMID: 39764140 PMCID: PMC11702818 DOI: 10.21203/rs.3.rs-5611378/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Background As a key inflammatory factor, the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome plays a crucial role in neuroinflammation and the progression of neurodegenerative diseases. Dysregulation of NLRP3 signaling can trigger various inflammatory responses in the brain, contributing to the development of neurodegenerative diseases such as ischemic stroke, vascular dementia (VaD), Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Therefore, the NLRP3 signaling pathway is a promising therapeutic target for the treatment of neurodegenerative diseases, including VaD. Methods In this study, we investigated the therapeutic effects of a synthetic sulfonylurea NLRP3 inhibitor, AMS-17, in a VaD mouse model using bilateral common carotid artery stenosis (BCAS) and elucidated the underlying mechanisms. All mice were randomly divided into three groups: Sham, VaD + Vehicle, and VaD + AMS-17. Cognitive function was assessed using the Y-maze and Morris water maze (MWM) on the 50th day after BCAS. Brain sections and blood serum samples were collected for biomarker analysis and immunohistochemistry. Neurodegeneration, expressions of the molecules involved in the NLRP3 signaling pathways, tight junction proteins, and myelination were assessed using western blotting and immunofluorescence (IF). The levels of Interleukin-1 beta (IL-1β), Tumor Necrosis Factor-alpha (TNF-α) and Interleukin-4 (IL-4) in the blood were measured using ELISA. Results AMS-17 treatment improved cognitive function, enhanced blood-brain barrier (BBB) integrity, and promoted remyelination in VaD mice. Additionally, AMS-17 reduced neurodegeneration and decreased the expression of NLRP3 and its associated proteins, Apoptosis-associated speck-like protein (ASC), and cleaved caspase-1 in the brain. It also lowered pro-inflammatory TNF-α and IL-1β levels, while increasing the anti-inflammatory IL-4 level in the blood. Conclusions The findings of this study provide the first promising evidence for the use of AMS-17 in VaD treatment in mice. This study introduces AMS-17 as a novel chemical scaffold with NLRP3 inhibitory activity, which can be further developed for the treatment of VaD in humans.
Collapse
Affiliation(s)
| | | | - Langni Liu
- The University of Texas Health Science Center at Houston
| | - Xiaotian Xu
- The Affiliated Hospital of Yangzhou University
| | | | | | | | | |
Collapse
|
5
|
Wang Y, Liu A, Hui F, Han N, Zhang W, Sang Y. Correlation between peripheral blood inflammatory markers and delayed cerebral ischemia after intracerebral hemorrhage. Am J Transl Res 2024; 16:7749-7756. [PMID: 39822513 PMCID: PMC11733345 DOI: 10.62347/zxpv4184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/31/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE To assess the predictive value of peripheral blood inflammatory markers for delayed cerebral ischemia (DCI) in patients with intracerebral hemorrhage (ICH) and explore methods for early intervention. METHODS This single-center retrospective study reviewed medical records of ICH patients admitted to Cangzhou People's Hospital over a 12-month period from January 2022 to December 2023. Of the 150 identified patients with ICH, including 80 patients without DCI (control group) and 70 with DCI (observation group). Demographics and blood biochemical markers during hospitalization were recorded. Binary logistic regression was used to identify independent factors influencing DCI occurrence. RESULTS Significant differences were observed between the two groups in neutrophil percentage, lymphocyte percentage, lymphocytes absolute and neutrophil-to-lymphocyte ratio (NLR) (all P<0.05). Logistic regression analysis identified mRS score, lymphocyte percentage, educational level, uric acid, neutrophils percentage, and NLR as independent risk factors for DCI. CONCLUSION Neutrophil percentage and NLR are independent factors influencing delayed cerebral ischemia after intracerebral hemorrhage. These markers are readily accessible and may provide valuable insights for early ICH management.
Collapse
Affiliation(s)
- Yuhua Wang
- Department of Neurosurgery, Cangzhou People’s HospitalCangzhou 061000, Hebei, China
| | - Aijun Liu
- Department of Neurosurgery, Cangzhou People’s HospitalCangzhou 061000, Hebei, China
| | - Fengju Hui
- Department of Neurosurgery, Cangzhou People’s HospitalCangzhou 061000, Hebei, China
| | - Ning Han
- Department of Neurosurgery, Cangzhou People’s HospitalCangzhou 061000, Hebei, China
| | - Wei Zhang
- Department of Neurosurgery, Cangzhou People’s HospitalCangzhou 061000, Hebei, China
| | - Yinzhou Sang
- Department of Pathology, Cangzhou People’s HospitalCangzhou 061000, Hebei, China
| |
Collapse
|
6
|
Song JQ, Shen LJ, Wang HJ, Liu QB, Ye LB, Liu K, Shi L, Cai B, Lin HS, Pang T. Discovery of Balasubramide Derivative with Tissue-Specific Anti-Inflammatory Activity Against Acute Lung Injury by Targeting VDAC1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2410550. [PMID: 39556713 DOI: 10.1002/advs.202410550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/29/2024] [Indexed: 11/20/2024]
Abstract
Macrophage-mediated inflammatory responses including pyroptosis are involved in the pathogenesis of sepsis and acute lung injury (ALI), for which there are currently no effective therapeutic treatments. The natural product (+)-Balasubramide is an eight-membered lactam compound extracted from the leaves of the Sri Lanka plant Clausena Indica and has shown anti-inflammatory activities, but its poor pharmacokinetic properties limit its further application for ALI. In this study, a compound (+)3C-20 is discovered with improved both pharmacokinetic properties and anti-inflammatory activity from a series of (+)-Balasubramide derivatives. The compound (+)3C-20 exhibits a markedly enhanced inhibitory effect against LPS-induced expressions of pro-inflammatory factors in mouse macrophages and human PBMCs from ALI patients and shows a preferable lung tissue distribution in mice. (+)3C-20 remarkably attenuates LPS-induced ALI through lung tissue-specific anti-inflammatory actions. Mechanistically, a chemical proteomics study shows that (+)3C-20 directly binds to mitochondrial VDAC1 and inhibits VDAC1 oligomerization to block mtDNA release, further preventing NLRP3 inflammasome activation. These findings identify (+)3C-20 as a novel VDAC1 inhibitor with promising therapeutic potential for ALI associated with inflammation.
Collapse
Affiliation(s)
- Jin-Qian Song
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Li-Juan Shen
- Intensive Care Unit, Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, P. R. China
| | - Hao-Jie Wang
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Qi-Bing Liu
- Department of Pharmacy, the First Affiliated Hospital of Hainan Medical University & Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, Hainan Medical University, Haikou, 571199, P.R. China
| | - Lian-Bao Ye
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Kui Liu
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Lei Shi
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian, 116044, P. R. China
| | - Bin Cai
- Intensive Care Unit, Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, P. R. China
| | - Han-Sen Lin
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, P. R. China
| |
Collapse
|
7
|
Kaur B, Biby S, Namme JN, More S, Xu Y, Zhang S. Biological and therapeutic significance of targeting NLRP3 inflammasome in the brain and the current efforts to develop brain-penetrant inhibitors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 102:103-157. [PMID: 39929578 PMCID: PMC11955958 DOI: 10.1016/bs.apha.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, a pivotal regulator of the innate immune system, orchestrates inflammatory responses implicated in neurodegenerative and inflammatory diseases. Over the past 20 years, the exploration of NLRP3 activation pathways has advanced significantly. Upon NLRP3 activation, it initiates the formation of a cytosolic multiprotein complex known as the inflammasome. This complex activates caspase-1, which then processes proinflammatory cytokines IL-1β and IL-18 and leads to gasdermin-mediated cell death, pyroptosis. Structural insights into NLRP3 inflammasome assembly and caspase-1 activation have spurred development of novel small molecule inhibitors targeting this pathway, aiming to mitigate excessive inflammation without compromising immune surveillance. The initial NLRP3 inhibitor reported was glyburide, an FDA-approved antidiabetic drug of the sulfonylurea class, which was found to inhibit the release of IL-1β induced by stimuli in human monocytes and murine macrophages. Subsequently, MCC950 (also known as CRID3), a direct NLRP3 inhibitor, was discovered. While showing promising results in preclinical and clinical trials for treating diseases, higher doses of MCC950 led to elevated transaminase levels and hepatotoxicity concerns. Recent studies using MCC950 as a research tool have prompted the development of safer and more effective NLRP3 inhibitors, including a series of compounds currently undergoing clinical trials, highlighting the potential of NLRP3 inhibitors in attenuating disease progression and improving therapeutic outcomes. In this chapter, we delve into the latest progress in understanding the mechanism of NLRP3 inflammasome activation and its roles in the pathophysiology of neurological diseases. We also summarize recent development of small molecule NLRP3 inhibitors along with the associated obstacles and concerns.
Collapse
Affiliation(s)
- Baljit Kaur
- Department of Medicinal Chemistry, VCU, Richmond, VA, United States
| | - Savannah Biby
- Department of Medicinal Chemistry, VCU, Richmond, VA, United States
| | - Jannatun N Namme
- Department of Medicinal Chemistry, VCU, Richmond, VA, United States
| | - Sayaji More
- Department of Medicinal Chemistry, VCU, Richmond, VA, United States
| | - Yiming Xu
- Department of Medicinal Chemistry, VCU, Richmond, VA, United States
| | - Shijun Zhang
- Department of Medicinal Chemistry, VCU, Richmond, VA, United States.
| |
Collapse
|
8
|
Zheng Y, Zhang X, Wang Z, Zhang R, Wei H, Yan X, Jiang X, Yang L. MCC950 as a promising candidate for blocking NLRP3 inflammasome activation: A review of preclinical research and future directions. Arch Pharm (Weinheim) 2024; 357:e2400459. [PMID: 39180246 DOI: 10.1002/ardp.202400459] [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: 06/06/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
The NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome is a key component of the innate immune system that triggers inflammation and pyroptosis and contributes to the development of several diseases. Therefore, blocking the activation of the NLRP3 inflammasome has therapeutic potential for the treatment of these diseases. MCC950, a selective small molecule inhibitor, has emerged as a promising candidate for blocking NLRP3 inflammasome activation. Ongoing research is focused on elucidating the specific targets of MCC950 as well as assessfing its metabolism and safety profile. This review discusses the diseases that have been studied in relation to MCC950, with a focus on stroke, Alzheimer's disease, liver injury, atherosclerosis, diabetes mellitus, and sepsis, using bibliometric analysis. It then summarizes the potential pharmacological targets of MCC950 and discusses its toxicity. Furthermore, it traces the progression from preclinical to clinical research for the treatment of these diseases. Overall, this review provides a solid foundation for the clinical therapeutic potential of MCC950 and offers insights for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Yujia Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Ziyu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Lin Yang
- School of Medicial Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, Jinghai, China
| |
Collapse
|
9
|
Yuan X, Xia Y, Jiang P, Chen J, Wang C. Neuroinflammation Targeting Pyroptosis: Molecular Mechanisms and Therapeutic Perspectives in Stroke. Mol Neurobiol 2024; 61:7448-7465. [PMID: 38383921 DOI: 10.1007/s12035-024-04050-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
Pyroptosis is a recently identified type of pro-inflammatory programmed cell death (PCD) mediated by inflammasomes and nucleotide oligomerization domain-like receptors (NLs) and dependent on members of the caspase family. Pyroptosis has been widely reported to participate in the occurrence and progression of various inflammatory diseases, including stroke, a frequently lethal disease with high prevalence and many complications. To date, there have been no effectively therapeutic strategies and methods for treating stroke. Pyroptosis is thought to be closely related to the occurrence and development of stroke. Understanding inflammatory responses induced by the activation of pyroptosis would be hopeful to provide feasible approaches and strategies. Targeting on molecules in the upstream or downstream of pyroptosis pathway has shown promise in the treatment of stroke. The present review summarizes current research on the characteristics of pyroptosis, the function and pathological phenomena of pyroptosis in stroke, the molecule mechanisms related to inflammatory pathways, and the drugs and other molecules that can affect outcomes after stroke. These findings may help identify possible targets or new strategies for the diagnosis and treatment of stroke.
Collapse
Affiliation(s)
- Xiwen Yuan
- Neurobiology Key Laboratory of Jining Medical University, Jining, 272067, China
| | - Yiwen Xia
- Neurobiology Key Laboratory of Jining Medical University, Jining, 272067, China
| | - Pei Jiang
- Institute of Clinical Pharmacy & Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, 272011, China
| | - Jing Chen
- Neurobiology Key Laboratory of Jining Medical University, Jining, 272067, China.
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| | - Chunmei Wang
- Neurobiology Key Laboratory of Jining Medical University, Jining, 272067, China.
| |
Collapse
|
10
|
Lei C, Chen K, Gu Y, Li Y, Wang L, Zhu X, Deng Q. HMGB1/TLR4 axis promotes pyroptosis after ICH by activating the NLRP3 inflammasome. J Neuroimmunol 2024; 393:578401. [PMID: 38996718 DOI: 10.1016/j.jneuroim.2024.578401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND We previously reported that the HMGB1/TLR4 axis promoted inflammation during the acute phase of intracerebral hemorrhage. Given that this phase is known to involve neuronal pyroptosis and neuroinflammation, here we explore whether HMGB1/TLR signaling activate inflammasome and pyroptosis after intracerebral hemorrhage. METHODS Autologous blood was injected into Sprague-Dawley rats to induce intracerebral hemorrhage. Neurological deficits were assessed using a modified neurological severity score. These expression and localization of NLRP1 and NLRP3 inflammasomes, as well as the levels of pyroptosis and pyroptosis-associated proteins were assessed using Western blot or immunocytochemistry. These experiments were repeated in animals that received treatment with short interfering RNAs against NLRP1 or NLRP3, with HMGB1 inhibitor ethyl pyruvate or TLR4 inhibitor TAK-242. RESULTS Intracerebral hemorrhage upregulated NLRP1 and NLRP3 in the ipsilateral striatum and increased the proportions of these cells that were pyroptosis-positive. Additionally, the levels of caspase protein family (e.g., pro-caspase-1 and caspase-1), apoptosis-associated speck-like protein (ASC), pro-interleukin-1β (IL-1β), and IL-1β were also elevated. These effects on pyroptosis and associated neurological deficit, were partially reversed by knockdown of NLRP1 or NLRP3, or by inhibition of HMGB1 or TLR4. Inhibition of HMGB1 or TLR4 resulted in the downregulation NLRP3 but not NLRP1. CONCLUSIONS The HMGB1/TLR4 signaling may activate the NLRP3 inflammasome during the acute phase of intracerebral hemorrhage, resulting in the inflammatory process known as pyroptosis. These insights suggest potential therapeutic targets for the mitigation tissue injury and associated neurological deficits following hemorrhagic stroke.
Collapse
Affiliation(s)
- Chunyan Lei
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| | - Keyang Chen
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yu Gu
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yongyu Li
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Lu Wang
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xiaoyan Zhu
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Qionghua Deng
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| |
Collapse
|
11
|
Zhou C, Qiu SW, Wang FM, Liu YC, Hu W, Yang ML, Liu WH, Li H. Gasdermin D could be lost in the brain parenchyma infarct core and a pyroptosis-autophagy inhibition effect of Jie-Du-Huo-Xue decoction after stroke. Front Pharmacol 2024; 15:1449452. [PMID: 39139639 PMCID: PMC11320715 DOI: 10.3389/fphar.2024.1449452] [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: 06/15/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Background The Chinese ethnic medicine Jie-Du-Huo-Xue Decoction (JDHXD) is used to alleviate neuroinflammation in cerebral ischemia (CI). Our previous studies have confirmed that JDHXD can inhibit microglial pyroptosis in CI. However, the pharmacological mechanism of JDHXD in alleviating neuroinflammation and pyroptosis needs to be further elucidated. New research points out that there is an interaction between autophagy and inflammasome NLRP3, and autophagy can help clear NLRP3. The NLRP3 is a key initiator of pyroptosis and autophagy. The effect of JDHXD promoting autophagy to clear NLRP3 to inhibit pyroptosis on cerebral ischemia-reperfusion inflammatory injury is currently unknown. We speculate that JDHXD can inhibit pyroptosis in CI by promoting autophagy to clear NLRP3. Methods Chemical characterization of JDHXD was performed using LC-MS. Model of middle cerebral artery occlusion/reperfusion (MCAO/R) was established in SD rats. Neurological deficits, neuron damage, and cerebral infarct volume were evaluated. Western Blot and immunofluorescence were used to detect neuronal pyroptosis and autophagy. Results 30 possible substance metabolites in JDHXD medicated serum were analyzed by LC-MS (Composite Score > 0.98). Furthermore, JDHXD protects rat neurological function and cerebral infarct size after CI. JDHXD inhibited the expression of pyroptosis and autophagy after CI. Our western blot and immunofluorescence results showed that JDHXD treatment can reduce the expression of autophagy-related factors ULK1, beclin1, and LC3-Ⅱ. The expression of NLRP3 protein was lower in the JDHXD group than in the I/R group. Compared with the I/R group, the expressions of pyroptosis-related factors caspase-1 P 10, GSDMD-NT, IL-18, and IL-1β decreased in the JDHXD group. Furthermore, we observed an unexpected result: immunofluorescence demonstrated that Gasdermin D (GSDMD) was significantly absent in the infarct core, and highly expressed in the peri-infarct and contralateral cerebral hemispheres. This finding challenges the prevailing view that GSDMD is elevated in the ischemic cerebral hemisphere. Conclusion JDHXD inhibited pyroptosis and autophagy after MCAO/R. JDHXD suppressed pyroptosis and autophagy by inhibiting NLRP3, thereby alleviating CI. In addition, we present a different observation from previous studies that the expression of GSDMD in the infarct core was lower than that in the peri-infarct and contralateral non-ischemic hemispheres on day 3 of CI.
Collapse
Affiliation(s)
- Chang Zhou
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, Hunan, China
| | - Shi-wei Qiu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Feng-ming Wang
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yu-chen Liu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wei Hu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Mei-lan Yang
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang-hua Liu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation & Medicated Diet and Dietotherapy, University of Chinese Medicine, Changsha, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hua Li
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
12
|
Jin X, Ma Y, Liu D, Huang Y. Role of pyroptosis in the pathogenesis and treatment of diseases. MedComm (Beijing) 2023; 4:e249. [PMID: 37125240 PMCID: PMC10130418 DOI: 10.1002/mco2.249] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/16/2023] [Accepted: 03/07/2023] [Indexed: 05/02/2023] Open
Abstract
Programmed cell death (PCD) is regarded as a pathological form of cell death with an intracellular program mediated, which plays a pivotal role in maintaining homeostasis and embryonic development. Pyroptosis is a new paradigm of PCD, which has received increasing attention due to its close association with immunity and disease. Pyroptosis is a form of inflammatory cell death mediated by gasdermin that promotes the release of proinflammatory cytokines and contents induced by inflammasome activation. Recently, increasing evidence in studies shows that pyroptosis has a crucial role in inflammatory conditions like cardiovascular diseases (CVDs), cancer, neurological diseases (NDs), and metabolic diseases (MDs), suggesting that targeting cell death is a potential intervention for the treatment of these inflammatory diseases. Based on this, the review aims to identify the molecular mechanisms and signaling pathways related to pyroptosis activation and summarizes the current insights into the complicated relationship between pyroptosis and multiple human inflammatory diseases (CVDs, cancer, NDs, and MDs). We also discuss a promising novel strategy and method for treating these inflammatory diseases by targeting pyroptosis and focus on the pyroptosis pathway application in clinics.
Collapse
Affiliation(s)
- Xiangyu Jin
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| | - Yinchu Ma
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| | - Didi Liu
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| | - Yi Huang
- Wuxi School of MedicineJiangnan UniversityJiangsuChina
| |
Collapse
|
13
|
Long J, Sun Y, Liu S, Yang S, Chen C, Zhang Z, Chu S, Yang Y, Pei G, Lin M, Yan Q, Yao J, Lin Y, Yi F, Meng L, Tan Y, Ai Q, Chen N. Targeting pyroptosis as a preventive and therapeutic approach for stroke. Cell Death Discov 2023; 9:155. [PMID: 37165005 PMCID: PMC10172388 DOI: 10.1038/s41420-023-01440-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/12/2023] Open
Abstract
Stroke has caused tremendous social stress worldwide, yet despite decades of research and development of new stroke drugs, most have failed and rt-PA (Recombinant tissue plasminogen activator) is still the accepted treatment for ischemic stroke. the complexity of the stroke mechanism has led to unsatisfactory efficacy of most drugs in clinical trials, indicating that there are still many gaps in our understanding of stroke. Pyroptosis is a programmed cell death (PCD) with inflammatory properties and are thought to be closely associated with stroke. Pyroptosis is regulated by the GSDMD of the gasdermin family, which when cleaved by Caspase-1/Caspase-11 into N-GSDMD with pore-forming activity can bind to the plasma membrane to form small 10-20 nm pores, which would allow the release of inflammatory factors IL-18 and IL-1β before cell rupture, greatly exacerbating the inflammatory response. The pyroptosis occurs mainly in the border zone of cerebral infarction, and glial cells, neuronal cells and brain microvascular endothelial cells (BMECs) all undergo pyroptosis after stroke, which largely exacerbates the breakdown of the blood-brain barrier (BBB) and thus aggravates brain injury. Therefore, pyroptosis may be a good direction for the treatment of stroke. In this review, we focus on the latest mechanisms of action of pyroptosis and the process by which pyroptosis regulates stroke development. We also suggest potential therapeutic stroke drugs that target the pyroptosis pathway, providing additional therapeutic strategies for the clinical management of stroke. The role of pyroptosis after stroke. After stroke, microglia first rush to the damaged area and polarize into M1 and M2 types. Under the influence of various stimuli, microglia undergo pyroptosis, release pro-inflammatory factors, and are converted to the M1 type; astrocytes and neuronal cells also undergo pyroptosis under the stimulation of various pro-inflammatory factors, leading to astrocyte death due to increased osmotic pressure in the membrane, resulting in water absorption and swelling until rupture. BMECs, the main structural component of the BBB, also undergo pyroptosis when stimulated by pro-inflammatory factors released from microglia and astrocytes, leading to the destruction of the structural integrity of the BBB, ultimately causing more severe brain damage. In addition, GSDMD in neutrophils mainly mediate the release of NETs rather than pyroptosis, which also aggravates brain injury. IL-10=interleukin-10; TGF-β = transforming growth factor-β; IL-18=interleukin-18; IL-1β = interleukin-1β; TNF-α = tumor necrosis factor-α; iNOS=induced nitrogen monoxide synthase; MMPs=Matrix metalloproteinases; GSDMD = gasdermin D; BMECs=brain microvascular endothelial cells; BBB = blood-brain barrier.
Collapse
Affiliation(s)
- Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal & Child Health Care, Changsha, P. R. China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, P. R. China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Gang Pei
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, P. R. China
| | - Lei Meng
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yong Tan
- Department of Nephrology, Xiangtan Central Hospital, Xiangtan, P. R. China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China.
| |
Collapse
|
14
|
Wu X, Wei J, Yi Y, Shu G, He Z, Gong Q, Gao J. Epimedium Aqueous Extract Ameliorates Cerebral Ischemia/Reperfusion Injury through Inhibiting ROS/NLRP3-Mediated Pyroptosis. Antioxidants (Basel) 2023; 12:999. [PMID: 37237865 PMCID: PMC10215306 DOI: 10.3390/antiox12050999] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Cerebral ischemia/reperfusion causes exacerbated neuronal damage involving excessive neuroinflammation and oxidative stress. ROS is considered a signal molecule to activate NLRP3; thus, the ROS/NLRP3/pyroptosis axis plays a vital role in the pathogenesis of cerebral ischemia/reperfusion injury (CIRI). Therefore, targeting the inhibition of the ROS/NLRP3/pyroptosis axis may be a promising therapeutic tactic for CIRI. Epimedium (EP) contains many active ingredients (ICA, ICS II, and ICT), which have a wide range of pharmacological activities. However, whether EP can protect against CIRI remains unknown. Thus, in this study, we designed to investigate the effect and possible underlying mechanism of EP on CIRI. The results showed that treatment with EP dramatically mitigated brain damage in rats following CIRI, which was achieved by suppressing mitochondrial oxidative stress and neuroinflammation. Furthermore, we identified the ROS/NLRP3/pyroptosis axis as a vital process and NLRP3 as a vital target in EP-mediated protection. Most interestingly, the main compounds of EP directly bonded with NLRP3, as reflected by molecular docking, which indicated that NLRP3 might be a promising therapeutic target for EP-elicited cerebral protection. In conclusion, our findings illustrate that ICS II protects against neuron loss and neuroinflammation after CIRI by inhibiting ROS/NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Jiajia Wei
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Yang Yi
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Guotao Shu
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Zhixu He
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration, Medicine of Zunyi Medical University, Zunyi 563000, China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|