1
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [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: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
2
|
Zhai Y, Pang X, Mei X, Pang Y, Shu J, Xiao Y, Ma W, Zou M, Yang P, Yue G, Lan D. Shuanglu tongnao formula alleviates cerebral ischemia/reperfusion injury by rebuilding inflammatory microenvironment after cerebral ischemia. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119640. [PMID: 40107474 DOI: 10.1016/j.jep.2025.119640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
ETHNOPHARMACOLOGICAL SIGNIFICANCE Shuanglu tongnao formula (SLTNF) has been clinically proven to have significant efficacy in the treatment of Ischemic stroke (IS), and is a promising formula for IS treatment. Still, the underlying mechanism is not clear. Whether SLTNF ameliorates ischemic brain injury by reversing the pro-inflammatory microenvironment after IS is an interesting field of investigation. AIM OF THE STUDY Based on the result of network pharmacology and single-cell RNA sequencing (scRNA-seq), whether SLTNF mitigates cerebral ischemia/reperfusion (I/R) injury by reversing the pro-inflammatory microenvironment was investigated in vivo for the first time. MATERIALS AND METHODS The mice middle cerebral artery occlusion (MCAO) model was established to induce focal cerebral I/R. Subsequently, the remission effects of SLTNF treatment for cerebral I/R injury were evaluated in the MCAO model. scRNA-seq data was used to analyze the immune microenvironment after IS in mice. scRNA-seq and Network pharmacology were applied to predict the mechanism of the treatment of IS by SLTNF. Western blot (WB) and immunofluorescence techniques were employed to validate the potential mechanism. RESULTS The experimental results demonstrated that SLTNF dosage-dependently attenuated the infarct volume, neurobehavioral, cell morphology and Nissl bodies damage, and inhibited the apoptosis in cerebral I/R mice. Moreover, scRNA-seq results revealed that the number of NK cells, neutrophils, monocytes, astrocytes and microglia significantly increased after IS. The cell-cell interactions dominated by microglia after IS, the cell-cell interactions between microglia and other immune cells significantly heightened. Furthermore, SLTNF promoted the transition of M1 microglia to M2 type, eventually reversing the pro-inflammatory microenvironment. Combined analysis of scRNA-seq and Network pharmacology results predicted that AGE-RAGE signaling pathway could involve in the regulation of microglia polarization by SLTNF. WB results revealed that SLTNF significantly inhibited the protein expression of CCND1, IL-1β and p-STAT3, which belong to crucial targets of SLTNF and AGE-RAGE signaling pathway. CONCLUSION SLTNF attenuated cerebral I/R injury by reversing the pro-inflammatory microenvironment via the AGE-RAGE signaling pathway in mice.
Collapse
Affiliation(s)
- Yang Zhai
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Xingwang Pang
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Xiaoping Mei
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China
| | - Yan Pang
- Department of Emergency, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, 530000, China
| | - Jianlong Shu
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Yuhan Xiao
- Department of Traditional Chinese Medicine, Nanning Seventh People's Hospital, Nanning, 530000, China
| | - Wei Ma
- Neurology Department, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Min Zou
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China
| | - Peng Yang
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China
| | - Guihua Yue
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China.
| | - Dazhi Lan
- Department of International Medical, Guangxi University of Traditional Chinese Medicine Affiliated International Zhuang Medicine Hospital, Nanning, 530000, China.
| |
Collapse
|
3
|
Balde A, Benjakul S, Nazeer RA. A review on NLRP3 inflammasome modulation by animal venom proteins/peptides: mechanisms and therapeutic insights. Inflammopharmacology 2025; 33:1013-1031. [PMID: 39934538 DOI: 10.1007/s10787-025-01656-7] [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: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025]
Abstract
The venom peptides from terrestrial as well as aquatic species have demonstrated potential in regulating the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a sophisticated assemblage present in immune cells responsible for detecting and responding to external mediators. The NLRP3 inflammasome plays a role in several pathological conditions such as type 2 diabetes, hyperglycemia, Alzheimer's disease, obesity, autoimmune disorders, and cardiovascular disorders. Venom peptides derived from animal venoms have been discovered to selectively induce certain signalling pathways, such as the NLRP3 inflammasome, mitogen-activated protein kinase (MAPK), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Experimental evidence has demonstrated that venom peptides can regulate the expression and activation of the NLRP3 inflammasome, resulting in the secretion of pro-inflammatory cytokines including interleukin (IL)-1β and IL-18. Furthermore, these peptides have been discovered to impede the activation of the NLRP3 inflammasome, therefore diminishing inflammation and tissue injury. The functional properties of venom proteins and peptides obtained from snakes, bees, wasps, and scorpions have been thoroughly investigated, specifically targeting the NLRP3 inflammasome pathway, venom proteins and peptides have shown promise as therapeutic agents for the treatment of certain inflammatory disorders. This review discusses the pathophysiology of NLRP3 inflammasome in the onset of various diseases, role of venom as therapeutics. Further, various venom components and their role in the modulation of NLRP3 inflammasome are discoursed. A substantial number of venomous animals and their toxins are yet unexplored, and to comprehensively grasp the mechanisms of action of them and their potential as therapeutic agents, additional research is required which can lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro Industry, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
- Department of Food and Nutrition, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India.
| |
Collapse
|
4
|
Luo Y, Dong W, Yuan L, Zhu YA, Zhang DD, Ni H, Zhu W. The Role of Thrombo-inflammation in Ischemic Stroke: Focus on the Manipulation and Clinical Application. Mol Neurobiol 2025; 62:2362-2375. [PMID: 39107669 DOI: 10.1007/s12035-024-04397-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/22/2024] [Indexed: 01/28/2025]
Abstract
Stroke leaves a great economic burden due to its high morbidity and mortality. Rapid revascularization of targeted vessel(s) is the effective treatment for ischemic stroke, but subsequent ischemia-reperfusion (I/R) injury is a common complication following revascularization, leading to microcirculation dysfunction and infarct volume increase. Thrombo-inflammation, the interaction between thrombosis and inflammation, plays a critical role in the pathophysiology of ischemic stroke. In the context of I/R injury, thrombo-inflammation consists of platelet activation, endothelial injury, and inflammatory cell infiltration. Numerous studies are devoted to exploring methods of regulating thrombo-inflammation to mitigate I/R injury post-stroke, including blocking activations of platelets and neutrophils. Drugs such as antiplatelet medications, anticoagulants, and glucocorticoids have been confirmed to have the potential to regulate thrombo-inflammation. Furthermore, several recently developed drugs have also shown promises in relieving I/R injury by manipulating thrombo-inflammation. However, the majority of these studies are still in the preclinical stage. Herein, in this review, we will address the mechanisms of thrombo-inflammation in ischemic stroke, related research advances, and particularly the clinical feasibility of thrombo-inflammation as a therapeutic strategy against I/R injury.
Collapse
Affiliation(s)
- Yuanfei Luo
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weichen Dong
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Linying Yuan
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yunqing Amelia Zhu
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
| | - Dachuan Dustin Zhang
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, M5G 2M1, Canada
- CCOA Therapeutics Inc., Toronto, ON, M5B 1W8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Department of Medicine, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Wusheng Zhu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
5
|
Santoro ML, Sachetto ATA, Rosa JG, Torquato RJS, Andrade-Silva D, Trevisan-Silva D, de Albuquerque CZ, Serrano SMT, de Moura Mattaraia VG, Tanaka AS, Peichoto ME. Jararaca GPIb-binding protein causes thrombocytopenia during Bothrops jararaca envenomation. Sci Rep 2024; 14:31769. [PMID: 39738271 PMCID: PMC11686094 DOI: 10.1038/s41598-024-81851-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
Inoculation of Bothrops jararaca snake venom (BjV) induces thrombocytopenia in humans and various animal species. Although several BjV toxins acting on hemostasis have been well characterized in vitro, it is not known which one is responsible for inducing thrombocytopenia in vivo. In previous studies, we showed that BjV incubated with metalloproteinase or serine proteinase inhibitors and/or anti-botrocetin antibodies still induced thrombocytopenia in rats and mice. Thus, herein we identified and characterized BjV toxins responsible for inducing thrombocytopenia. Initially, by filtering BjV on ultrafiltration systems, proteins with molecular masses between 30 and 50 kDa were shown to induce thrombocytopenia in mice, but they were not associated with hemorrhagic or coagulating activities. The 50 kDa ultrafiltrate was chromatographed, and two proteins (named fraction D and fraction E) induced thrombocytopenia in mice. However, neither fraction D nor fraction E induced platelet aggregation in platelet-rich plasma or whole blood from humans or mice. By mass spectrometry analysis, fraction E was identified as jararaca glycoprotein Ib (GPIb)-binding protein. Injection of these fractions caused thrombocytopenia in control or Vwf-/- mice, showing that the axis platelet GPIb - von Willebrand factor is not involved in their biological action in vivo. New studies are necessary to understand how these proteins act in vivo.
Collapse
Affiliation(s)
- Marcelo Larami Santoro
- Biotério Central, Instituto Butantan, Av. Dr. Vital Brasil, 1500, São Paulo, SP, 05503 - 900, Brazil.
- Escola Superior do Instituto Butantan, Av. da Universidade, São Paulo, SP, Brazil.
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Ana Teresa Azevedo Sachetto
- Biotério Central, Instituto Butantan, Av. Dr. Vital Brasil, 1500, São Paulo, SP, 05503 - 900, Brazil
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Jaqueline Gomes Rosa
- Biotério Central, Instituto Butantan, Av. Dr. Vital Brasil, 1500, São Paulo, SP, 05503 - 900, Brazil
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ricardo José Soares Torquato
- Departmento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Débora Andrade-Silva
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, SP, Brazil
| | - Dilza Trevisan-Silva
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, SP, Brazil
| | | | - Solange M T Serrano
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, SP, Brazil
| | | | - Aparecida Sadae Tanaka
- Departmento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Maria Elisa Peichoto
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto Nacional de Medicina Tropical (INMeT)-ANLIS "Dr. Carlos G Malbrán", Almafuerte y Ambar s/n, 3370, Puerto Iguazú, Argentina
- Universidade Federal da Integração Latino-Americana (UNILA), Foz do Iguaçu, PR, CEP 85870-901, Brazil
| |
Collapse
|
6
|
Tian X, Zeng Y, Tu Q, Jiao Y, Yao S, Chen Y, Sun L, Xia Q, Luo Y, Yuan L, Jiang Q. Butyrate alleviates renal fibrosis in CKD by regulating NLRP3-mediated pyroptosis via the STING/NF-κB/p65 pathway. Int Immunopharmacol 2023; 124:111010. [PMID: 37852118 DOI: 10.1016/j.intimp.2023.111010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023]
Abstract
Chronic kidney disease (CKD) is a serious and irreversible disease primarily characterized by chronic inflammation and renal fibrosis. Recent studies have suggested that gut microbiota-related metabolites, particularly short-chain fatty acids (SCFAs) are significantly associated with kidney diseases. Notably, butyrate, a type of SCFAs, plays a crucial role in this correlation. However, the effect of butyrate on renal fibrosis in patients with CKD and its potential mechanisms remain unclear. In this study, we demonstrated that butyrate levels are reduced as CKD progresses using a CKD C57BL/6 mouse model established by a 0.2% adenine diet. Exogenous supplementation of butyrate effectively alleviated renal fibrosis and repressed the levels of proteins associated with NLRP3-mediated pyroptosis (NLRP3, IL-1β, caspase-1, and GSDMD). Additionally, we conducted an in vitro experiment using HK-2 cells, which also confirmed that the elevated levels of NLRP3-mediated pyroptosis proteins in TGF-β1-stimulated HK-2 cells are reversed by butyrate intervention. Further, butyrate mitigated the activity of the STING/NF-κB/p65 pathway, and STING overexpression impaired the protective function of butyrate in CKD. Hence, we suggest that butyrate may have a renoprotective role in CKD, alleviating renal fibrosis possibly by regulating NLRP3-mediated pyroptosis via the STING/NF-κB/p65 pathway.
Collapse
Affiliation(s)
- Xiaofang Tian
- Medical College of Soochow University, 215123 Suzhou, Jiangsu, China; The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Yizhou Zeng
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Qingxian Tu
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Yang Jiao
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Song Yao
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Ying Chen
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Li Sun
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Qianhang Xia
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Yadan Luo
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Liying Yuan
- The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China
| | - Qianfeng Jiang
- Medical College of Soochow University, 215123 Suzhou, Jiangsu, China; The First People's Hospital of Zunyi (the Third Affiliated Hospital of Zunyi Medical University), 563000 Zunyi, Guizhou, China; Guizhou Aerospace Hospital, 563000 Zunyi, Guizhou, China.
| |
Collapse
|
7
|
Luo P, Ji Y, Liu X, Zhang W, Cheng R, Zhang S, Qian X, Huang C. Affected inflammation-related signaling pathways in snake envenomation: A recent insight. Toxicon 2023; 234:107288. [PMID: 37703930 DOI: 10.1016/j.toxicon.2023.107288] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
Snake envenomation is well known to cause grievous pathological signs, including haemorrhagic discharge, necrosis, and respiratory distress. However, inflammatory reactions are also common envenoming manifestations that lead to successive damage, such as oedema, ulceration, lymphadenectasis, systemic inflammatory response syndrome (SIRS) and even multiple organ dysfunction syndrome (MODS). Interference with the inflammatory burst is hence important in the clinical treatment of snake envenomation. Here, we summarize the typical snake toxins (or venoms) that cause inflammatory reactions and the underlying signaling pathways. In brief, inflammatory reactions are usually triggered by snake venom phospholipase A2 (svPLA2), snake venom metalloprotease (SVMP), snake venom serine protease (SVSP) and C-type lectin/snaclec (CTL) as well as disintegrin (DIS) via multiple signaling pathways. They are nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3), nuclear factor kappa-B (NF-κB), mitogen-activated protein kinase (MAPK), janus kinase/signal transducer and activator of transcription (JAK-STAT) and phosphoinositide 3-Kinase/protein kinase B (PI3K/PKB also called PI3K-AKT) signaling pathways. Activation of these pathways promotes the expression of pro-inflammatory molecules such as cytokines, especially interleukin-1β (IL-1β) which causes further inflammatory cascades and manifestations, such as swelling, fever, pain, and severe complications. Remarkably, almost half of introduced snake toxins (or venoms) have anti-inflammatory effects through blocking these pathways and suppressing the expression of pro-inflammatory molecules. Investigation of affected inflammation-related signaling pathways is meaningful to achieve better clinical treatment.
Collapse
Affiliation(s)
- Peiyi Luo
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Yuxin Ji
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Xiaohan Liu
- Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China.
| | - Weiyun Zhang
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Ruoxi Cheng
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Shuxian Zhang
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Xiao Qian
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Chunhong Huang
- College of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| |
Collapse
|
8
|
Chen J, Liu X, Bi R, Liu P, Gong W. NDUFC2 deficiency exacerbates endothelial mesenchymal transformation during ischemia-reperfusion via NLRP3. Neuroreport 2023; 34:670-676. [PMID: 37506315 DOI: 10.1097/wnr.0000000000001940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Ischemic stroke is the main type of cerebrovascular disease. Emergency thrombectomy combined with medication therapy is currently the primary treatment for stroke. Inflammation and oxidative stress induced by ischemia-reperfusion cause secondary damage to blood vessels, especially endothelial mesenchymal transformation (EndoMT). However, much is still unclear about the role of EndoMT in ischemia-reperfusion. In this study, an in vivo ischemia-reperfusion model was established by transient middle cerebral artery occlusion (tMCAO) in wild-type (WT) C57BL/6 mice and NLRP3 (NOD-like receptor thermal protein domain associated protein 3) knockout (KO) C57BL/6 mice. An in vitro ischemia-reperfusion model was established by oxygen glucose deprivation and reoxygenation (OGD/R) of human brain microvascular endothelial cells (HBMECs). α-SMA (alpha smooth muscle actin), CD31 (platelet endothelial cell adhesion molecule-1, PECAM-1/CD31), NDUFC2 (NADH: ubiquinone oxidoreductase subunit C2), and NLRP3 were used to evaluate EndoMT and inflammation. Real-time PCR measured superoxide dismutase 1 (SOD1) and catalase (CAT) mRNA expression to evaluate oxidative stress levels. NLRP3 was activated by ischemia-reperfusion injury and NLRP3 inactivation inhibited the EndoMT in tMCAO mice. Further experiments demonstrated that OGD/R treatment induced NLRP3 activation and EndoMT in HBMECs, which resulted in NDUFC2 deficiency. NDUFC2 overexpression suppressed NLRP3 activation and EndoMT in HBMECs induced by OGD/R. Moreover, NDUFC2 overexpression rescued SOD1 and CAT mRNA expression. These results demonstrated that NDUFC2 deficiency decreased the antioxidant levels, leading to NLRP3 activation and EndoMT during ischemia-reperfusion injury and suggesting that NDUFC2 is a potential drug target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jianwei Chen
- Interventional Medicine Center, Xi'an People's Hospital, Xi'an, China
| | | | | | | | | |
Collapse
|
9
|
Zhao H, Lv J, Meng L, Lv J, Li Z. Dual-specificity phosphatase 26-dificient neurons are susceptible to oxygen-glucose deprivation/reoxygenation-evoked apoptosis and proinflammatory response by affecting the TAK1-medaited JNK/P38 MAPK pathway. Int Immunopharmacol 2023; 117:109980. [PMID: 37012870 DOI: 10.1016/j.intimp.2023.109980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023]
Abstract
Dual-specificity phosphatase 26 (DUSP26) is linked to a broad range of human disorders as it affects numerous signaling cascades. However, the involvement of DUSP26 in ischemic stroke has not been explored. Here, we investigated DUSP26 as a key mediator of oxygen-glucose deprivation/reoxygenation (OGD/R)-associated neuronal injury, an in vitro model for investigating ischemic stroke. A decline in DUSP26 occurred in neurons suffering from OGD/R. A deficiency in DUSP26 rendered neurons more susceptible to OGD/R by aggravating neuronal apoptosis and inflammation, while the overexpression of DUSP26 blocked OGD/R-evoked neuronal apoptosis and inflammation. Mechanistically, enhanced phosphorylation of transforming growth factor-β-activated kinase 1 (TAK1), c-Jun N-terminal kinase (JNK) and P38 mitogen-activated protein kinase (MAPK) was evidenced in DUSP26-deficient neurons suffering from OGD/R, whereas the opposite effects were observed in DUSP26-overexpressed neurons. Moreover, the inhibition of TAK1 abolished the DUSP26-deficiency-elicited activation of JNK and P38 MAPK and exhibited anti-OGD/R injury effects in DUSP26-deficiency neurons. Results from these experiments show that DUSP26 is essential for neurons in defending against OGD/R insult, while neuroprotection is achieved by restraining the TAK1-mediated JNK/P38 MAPK pathway. Therefore, DUSP26 may serve as a therapeutic target for the management of ischemic stroke.
Collapse
|
10
|
Chiarini A, Gui L, Viviani C, Armato U, Dal Prà I. NLRP3 Inflammasome’s Activation in Acute and Chronic Brain Diseases—An Update on Pathogenetic Mechanisms and Therapeutic Perspectives with Respect to Other Inflammasomes. Biomedicines 2023; 11:biomedicines11040999. [PMID: 37189617 DOI: 10.3390/biomedicines11040999] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Increasingly prevalent acute and chronic human brain diseases are scourges for the elderly. Besides the lack of therapies, these ailments share a neuroinflammation that is triggered/sustained by different innate immunity-related protein oligomers called inflammasomes. Relevant neuroinflammation players such as microglia/monocytes typically exhibit a strong NLRP3 inflammasome activation. Hence the idea that NLRP3 suppression might solve neurodegenerative ailments. Here we review the recent Literature about this topic. First, we update conditions and mechanisms, including RNAs, extracellular vesicles/exosomes, endogenous compounds, and ethnic/pharmacological agents/extracts regulating NLRP3 function. Second, we pinpoint NLRP3-activating mechanisms and known NLRP3 inhibition effects in acute (ischemia, stroke, hemorrhage), chronic (Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, MS, ALS), and virus-induced (Zika, SARS-CoV-2, and others) human brain diseases. The available data show that (i) disease-specific divergent mechanisms activate the (mainly animal) brains NLRP3; (ii) no evidence proves that NLRP3 inhibition modifies human brain diseases (yet ad hoc trials are ongoing); and (iii) no findings exclude that concurrently activated other-than-NLRP3 inflammasomes might functionally replace the inhibited NLRP3. Finally, we highlight that among the causes of the persistent lack of therapies are the species difference problem in disease models and a preference for symptomatic over etiologic therapeutic approaches. Therefore, we posit that human neural cell-based disease models could drive etiological, pathogenetic, and therapeutic advances, including NLRP3’s and other inflammasomes’ regulation, while minimizing failure risks in candidate drug trials.
Collapse
|
11
|
Sun J, Gan L, Lv S, Wang T, Dai C, Sun J. Exposure to Di-(2-Ethylhexyl) phthalate drives ovarian dysfunction by inducing granulosa cell pyroptosis via the SLC39A5/NF-κB/NLRP3 axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114625. [PMID: 36774801 DOI: 10.1016/j.ecoenv.2023.114625] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) have been reported to affect populations by disrupting the human endocrine system. Di-(2-ethylhexyl) phthalate (DEHP) is an EDC that is present in various consumer products. Exposure to DEHP could contribute to reproductive system dysfunction, with subsequent adverse female reproductive outcomes. Granulosa cells (GCs) play essential roles in ovarian function and fertility. To further reveal the underlying mechanism by which DEHP impairs female fertility and affects the normal function of GCs, in vivo and in vitro experiments were performed. Transcript sequencing was used to identify genes that were differentially expressed in GCs after DEHP treatment. SLC39A5 was shown to be overexpressed in the DEHP group compared to the normal control group. DEHP treatment and overexpression of SLC39A5 activated NF-κB-related factors, followed by an increase in the transcript expression level of NLRP3. NLRP3 inflammasomes play crucial roles in pyroptosis by acting as sensors. Pyroptosis is a type of inflammation-related cell death associated with various diseases, including ovarian cancer and polycystic ovary syndrome. Activation of NF-κB contributed to the upregulation of pyroptosis in GCs, while pyroptosis factors were downregulated after the inhibition of NF-κB with JSH-23. The same phenomenon was also observed in a mouse model in which DEHP-treated mice had higher expression levels of NF-κB and pyroptosis markers in GCs. Moreover, this phenomenon could be partially reversed by the NF-κB inhibitor JSH-23. DEHP treatment also disrupted the normal expression of ovarian function-related genes and inhibited the proliferation of GCs. Reproductive system impairment was observed in mice exposed to DEHP. DEHP-treated mice had a lower body weight, smaller reproductive organs, fewer healthy follicles, and diminished ovarian reserve. Thus, DEHP contributes to ovarian dysfunction by inducing pyroptosis via the SLC39A5/NF-κB/NLRP3 axis in GCs.
Collapse
Affiliation(s)
- Jiani Sun
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lei Gan
- Department of Gynaecology and Obstetrics, Ningbo First Hospital, Ningbo, Zhejiang 315010, China
| | - Siji Lv
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Tao Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Chaoqun Dai
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jing Sun
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
12
|
Zhang Y, Zhao X, Zhang Y, Zeng F, Yan S, Chen Y, Li Z, Zhou D, Liu L. The role of circadian clock in astrocytes: From cellular functions to ischemic stroke therapeutic targets. Front Neurosci 2022; 16:1013027. [PMID: 36570843 PMCID: PMC9772621 DOI: 10.3389/fnins.2022.1013027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence suggests that astrocytes, the abundant cell type in the central nervous system (CNS), play a critical role in maintaining the immune response after cerebral infarction, regulating the blood-brain barrier (BBB), providing nutrients to the neurons, and reuptake of glutamate. The circadian clock is an endogenous timing system that controls and optimizes biological processes. The central circadian clock and the peripheral clock are consistent, controlled by various circadian components, and participate in the pathophysiological process of astrocytes. Existing evidence shows that circadian rhythm controls the regulation of inflammatory responses by astrocytes in ischemic stroke (IS), regulates the repair of the BBB, and plays an essential role in a series of pathological processes such as neurotoxicity and neuroprotection. In this review, we highlight the importance of astrocytes in IS and discuss the potential role of the circadian clock in influencing astrocyte pathophysiology. A comprehensive understanding of the ability of the circadian clock to regulate astrocytes after stroke will improve our ability to predict the targets and biological functions of the circadian clock and gain insight into the basis of its intervention mechanism.
Collapse
Affiliation(s)
- Yuxing Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yao Chen
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhong Li
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,Desheng Zhou,
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,*Correspondence: Lijuan Liu,
| |
Collapse
|
13
|
Xin C, Zhang J, Hao N, Wang J, Liu H, Wei H, Wang Y, Wang C, Wang S, Zheng C, Zhang Z, Jin Z. Irisin inhibits NLRP3 inflammasome activation in HG/HF incubated cardiac microvascular endothelial cells with H/R injury. Microcirculation 2022; 29:e12786. [PMID: 36151930 DOI: 10.1111/micc.12786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE NLRP3 inflammasome mediates myocardial ischemia/reperfusion (MI/R) injury and diabetic vascular endothelia dysfunction. However, the role of NLRP3 inflammasome in MI/R injury with diabetes has not been fully described. Irisin plays an important role in anti-inflammation and improves endothelial function in type 2 diabetes. The current study aimed to investigate the effect of irisin on regulating NLRP3 inflammasome activation in diabetic vascular endothelia dysfunction. METHODS Cardiac microvascular endothelial cells (CMECs) were cultured and subjected to high glucose/high fat (HG/HF) receiving hypoxia/reoxygenation (H/R) with irisin incubation or not. Then, apoptosis, viability, migration, NO secretion, and inflammasome activation were examined. RESULTS The hypoxic CMECs exhibited increased apoptosis, impaired viability, and migration, even decreased NO secretion and enhanced inflammasome activation. Moreover, irisin incubation decreased NLRP3 activation and attenuated cell injury in HG/HF cultured CMECs subjected to H/R injury, which was abolished by NLRP3 inflammasome activation. Meanwhile, NLRP3 inflammasome siRNA also attenuated H/R injury in CMECs under HG/HF condition. CONCLUSION The current study demonstrated for the first time that irisin inhibits NLRP3 inflammasome activation in CMECs as a novel mechanism in myocardial ischemia/reperfusion injury in diabetes.
Collapse
Affiliation(s)
- Chao Xin
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jinglong Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ningbo Hao
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jianan Wang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hui Liu
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hanwen Wei
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yong Wang
- The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Chengzhu Wang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Shuo Wang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Chengrong Zheng
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zheng Zhang
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zhitao Jin
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
14
|
The Role of Concomitant Nrf2 Targeting and Stem Cell Therapy in Cerebrovascular Disease. Antioxidants (Basel) 2022; 11:antiox11081447. [PMID: 35892653 PMCID: PMC9332234 DOI: 10.3390/antiox11081447] [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: 07/12/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the reality that a death from cerebrovascular accident occurs every 3.5 min in the United States, there are few therapeutic options which are typically limited to a narrow window of opportunity in time for damage mitigation and recovery. Novel therapies have targeted pathological processes secondary to the initial insult, such as oxidative damage and peripheral inflammation. One of the greatest challenges to therapy is the frequently permanent damage within the CNS, attributed to a lack of sufficient neurogenesis. Thus, recent use of cell-based therapies for stroke have shown promising results. Unfortunately, stroke-induced inflammatory and oxidative damage limit the therapeutic potential of these stem cells. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been implicated in endogenous antioxidant and anti-inflammatory activity, thus presenting an attractive target for novel therapeutics to enhance stem cell therapy and promote neurogenesis. This review assesses the current literature on the concomitant use of stem cell therapy and Nrf2 targeting via pharmaceutical and natural agents, highlighting the need to elucidate both upstream and downstream pathways in optimizing Nrf2 treatments in the setting of cerebrovascular disease.
Collapse
|