1
|
Wang X, Shang Z, Zhao J, Hou H, Li Y, Song J. Efficacy and Safety of Early Use of Naoxueshu Within 72 hours in the Treatment of Spontaneous Intracerebral Hemorrhage: A Real-World Retrospective Cohort Study. Int J Gen Med 2025; 18:2057-2065. [PMID: 40226803 PMCID: PMC11994074 DOI: 10.2147/ijgm.s511802] [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: 12/31/2024] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Background Naoxueshu Oral Liquid (NXS) is the only traditional Chinese medicine approved for the treatment of spontaneous intracerebral hemorrhage (ICH). While randomized controlled trials have demonstrated its ability to promote hematoma absorption and improve neurological function prognosis, the efficacy and safety of NXS early use (within 72 hours) remain unclear. This study aims to evaluate the efficacy and safety of early NXS administration (within 72 hours) in a real-world setting. Methods Data were collected from 34 tertiary hospitals in China. Patients were enrolled from March 25, 2019 to December 31, 2023. NXS administration was defined as the exposure. The primary outcome was hematoma volume at 14 days after onset. We employed the 1:1 propensity score matching (PSM) method to deal with confounding factors. Results A total of 1602 patients were enrolled after PSM, including 872 NXS users (exposed group) and 730 non-NXS users (control group). At baseline, there was no significant difference in hematoma volumes between the two groups (21.46±19.47 vs 22.01±14.26mL, P=0.55), the NXS group showed significantly less hematoma volume by day 14 (6.87±8.62 vs 5.43±5.35mL, P<0.001). There was no statistically significant difference in the incidence rate of serious adverse events between the two groups. Subgroup analysis indicated that NXS might have a more pronounced effect on hematoma absorption in supratentorial hemorrhage patients, with earlier administration potentially enhancing efficacy. Conclusion This retrospective study explored the efficacy and safety of NXS in promoting hematoma absorption within 72 hours in real-world ICH patients, but its effect on short-term neurological improvement remains inconclusive. Further studies with longer follow-up periods and more comprehensive functional assessments are warranted to explore the long-term neurological benefits of NXS in ICH patients.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhiyuan Shang
- Department of Neurosurgery, Beijing Fengtai You’anmen Hospital, Beijing, People’s Republic of China
| | - Jiapeng Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Hongping Hou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Ye Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, People’s Republic of China
| | - Juexian Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Tang X, Zheng N, Lin Q, You Y, Gong Z, Zhuang Y, Wu J, Wang Y, Huang H, Ke J, Chen F. Hypoxia-preconditioned bone marrow-derived mesenchymal stem cells protect neurons from cardiac arrest-induced pyroptosis. Neural Regen Res 2025; 20:1103-1123. [PMID: 38845218 PMCID: PMC11438345 DOI: 10.4103/nrr.nrr-d-23-01922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/28/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00027/figure1/v/2024-07-06T104127Z/r/image-tiff Cardiac arrest can lead to severe neurological impairment as a result of inflammation, mitochondrial dysfunction, and post-cardiopulmonary resuscitation neurological damage. Hypoxic preconditioning has been shown to improve migration and survival of bone marrow-derived mesenchymal stem cells and reduce pyroptosis after cardiac arrest, but the specific mechanisms by which hypoxia-preconditioned bone marrow-derived mesenchymal stem cells protect against brain injury after cardiac arrest are unknown. To this end, we established an in vitro co-culture model of bone marrow-derived mesenchymal stem cells and oxygen-glucose deprived primary neurons and found that hypoxic preconditioning enhanced the protective effect of bone marrow stromal stem cells against neuronal pyroptosis, possibly through inhibition of the MAPK and nuclear factor κB pathways. Subsequently, we transplanted hypoxia-preconditioned bone marrow-derived mesenchymal stem cells into the lateral ventricle after the return of spontaneous circulation in an 8-minute cardiac arrest rat model induced by asphyxia. The results showed that hypoxia-preconditioned bone marrow-derived mesenchymal stem cells significantly reduced cardiac arrest-induced neuronal pyroptosis, oxidative stress, and mitochondrial damage, whereas knockdown of the liver isoform of phosphofructokinase in bone marrow-derived mesenchymal stem cells inhibited these effects. To conclude, hypoxia-preconditioned bone marrow-derived mesenchymal stem cells offer a promising therapeutic approach for neuronal injury following cardiac arrest, and their beneficial effects are potentially associated with increased expression of the liver isoform of phosphofructokinase following hypoxic preconditioning.
Collapse
Affiliation(s)
- Xiahong Tang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Nan Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Qingming Lin
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Yan You
- The Second Department of Intensive Care Unit, Fujian Provincial Hospital South Branch, Fuzhou, Fujian Province, China
| | - Zheng Gong
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Yangping Zhuang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Jiali Wu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Yu Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Hanlin Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Jun Ke
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Feng Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| |
Collapse
|
3
|
Sun KY, Bai XY, Zhang L, Zhang X, Hu QQ, Song YX, Qiang RR, Zhang N, Zou JL, Yang YL, Xiang Y. A new strategy for the treatment of intracerebral hemorrhage: Ferroptosis. Exp Neurol 2024; 382:114961. [PMID: 39288829 DOI: 10.1016/j.expneurol.2024.114961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Intracerebral hemorrhage, is a cerebrovascular disease with high morbidity, mortality, and disability. Due to the lack of effective clinical treatments, the development of new drugs to treat intracerebral hemorrhage is necessary. In recent years, ferroptosis has been found to play an important role in the pathophysiological process of intracerebral hemorrhage, which can be treated by inhibiting ferroptosis and thus intracerebral hemorrhage. This article aims to explain the mechanism of ferroptosis and its relationship to intracerebral hemorrhage. In the meantime, it briefly discusses the molecules identified to alleviate intracerebral hemorrhage by inhibiting ferroptosis, along with other clinical agents that are expected to treat intracerebral hemorrhage through this mechanism. In addition, a brief overview of the morphological alterations of different forms of cell death and their role in ICH is provided. Finally, the challenges that may arise in translating ferroptosis inhibitors from basic research to clinical use are presented. This article serves as a reference and provides insights to aid in the treatment of intracerebral hemorrhage in the clinic.
Collapse
Affiliation(s)
- Ke Yao Sun
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Qian Qian Hu
- School of Medicine, Yan'an University, Yan'an, China
| | - Yu Xuan Song
- School of Medicine, Yan'an University, Yan'an, China
| | | | - Ning Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Jia Lun Zou
- School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- School of Medicine, Yan'an University, Yan'an, China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, China; College of Physical Education, Yan'an University, Yan'an, China.
| |
Collapse
|
4
|
Zhang MJ, Yang L, Li ZY, Zhou LY, Wang YJ, Wang HS, Cui XJ, Yao M. NLRP1 inflammasome in neurodegenerative disorders: From pathology to therapies. Cytokine Growth Factor Rev 2024; 80:138-155. [PMID: 39443194 DOI: 10.1016/j.cytogfr.2024.10.004] [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: 08/13/2024] [Revised: 10/06/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
Neuroinflammation is a critical component in neurodegenerative disorders. The inflammasome, facilitates the cleavage of caspase-1, leading to the maturation and subsequent secretion of inflammatory factors interleukin (IL)-1β and IL-18. Consequently, pyroptosis mediated by gasdermin D, exacerbates neuroinflammation. Among the inflammasomes, NLRP1/3 are predominant in the central nervous system (CNS), Although NLRP1 was the earliest discovered inflammasome, the specific involvement of NLRP1 in neurodegenerative diseases remains to be fully elucidated. Recently, the discovery of an endogenous inhibitor of NLRP1, dipeptidyl peptidase 9, suggests the feasibility of producing of small-molecule drugs targeting NLRP1. This review describes the latest findings on the role of the NLRP1 inflammasome in the pathology of neurodegenerative disorders, including Alzheimer's disease, and summarises the regulatory mechanisms of NLRP1 inflammasome activation in the CNS. Furthermore, we highlight the recent progress in developing small-molecule and biological inhibitors that modulate the NLRP1 infammasome for the treatment of neurodegenerative disorders, some of which are advancing to preclinical testing. SIGNIFICANCE STATEMENT: The objective of this review is to synthesise the research on the structure, activation, and regulatory mechanisms of the NLRP1 inflammasome, along with its potential impact on both acute and chronic neurodegenerative conditions. The discovery of endogenous inhibitors, such as dipeptidyl peptidase 9 and thioredoxin, and their interaction with NLRP1 suggest the possibility of developing NLRP1-targeted small-molecule drugs for the treatment of neurodegenerative disorders. This review also discusses the use of both direct and indirect NLRP1 inhibitors as prospective therapeutic strategies for these conditions.
Collapse
Affiliation(s)
- Meng-Jie Zhang
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Long Yang
- Rehabilitation Medicine Department, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Zhuo-Yao Li
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Long-Yun Zhou
- Rehabilitation Medicine Center, Jiangsu Provincial People's Hospital, Jiangsu 210029, China
| | - Yong-Jun Wang
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Hong-Shen Wang
- Orthopedics Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Xue-Jun Cui
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Min Yao
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
5
|
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
|
6
|
Jin Z, Lu J, Xu H, Zhang Y, Zhang S, Zhang D, Hu J, Shi Z, Li Z, Wang J. Exploring the correlation between innate immune activation of inflammasome and regulation of pyroptosis after intracerebral hemorrhage: From mechanism to treatment. Biomed Pharmacother 2024; 179:117382. [PMID: 39241565 DOI: 10.1016/j.biopha.2024.117382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024] Open
Abstract
Stroke has emerged as the primary cause of disability and death globally in recent years. Intracerebral hemorrhage (ICH), a particularly severe kind of stroke, is occurring in an increasing number of people. The two main clinical treatments for ICH now in use are conservative pharmaceutical therapy and surgical intervention, both of which have risks and drawbacks. Consequently, it is crucial to look into the pathophysiology of ICH and consider cutting-edge therapeutic approaches. Recent research has revealed that pyroptosis is a newly identified type of cell death distinguished by the break of the cell membrane and the discharge of pro-inflammatory substances through different routes. Following ICH, glial cells experience pyroptosis, which worsens neuroinflammation. Hence, the onset and progression of ICH are strongly linked to pyroptosis, which is facilitated by different inflammasomes. It is essential to conduct a comprehensive investigation of ICH damage processes and uncover new targets for treatment. The impact and function of pyroptosis in ICH, as well as the activation and regulation of inflammasomes and their mediated pyroptosis pathways will be fully discussed in this review.
Collapse
Affiliation(s)
- Ziqi Jin
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130017, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Ying Zhang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Shanshan Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130017, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Jing Hu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130017, China
| | - Zhao Shi
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130017, China
| | - Zhuyang Li
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130017, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, China.
| |
Collapse
|
7
|
Xiang C, Chen L, Zhu S, Chen Y, Huang H, Yang C, Chi Y, Wang Y, Lei Y, Cai X. CRLF1 bridges AKT and mTORC2 through SIN1 to inhibit pyroptosis and enhance chemo-resistance in ovarian cancer. Cell Death Dis 2024; 15:662. [PMID: 39256356 PMCID: PMC11387770 DOI: 10.1038/s41419-024-07035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024]
Abstract
Ovarian cancer, the second most leading cause of gynecologic cancer mortality worldwide, is challenged by chemotherapy resistance, presenting a significant hurdle. Pyroptosis, an inflammation-linked programmed cell death mediated by gasdermins, has been shown to impact chemoresistance when dysregulated. However, the mechanisms connecting pyroptosis to chemotherapy resistance in ovarian cancer are unclear. We found that cytokine receptor-like factor 1 (CRLF1) is a novel component of mTORC2, enhancing AKT Ser473 phosphorylation through strengthening the interaction between AKT and stress-activated protein kinase interacting protein 1 (SIN1), which in turn inhibits the mitogen-activated protein kinase kinase kinase 5 (ASK1)-JNK-caspase-3-gasdermin E pyroptotic pathway and ultimately confers chemoresistance. High CRLF1-expressing tumors showed sensitivity to AKT inhibition but tolerance to cisplatin. Remarkably, overexpression of binding-defective CRLF1 variants impaired AKT-SIN1 interaction, promoting pyroptosis and chemosensitization. Thus, CRLF1 critically regulates chemoresistance in ovarian cancer by modulating AKT/SIN1-dependent pyroptosis. Binding-defective CRLF1 variants could be developed as tumor-specific polypeptide drugs to enhance chemotherapy for ovarian cancer.
Collapse
Affiliation(s)
- Cong Xiang
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, China
| | - Li Chen
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, China
| | - Shilei Zhu
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yue Chen
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, China
| | - Haodong Huang
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, China
| | - Chunmao Yang
- Department of Gynecology, Southwest Hospital, Chongqing, China
| | - Yugang Chi
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, China
| | - Yanzhou Wang
- Department of Gynecology, Southwest Hospital, Chongqing, China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Molecular Medicine and Cancer Research Center, College of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Xiongwei Cai
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, China.
| |
Collapse
|
8
|
Chen CC, Ke CH, Wu CH, Lee HF, Chao Y, Tsai MC, Shyue SK, Chen SF. Transient receptor potential vanilloid 1 inhibition reduces brain damage by suppressing neuronal apoptosis after intracerebral hemorrhage. Brain Pathol 2024; 34:e13244. [PMID: 38308041 PMCID: PMC11328348 DOI: 10.1111/bpa.13244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/19/2024] [Indexed: 02/04/2024] Open
Abstract
Intracerebral hemorrhage (ICH) induces a complex sequence of apoptotic cascades and inflammatory responses, leading to neurological impairment. Transient receptor potential vanilloid 1 (TRPV1), a nonselective cation channel with high calcium permeability, has been implicated in neuronal apoptosis and inflammatory responses. This study used a mouse ICH model and neuronal cultures to examine whether TRPV1 activation exacerbates brain damage and neurological deficits by promoting neuronal apoptosis and neuroinflammation. ICH was induced by injecting collagenase in both wild-type (WT) C57BL/6 mice and TRPV1-/- mice. Capsaicin (CAP; a TRPV1 agonist) or capsazepine (a TRPV1 antagonist) was administered by intracerebroventricular injection 30 min before ICH induction in WT mice. The effects of genetic deletion or pharmacological inhibition of TRPV1 using CAP or capsazepine on motor deficits, histological damage, apoptotic responses, blood-brain barrier (BBB) permeability, and neuroinflammatory reactions were explored. The antiapoptotic mechanisms and calcium influx induced by TRPV1 inactivation were investigated in cultured hemin-stimulated neurons. TRPV1 expression was upregulated in the hemorrhagic brain, and TRPV1 was expressed in neurons, microglia, and astrocytes after ICH. Genetic deletion of TRPV1 significantly attenuated motor deficits and brain atrophy for up to 28 days. Deletion of TRPV1 also reduced brain damage, neurodegeneration, microglial activation, cytokine expression, and cell apoptosis at 1 day post-ICH. Similarly, the administration of CAP ameliorated brain damage, neurodegeneration, brain edema, BBB permeability, and cytokine expression at 1 day post-ICH. In primary neuronal cultures, pharmacological inactivation of TRPV1 by CAP attenuated neuronal vulnerability to hemin-induced injury, suppressed apoptosis, and preserved mitochondrial integrity in vitro. Mechanistically, CAP reduced hemin-stimulated calcium influx and prevented the phosphorylation of CaMKII in cultured neurons, which was associated with reduced activation of P38 and c-Jun NH2-terminal kinase mitogen-activated protein kinase signaling. Our results suggest that TRPV1 inhibition may be a potential therapy for ICH by suppressing mitochondria-related neuronal apoptosis.
Collapse
Affiliation(s)
- Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Taoyuan, Taiwan, Republic of China
| | - Chia-Hua Ke
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Chun-Hu Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Hung-Fu Lee
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, Republic of China
| | - Yuan Chao
- Department of Medical Education, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
9
|
Wang Y, Fang N, Wang Y, Geng Y, Li Y. Activating MC4R Promotes Functional Recovery by Repressing Oxidative Stress-Mediated AIM2 Activation Post-spinal Cord Injury. Mol Neurobiol 2024; 61:6101-6118. [PMID: 38277117 DOI: 10.1007/s12035-024-03936-9] [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: 09/22/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
Spinal cord injury (SCI) is a destructive neurological trauma that induces permanent sensory and motor impairment as well as a deficit in autonomic physiological function. Melanocortin receptor 4 (MC4R) is a G protein-linked receptor that is extensively expressed in the neural system and contributes to inhibiting inflammation, regulating mitochondrial function, and inducing programmed cell death. However, the effect of MC4R in the modulation of oxidative stress and whether this mechanism is related to the role of absent in melanoma 2 (AIM2) in SCI are not confirmed yet. In the current study, we demonstrated that MC4R is significantly increased in the neurons of spinal cords after trauma and oxidative stimulation of cells. Further, activation of MC4R by RO27-3225 effectively improved functional recovery, inhibited AIM2 activation, maintained mitochondrial homeostasis, repressed oxidative stress, and prevented Drp1 translocation to the mitochondria. Meanwhile, treating Drp1 inhibitors would be beneficial in reducing AIM2 activation, and activating AIM2 could abolish the protective effect of MC4R on neuron homeostasis. In conclusion, we demonstrated that MC4R protects against neural injury through a novel process by inhibiting mitochondrial dysfunction, oxidative stress, as well as AIM2 activation, which may serve as an available candidate for SCI therapy.
Collapse
Affiliation(s)
- Yongli Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Orthopaedics, Huzhou Central Hospital, Huzhou Basic and Clinical Translation of Orthopaedics Key Laboratory, Huzhou, Zhejiang, China
| | - Nongtao Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yikang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yibo Geng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
10
|
Wang J, Gu D, Jin K, Shen H, Qian Y. Egr1 promotes Nlrc4-dependent neuronal pyroptosis through phlda1 in an in-vitro model of intracerebral hemorrhage. Neuroreport 2024; 35:590-600. [PMID: 38652514 DOI: 10.1097/wnr.0000000000002035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Intracerebral hemorrhage (ICH) is a fatal brain injury, but the current treatments for it are inadequate to reduce the severity of secondary brain injury. Our study aims to explore the molecular mechanism of Egr1 and Phlda1 in regulating hemin-induced neuronal pyroptosis, and hope to provide novel therapeutic targets for ICH treatment. Mouse hippocampal neuron cells treated with hemin were used to simulate an in-vitro ICH model. Using qRT-PCR and western blot to evaluate mRNA and protein concentrations. MTT assay was utilized to assess cell viability. LDH levels were determined by lactate Dehydrogenase Activity Assay Kit. IL-1β and IL-18 levels were examined by ELISA. The interaction of Egr1 and Phlda1 promoter was evaluated using chromatin immunoprecipitation and dual-luciferase reporter assays. Egr1 and Phlda1 were both upregulated in HT22 cells following hemin treatment. Hemin treatment caused a significant reduction in HT22 cell viability, an increase in Nlrc4 and HT22 cell pyroptosis, and heightened inflammation. However, knocking down Egr1 neutralized hemin-induced effects on HT22 cells. Egr1 bound to the promoter of Phlda1 and transcriptionally activated Phlda1. Silencing Phlda1 significantly reduced Nlrc4-dependent neuronal pyroptosis. Conversely, overexpressing Phlda1 mitigated the inhibitory effects of Egr1 knockdown on Nlrc4 and neuronal pyroptosis during ICH. Egr1 enhanced neuronal pyroptosis mediated by Nlrc4 under ICH via transcriptionally activating Phlda1.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurosurgery, Taicang Hospital of Traditional Chinese Medicine, Taicang, Jiangsu Province, China
| | | | | | | | | |
Collapse
|
11
|
Jin MH, Liu XD, Sun HN, Han YH, Kwon T. Peroxiredoxin II exerts neuroprotective effects by inhibiting endoplasmic reticulum stress and oxidative stress-induced neuronal pyroptosis. Mol Biol Rep 2024; 51:607. [PMID: 38704801 DOI: 10.1007/s11033-024-09568-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a critical neurological condition with few treatment options, where secondary immune responses and specific cell death forms, like pyroptosis, worsen brain damage. Pyroptosis involves gasdermin-mediated membrane pores, increasing inflammation and neural harm, with the NLRP3/Caspase-1/GSDMD pathway being central to this process. Peroxiredoxin II (Prx II), recognized for its mitochondrial protection and reactive oxygen species (ROS) scavenging abilities, appears as a promising neuronal pyroptosis modulator. However, its exact role and action mechanisms need clearer definition. This research aims to explore Prx II impact on neuronal pyroptosis and elucidate its mechanisms, especially regarding endoplasmic reticulum (ER) stress and oxidative stress-induced neuronal damage modulation. METHODS AND RESULTS Utilizing MTT assays, Microscopy, Hoechst/PI staining, Western blotting, and immunofluorescence, we found Prx II effectively reduces LPS/ATP-induced pyroptosis and neuroinflammation in HT22 hippocampal neuronal cells. Our results indicate Prx II's neuroprotective actions are mediated through PI3K/AKT activation and ER stress pathway inhibition, diminishing mitochondrial dysfunction and decreasing neuronal pyroptosis through the ROS/MAPK/NF-κB pathway. These findings highlight Prx II potential therapeutic value in improving intracerebral hemorrhage outcomes by lessening secondary brain injury via critical signaling pathway modulation involved in neuronal pyroptosis. CONCLUSIONS Our study not only underlines Prx II importance in neuroprotection but also opens new therapeutic intervention avenues in intracerebral hemorrhage, stressing the complex interplay between redox regulation, ER stress, and mitochondrial dynamics in neuroinflammation and cell death management.
Collapse
Affiliation(s)
- Mei-Hua Jin
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Xiao-Dong Liu
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Hu-Nan Sun
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Ying-Hao Han
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup-si, Jeonbuk, 56216, Republic of Korea.
- Department of Applied Biological Engineering, KRIBB School of Biotechnology, Korea National University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
12
|
Gao L, Peng L, Wang J, Zhang JH, Xia Y. Mitochondrial stress: a key role of neuroinflammation in stroke. J Neuroinflammation 2024; 21:44. [PMID: 38321473 PMCID: PMC10845693 DOI: 10.1186/s12974-024-03033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/27/2024] [Indexed: 02/08/2024] Open
Abstract
Stroke is a clinical syndrome characterized by an acute, focal neurological deficit, primarily caused by the occlusion or rupture of cerebral blood vessels. In stroke, neuroinflammation emerges as a pivotal event contributing to neuronal cell death. The occurrence and progression of neuroinflammation entail intricate processes, prominently featuring mitochondrial dysfunction and adaptive responses. Mitochondria, a double membrane-bound organelle are recognized as the "energy workshop" of the body. Brain is particularly vulnerable to mitochondrial disturbances due to its high energy demands from mitochondria-related energy production. The interplay between mitochondria and neuroinflammation plays a significant role in the pathogenesis of stroke. The biological and pathological consequences resulting from mitochondrial stress have substantial implications for cerebral function. Mitochondrial stress serves as an adaptive mechanism aimed at mitigating the stress induced by the import of misfolded proteins, which occurs in response to stroke. This adaptive response involves a reduction in misfolded protein accumulation and overall protein synthesis. The influence of mitochondrial stress on the pathological state of stroke is underscored by its capacity to interact with neuroinflammation. The impact of mitochondrial stress on neuroinflammation varies according to its severity. Moderate mitochondrial stress can bolster cellular adaptive defenses, enabling cells to better withstand detrimental stressors. In contrast, sustained and excessive mitochondrial stress detrimentally affects cellular and tissue integrity. The relationship between neuroinflammation and mitochondrial stress depends on the degree of mitochondrial stress present. Understanding its role in stroke pathogenesis is instrumental in excavating the novel treatment of stroke. This review aims to provide the evaluation of the cross-talk between mitochondrial stress and neuroinflammation within the context of stroke. We aim to reveal how mitochondrial stress affects neuroinflammation environment in stroke.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Li Peng
- Department of Ophthalmology, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
| | - Jian Wang
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA.
- Department of Neurosurgery and Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, 92354, USA.
| | - Ying Xia
- Department of Neurosurgery, Xiangya School of Medicine, Affiliated Haikou Hospital, Central South University, Haikou, 570208, China.
| |
Collapse
|
13
|
Pan Q, Chen C, Yang YJ. Top Five Stories of the Cellular Landscape and Therapies of Atherosclerosis: Current Knowledge and Future Perspectives. Curr Med Sci 2024; 44:1-27. [PMID: 38057537 DOI: 10.1007/s11596-023-2818-2] [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: 07/25/2023] [Accepted: 10/22/2023] [Indexed: 12/08/2023]
Abstract
Atherosclerosis (AS) is characterized by impairment and apoptosis of endothelial cells, continuous systemic and focal inflammation and dysfunction of vascular smooth muscle cells, which is documented as the traditional cellular paradigm. However, the mechanisms appear much more complicated than we thought since a bulk of studies on efferocytosis, transdifferentiation and novel cell death forms such as ferroptosis, pyroptosis, and extracellular trap were reported. Discovery of novel pathological cellular landscapes provides a large number of therapeutic targets. On the other side, the unsatisfactory therapeutic effects of current treatment with lipid-lowering drugs as the cornerstone also restricts the efforts to reduce global AS burden. Stem cell- or nanoparticle-based strategies spurred a lot of attention due to the attractive therapeutic effects and minimized adverse effects. Given the complexity of pathological changes of AS, attempts to develop an almighty medicine based on single mechanisms could be theoretically challenging. In this review, the top stories in the cellular landscapes during the initiation and progression of AS and the therapies were summarized in an integrated perspective to facilitate efforts to develop a multi-targets strategy and fill the gap between mechanism research and clinical translation. The future challenges and improvements were also discussed.
Collapse
Affiliation(s)
- Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Cheng Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
14
|
Li M, Shi X, Wu Y, Qi B, Zhang C, Wang B, Zhang B, Xu Y. Pmepa1 knockdown alleviates SpA-induced pyroptosis and osteogenic differentiation inhibition of hBMSCs via p38MAPK/NLRP3 axis. Int Immunopharmacol 2023; 124:110843. [PMID: 37634444 DOI: 10.1016/j.intimp.2023.110843] [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: 04/07/2023] [Revised: 08/12/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Osteomyelitis is a refractory bone infectious disease, which usually results in progressive bone destruction and bone loss. The invasion of pathogens and subsequent inflammatory response could damage bone marrow mesenchymal stem cells (BMSCs) and inhibit osteogenic differentiation, and finally aggravate uncontrolled bone remodeling in osteomyelitis by affecting bone formation. Exploring the mechanisms of BMSCs injury and osteogenic differentiation inhibition may would help us to find potential therapeutic targets. METHOD Firstly, staphylococcal protein A (SpA)-treated human bone marrow mesenchymal stem cells (hBMSCs) were used to construct cell models of osteomyelitis. Secondly, transcriptome sequencing was performed to screen differentially expressed genes and then verified the expression of target genes. Next, in vitro experiments were conducted to explore the functions and mechanisms of prostate transmembrane protein androgen induced 1 (Pmepa1) in SpA-treated hBMSCs. Finally, the rat model of osteomyelitis was established to provide an auxiliary validation of the in vitro experimental results. RESULTS We found that SpA treatment induced inflammatory injury and inhibited osteogenic differentiation in hBMSCs, then the transcriptome sequencing and further detection results showed that Pmepa1 was significantly upregulated in this process. Functionally, Pmepa1 knockdown alleviated inflammatory injury and promoted osteogenic differentiation in SpA-treated hBMSCs. Among them, it was demonstrated that Pmepa1 knockdown exerted cytoprotective effects by alleviating pyroptosis of SpA-infected hBMSCs. Furthermore, recovery experiments revealed that Pmepa1 knockdown reversed SpA-mediated adverse effects by downregulating the p38MAPK/NLRP3 axis. Finally, the detection results of rat femoral osteomyelitis showed that the expression of Pmepa1 was up-regulated, and the expression trends of other indicators including p38MAPK, NLRP3, and caspase-1 were also consistent with the in vitro model. CONCLUSION Pmepa1 knockdown alleviates SpA-induced pyroptosis and inhibition of osteogenic differentiation in hBMSCs by downregulating p38MAPK/NLRP3 signaling axis. Modulating the expression of Pmepa1 may be a potential strategy to ameliorate osteomyelitis.
Collapse
Affiliation(s)
- Mingjun Li
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Xiangwen Shi
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yipeng Wu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Baochuang Qi
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Chaoqun Zhang
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bin Wang
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bihuan Zhang
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yongqing Xu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force, Kunming, China.
| |
Collapse
|
15
|
Xie Y, He W, Ma L, Ren R, Yang S, Lu Q. Endothelial TREM-1 receptor regulates the blood-brain barrier integrity after intracerebral hemorrhage in mice via SYK/β-catenin signaling. CNS Neurosci Ther 2023; 29:3228-3238. [PMID: 37170484 PMCID: PMC10580358 DOI: 10.1111/cns.14255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a high mortality and disability stroke subtype. Destruction of the blood-brain barrier (BBB) is a crucial contributor to brain edema and neurological deficit after ICH. Triggering receptor expressed on myeloid cells 1 (TREM-1) has been reported to be expressed in endothelial cells, but its role in ICH remains unclear. This study aims to evaluate the role of TREM-1 on BBB permeability after ICH in mice. METHODS Two hundred and forty-two CD1 mice were used in this study. The ICH model was established by collagenase injection. LP17 was administered intranasally at 2 or 8 h after ICH to inhibit TREM-1. To explore the underlying mechanism, SYK activation CRISPR was administered intracerebroventricularly with LP17, and Anti-mouse TREM-1 rat IgG2a (a specific TREM-1 agonist) was injected intracerebroventricularly with R406 (a specific SYK inhibitor) intraperitoneally. Neurobehavioral outcome, brain water content, BBB permeability, and protein expression were evaluated. RESULTS The expression level of the TREM-1 receptor increased rapidly as early as 6 h after ICH, and it was mainly expressed on the endotheliocytes in the neurovascular unit. Early and delayed administration of LP17 significantly decreased brain edema and improved neurobehavioral outcomes at 24 h after ICH. LP17 reduced the BBB permeability by increasing β-catenin, claudin-5 and ZO-1 expression. Furthermore, SYK activation CRISPR abolished the beneficial effect of LP17 on the expression of the above junction molecules. Meanwhile, R406 reversed the impact of the TREM-1 activator on the downregulation of β-catenin, claudin-5 and ZO-1 expression. CONCLUSIONS This study demonstrated that TREM-1 deteriorated BBB permeability via modulating the expression of interendothelial junction molecules after ICH, and this regulation is partly mediated by the SYK/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yonglin Xie
- Department of Emergency, Sir Run Run Shaw HospitalZhejiang University, School of MedicineHangzhouChina
| | - Wei He
- Department of Pharmacy, Second Affiliated HospitalZhejiang University, School of MedicineHangzhouChina
| | - Li Ma
- Department of Neurosurgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Reng Ren
- Department of Neurointensive Care Unit, The Second Affiliated HospitalZhejiang University, School of MedicineHangzhouChina
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Qin Lu
- Department of Neurosurgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
16
|
Meng S, Cao H, Huang Y, Shi Z, Li J, Wang Y, Zhang Y, Chen S, Shi H, Gao Y. ASK1-K716R reduces neuroinflammation and white matter injury via preserving blood-brain barrier integrity after traumatic brain injury. J Neuroinflammation 2023; 20:244. [PMID: 37875988 PMCID: PMC10594934 DOI: 10.1186/s12974-023-02923-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/05/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a significant worldwide public health concern that necessitates attention. Apoptosis signal-regulating kinase 1 (ASK1), a key player in various central nervous system (CNS) diseases, has garnered interest for its potential neuroprotective effects against ischemic stroke and epilepsy when deleted. Nonetheless, the specific impact of ASK1 on TBI and its underlying mechanisms remain elusive. Notably, mutation of ATP-binding sites, such as lysine residues, can lead to catalytic inactivation of ASK1. To address these knowledge gaps, we generated transgenic mice harboring a site-specific mutant ASK1 Map3k5-e (K716R), enabling us to assess its effects and elucidate potential underlying mechanisms following TBI. METHODS We employed the CRIPR/Cas9 system to generate a transgenic mouse model carrying the ASK1-K716R mutation, aming to investigate the functional implications of this specific mutant. The controlled cortical impact method was utilized to induce TBI. Expression and distribution of ASK1 were detected through Western blotting and immunofluorescence staining, respectively. The ASK1 kinase activity after TBI was detected by a specific ASK1 kinase activity kit. Cerebral microvessels were isolated by gradient centrifugation using dextran. Immunofluorescence staining was performed to evaluate blood-brain barrier (BBB) damage. BBB ultrastructure was visualized using transmission electron microscopy, while the expression levels of endothelial tight junction proteins and ASK1 signaling pathway proteins was detected by Western blotting. To investigate TBI-induced neuroinflammation, we conducted immunofluorescence staining, quantitative real-time polymerase chain reaction (qRT-PCR) and flow cytometry analyses. Additionally, immunofluorescence staining and electrophysiological compound action potentials were conducted to evaluate gray and white matter injury. Finally, sensorimotor function and cognitive function were assessed by a battery of behavioral tests. RESULTS The activity of ASK1-K716R was significantly decreased following TBI. Western blotting confirmed that ASK1-K716R effectively inhibited the phosphorylation of ASK1, JNKs, and p38 in response to TBI. Additionally, ASK1-K716R demonstrated a protective function in maintaining BBB integrity by suppressing ASK1/JNKs activity in endothelial cells, thereby reducing the degradation of tight junction proteins following TBI. Besides, ASK1-K716R effectively suppressed the infiltration of peripheral immune cells into the brain parenchyma, decreased the number of proinflammatory-like microglia/macrophages, increased the number of anti-inflammatory-like microglia/macrophages, and downregulated expression of several proinflammatory factors. Furthermore, ASK1-K716R attenuated white matter injury and improved the nerve conduction function of both myelinated and unmyelinated fibers after TBI. Finally, our findings demonstrated that ASK1-K716R exhibited favorable long-term functional and histological outcomes in the aftermath of TBI. CONCLUSION ASK1-K716R preserves BBB integrity by inhibiting ASK1/JNKs pathway in endothelial cells, consequently reducing the degradation of tight junction proteins. Additionally, it alleviates early neuroinflammation by inhibiting the infiltration of peripheral immune cells into the brain parenchyma and modulating the polarization of microglia/macrophages. These beneficial effects of ASK1-K716R subsequently result in a reduction in white matter injury and promote the long-term recovery of neurological function following TBI.
Collapse
Affiliation(s)
- Shan Meng
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Hui Cao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ziyu Shi
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yana Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yue Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shuning Chen
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Hong Shi
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Cai W, Wu Z, Lai J, Yao J, Zeng Y, Fang Z, Lin W, Chen J, Xu C, Chen X. LDC7559 inhibits microglial activation and GSDMD-dependent pyroptosis after subarachnoid hemorrhage. Front Immunol 2023; 14:1117310. [PMID: 37063846 PMCID: PMC10090682 DOI: 10.3389/fimmu.2023.1117310] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/03/2023] [Indexed: 03/31/2023] Open
Abstract
Mounting evidence indicates that inhibition of microglial activation and neuronal pyroptosis plays important roles in brain function recovery after subarachnoid hemorrhage (SAH). LDC7559 is a newly discovered gasdermin D (GSDMD) inhibitor. Previous studies have demonstrated that LDC7559 could inhibit microglial proliferation and pyroptosis. However, the beneficial effects of LDC7559 on SAH remain obscure. Based on this background, we investigated the potential role and the mechanism of LDC7559 on SAH-induced brain damage both in vivo and in vitro. The findings revealed that microglial activation and neuronal pyroptosis were evidently increased after SAH, which could be markedly suppressed by LDC7559 both in vivo and in vitro. Meanwhile, LDC7559 treatment reduced neuronal apoptosis and improved behavior function. Mechanistically, LDC7559 decreased the levels of GSDMD and cleaved GSDMD after SAH. In contrast, nod-like receptor pyrin domain-containing 3 (NLRP3) inflammasome activation by nigericin increased GSDMD-mediated pyroptosis and abated the beneficial effects of LDC7559 on SAH-induced brain damage. However, LDC7559 treatment did not significantly affect the expression of NLRP3 after SAH. Taken together, LDC7559 might suppress neuronal pyroptosis and microglial activation after SAH by inhibiting GSDMD, thereby promoting brain functional recovery.
Collapse
Affiliation(s)
- Wenhua Cai
- Department of Neurosurgery, The Jinjiang Municipal Hospital, Quanzhou, Fujian, China
| | - Zhe Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jinqing Lai
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jieran Yao
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yile Zeng
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhongning Fang
- Department of Neurosurgery, The Jinjiang Municipal Hospital, Quanzhou, Fujian, China
| | - Weibin Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Junyan Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Chaoyang Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- *Correspondence: Xiangrong Chen, ; Chaoyang Xu,
| | - Xiangrong Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- *Correspondence: Xiangrong Chen, ; Chaoyang Xu,
| |
Collapse
|
18
|
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: 6] [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
|
19
|
Wu Y, Sun Y, Wang X, Zhu C. The Regulated Cell Death and Potential Interventions in Preterm Infants after Intracerebral Hemorrhage. Curr Neuropharmacol 2023; 21:1488-1503. [PMID: 36397619 PMCID: PMC10472811 DOI: 10.2174/1570159x21666221117155209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/21/2022] Open
Abstract
Intracerebral hemorrhage (ICH) in preterm infants is one of the major co-morbidities of preterm birth and is associated with long-term neurodevelopmental deficits. There are currently no widely accepted treatments to prevent ICH or therapies for the neurological sequelae. With studies broadening the scope of cell death, the newly defined concept of regulated cell death has enriched our understanding of the underlying mechanisms of secondary brain injury after ICH and has suggested potential interventions in preterm infants. In this review, we will summarize the current evidence for regulated cell death pathways in preterm infants after ICH, including apoptosis, necroptosis, pyroptosis, ferroptosis, autophagy, and PANoptosis as well as several potential intervention strategies that may protect the immature brain from secondary injury after ICH through regulating regulated cell death.
Collapse
Affiliation(s)
- Yanan Wu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
| | - Yanyan Sun
- Department of Human Anatomy, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
20
|
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: 0.5] [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
|
21
|
Wang J, Lu S, Yuan Y, Huang L, Bian M, Yu J, Zou J, Jiang L, Meng D, Zhang J. Inhibition of Schwann Cell Pyroptosis Promotes Nerve Regeneration in Peripheral Nerve Injury in Rats. Mediators Inflamm 2023; 2023:9721375. [PMID: 37144237 PMCID: PMC10154099 DOI: 10.1155/2023/9721375] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/14/2022] [Accepted: 03/24/2023] [Indexed: 05/06/2023] Open
Abstract
Background Peripheral nerve injury (PNI) is one of the most debilitating injuries, but therapies for PNI are still far from satisfactory. Pyroptosis, a recently identified form of cell death, has been demonstrated to participate in different diseases. However, the role of pyroptosis of Schwann cells in PNI remains unclear. Methods We established a rat PNI model, and western blotting, transmission electron microscopy, and immunofluorescence staining were used to confirm pyroptosis of Schwann cells in PNI in vivo. In vitro, pyroptosis of Schwann cells was induced by lipopolysaccharides (LPS)+adenosine triphosphate disodium (ATP). An irreversible inhibitor of pyroptosis, acetyl (Ac)-Tyr-Val-Ala-Asp-chloromethyl ketone (Ac-YVAD-cmk), was used to attenuate Schwann cell pyroptosis. Moreover, the influence of pyroptotic Schwann cells on the function of dorsal root ganglion neurons (DRGns) was analyzed by a coculture system. Finally, the rat PNI model was intraperitoneally treated with Ac-YVAD-cmk to observe the effect of pyroptosis on nerve regeneration and motor function. Results Schwann cell pyroptosis was notably observed in the injured sciatic nerve. LPS+ATP treatment effectively induced Schwann cell pyroptosis, which was largely attenuated by Ac-YVAD-cmk. Additionally, pyroptotic Schwann cells inhibited the function of DRGns by secreting inflammatory factors. A decrease in pyroptosis in Schwann cells promoted regeneration of the sciatic nerve and recovery of motor function in rats. Conclusion Given the role of Schwann cell pyroptosis in PNI progression, inhibition of Schwann cell pyroptosis might be a potential therapeutic strategy for PNI in the future.
Collapse
Affiliation(s)
- Jiayi Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ya Yuan
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Huang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieqin Yu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiapeng Zou
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dehua Meng
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Cao Y, Shi M, Liu L, Zuo Y, Jia H, Min X, Liu X, Chen Z, Zhou Y, Li S, Yang G, Liu X, Deng Q, Chen F, Chen X, Zhang S, Zhang J. Inhibition of neutrophil extracellular trap formation attenuates NLRP1-dependent neuronal pyroptosis via STING/IRE1α pathway after traumatic brain injury in mice. Front Immunol 2023; 14:1125759. [PMID: 37143681 PMCID: PMC10152368 DOI: 10.3389/fimmu.2023.1125759] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction Increased neutrophil extracellular trap (NET) formation has been reported to be associated with cerebrovascular dysfunction and neurological deficits in traumatic brain injury (TBI). However, the biological function and underlying mechanisms of NETs in TBI-induced neuronal cell death are not yet fully understood. Methods First, brain tissue and peripheral blood samples of TBI patients were collected, and NETs infiltration in TBI patients was detected by immunofluorescence staining and Western blot. Then, a controlled cortical impact device was used to model brain trauma in mice, and Anti-Ly6G, DNase, and CL-amidine were given to reduce the formation of neutrophilic or NETs in TBI mice to evaluate neuronal death and neurological function. Finally, the pathway changes of neuronal pyroptosis induced by NETs after TBI were investigated by administration of peptidylarginine deiminase 4 (a key enzyme of NET formation) adenovirus and inositol-requiring enzyme-1 alpha (IRE1α) inhibitors in TBI mice. Results We detected that both peripheral circulating biomarkers of NETs and local NETs infiltration in the brain tissue were significantly increased and had positive correlations with worse intracranial pressure (ICP) and neurological dysfunction in TBI patients. Furthermore, the depletion of neutrophils effectively reduced the formation of NET in mice subjected to TBI. we found that degradation of NETs or inhibition of NET formation significantly inhibited nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 1 (NLRP1) inflammasome-mediated neuronal pyroptosis after TBI, whereas these inhibitory effects were abolished by cyclic GMP-AMP (cGAMP), an activator of stimulating Interferon genes (STING). Moreover, overexpression of PAD4 in the cortex by adenoviruses could aggravate NLRP1-mediated neuronal pyroptosis and neurological deficits after TBI, whereas these pro-pyroptotic effects were rescued in mice also receiving STING antagonists. Finally, IRE1α activation was significantly upregulated after TBI, and NET formation or STING activation was found to promote this process. Notably, IRE1α inhibitor administration significantly abrogated NETs-induced NLRP1 inflammasome-mediated neuronal pyroptosis in TBI mice. Discussion Our findings indicated that NETs could contribute to TBI-induced neurological deficits and neuronal death by promoting NLRP1-mediated neuronal pyroptosis. Suppression of the STING/ IRE1α signaling pathway can ameliorate NETs-induced neuronal pyroptotic death after TBI.
Collapse
Affiliation(s)
- Yiyao Cao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Mingming Shi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Liang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Yan Zuo
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Haoran Jia
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Xiaobin Min
- Baodi Clinical College, Tianjin Medical University, Tianjin, China
| | - Xilei Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Zhijuan Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Yuan Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Shenghui Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Guili Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Xiao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Quanjun Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Fanglian Chen
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
- *Correspondence: Jianning Zhang, ; Xin Chen, ; Shu Zhang,
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
- *Correspondence: Jianning Zhang, ; Xin Chen, ; Shu Zhang,
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin, China
- *Correspondence: Jianning Zhang, ; Xin Chen, ; Shu Zhang,
| |
Collapse
|
23
|
Gu L, Sun M, Li R, Tao Y, Luo X, Xu J, Wu X, Xie Z. Activation of RKIP Binding ASC Attenuates Neuronal Pyroptosis and Brain Injury via Caspase-1/GSDMD Signaling Pathway After Intracerebral Hemorrhage in Mice. Transl Stroke Res 2022; 13:1037-1054. [PMID: 35355228 DOI: 10.1007/s12975-022-01009-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/23/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
Abstract
Pyroptosis has been proven to be responsible for secondary brain injury after intracerebral hemorrhage (ICH). A recent study reported that Raf kinase inhibitor protein (RKIP) inhibited assembly and activation of inflammasome in macrophages. Our present study aimed to investigate the effects of RKIP on inflammasome-mediated neuronal pyroptosis and underlying neuroprotective mechanisms in experimental ICH. Here, we showed that RKIP expression was decreased both in cerebrospinal fluid (CSF) samples from patients with ICH and in the peri-hematoma tissues after experimental ICH. In mouse ICH model, activation of RKIP remarkably improved neurological deficits, reduced brain water content and BBB disruption, and promoted hematoma absorption at 24 h after ICH, as well as alleviated neuronal degeneration, reduced membrane pore formation, and downregulated pyroptotic molecules NLRP3, caspase-1 P20, GSDMD-N, and mature IL-1β. Besides, RKIP activation decreased the number of caspase-1 P20-positive neurons after ICH. However, RKIP inhibitor reserved the neuroprotective effects of RKIP at 24 h following ICH. Moreover, RKIP could bind with ASC, then interrupt the assembly of NLRP3 inflammasome. Mechanistically, inhibiting the caspase-1 by VX-765 attenuated brain injury and suppressed neuronal pyroptosis after RKIP inhibitor-pretreated ICH. In conclusion, our findings indicated that activation of RKIP could attenuate neuronal pyroptosis and brain injury after ICH, to some extent, through ASC/Caspase-1/GSDMD pathway. Thus, RKIP may be a potential target to attenuate brain injury via its anti-pyroptosis effect after ICH.
Collapse
Affiliation(s)
- Lingui Gu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, China
| | - Mingjiang Sun
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, China
| | - Ruihao Li
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, China
| | - Xu Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, China
| | - Jing Xu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, China
| | - Xuan Wu
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, China.
| |
Collapse
|
24
|
Lu J, Tang X, Zhang D, Lan T, Huang Q, Xu P, Liu M, Liu L, Wang J. Didang Tang inhibits intracerebral hemorrhage-induced neuronal injury via ASK1/MKK7/JNK signaling pathway, network pharmacology-based analyses combined with experimental validation. Heliyon 2022; 8:e11407. [PMID: 36387497 PMCID: PMC9647469 DOI: 10.1016/j.heliyon.2022.e11407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is an acute cerebrovascular disease, which is also a principal consideration for disability. Didang tang (DDT) is a classic traditional Chinese medicine formula for treating ICH. However, its pharmacological mechanism of action has not been elucidated. Materials and methods The TCMSP and BATMAN-TCM databases were used to collect chemical compounds and predict targets of DDT. Protein targets in ICH were identified by GeneCards, OMIM, and DrugBank databases. DDT compounds-ICH targets and protein-protein interaction (PPI) networks were constructed for topological analysis and hub-targets screening. Further, Key biological processes and signaling pathways were identified by GO and KEGG enrichment analyses. Then, an ICH rat model and a Cobaltous Chloride (CoCl2)-induced PC12 cells model were established. Cell viability and lactate dehydrogenase (LDH) release were detected using cck8 and LDH kits. Apoptosis levels were detected by TUNEL assessment and flow cytometry. IL-1β levels were detected by ELISA, while key protein expressions were determined by Western blot. Results A total of 126 active compounds related to DDT and 3,263 therapeutic targets for ICH were predicted. The functional enrichment of the GO and KEGG pathways combined with literature studies suggested that DDT is most likely to influence MAPK and apoptotic signaling pathways for ICH treatment. In vitro and in vivo experiments have shown that DDT remarkably inhibited apoptosis and increased the expression of Bcl-2, while inhibiting Bax and cleaved-Caspase 3. For other enriched core proteins, DDT suppressed the phosphorylation of Src and the expression of c-Myc and IL-1β, and up-regulated the level of MMP-9. The further results showed that, DDT decreased the phosphorylation of ASK1, MKK7, JNK and c-JUN. Conclusion Based on network pharmacology and experimental validation results, our in vivo and in vitro study indicated that ASK1/MKK7/JNK pathway might be the critical target for DDT against ICH.
Collapse
|
25
|
Wang M, Zhang Y, Chang W, Zhang L, Syrigos KN, Li P. Noncoding RNA-mediated regulation of pyroptotic cell death in cancer. Front Oncol 2022; 12:1015587. [PMID: 36387211 PMCID: PMC9659888 DOI: 10.3389/fonc.2022.1015587] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/18/2022] [Indexed: 07/30/2023] Open
Abstract
Pyroptosis is a newly discovered form of programmed cell death, which is manifested by DNA fragmentation, cell swelling, cell membrane rupture and leakage of cell contents. Previous studies have demonstrated that pyroptosis is tightly associated with the initiation and development of various cancers, whereas the molecular mechanisms underlying pyroptosis remain obscure. Noncoding RNAs (ncRNAs) are a type of heterogeneous transcripts that are broadly expressed in mammalian cells. Owing to their potency of regulating gene expression, ncRNAs play essential roles in physiological and pathological processes. NcRNAs are increasingly acknowledged as important regulators of the pyroptosis process. Importantly, the crosstalk between ncRNAs and pyroptosis affects various hallmarks of cancer, including cell growth, survival, metastasis and therapeutic resistance. The study of the involvement of pyroptosis-associated ncRNAs in cancer pathobiology has become a hot area in recent years, while there are limited reviews on this topic. Herein, we provide an overview of the complicated roles of ncRNAs, mainly including microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), in modulating pyroptosis, with a focus on the underlying mechanisms of the ncRNA-pyroptosis axis in cancer pathogenesis. Finally, we discuss the potential applications and challenges of exploiting pyroptosis-regulating ncRNAs as molecular biomarkers and therapeutic targets in cancer.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Konstantinos N. Syrigos
- Third Department of Internal Medicine and Laboratory, National & Kapodistrian University of Athens, Athens, Greece
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Gu L, Sun M, Li R, Tao Y, Luo X, Zhang X, Yuan Y, Xie Z. Microglial pyroptosis: Therapeutic target in secondary brain injury following intracerebral hemorrhage. Front Cell Neurosci 2022; 16:971469. [PMID: 36159393 PMCID: PMC9507402 DOI: 10.3389/fncel.2022.971469] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a major cerebrovascular illness that causes substantial neurological sequelae and dysfunction caused by secondary brain injury (SBI), and there are no effective therapies to mitigate the disability. Microglia, the brain-resident macrophage, participates in the primary inflammatory response, and activation of microglia to an M1-like phenotype largely takes place in the acute phase following ICH. A growing body of research suggests that the pathophysiology of SBI after ICH is mediated by an inflammatory response mediated by microglial-pyroptotic inflammasomes, while inhibiting the activation of microglial pyroptosis could suppress the inflammatory cascade reaction, thus attenuating the brain injury after ICH. Pyroptosis is characterized by rapid plasma membrane disruption, followed by the release of cellular contents and pro-inflammatory mediators. In this review, we outline the molecular mechanism of microglial pyroptosis and summarize the up-to-date evidence of its involvement in the pathological process of ICH, and highlight microglial pyroptosis-targeted strategies that have the potential to cure intracerebral hemorrhage. This review contributes to a better understanding of the function of microglial pyroptosis in ICH and assesses it as a possible therapeutic target.
Collapse
|
27
|
Song D, Yeh CT, Wang J, Guo F. Perspectives on the mechanism of pyroptosis after intracerebral hemorrhage. Front Immunol 2022; 13:989503. [PMID: 36131917 PMCID: PMC9484305 DOI: 10.3389/fimmu.2022.989503] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/17/2022] [Indexed: 12/18/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a highly harmful neurological disorder with high rates of mortality, disability, and recurrence. However, effective therapies are not currently available. Secondary immune injury and cell death are the leading causes of brain injury and a poor prognosis. Pyroptosis is a recently discovered form of programmed cell death that differs from apoptosis and necrosis and is mediated by gasdermin proteins. Pyroptosis is caused by multiple pathways that eventually form pores in the cell membrane, facilitating the release of inflammatory substances and causing the cell to rupture and die. Pyroptosis occurs in neurons, glial cells, and endothelial cells after ICH. Furthermore, pyroptosis causes cell death and releases inflammatory factors such as interleukin (IL)-1β and IL-18, leading to a secondary immune-inflammatory response and further brain damage. The NOD-like receptor protein 3 (NLRP3)/caspase-1/gasdermin D (GSDMD) pathway plays the most critical role in pyroptosis after ICH. Pyroptosis can be inhibited by directly targeting NLRP3 or its upstream molecules, or directly interfering with caspase-1 expression and GSDMD formation, thus significantly improving the prognosis of ICH. The present review discusses key pathological pathways and regulatory mechanisms of pyroptosis after ICH and suggests possible intervention strategies to mitigate pyroptosis and brain dysfunction after ICH.
Collapse
Affiliation(s)
- Dengpan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| |
Collapse
|
28
|
Zhang H, Wang J, Ruan C, Gao Z, Zhu Q, Li S. Co-exposure of chronic stress and alumina nanoparticles aggravates hippocampal microglia pyroptosis by activating cathepsin B/NLRP3 signaling pathway. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129093. [PMID: 35569374 DOI: 10.1016/j.jhazmat.2022.129093] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/26/2022] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Combined exposure of chronic stress and alumina nanoparticles (AlNPs) aggravates hippocampal injury, but the pathogenesis is unevaluated. This study aimed to investigate the effect and mechanism of co-exposure to chronic stress and AlNPs on hippocampal microglia pyroptosis. In this study, chronic restraint stress (CRS) alone caused NLRP3-mediated hippocampal microglia pyroptosis, but AlNPs did not. Moreover, co-exposure to CRS and AlNPs exacerbated hippocampal microglia pyroptosis, resulting in more severe hippocampal damage and behavioral deficits in rats. Protein-protein interaction network predicted that cathepsin B was a potential regulatory protein of NLRP3. CRS up-regulated cathepsin B expression which had a more pronounced increase in co-exposure group. Whereas, caspase-1 inhibitor VX-765 alleviated hippocampal microglia pyroptosis and behavioral deficits in rats. Consistent with in vivo results, co-exposure of corticosterone and AlNPs aggravated NLRP3-mediated pyroptosis and cathepsin B expression in HAPI cells. Nevertheless, the pyroptosis of HAPI cells was inhibited by cathepsin B inhibitor CA-074Me and NLRP3 knockout, respectively. NLRP3 agonist nigericin failed to promote the pyroptosis of HAPI cells in the presence of cathepsin B inhibition. These results demonstrated that co-exposure to chronic stress and AlNPs could aggravate hippocampal microglia pyroptosis by activating cathepsin B/NLRP3 signaling pathway, resulting in hippocampal damage and behavioral deficits.
Collapse
Affiliation(s)
- Haiyang Zhang
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangdong Technological Engineering Research Center for Pets, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, People's Republic of China.
| | - Jibin Wang
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangdong Technological Engineering Research Center for Pets, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, People's Republic of China
| | - Chuqian Ruan
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangdong Technological Engineering Research Center for Pets, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, People's Republic of China
| | - Zhicheng Gao
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangdong Technological Engineering Research Center for Pets, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, People's Republic of China
| | - Qiuxiang Zhu
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangdong Technological Engineering Research Center for Pets, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, People's Republic of China
| | - Shoujun Li
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangdong Technological Engineering Research Center for Pets, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, People's Republic of China.
| |
Collapse
|
29
|
Jin P, Qi D, Cui Y, Lenahan C, Zhang JH, Tao X, Deng S, Tang J. Aprepitant attenuates NLRC4-dependent neuronal pyroptosis via NK1R/PKCδ pathway in a mouse model of intracerebral hemorrhage. J Neuroinflammation 2022; 19:198. [PMID: 35922848 PMCID: PMC9351153 DOI: 10.1186/s12974-022-02558-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 07/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pyroptosis is a programmed cell death mediated by inflammasomes. Previous studies have reported that inhibition of neurokinin receptor 1 (NK1R) exerted neuroprotection in several neurological diseases. Herein, we have investigated the role of NK1R receptor inhibition using Aprepitant to attenuate NLRC4-dependent neuronal pyroptosis after intracerebral hemorrhage (ICH), as well as the underlying mechanism. METHODS A total of 182 CD-1 mice were used. ICH was induced by injection of autologous blood into the right basal ganglia. Aprepitant, a selective antagonist of NK1R, was injected intraperitoneally at 1 h after ICH. To explore the underlying mechanism, NK1R agonist, GR73632, and protein kinase C delta (PKCδ) agonist, phorbol 12-myristate 13-acetate (PMA), were injected intracerebroventricularly at 1 h after ICH induction, and small interfering ribonucleic acid (siRNA) for NLRC4 was administered via intracerebroventricular injection at 48 h before ICH induction, respectively. Neurobehavioral tests, western blot, and immunofluorescence staining were performed. RESULTS The expression of endogenous NK1R and NLRC 4 were gradually increased after ICH. NK1R was expressed on neurons. Aprepitant significantly improved the short- and long-term neurobehavioral deficits after ICH, which was accompanied with decreased neuronal pyroptosis, as well as decreased expression of NLRC4, Cleaved-caspase-1, GSDMD (gasdermin D), IL-1β, and IL-18. Activation of NK1R or PKCδ abolished these neuroprotective effects of Aprepitant after ICH. Similarly, knocking down NLRC4 using siRNA produced similar neuroprotective effects. CONCLUSION Aprepitant suppressed NLRC4-dependent neuronal pyroptosis and improved neurological function, possibly mediated by inhibition of NK1R/PKCδ signaling pathways after ICH. The NK1R may be a promising therapeutic target for the treatment of ICH.
Collapse
Affiliation(s)
- Peng Jin
- Department of Intensive Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.,Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - Dongqing Qi
- Department of Rehabilitation Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yuhui Cui
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92354, USA.,Department of Neurosurgery, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200040, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM, 88001, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92354, USA.,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA.,Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Xiaogen Tao
- Department of Intensive Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Shuixiang Deng
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92354, USA. .,Department of Intensive Care Unit, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai, 200040, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92354, USA.
| |
Collapse
|
30
|
Yan WT, Yang YD, Hu XM, Ning WY, Liao LS, Lu S, Zhao WJ, Zhang Q, Xiong K. Do pyroptosis, apoptosis, and necroptosis (PANoptosis) exist in cerebral ischemia? Evidence from cell and rodent studies. Neural Regen Res 2022; 17:1761-1768. [PMID: 35017436 PMCID: PMC8820688 DOI: 10.4103/1673-5374.331539] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/15/2021] [Accepted: 11/01/2021] [Indexed: 11/04/2022] Open
Abstract
Some scholars have recently developed the concept of PANoptosis in the study of infectious diseases where pyroptosis, apoptosis and necroptosis act in consort in a multimeric protein complex, PANoptosome. This allows all the components of PANoptosis to be regulated simultaneously. PANoptosis provides a new way to study the regulation of cell death, in that different types of cell death may be regulated at the same time. To test whether PANoptosis exists in diseases other than infectious diseases, we chose cerebral ischemia/reperfusion injury as the research model, collected articles researching cerebral ischemia/reperfusion from three major databases, obtained the original research data from these articles by bibliometrics, data mining and other methods, then integrated and analyzed these data. We selected papers that investigated at least two of the components of PANoptosis to check its occurrence in ischemia/reperfusion. In the cell model simulating ischemic brain injury, pyroptosis, apoptosis and necroptosis occur together and this phenomenon exists widely in different passage cell lines or primary neurons. Pyroptosis, apoptosis and necroptosis also occurred in rat and mouse models of ischemia/reperfusion injury. This confirms that PANoptosis is observed in ischemic brain injury and indicates that PANoptosis can be a target in the regulation of various central nervous system diseases.
Collapse
Affiliation(s)
- Wei-Tao Yan
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Yan-Di Yang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Xi-Min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wen-Ya Ning
- Department of Human Resources, Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Lyu-Shuang Liao
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Shuang Lu
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Wen-Juan Zhao
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Qi Zhang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Kun Xiong
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan Province, China
| |
Collapse
|
31
|
Liu Y, Tao X, Tao J. Strategies of Targeting Inflammasome in the Treatment of Systemic Lupus Erythematosus. Front Immunol 2022; 13:894847. [PMID: 35664004 PMCID: PMC9157639 DOI: 10.3389/fimmu.2022.894847] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple organ dysfunction resulting from the production of multiple autoantibodies and adaptive immune system abnormalities involving T and B lymphocytes. In recent years, inflammasomes have been recognized as an important component of innate immunity and have attracted increasing attention because of their pathogenic role in SLE. In short, inflammasomes regulate the abnormal differentiation of immune cells, modulate pathogenic autoantibodies, and participate in organ damage. However, due to the clinical heterogeneity of SLE, the pathogenic roles of inflammasomes are variable, and thus, the efficacy of inflammasome-targeting therapies is uncertain. To provide a foundation for the development of such therapeutic strategies, in this paper, we review the role of different inflammasomes in the pathogenesis of SLE and their correlation with clinical phenotypes and propose some corresponding treatment strategies.
Collapse
Affiliation(s)
- Yaling Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinyu Tao
- Department of Clinical Medicine "5 + 3" Integration, The First Clinical College, Anhui Medical University, Hefei, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
32
|
Zhang LQ, Gao SJ, Sun J, Li DY, Wu JY, Song FH, Liu DQ, Zhou YQ, Mei W. DKK3 ameliorates neuropathic pain via inhibiting ASK-1/JNK/p-38-mediated microglia polarization and neuroinflammation. J Neuroinflammation 2022; 19:129. [PMID: 35658977 PMCID: PMC9164405 DOI: 10.1186/s12974-022-02495-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/23/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Neuropathic pain is a common and severely disabling state that affects millions of people worldwide. Microglial activation in the spinal cord plays a critical role in the pathogenesis of neuropathic pain. However, the mechanisms underlying spinal microglial activation during neuropathic pain remain incompletely understood. Here, we investigated the role of Dickkopf (DKK) 3 and its interplay with microglial activation in the spinal cord in neuropathic pain. METHODS In this study, we investigated the effects of intrathecal injection of recombinant DKK3 (rDKK3) on mechanical allodynia and microglial activation in the spinal cord after spared nerve injury (SNI) in rats by western blot (WB), immunofluorescence (IF), quantitative polymerase chain reaction (qPCR), and enzyme-linked immunosorbent assay (ELISA). RESULTS We found that SNI induced a significant decrease in the levels of DKK3, Kremen-1 and Dishevelled-1 (DVL-1) and up-regulated the expression of phosphorylated apoptosis signal-regulating kinase 1 (p-ASK1), phosphorylated c-JUN N-terminal kinase (p-JNK), phosphorylated p38 (p-p38) in the spinal cord. Moreover, our results showed that exogenous intrathecal administration of rDKK3 inhibited expression of p-ASK1, p-JNK, p-p38, promoted the transformation of microglia from M1 type to M2 type, and decreased the production of pro-inflammatory cytokines compared to the rats of SNI + Vehicle. However, these effects were reversed by intrathecal administration of Kremen-1 siRNA or Dishevelled-1 (DVL-1) siRNA. CONCLUSIONS These results suggest that DKK3 ameliorates neuropathic pain via inhibiting ASK-1/JNK/p-38-mediated microglia polarization and neuroinflammation, at least partly, by the Kremen-1 and DVL-1 pathways.
Collapse
Affiliation(s)
- Long-Qing Zhang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Shao-Jie Gao
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Jia Sun
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Dan-Yang Li
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Jia-Yi Wu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Fan-He Song
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Dai-Qiang Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
| | - Wei Mei
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji MedicalCollege, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
33
|
Zhang J, Li S, Yang Z, Liu C, Chen X, Zhang Y, Zhang F, Shi H, Chen X, Tao L, Shan H, Zhang M. Implantation of injectable SF hydrogel with sustained hydrogen sulfide delivery reduces neuronal pyroptosis and enhances functional recovery after severe intracerebral hemorrhage. BIOMATERIALS ADVANCES 2022; 135:212743. [PMID: 35929216 DOI: 10.1016/j.bioadv.2022.212743] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/15/2022] [Accepted: 02/27/2022] [Indexed: 06/15/2023]
Abstract
Hydrogen sulfide (H2S), an important endogenous signaling molecule, plays an important neuroprotective role in the central nervous system. However, there is no ideal delivery material or method involving the sustained and controlled release of H2S for clinical application in brain diseases. Silk fibroin (SF)-based hydrogels have become a potentially promising strategy for local, controlled, sustained drug release in the treatment of various disorders. Here, we show a silk fibroin (SF)-based hydrogel with sustained H2S delivery (H2S@SF hydrogel) is effective in treating brain injury through stereotactic orthotopic injection in a severe intracerebral hemorrhage (ICH) mouse model. In this study, we observed H2S@SF hydrogel sustained H2S release in vitro and in vivo. The physicochemical properties of H2S@SF hydrogel were studied using FE-SEM, Raman spectroscopy and Rheological analysis. Treatment with H2S@SF hydrogel attenuated brain edema, reduced hemorrhage volume and improved the recovery of neurological deficits after severe ICH following stereotactic orthotopic injection. Double immunofluorescent staining also revealed that H2S@SF hydrogel may reduce cell pyroptosis in the striatum, cortex and hippocampus. However, when using endogenous H2S production inhibitor AOAA, H2S@SF hydrogel could not suppress ICH-induced cell pyroptosis. Hence, the therapeutic effect of the H2S@SF hydrogel may be partly the result of the slow-release of H2S and/or the effect of the SF hydrogel on the production of endogenous H2S. Altogether, the results exhibit promising attributes of injectable silk fibroin hydrogel and the utility of H2S-loaded injectable SF hydrogel as an alternative biomaterial toward brain injury treatment for clinical application.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Institute of Forensic Sciences, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Sunao Li
- Institute of Forensic Sciences, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Zhenbei Yang
- Department of Textile Engineering, College of Textile and Clothing Engineering, Soochow University, Suzhou 215001, China
| | - Chao Liu
- Institute of Forensic Sciences, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Xueshi Chen
- Institute of Forensic Sciences, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Feng Zhang
- Department of Textile Engineering, College of Textile and Clothing Engineering, Soochow University, Suzhou 215001, China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xiping Chen
- Institute of Forensic Sciences, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Luyang Tao
- Institute of Forensic Sciences, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China.
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215008, China.
| | - Mingyang Zhang
- Institute of Forensic Sciences, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
34
|
A new cell death program regulated by toll-like receptor 9 through p38 mitogen-activated protein kinase signaling pathway in a neonatal rat model with sepsis associated encephalopathy. Chin Med J (Engl) 2022; 135:1474-1485. [PMID: 35261352 PMCID: PMC9481440 DOI: 10.1097/cm9.0000000000002010] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: Sepsis, a serious condition with high mortality, usually causes sepsis associated encephalopathy (SAE) that involves neuronal cell death. However, the cell death programs involved and their underlying mechanisms are not clear. This study aimed to explore the regulatory mechanisms of different cell death programs in SAE. Methods: A neonatal rat model of SAE was established by cecal ligation and perforation. Survival rate and vital signs (mean arterial pressure and heart rate) were monitored, nerve reflexes were evaluated, and cortical pathological changes were observed by hematoxylin and eosin staining. The expression of pyroptosis, apoptosis, and necroptosis (PANoptosis)-related proteins, mitogen- activated protein kinase (MAPK), and its upstream regulator toll-like receptor 9 (TLR9) were detected. The expression of TLR9 in neurons was observed by immunofluorescence staining. The ultrastructure of neurons was observed by transmission electron microscope. Results: First, PANoptosis was found in cortical nerve cells of the SAE rats. Meanwhile, the subunits of MAPKs, p38 MAPK, Jun N- terminal kinase, and extracellular signal-regulated kinase (ERK) were activated. After pharmacologically inhibiting each of the subunits, only p38 MAPK was found to be associated with PANoptosis. Furthermore, blocking the p38 MAPK signaling pathway activated necroptosis but inhibited apoptosis and pyroptosis. When necroptosis was pharmacologically inhibited, apoptosis and pyroptosis were reactivated. Finally, we found that the expression of TLR9, a regulator of MAPKs, was significantly increased in this model. After down-regulation of TLR9, p38 MAPK, and ERK signaling pathways were inhibited, which led to the inhibition of PANoptosis. Further analysis found that down-regulation of TLR9 improved the survival rate and reduced the pathological changes in SAE rats. Conclusions: Our study showed that the programs comprising PANoptosis are activated simultaneously in SAE rats. TLR9 activated PANoptosis through the p38 MAPK signaling pathway. TLR9 may work as a potential target for SAE treatment.
Collapse
|
35
|
Interleukin-13 Affects the Recovery Processes in a Mouse Model of Hemorrhagic Stroke with Bilateral Tibial Fracture. Mol Neurobiol 2022; 59:3040-3051. [PMID: 35258849 DOI: 10.1007/s12035-021-02650-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/16/2021] [Indexed: 10/18/2022]
Abstract
As one form of stroke, intracerebral hemorrhage (ICH) is a fatal cerebrovascular disease, which has high morbidity and mortality and lacks effective medical treatment. Increased infiltration of inflammatory cytokines coupled with pyroptotic cell death is involved in the pathophysiological process of ICH. However, little is known about whether concomitant fracture patients have the same progression of inflammation and pyroptosis. Hence, we respectively established the mouse ICH model and ICH with bilateral tibial fracture model (MI) to explore the potential cross-talk between the above two injuries. We found that MI obviously reversed the expressions of pyroptosis-associated proteins, which were remarkably up-regulated at the acute phase after ICH. Similar results were observed in neuronal expressions via double immunostaining. Furthermore, brain edema was also significantly alleviated in mice who suffered MI, when compared with ICH alone. To better clarify the potential mechanisms that mediated this cross-talk, recombinant mouse interleukin-13 (IL-13) was used to investigate its effect on pyroptosis in the mouse MI model, in which a lower level of IL-13 was observed. Remarkably, IL-13 administration re-awakened cell death, which was mirrored by the re-upregulation of pyroptosis-associated proteins and PI-positive cell counts. The results of hemorrhage volume and behavioral tests further confirmed its critical role in regulating neurological functions. Besides, the IL-13-treated MI group showed poor outcomes of fracture healing. To sum up, our research indicates that controlling the IL-13 content in the acute phase would be a promising target in influencing the outcomes of brain injury and fracture, and meanwhile, provides new evidence in repairing compound injuries in clinics.
Collapse
|
36
|
Yao X, Song Y, Wang Z, Bai S, Yu H, Wang Y, Guan Y. Proteinase-activated receptor-1 antagonist attenuates brain injury via regulation of FGL2 and TLR4 after intracerebral hemorrhage in mice. Neuroscience 2022; 490:193-205. [PMID: 35182700 DOI: 10.1016/j.neuroscience.2022.02.012] [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: 09/28/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 11/25/2022]
Abstract
Proteinase-activated receptor-1 (PAR1) antagonist plays a protective effect in brain injury. We investigated the potential function and mechanisms of PAR1 antagonist in ICH-induced brain injury. Results showed that PAR1 antagonist protected against neurobehavior deficits, brain edema and BBB integrity in ICH mice via activating JNK/ERK/p38 MAPK signaling pathway at 24h after ICH. In addition, ICH resulted in the increase of FGL2 and TLR4 expression over time, and phosphorylated JNK, ERK and p38 MAPK expression. Suppression of FGL2 and TLR4 alleviated brain injury and decreased the expression of p-JNK, p-ERK, p-p38 MAPK and p-IKKα at 24 h after ICH; while overexpression of them showed the opposite result. Moreover, the protective effect of PAR1 antagonist on ICH-induced brain injury was blocked by FGL2 or TLR4 overexpression, and the levels of p-JNK, p-ERK and p-p38 MAPK were inhibited. Furthermore, PAR1 antagonist combined with TLR4 antagonist markedly alleviated brain injury after ICH at 72h. Overall, PAR1 antagonist protected against short-term brain injury, and the effect of PAR1 antagonist on ICH-induced brain injury was mediated by FGL2 or TLR4.
Collapse
Affiliation(s)
- Xiaoying Yao
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yaying Song
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ze Wang
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuwei Bai
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Haojun Yu
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yishu Wang
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yangtai Guan
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
37
|
Zhang Y, Khan S, Liu Y, Zhang R, Li H, Wu G, Tang Z, Xue M, Yong VW. Modes of Brain Cell Death Following Intracerebral Hemorrhage. Front Cell Neurosci 2022; 16:799753. [PMID: 35185473 PMCID: PMC8851202 DOI: 10.3389/fncel.2022.799753] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high rates of mortality and morbidity. It induces cell death that is responsible for neurological deficits postinjury. There are no therapies that effectively mitigate cell death to treat ICH. This review aims to summarize our knowledge of ICH-induced cell death with a focus on apoptosis and necrosis. We also discuss the involvement of ICH in recently described modes of cell death including necroptosis, pyroptosis, ferroptosis, autophagy, and parthanatos. We summarize treatment strategies to mitigate brain injury based on particular cell death pathways after ICH.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Ruiyi Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Hongmin Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhouping Tang
- Department of Neurology, Affiliated Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
- *Correspondence: Mengzhou Xue,
| | - V. Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- V. Wee Yong,
| |
Collapse
|
38
|
He Q, Qu M, Xu C, Shi W, Hussain M, Jin G, Zhu H, Zeng LH, Wu X. The emerging roles of nitric oxide in ferroptosis and pyroptosis of tumor cells. Life Sci 2021; 290:120257. [PMID: 34952041 DOI: 10.1016/j.lfs.2021.120257] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/06/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Tumor cells can develop resistance to cell death which is divided into necrosis and programmed cell death (PCD). PCD, including apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis. Ferroptosis and pyroptosis, two new forms of cell death, have gradually been of interest to researchers. Boosting ferroptosis and pyroptosis of tumor cells could be a potential cancer therapy. Nitric oxide (NO) is a ubiquitous, lipophilic, highly diffusible, free-radical signaling molecule that plays various roles in tumorigenesis. In addition, NO also has regulatory mechanisms through S-nitrosylation that do not depend on the classic NO/sGC/cGMP signaling. The current tumor treatment strategy for NO is to promote cell death through promoting S-nitrosylation-induced apoptosis while multiple drawbacks dampen this tumor therapy. However, numerous studies have suggested that suppression of NO is perceived to active ferroptosis and pyroptosis, which could be a better anti-tumor treatment. In this review, ferroptosis and pyroptosis are described in detail. We summarize that NO influences ferroptosis and pyroptosis and infer that S-nitrosylation mediates ferroptosis- and pyroptosis-related signaling pathways. It could be a potential cancer therapy different from NO-induced apoptosis of tumor cells. Finally, the information shows the drugs that manipulate endogenous production and exogenous delivery of NO to modulate the levels of S-nitrosylation.
Collapse
Affiliation(s)
- Qiangqiang He
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China
| | - Meiyu Qu
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Shi
- Department of Biology and Genetics, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Musaddique Hussain
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Guojian Jin
- Department of Internal Medicine, Shaoxing Central Hospital Anchang Branch, Shaoxing City 312080, China
| | - Haibin Zhu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
| | - Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
39
|
Rh-CXCL-12 Attenuates Neuronal Pyroptosis after Subarachnoid Hemorrhage in Rats via Regulating the CXCR4/NLRP1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6966394. [PMID: 34795842 PMCID: PMC8595028 DOI: 10.1155/2021/6966394] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022]
Abstract
Subarachnoid hemorrhage (SAH) is a cerebrovascular disease associated with high morbidity and mortality. CXCR4 provides neuroprotective effects, which can alleviate brain injury and inflammation induced by stroke. Previous studies have suggested that CXCR4 reduces the pyroptosis of LPS-stimulated BV2 cells. The purpose of this study was to evaluate the antipyroptosis effects and mechanisms of CXCR4 after SAH. SAH animal model was induced via endovascular perforation. A total of 136 male Sprague-Dawley rats were used. Recombinant human cysteine-X-cysteine chemokine ligand 12 (rh-CXCL-12) was administered intranasally at 1 h after SAH induction. To investigate the underlying mechanism, the inhibitor of CXCR4, AMD3100, was administered intraperitoneally at 1 h before SAH. The neurobehavior tests were assessed, followed by performing Western blot and immunofluorescence staining. The Western blot results suggested that the expressions of endogenous CXCL-12, CXCR4, and NLRP1 were increased and peaked at 24 h following SAH. Immunofluorescence staining showed that CXCR4 was expressed on neurons, microglia, and astrocytes. Rh-CXCL-12 treatment improved the neurological deficits and reduced the number of FJC-positive cells, IL-18-positive neurons, and cleaved caspase-1(CC-1)-positive neurons after SAH. Meanwhile, rh-CXCL-12 treatment increased the levels of CXCL-12 and CXCR4, and reduced the levels of NLRP1, IL-18, IL-1β, and CC-1. Moreover, the administration of AMD3100 abolished antipyroptosis effects of CXCL-12 and its regulation of CXCR4 post-SAH. The CXCR4/NLRP1 signaling pathway may be involved in CXCL-12-mediated neuronal pyroptosis after SAH. Early administration of CXCL-12 may be a preventive and therapeutic strategy against brain injury after SAH.
Collapse
|
40
|
Jung KH, Seong SY. Role of inflammasomes in neuroinflammation after ischemic stroke. ENCEPHALITIS 2021; 1:89-97. [PMID: 37470048 PMCID: PMC10295893 DOI: 10.47936/encephalitis.2021.00073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 07/21/2023] Open
Abstract
Ischemic stroke is a devastating disease for which there is no effective medical treatment. In the era of extensive reperfusion strategies, established neuroprotectant candidates and novel therapeutic drugs with better targets are promising for treatment of acute ischemic stroke. Such targets include the inflammasome pathway, which contributes significantly to the pathogenesis of ischemic stroke. Following ischemic stroke, damage-associated molecular patterns from damaged cells activate inflammasomes, incur inflammatory responses, and induce cell death. Therefore, inhibiting inflammasome pathways has great promise for treatment of ischemic stroke. However, the efficacy and safety of inflammasome inhibitors remain controversial, and better upstream targets are needed for effective modulation. Herein, the roles of the inflammasome in ischemic injury caused by stroke are reviewed and the potential of neuroprotectants targeting the inflammasome is discussed.
Collapse
Affiliation(s)
- Keun-Hwa Jung
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Yong Seong
- Department of Microbiology and Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
Zeng P, Wang XM, Su HF, Zhang T, Ning LN, Shi Y, Yang SS, Lin L, Tian Q. Protective effects of Da-cheng-qi decoction in rats with intracerebral hemorrhage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153630. [PMID: 34217968 DOI: 10.1016/j.phymed.2021.153630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/15/2021] [Accepted: 06/10/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH), the most fatal subtype of stroke, has no disease-modifying treatment. Da-cheng-qi decoction (DCQ), composed of rhubarb, is one of the most commonly used Chinese traditional decoctions in ICH treatment. But the mechanism is not clear. Emodin is an active compound found in rhubarb. PURPOSE To study the protective effects of DCQ on ICH and its possible mechanisms of action. METHODS The ICH model was reproduced by injecting collagenase-VII into the left caudate putamen (CPu) of rats. DCQ and emodin were used to treat the ICH rats for 7 days. Behavior tests, proteomic analysis, morphological studies, and western blotting were performed. RESULTS The neurological deficits in the ICH rats recovered with DCQ and emodin on the 14th day after ICH. The proteomics data revealed that DCQ significantly corrected the pathological signals in the CPu and hippocampus after ICH. The numbers of amoebic microglia in the CPu and M2 microglia in both CPu and hippocampus were significantly increased after DCQ and emodin treatment. The increase in GluN2B-containing NMDA receptor (NR2B) and postsynaptic density protein-95, activation of mitogen-activated protein kinase (MAPK) signals in the CPu, and secondary neurodegeneration (SND) in the hippocampus were significantly recovered in DCQ-treated rats. Inhibition of MAPK p38 (p38) in the hippocampus was observed after DCQ and emodin treatment. CONCLUSION The protective effects of DCQ on ICH were confirmed in this study, and its mechanism may be related to the inhibition of MAPK and activation of M2 microglia. These results are beneficial to the development of ICH therapeutic targets.
Collapse
Affiliation(s)
- Peng Zeng
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Ming Wang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong-Fei Su
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Teng Zhang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lin-Na Ning
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Shi
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shu-Sheng Yang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Traditional Chinese Medicine, Wuhan Red Cross Hospital, Wuhan 430015, China.
| | - Li Lin
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China; Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
42
|
Jin P, Deng S, Sherchan P, Cui Y, Huang L, Li G, Lian L, Xie S, Lenahan C, Travis ZD, Zhang JH, Gong Y, Tang J. Neurokinin Receptor 1 (NK1R) Antagonist Aprepitant Enhances Hematoma Clearance by Regulating Microglial Polarization via PKC/p38MAPK/NFκB Pathway After Experimental Intracerebral Hemorrhage in Mice. Neurotherapeutics 2021; 18:1922-1938. [PMID: 34244927 PMCID: PMC8608951 DOI: 10.1007/s13311-021-01077-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 02/04/2023] Open
Abstract
Hematoma clearance is an important therapeutic target to improve outcome following intracerebral hemorrhage (ICH). Recent studies showed that Neurokinin receptor-1 (NK1R) inhibition exerts protective effects in various neurological disease models, but its role in ICH has not been explored. The objective of this study was to investigate the role of NK1R and its relation to hematoma clearance after ICH using an autologous blood injection mouse model. A total of 332 adult male CD1 mice were used. We found that the expression levels of NK1R and its endogenous ligand, substance P (SP), were significantly upregulated after ICH. Intraperitoneal administration of the NK1R selective antagonist, Aprepitant, significantly improved neurobehavior, reduced hematoma volume and hemoglobin levels after ICH, and promoted microglia polarization towards M2 phenotype. Aprepitant decreased phosphorylated PKC, p38MAPK, and NFκB p65, and downregulated M1 markers while upregulating M2 markers after ICH. Intracerebroventricular administration of the NK1R agonist, GR73632 or PKC agonist, phorbol 12-myristate 13-acetate (PMA) reversed the effects of Aprepitant. To demonstrate the upstream mediator of NK1R activation, we performed thrombin injection and found that it increased SP. Inhibiting thrombin suppressed SP and decreased M1 markers while increasing M2 microglia polarization. Thus, NK1R inhibition promoted hematoma clearance after ICH by increasing M2 microglial polarization via downregulating PKC/p38MAPK/NFκB signaling pathway, and thrombin may be a key upstream mediator of NK1R activation. Therapeutic interventions inhibiting NK1R signaling may be a new target for the treatment of ICH.
Collapse
Affiliation(s)
- Peng Jin
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Shuixiang Deng
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Yuhui Cui
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Gaigai Li
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Lifei Lian
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Shucai Xie
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
- Burrell College of Osteopathic Medicine, Las Cruces, NM, 88001, USA
| | - Zachary D Travis
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Ye Gong
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA.
| |
Collapse
|
43
|
Liu T, Li X, Cui Y, Meng P, Zeng G, Wang Y, Wang Q. Bioinformatics Analysis Identifies Potential Ferroptosis Key Genes in the Pathogenesis of Intracerebral Hemorrhage. Front Neurosci 2021; 15:661663. [PMID: 34163322 PMCID: PMC8215678 DOI: 10.3389/fnins.2021.661663] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a dangerous neurological disease. The mechanism of ferroptosis in ICH remains unclear. Using bioinformatics analysis, we aimed to identify the key molecules involved in ferroptosis and provide treatment targets for ICH to further explore the mechanism of ferroptosis in ICH. GSE24265 was downloaded from the Gene Expression Omnibus (GEO) dataset and intersected with ferroptosis genes. A total of 45 differentially expressed genes (DEGs) were selected, most of which were involved in the TNF signaling pathway and oxidative stress response. Key modules constructed by the protein–protein interaction (PPI) network analysis and screening of genes related to the TNF signaling pathway led to the confirmation of the following genes of interest: MAPK1, MAPK8, TNFAIP3, ATF4, and SLC2A1. Moreover, MAPK1 was one of the key genes related to TNF signaling and oxidative stress, and it may play an important role in ferroptosis after cerebral hemorrhage. The MAPK1-related molecules included hsa-miR-15b-5P, hsa-miR-93-5P, miR-20b-5p, SNHG16, XIST, AC084219.4, RP11-379K17.11, CTC-444N24.11, GS1-358P8.4, CTB-89H12.4, RP4-773N10.5, and FGD5-AS1. We also generated a hemorrhage rat model, which was used to conduct exercise intervention in ICH rats, and qRT-PCR was used to assess the expression levels of our genes of interest. The mRNA levels after cerebral hemorrhage showed that MAPK1, ATF4, SLC2A1, and TNFAIP3 were upregulated, whereas MAPK8 was downregulated. Treadmill training increased the expression of anti-inflammatory molecules TNFAIP3 and SLC2A1 and reduced the expression of MAPK1, ATF4, and MAPK8, indicating that treadmill training may be utilized as antioxidant therapy to decrease neuronal ferroptosis. The results of this study indicated that the MAPK1-related mRNA–miRNA–lncRNA interaction chain could be potentially employed as a biomarker of the inception and progression of ferroptosis after cerebral hemorrhage.
Collapse
Affiliation(s)
- Tongye Liu
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinhe Li
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yiteng Cui
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pingping Meng
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guanghui Zeng
- Department of Sports Medicine, Qingdao University Medical College, Qingdao, China
| | - Yuyang Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiang Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
44
|
Zhang H, Jiao W, Cui H, Sun Q, Fan H. Combined exposure of alumina nanoparticles and chronic stress exacerbates hippocampal neuronal ferroptosis via activating IFN-γ/ASK1/JNK signaling pathway in rats. JOURNAL OF HAZARDOUS MATERIALS 2021; 411:125179. [PMID: 33858114 DOI: 10.1016/j.jhazmat.2021.125179] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/23/2020] [Accepted: 01/17/2021] [Indexed: 06/12/2023]
Abstract
Alumina nanoparticles (AlNPs) exposure causes hippocampal-dependent cognitive dysfunction. However, whether chronic stress exacerbates AlNPs-induced hippocampal lesion and its mechanism remains unclear. This study was aimed to investigate the combined effects and mechanisms of AlNPs and chronic stress on the hippocampal lesion. The behavioral tests demonstrated that combined exposure to AlNPs and chronic restraint stress (CRS) worsened both cognition and depression-like behavior than exposed to AlNPs and CRS alone. Microstructural and ultrastructural observations showed that combined exposure to AlNPs and CRS exacerbated hippocampal damage. Both AlNPs and CRS induced hippocampal neuronal ferroptosis, presenting as iron and glutamate metabolism disorder, GPX4 fluorescence of neurons decrease, LPO and ROS levels increase, and FJB-positive neurons increase. Meanwhile, combined exposure to AlNPs and CRS exacerbated hippocampal neuronal ferroptosis. Mechanism investigation revealed that combined exposure to AlNPs and CRS activated IFN-γ/ASK1/JNK signaling pathway. Furthermore, IFN-γ neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-γ/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. Together, these results demonstrate that combined exposure to AlNPs and CRS exacerbates hippocampal neuronal ferroptosis via activating IFN-γ/ASK1/JNK signaling pathway.
Collapse
Affiliation(s)
- Haiyang Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Wenjing Jiao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hailin Cui
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Qinghong Sun
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
45
|
Zhang CS, Han Q, Song ZW, Jia HY, Shao TP, Chen YP. Hydrogen gas post-conditioning attenuates early neuronal pyroptosis in a rat model of subarachnoid hemorrhage through the mitoK ATP signaling pathway. Exp Ther Med 2021; 22:836. [PMID: 34149882 PMCID: PMC8200808 DOI: 10.3892/etm.2021.10268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 05/12/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal pyroptosis serves an important role in the progress of neurologic dysfunction following subarachnoid hemorrhage (SAH), which is predominantly caused by a ruptured aneurysm. Hydrogen gas has been previously reported to be an effective anti-inflammatory agent against ischemia-associated diseases by regulating mitochondrial function. The objective of the present study was to investigate the potential neuroprotective effects of hydrogen gas post-conditioning against neuronal pyroptosis after SAH, with specific focus on the mitochondrial ATP-sensitive K+ (mitoKATP) channels. Following SAH induction by endovascular perforation, rats were treated with inhalation of 2.9% hydrogen gas for 2 h post-perforation. Neurologic deficits, brain water content, reactive oxygen species (ROS) levels, neuronal pyroptosis, phosphorylation of ERK1/2, p38 MAPK and pyroptosis-associated proteins IL-1β and IL-18 were evaluated 24 h after perforation by a modified Garcia method, ratio of wet/dry weight, 2',7'-dichlorofluorescin diacetate, immunofluorescence and western blot assays, respectively. An inhibitor of the mitoKATP channel, 5-hydroxydecanoate sodium (5-HD), was used to assess the potential role of the mitoKATP-ERK1/2-p38 MAPK signal pathway. Hydrogen gas post-conditioning significantly alleviated brain edema and improved neurologic function, reduced ROS production and neuronal pyroptosis, suppressed the expression of IL-1β and IL-18 whilst upregulating ERK1/2 phosphorylation, but downregulated p38 MAPK activation 24 h post-SAH. These aforementioned effects neuroprotective were partially reversed by 5-HD treatment. Therefore, these observations suggest that post-conditioning with hydrogen gas ameliorated SAH-induced neuronal pyroptosis at least in part through the mitoKATP/ERK1/2/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Chuan-Suo Zhang
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Qian Han
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Zhao-Wei Song
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Hong-Yan Jia
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Tian-Peng Shao
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yan-Peng Chen
- Department of Radioactive Intervention, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
46
|
Tan C, Liu W, Zheng ZH, Wan XG. LncRNA HOTTIP inhibits cell pyroptosis by targeting miR-148a-3p/AKT2 axis in ovarian cancer. Cell Biol Int 2021; 45:1487-1497. [PMID: 33710684 DOI: 10.1002/cbin.11588] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/26/2021] [Accepted: 03/07/2021] [Indexed: 12/28/2022]
Abstract
Long noncoding RNA HOTTIP is a crucial regulator in multiple types of cancer, including ovarian cancer (OC). However, the biological roles and underlying mechanisms of HOTTIP in OC have rarely been studied. Hence, this study aimed to investigate the functional correlation between HOTTIP and pyroptosis in OC progression. The expression of HOTTIP in OC tissues and cell lines was characterized by quantitative real-time PCR. Cell proliferation was evaluated using Cell Counting Kit-8 and clone formation assays. Western blot was performed to quantify protein levels. A dual-luciferase reporter assay was used to analyze the molecular interaction among HOTTIP, miR-148a-3p, and AKT2. The expression of HOTTIP was significantly upregulated in OC tissue samples and cell lines. The silencing of HOTTIP led to the inhibition of cell proliferation and NLRP1 inflammasome-mediated pyroptosis. In addition, HOTTIP increased AKT2 expression by negatively regulating miR-148a-3p and then inhibited ASK1/JNK signaling. Further rescue experiments revealed that downregulation of miR-148a-3p and overexpression of AKT2 obviously diminished the effects of HOTTIP downregulation in OC cells. Thus, our study elucidated a novel pyroptosis-related mechanism by which HOTTIP participated in OC progression, which might provide a theoretical reference for clinical treatment.
Collapse
Affiliation(s)
- Cai Tan
- Department of Health, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, PR China
| | - Wei Liu
- Department of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, PR China
| | - Zhi-Hua Zheng
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Xiao-Gang Wan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| |
Collapse
|
47
|
Gou X, Xu D, Li F, Hou K, Fang W, Li Y. Pyroptosis in stroke-new insights into disease mechanisms and therapeutic strategies. J Physiol Biochem 2021; 77:511-529. [PMID: 33942252 DOI: 10.1007/s13105-021-00817-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
Stroke is a common disease with high mortality and disability worldwide. Different forms of cell deaths, including apoptosis and necrosis, occur in ischemic or hemorrhagic brain tissue, among which pyroptosis, a newly discovered inflammation-related programmed cell death, is generally divided into two main pathways, the canonical inflammasome pathway and the non-canonical inflammasome pathway. Caspase-mediated pyroptosis requires the assembly of inflammasomes such as NLRP3, which leads to the release of inflammatory cytokines IL-1β and IL-18 through the pores formed in the plasma membrane by GSDMD followed by neuroinflammation. Recently, pyroptosis and its relationship with inflammation have attracted more and more attention in the study of cerebral ischemia or hemorrhage. In addition, many inhibitors of pyroptosis targeting caspase, NLRP3, and the upstream pathway have been found to reduce brain tissue damage after stroke. In this review, we mainly introduce the pathology of stroke, the molecular mechanism, and process of pyroptosis, as well as the pivotal roles of pyroptosis in stroke, in order to provide new insights for the treatment of stroke.
Collapse
Affiliation(s)
- Xue Gou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China
| | - Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China.,Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
48
|
Dinparastisaleh R, Mirsaeidi M. Antifibrotic and Anti-Inflammatory Actions of α-Melanocytic Hormone: New Roles for an Old Player. Pharmaceuticals (Basel) 2021; 14:ph14010045. [PMID: 33430064 PMCID: PMC7827684 DOI: 10.3390/ph14010045] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
The melanocortin system encompasses melanocortin peptides, five receptors, and two endogenous antagonists. Besides pigmentary effects generated by α-Melanocytic Hormone (α-MSH), new physiologic roles in sexual activity, exocrine secretion, energy homeostasis, as well as immunomodulatory actions, exerted by melanocortins, have been described recently. Among the most common and burdensome consequences of chronic inflammation is the development of fibrosis. Depending on the regenerative capacity of the affected tissue and the quality of the inflammatory response, the outcome is not always perfect, with the development of some fibrosis. Despite the heterogeneous etiology and clinical presentations, fibrosis in many pathological states follows the same path of activation or migration of fibroblasts, and the differentiation of fibroblasts to myofibroblasts, which produce collagen and α-SMA in fibrosing tissue. The melanocortin agonists might have favorable effects on the trajectories leading from tissue injury to inflammation, from inflammation to fibrosis, and from fibrosis to organ dysfunction. In this review we briefly summarized the data on structure, receptor signaling, and anti-inflammatory and anti-fibrotic properties of α-MSH and proposed that α-MSH analogues might be promising future therapeutic candidates for inflammatory and fibrotic diseases, regarding their favorable safety profile.
Collapse
Affiliation(s)
- Roshan Dinparastisaleh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL 33146, USA
- Correspondence: ; Tel.: +1-305-243-1377
| |
Collapse
|
49
|
Hu X, Yan J, Huang L, Araujo C, Peng J, Gao L, Liu S, Tang J, Zuo G, Zhang JH. INT-777 attenuates NLRP3-ASC inflammasome-mediated neuroinflammation via TGR5/cAMP/PKA signaling pathway after subarachnoid hemorrhage in rats. Brain Behav Immun 2021; 91:587-600. [PMID: 32961266 PMCID: PMC7749833 DOI: 10.1016/j.bbi.2020.09.016] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Inflammasome-mediated neuroinflammation plays an important role in the pathogenesis of early brain injury (EBI) following subarachnoid hemorrhage (SAH). The activation of the TGR5 receptor has been shown to be neuroprotective in a variety of neurological diseases. This study aimed to investigate the effects of the specific synthetic TGR5 agonist, INT-777, in attenuating NLRP3-ASC inflammasome activation and reducing neuroinflammation after SAH. METHODS One hundred and eighty-four male Sprague Dawley rats were used. SAH was induced by the endovascular perforation. INT-777 was administered intranasally at 1 h after SAH induction. To elucidate the signaling pathway involved in the effect of INT-777 on inflammasome activation during EBI, TGR5 knockout CRISPR and PKA inhibitor H89 were administered intracerebroventricularly and intraperitoneally at 48 h and 1 h before SAH. The SAH grade, short- and long-term neurobehavioral assessments, brain water content, western blot, immunofluorescence staining, and Nissl staining were performed. RESULTS The expressions of endogenous TGR5, p-PKA, and NLRP3-ASC inflammasome were increased after SAH. INT-777 administration significantly decreased NLRP3-ASC inflammasome activation in microglia, reduced brain edema and neuroinflammation, leading to improved short-term neurobehavioral functions at 24 h after SAH. The administration of TGR5 CRISPR or PKA inhibitor (H89) abolished the anti-inflammation effects of INT-777, on NLRP3-ASC inflammasome, pro-inflammatory cytokines (IL-6, IL-1β, and TNF-a), and neutrophil infiltration at 24 h after SAH. Moreover, early administration of INT-777 attenuated neuronal degeneration in hippocampus on 28 d after SAH. CONCLUSIONS INT-777 attenuated NLRP3-ASC inflammasome-dependent neuroinflammation in the EBI after SAH, partially via TGR5/cAMP/PKA signaling pathway. Early administration of INT-777 may serve as a potential therapeutic strategy for EBI management in the setting of SAH.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jun Yan
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Camila Araujo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jun Peng
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570000, China
| | - Ling Gao
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570000, China
| | - Shengpeng Liu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Taicang Hospital Affiliated to Soochow University, Taicang, Suzhou, Jiangsu 215400, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA; Department of Anesthesiology, Loma Linda University, Loma Linda, CA 92350, USA.
| |
Collapse
|
50
|
Dinesh P, Kalaiselvan S, Sujitha S, Rasool M. MicroRNA-532-3p Regulates Pro-Inflammatory Human THP-1 Macrophages by Targeting ASK1/p38 MAPK Pathway. Inflammation 2020; 44:229-242. [PMID: 32876895 DOI: 10.1007/s10753-020-01325-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/09/2020] [Accepted: 08/18/2020] [Indexed: 01/14/2023]
Abstract
Inflammation is a complex biological process which alters the normal physiological function of the immune system resulting in an abnormal microenvironment that leads to several clinical complications. The process of inflammation is mediated through various intracellular signaling factors inside the cells. Apoptosis signal-regulating kinase 1 (ASK1) is an inflammation-derived kinase that controls the activation of other family of kinases such as p38 mitogen-activated protein kinases (p38 MAPKs), which mediates various the inflammatory processes. In this study, we cultured THP-1 macrophage cells to undergo inflammatory proliferation with LPS (1 μg/ml) and TNFα (10 ng/ml) stimulation. Initial in silico analysis was utilized to predict novel microRNAs (miRNAs) that target ASK1 signaling and its expression levels in LPS and TNFα stimulated THP-1 cells were estimated. Among the miRNAs, miR-532-3p showcased the highest binding affinity towards ASK1 kinase. We witnessed that transient transfection of miR-532-3p diminished the levels of ASK1 and downstream phosphorylation/translocation of p38 MAPK. Furthermore, direct targeting of ASK1 resulted in regulation of uncontrolled release of cytokines (TNFα, IL-6, and IL-23) and chemokines (GM-CSF and MIP-2α). Overall, we suggest that miR-532-3p attenuates the pro-inflammatory nature of macrophages by targeting ASK1/p38 MAPK signaling pathway and can be used as a molecular intervention for treating inflammatory diseases.
Collapse
Affiliation(s)
- Palani Dinesh
- SMV 240, Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632 014, India
| | - Sowmiya Kalaiselvan
- SMV 240, Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632 014, India
| | - Sali Sujitha
- SMV 240, Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632 014, India
| | - Mahaboobkhan Rasool
- SMV 240, Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632 014, India.
| |
Collapse
|