1
|
Cui C, Wu X, Dong S, Chen B, Zhang T. Remifentanil-induced inflammation in microglial cells: Activation of the PAK4-mediated NF-κB/NLRP3 pathway and onset of hyperalgesia. Brain Behav Immun 2025; 123:334-352. [PMID: 39322089 DOI: 10.1016/j.bbi.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND The perioperative use of remifentanil is associated with postoperative hyperalgesia, which can impair recovery and extend hospitalization. Recent studies have revealed that microglia-mediated activation of the NLRP3 inflammasome plays a critical role in opioid-induced hyperalgesia, with NF-κB acting as a pivotal activation point for NLRP3. Despite these findings, the specific molecular mechanisms underlying remifentanil-induced postoperative hyperalgesia remain unclear. This study aims to develop a model of remifentanil-induced hyperalgesia and investigate the molecular mechanisms, focusing on the NF-κB/NLRP3 pathway, using both in vitro and in vivo approaches. METHOD We established a remifentanil-induced hyperalgesia model and performed proteomic analysis to identify differential protein expression in the spinal cord tissue of rats. NLRP3 or PAK4 antagonists were administered intrathecally in vivo, and mechanical pain thresholds in the hind paws were measured using Von Frey testing. In vitro, we applied NLRP3 or PAK4 inhibitors or used lentivirus infection to silence PAK4, NF-κB, and NLRP3 genes. Protein expression was assessed through immunohistochemistry, immunofluorescence, and Western blotting. Additionally, ELISA was performed to measure IL-1β and IL-18 levels, and RT-qPCR was conducted to evaluate the transcription of target genes. RESULTS Proteomic analysis revealed that remifentanil upregulates PAK4 protein in spinal cord tissue two hours after the surgery. In addition, remifentanil induces morphological changes in the spinal cord dorsal horn, characterized by increased expression of PAK4, p-p65, NLRP3 and Iba-1 proteins, which in turn leads to elevated IL-1β and IL-18 levels and an inflammatory response. Intrathecal injection of NLRP3 or PAK4 inhibitors mitigates remifentanil-induced hyperalgesia and associated changes. In vitro, downregulation of PAK4 inhibits the increase in PAK4, p-p65, NLRP3 and Caspase-1 induced by LPS. Conversely, the downregulation of NLRP3 does not impact the levels of PAK4 and p-p65 proteins, aligning with the in vivo results and suggesting that PAK4 acts as an upstream signaling molecule of NLRP3. CONCLUSION Remifentanil can increase PAK4 expression in spinal cord dorsal horn cells by activating the NF-κB/NLRP3 pathway and mediating microglial activation, thereby contributing to postoperative hyperalgesia.
Collapse
Affiliation(s)
- Chang Cui
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China; Department of Anesthesiology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610500, Sichuan Province, China
| | - Xiaochu Wu
- West China Hospital of Sichuan University, Chengdu 610500, Sichuan Province, China
| | - Shuhua Dong
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China; Department of Anesthesiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Benzhen Chen
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China; Department of Anesthesiology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu 610500, Sichuan Province, China
| | - Tianyao Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, Sichuan Province, China; Department of Anesthesiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China.
| |
Collapse
|
2
|
Sabrie Z, Temiz-Resitoglu M, Kalkan T, Kilic B, Tunctan B, Malik KU, Sahan-Firat S. Protection by selective mTORC2 inhibition of Zymosan-induced hypotension and systemic inflammation mediated via IKKα/IκB-α/NF-κB activation. Prostaglandins Other Lipid Mediat 2024; 175:106918. [PMID: 39461547 DOI: 10.1016/j.prostaglandins.2024.106918] [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: 06/03/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Non-septic shock is a serious condition leading to multiple organ dysfunction. Although targeting the mammalian target of the rapamycin complex 1 (mTORC1) signaling pathway exerts potent anti-inflammatory activity, little is known about mTORC2's contribution to non-septic shock. Thus, our research aims to investigate mTORC2's contribution and associated changes of IκB kinase (IKKα)/inhibitor κB (IκB-α)/nuclear factor-ĸB (NF-κB) pathway on Zymosan (ZYM)-induced non-septic rat model using the novel mTORC2 selective inhibitor JR-AB2-011. Rats were given saline (4 ml/kg), dimethylsulfoxide (DMSO) (4 ml/kg), ZYM (500 mg/kg), and (or) JR-AB2-011 (1 mg/kg). Mean arterial pressure (MAP) and heart rate (HR) of rats were recorded. JR-AB2-011 reversed both ZYM-induced reduction in MAP and increase in HR. Protein expression and/or phosphorylation of rictor, protein kinase B (Akt), IκB-α, IKKα, NF-κB p65, inducible nitric oxide synthase (iNOS), nitrotyrosine, cyclooxygenase 2 (COX-2), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, besides prostaglandin (PG) E2 levels were measured. The enhanced expression of the proteins mentioned above has been inhibited by JR-AB2-011. These data suggest mTORC2's promising role in ZYM-induced hypotension and systemic inflammation mediated via IKKα/IκB-α/NF-κB pathway.
Collapse
Affiliation(s)
- Zainab Sabrie
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Taskin Kalkan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Banu Kilic
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Center for Health Sciences, Memphis, TN, USA
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey.
| |
Collapse
|
3
|
Vizuete AFK, Fróes F, Seady M, Caurio AC, Ramires Junior OV, Leite AKO, Farias CP, Wyse AT, Gonçalves CA. Targeting glycolysis for neuroprotection in early LPS-induced neuroinflammation. Brain Behav Immun Health 2024; 42:100901. [PMID: 39583162 PMCID: PMC11582448 DOI: 10.1016/j.bbih.2024.100901] [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: 07/01/2024] [Revised: 09/21/2024] [Accepted: 10/27/2024] [Indexed: 11/26/2024] Open
Abstract
Neuroinflammation is a pathophysiological feature of numerous neurological and psychiatric disorders. The immune response in the central nervous system, driven by microglia and astrocytes, leads to metabolic reprogramming towards aerobic glycolysis, a phenomenon known as the Warburg effect. The control of metabolic reprogramming via immunomodulation may represent a potential therapeutic target for providing protection against neuroinflammation, which contributes to neuronal dysfunction and death in several neurological disorders. For this purpose, we investigated putative neuroprotective effects of the downregulation of aerobic glycolysis using the 3PO inhibitor, and the downregulation of neuroinflammation using MCC950, in the early LPS-induced neuroinflammation model. The LPS-induced shift towards glycolysis, inflammatory and glial changes (IL-1β, NF-κB, COX2, Iba1, GFAP) were reversed by 3PO, which improved animal behavior. Additionally, MCC950 (an NLRP3 inhibitor) downregulated TLR4/Akt/p38 MAPK/NF-κB/STAT3 signaling, expressions of COX2 and IL-1β, and the astrocyte reactivity (decreasing GFAP) induced by early neuroinflammation, resulting in low glucose uptake. Our data support the occurrence of the Warburg effect during early neuroinflammation and suggest potential new approaches for the treatment of brain injury, given the role of neuroinflammation in such events.
Collapse
Affiliation(s)
- Adriana Fernanda K. Vizuete
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Fróes
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marina Seady
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Aline Castro Caurio
- Post Graduate Program in Biochemistry, Unipampa (Universidade Federal do Pampa) Campus Uruguaiana, Uruguaina, Rio Grande do Sul, Brazil
| | - Osmar Vieira Ramires Junior
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Karla Oliveira Leite
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Translational Neuroscience (PGNET), National Institute of Translational Neuroscience, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Clarissa Penha Farias
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Postgraduate Program in Translational Neuroscience (PGNET), National Institute of Translational Neuroscience, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Angela T.S. Wyse
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Neuroprotection and Neurometabolic Diseases Laboratory (Wyse's Lab), Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos-Alberto Gonçalves
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
4
|
Yang Z, Li X, Wei L, Bao L, Hu H, Liu L, Tan W, Tong X, Huang F. Involucrasin B suppresses airway inflammation in obese asthma by inhibiting the TLR4-NF-κB-NLRP3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155850. [PMID: 39029138 DOI: 10.1016/j.phymed.2024.155850] [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: 03/08/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Obese asthma is an asthma phenotype that causes more severe lung inflammation and airway hyperresponsiveness than allergic asthma and it is resistant to conventional therapy. Involucrasin B (IB) is a dihydroflavonoid isolated from Shuteria involucrata (Wall.) Wight & Arn., a traditional "Dai" and "Wa" medicine was used in southern China to treat the "phlegm and wetness of sputum" (obesity disease) as well as lung inflammation. However, whether IB can ameliorate obese asthma remains unclear, and the underlying mechanisms and molecular expression in obese asthma specifically targeted by IB are still not fully understood. METHODS An in vivo C57BL/6 J mouse model of obese asthma was established using house dust mites (HDMs) and high-fat diet (HFD) as inducers to evaluate the therapeutic effect of IB. An in vitro cell culture of human THP-1 monocytic cell culture was used to investigate the effect of IB after the treatment with lipopolysaccharide (LPS) and palmitic acid (PA). RESULTS In vivo, we found that intervention with IB improved airway hyperresponsiveness and lung histopathology and significantly inhibited the secretion of relevant inflammatory factors, such as interleukin (IL)-1β, IL-17A, and IL-22 in bronchoalveolar lavage fluid, and total-IgE and HDM-IgE in serum compared with the model group (HFD+HDM). The findings indicate that IB could decrease the expression of granulocyte receptor 1 (Gr-1) and neutrophil extracellular traps (NETs) in lung tissue, as well as the expression of NLR family pyrin domain containing 3 (NLRP3) and inducible nitric oxide synthase in M1 macrophages (M1). IB also reduced the population of ILC3/Th17 cells, which are responsible for producing IL-17A, a crucial mediator of neutrophil-mediated inflammation, confirming that the therapeutic effect of IB in obesity-related asthma was related to neutrophils and M1 cells. In addition, IB regulated lipid metabolism and inhibited the production of macrophages in adipose tissue. The in vitro results revealed that IB inhibited the secretion of IL-1β, IL-18, and tumor necrosis factor-α (TNF-α) from THP-1 cells, and the expression of NLRP3-related protein in THP-1 cells compared with the model groups (LPS, PA, and LPS+PA), confirming that the action of IB involved the TLR4-NF-κB-NLRP3 pathway. CONCLUSION This study demonstrated the therapeutic effect of IB in obese asthma for the first time and further clarified its mechanistic pathway as the TLR4-NF-κB-NLRP3 pathway.
Collapse
Affiliation(s)
- Zhuya Yang
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiaohong Li
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lisha Wei
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lue Bao
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Huiling Hu
- Key Laboratory of Standardization of Chinese Herbal Medicine, Ministry of Education, State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Lu Liu
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Wenhong Tan
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiaoyun Tong
- The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming 650021, China.
| | - Feng Huang
- Key Laboratory of Yunnan Provincial Department of Education on Substance Benchmark Research of Ethnic Medicines, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
5
|
Henedak NT, El-Abhar HS, Abdallah DM, Ahmed KA, Soubh AA. Demotion of canonical/non-canonical inflammasome and pyroptosis alleviates ischemia/reperfusion-induced acute kidney injury: Novel role of the D2/D3 receptor agonist ropinirole. Eur J Pharmacol 2024; 969:176460. [PMID: 38402931 DOI: 10.1016/j.ejphar.2024.176460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Ropinirole used to treat Parkinson's disease highly targets the dopaminergic receptor D3 over the D2 receptor but although both are expressed in the kidneys the ropinirole potential to treat kidney injury provoked by ischemia/reperfusion (I/R) is undraped. We investigated whether ropinirole can alleviate renal I/R by studying its anti-inflammatory, antioxidant, and anti-pyroptotic effects targeting its aptitude to inhibit the High-mobility group box 1/Toll-like receptor 4/Nuclear factor-kappa B (HMGB1/TLR4/NF-κB) cue and the canonical/non-canonical NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome trajectories. Herein, bilateral I/R surgery was induced in animals to be either untreated or treated with ropinirole for three days after the insult. Ropinirole successfully improved the histopathological picture and renal function which was confirmed by reducing cystatin C and the standard parameters creatinine and blood urea nitrogen (BUN). Ropinirole achieved this through its anti-inflammatory capacity mediated by reducing the HMGB1/TLR4 axis and inactivating NF-κB, which are upstream regulators of the NLRP3 pathway. As a result, the injurious inflammasome markers (NLRP3, apoptosis-associated speck-like protein (ASC), active caspase-1) and their target cytokines interleukin-1 beta (IL-1β) and IL-18 were decreased. Ropinirole also reduced the pyroptotic cell death markers caspase-11 and gasdermin-D. Furthermore, ropinirole by replenishing antioxidants and decreasing malondialdehyde helped to reduce oxidative stress in the kidneys. The docking findings confirmed that ropinirole highly binds to the dopaminergic D3 receptor more than to the D2 receptor. In conclusion, ropinirole has the potential to be a reno-therapeutic treatment against I/R insult by abating the inflammatory NLRP3 inflammasome signal, pyroptosis, and oxidative stress.
Collapse
Affiliation(s)
- Nada T Henedak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, 11835, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ayman A Soubh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, Egypt
| |
Collapse
|
6
|
Liu Q, Zhang MM, Guo MX, Zhang QP, Li NZ, Cheng J, Wang SL, Xu GH, Li CF, Zhu JX, Yi LT. Inhibition of Microglial NLRP3 with MCC950 Attenuates Microglial Morphology and NLRP3/Caspase-1/IL-1β Signaling In Stress-induced Mice. J Neuroimmune Pharmacol 2022; 17:503-514. [PMID: 34978026 DOI: 10.1007/s11481-021-10037-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/24/2021] [Indexed: 01/13/2023]
Abstract
Major depressive disorder is characterized by the deficiencies of monoamine neurotransmitters, neurotrophic factors and persistent neuroinflammation. Microglial activation has been associated with neuroinflammation-related mental diseases, accompanied by NLR family pyrin domain containing 3 (NLRP3) inflammasome. Here, we investigated the effect of NLRP3 inhibition by its small molecular inhibitor MCC950 on inflammatory activity and depressive-like mice induced by chronic unpredictable mild stress (CUMS), followed by the behavioral tests including sucrose preference test and forced swimming test. NLRP3/caspase-1/IL-1β signaling and microglial morphology in the prefrontal cortex were measured. The results showed that CUMS caused a decrease in sucrose preference and an increase in immobility time, which were reversed by NLRP3 inhibitor MCC950. In addition, NLRP3 inhibition decreased the number of microglia and changed the activated state of microglia to a resting state by morphology 3D reconstruction. Moreover, NLRP3 inhibition inactivated NLRP3/caspase-1/IL-1β signaling in the prefrontal cortex. The results from immunofluorescence demonstrated that NLRP3 and IL-1β expression was decreased in microglia in response to MCC950 treatment. Accordingly, proinflammatory cytokines were also decreased by NLRP3 inhibition. In conclusion, this study demonstrates that microglial NLRP3 inhibition prevents stress-induced neuroinflammation in the prefrontal cortex and suggests that microglial NLRP3 could be one of the potential therapeutic targets for depression treatment.
Collapse
Affiliation(s)
- Qing Liu
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Fujian province, Xiamen, 361021, PR China
| | - Man-Man Zhang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Fujian province, Xiamen, 361021, PR China
| | - Min-Xia Guo
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, PR China
| | - Qiu-Ping Zhang
- Xiamen Hospital of Traditional Chinese Medicine, Fujian province, Xiamen, 361009, PR China
| | - Na-Zhi Li
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, PR China
| | - Jie Cheng
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Fujian province, Xiamen, 361021, PR China
| | - Shi-Le Wang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Fujian province, Xiamen, 361021, PR China
| | - Guang-Hui Xu
- Xiamen Medicine Research Institute, Fujian province, Xiamen, 361008, PR China
| | - Cheng-Fu Li
- Xiamen Hospital of Traditional Chinese Medicine, Fujian province, Xiamen, 361009, PR China
| | - Ji-Xiao Zhu
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, PR China.
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Fujian province, Xiamen, 361021, PR China.
- Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen, 361021, PR China.
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, PR China.
| |
Collapse
|
7
|
Guo L, Wang Z, Li J, Cui L, Dong J, Meng X, Zhu G, Li J, Wang H. MCC950 attenuates inflammation-mediated damage in canines with Staphylococcus pseudintermedius keratitis by inhibiting the NLRP3 inflammasome. Int Immunopharmacol 2022; 108:108857. [PMID: 35597123 DOI: 10.1016/j.intimp.2022.108857] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Bacterial keratitis is a common eye disease in dogs and can seriously affect vision. This study investigated the anti-inflammatory effect of MCC950 in the cornea of canines infected with Staphylococcus pseudintermedius (S. pseudintermedius). METHODS In vitro, canine cornea epithelial cells were pretreated with MCC950 and PDTC and then infected with S. pseudintermedius. The key proteins of the NF-κB pathway and NLRP3 inflammasome were detected by Western blotting, the levels of inflammatory factors were detected by qPCR, and the levels of MDA and LDH were detected by assay kit. In vivo, the canine keratitis model was established by injecting S. pseudintermedius into the corneal stroma layer. After treatment with MCC950, slit-lamp examinations were performed. Cornea tissue protein and RNA were extracted, and Western blotting was used to detect key proteins of the NF-κB pathway and NLRP3 inflammasome. qPCR was used to detect the inflammatory factors. Paraffin sections of corneal tissue were prepared for HE staining and immunohistochemical staining. RESULTS After MCC950 treatment, the expression levels of key proteins in the NF-κB pathway and NLRP3 inflammasome in canine cornea epithelial cells and corneal tissues were decreased, and the expression levels of IL-1β, IL-6, IL-8, IL-18 and TNF-α were reduced. Cellular MDA and LDH levels were decreased. In vivo, the degree of corneal opacity, edema, neovascularization and corneal injury area decreased after MCC950 treatment. Canine corneal sections showed that MCC950 attenuated neutrophil infiltration. CONCLUSION MCC950 alleviates the inflammatory response to canine keratitis caused by S. pseudintermedius by inhibiting the activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Long Guo
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Zhihao Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Jun Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Luying Cui
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Xia Meng
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
8
|
Wu Z, Deng J, Zhou H, Tan W, Lin L, Yang J. Programmed Cell Death in Sepsis Associated Acute Kidney Injury. Front Med (Lausanne) 2022; 9:883028. [PMID: 35655858 PMCID: PMC9152147 DOI: 10.3389/fmed.2022.883028] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 01/15/2023] Open
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is common in patients with severe sepsis, and has a high incidence rate and high mortality rate in ICU patients. Most patients progress to AKI before drug treatment is initiated. Early studies suggest that the main mechanism of SA-AKI is that sepsis leads to vasodilation, hypotension and shock, resulting in insufficient renal blood perfusion, finally leading to renal tubular cell ischemia and necrosis. Research results in recent years have shown that programmed cell death such as apoptosis, necroptosis, pyroptosis and autophagy play important roles. In the early stage of sepsis-related AKI, autophagy bodies form and inhibit various types of programmed cell death. With the progress of disease, programmed cell death begins. Apoptosis promoter represents caspase-8-induced apoptosis and apoptosis effector represents caspase-3-induced apoptosis, however, caspase-11 and caspase-1 regulate gasdermin D-mediated pyroptosis. Caspase-8 and receptor interacting kinase 1 bodies mediate necroptosis. This review focuses on the pathophysiological mechanisms of various programmed cell death in sepsis-related AKI.
Collapse
Affiliation(s)
- Zhifen Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junhui Deng
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongwen Zhou
- Department of Nephrology, Chongqing Liangping District People's Hospital, Chongqing, China
| | - Wei Tan
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lirong Lin
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jurong Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Shi C, Wang J, Zhang R, Ishfaq M, Li Y, Zhang R, Si C, Li R, Li C, Liu F. Dihydromyricetin alleviates Escherichia coli lipopolysaccharide-induced hepatic injury in chickens by inhibiting the NLRP3 inflammasome. Vet Res 2022; 53:6. [PMID: 35073994 PMCID: PMC8785529 DOI: 10.1186/s13567-022-01024-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022] Open
Abstract
Dihydromyricetin (DHM), a flavonoid in vine tea, has many pharmacological activities, including anti-inflammatory and antibacterial effects. Lipopolysaccharide is the key inducer of inflammation in avian pathogenic Escherichia coli (E. coli) infection; however, the effect of DHM on E. coli lipopolysaccharide-induced hepatic injury remains unknown. The present study aimed to explore the role of the NLRP3 inflammasome in hepatic injury and the possible protective mechanisms of DHM against hepatic injury in chickens. The results showed that when chickens were administered lipopolysaccharide, liver damage was observed, accompanied by increased levels of serum transaminases and direct bilirubin. Additionally, hepatic expression levels of NLRP3 and caspase-1 p20, the subunit of caspase-1 that is cleaved after NLRP3 activation, significantly increased in liver injury. We found that treatment with MCC950, a specific NLRP3 inhibitor, significantly decreased serum transaminase activities, direct bilirubin content, and hepatic NLRP3 and caspase-1 p20 expression levels. DHM significantly reduced serum transaminase activities and direct bilirubin content and ameliorated histopathological and ultrastructural changes in the liver. DHM decreased hepatic levels of H2O2 and malondialdehyde and increased the activities of superoxide dismutase and glutathione peroxidase. Furthermore, DHM significantly decreased the expression levels of NLRP3, pro-caspase-1 and caspase-1 p20. Moreover, DHM reduced serum lactate dehydrogenase, IL-1β and IL-18 levels and repressed hepatic IL-1β, IL-18 and gasdermin A expression. The results demonstrated that the NLRP3 inflammasome was involved in the mechanism of lipopolysaccharide-induced hepatic injury. Furthermore, DHM could inhibit NLRP3 inflammasome activation and subsequent pyroptosis, eventually ameliorating E. coli lipopolysaccharide-induced liver injury.
Collapse
Affiliation(s)
- Chenxi Shi
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jiaqi Wang
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ruichen Zhang
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Muhammad Ishfaq
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ying Li
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ruihui Zhang
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chuanbiao Si
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Rui Li
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Changwen Li
- Laboratory Animal Base, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Fangping Liu
- Basic Veterinary Department, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China. .,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China.
| |
Collapse
|
10
|
Li C, Wang W, Xie SS, Ma WX, Fan QW, Chen Y, He Y, Wang JN, Yang Q, Li HD, Jin J, Liu MM, Meng XM, Wen JG. The Programmed Cell Death of Macrophages, Endothelial Cells, and Tubular Epithelial Cells in Sepsis-AKI. Front Med (Lausanne) 2021; 8:796724. [PMID: 34926535 PMCID: PMC8674574 DOI: 10.3389/fmed.2021.796724] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome caused by infection, following with acute injury to multiple organs. Sepsis-induced acute kidney injury (AKI) is currently recognized as one of the most severe complications related to sepsis. The pathophysiology of sepsis-AKI involves multiple cell types, including macrophages, vascular endothelial cells (ECs) and renal tubular epithelial cells (TECs), etc. More significantly, programmed cell death including apoptosis, necroptosis and pyroptosis could be triggered by sepsis in these types of cells, which enhances AKI progress. Moreover, the cross-talk and connections between these cells and cell death are critical for better understanding the pathophysiological basis of sepsis-AKI. Mitochondria dysfunction and oxidative stress are traditionally considered as the leading triggers of programmed cell death. Recent findings also highlight that autophagy, mitochondria quality control and epigenetic modification, which interact with programmed cell death, participate in the damage process in sepsis-AKI. The insightful understanding of the programmed cell death in sepsis-AKI could facilitate the development of effective treatment, as well as preventive methods.
Collapse
Affiliation(s)
- Chao Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wei Wang
- Anhui Province Key Laboratory of Genitourinary Diseases, Department of Urology and Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Shuai-Shuai Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wen-Xian Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Qian-Wen Fan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ying Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yuan He
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Qin Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan Jin
- Key Laboratory of Anti-inflammatory and Immunopharmacology (Ministry of Education), Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Chen G, Xu Y, Fan R, Liu Y, Yao Y, Jiang H, Wu Q, Li L, Chen W, Chen X. IKKε protects against starvation-induced NLRP3 inflammasome and pyroptosis in H9c2 cells by alleviating mitochondrial injury. Biochem Biophys Res Commun 2021; 589:267-274. [PMID: 34933200 DOI: 10.1016/j.bbrc.2021.12.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 11/26/2022]
Abstract
The deprivation of myocardial nutrition causes cardiomyocyte death and disturbance of energy metabolism. IKKε plays an important regulatory role in many biological events such as inflammation, redox reaction, cell death, etc. However, the more in-depth mechanism by which IKKε contributes to cardiomyocytes death in nutrition deprivation remains poorly understood. IKKε expression was knocked down by siRNA in H9c2 cells, and cells were cultured under starvation conditions to simulate ischemic conditions. Starvation triggered greater NLRP3 activation, accompanied by more IL-1β, IL-18 and caspase-1 release in the siIKKε H9c2 cells compared with the control H9c2 cells. Western blot and immunofluorescence showed that the IKKε konckdown promoted NLRP3 expressions and ROS release under starvation conditions. Furthermore, electron micrography and JC-1 analysis revealed that IKKε konckdown resulted in aggravated mitochondrial damage and more mitochondrial ROS (mtROS) released in vitro. Notably, Western blot analysis showed that IKKε deficiency activated the TBK1 and IRF3 signaling pathways to promote pyroptosis in vitro. Collectively, our results indicate that IKKε protects against cardiomyocyte injury by reducing mitochondrial damage and NLRP3 expression following nutrition deprivation via regulation of the TBK1/IRF3 signaling pathway. This study further revealed the mechanism of IKKε in inflammation and myocardial nutrition deprivation.
Collapse
Affiliation(s)
- Ganyi Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Yueyue Xu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Rui Fan
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210006, China
| | - Yafeng Liu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Yiwei Yao
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Hongwei Jiang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Qiyong Wu
- Department of Thoracic and Cardiovascular Surgery, Changzhou Second People's Hospital, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Liangpeng Li
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China.
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China.
| |
Collapse
|
12
|
Li Y, Wang Y, Guo H, Wu Q, Hu Y. IRF2 contributes to myocardial infarction via regulation of GSDMD induced pyroptosis. Mol Med Rep 2021; 25:40. [PMID: 34878155 PMCID: PMC8674697 DOI: 10.3892/mmr.2021.12556] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022] Open
Abstract
Interferon regulatory factor (IRF) 2 is a transcription factor belonging to the IRF family, which is essential for gasdermin D (GSDMD)‑induced pyroptosis. Decreasing myocardial cell pyroptosis confers protection against heart damage and cardiac dysfunction caused by myocardial infarction (MI). The aim of the present study was to investigate the involvement of IRF2 in MI and the underlying mechanism of IRF2 in pyroptosis. To mimic MI, ligation of the left anterior descending coronary artery was performed to establish an in vivo mouse model and rat cardiomyocytes H9c2 cells were cultured under hypoxic conditions to establish an in vitro model. Transthoracic echocardiography was used to assess cardiac function. Hematoxylin and eosin staining was used to observe histopathological changes in the myocardial tissue. Immunohistochemistry and western blotting were performed to detect IRF2 expression levels. TUNEL staining and flow cytometry were used to detect apoptosis in myocardial tissue and cells. Chromatin immunoprecipitation and dual luciferase reporter assay were used to verify the effect of IRF2 on GSDMD transcription. IRF2 was upregulated in MI mice. MI induced pyroptosis, as evidenced by increased GSDMD, N‑terminal GSDMD (GSDMD‑N), and cleaved (c‑) caspase‑1 levels. MI increased IL‑1β and IL‑18 levels. These alterations were alleviated by IRF2 silencing. Furthermore, in hypoxia‑treated H9c2 cells, IRF2 silencing significantly decreased the elevated levels of IL‑1β and IL‑18 and pyroptosis‑associated proteins, including GSDMD, GSDMD‑N and c‑caspase1. Moreover, in hypoxia‑treated H9c2 cells, IRF2 directly bound to the GSDMD promoter to drive GSDMD transcription and promote pyroptosis and IRF2 expression may be regulated via the hypoxia inducible factor 1 signaling pathway. In conclusion, the present results demonstrated that IRF2 is a key regulator of MI by mediating pyroptosis, which triggers GSDMD activation.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Yan Wang
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Hua Guo
- Department of Cardiovascular Medicine, Hebei Province Cangzhou Hospital of Integrated Traditional and Western, Cangzhou, Hebei 061001, P.R. China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Yamin Hu
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
13
|
Wilson SH, Hellman KM, James D, Adler AC, Chandrakantan A. Mechanisms, Diagnosis, and Medical Management of Hyperalgesia: an Educational Review. CURRENT ANESTHESIOLOGY REPORTS 2021. [DOI: 10.1007/s40140-021-00485-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Chen J, Yan J, Li S, Zhu J, Zhou J, Li J, Zhang Y, Huang Z, Yuan L, Xu K, Chen W, Ye W. Atorvastatin inhibited TNF-α induced matrix degradation in rat nucleus pulposus cells by suppressing NLRP3 inflammasome activity and inducing autophagy through NF-κB signaling. Cell Cycle 2021; 20:2160-2173. [PMID: 34494933 DOI: 10.1080/15384101.2021.1973707] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is one of the main causes of lower back pain (LBP). It results from an imbalance between the degradation and synthesis of extracellular matrix (ECM) components in nucleus pulposus (NP) cells. Atorvastatin, an HMG-CoA reductase inhibitor, plays a vital role in many diseases, such as cardiovascular disease and osteoarthritis. However, the effect of atorvastatin on IDD is unclear. Herein, we demonstrated that atorvastatin affects matrix degradation induced by TNF-α and demonstrated the mechanism by which TNF-α modulates matrix metabolism in rat NP cells. Real-time PCR, western blotting and immunofluorescence staining were performed to detect the mRNA and protein expression of related genes. mRFP-GFP-LC3 adenovirus plasmid transfection and transmission electron microscopy (TEM) were used to detect cell autophagy. NLRP3 inhibitor and lentiviral vectors containing shRNA-NLRP3 were used to show the effect of NLRP3 on autophagic flux and the NF-κB signaling pathway. The results revealed that atorvastatin might suppress matrix degradation induced by TNF-α by suppressing NLRP3 inflammasome activity and inducing autophagic flux. Moreover, atorvastatin suppressed NF-κB signaling induced by TNF-α. NF-κB signaling inhibition suppressed NLRP3 inflammasome activity, and NLRP3 inhibition suppressed NF-κB signaling activation induced by TNF-α. NLRP3 inhibition or NLRP3 knockdown induced autophagic flux in the presence of TNF-α. Overall, the present study demonstrated that atorvastatin might suppress matrix degradation induced by TNF-α and further revealed the crosstalk among NLRP3 inflammasome activity, autophagy and NF-κB signaling.
Collapse
Affiliation(s)
- Jiancong Chen
- Department of Orthopedics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiansen Yan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuangxing Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianxiong Zhu
- Department of Orthopedics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jie Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Breast Cancer Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jun Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Orthopedics, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yangyang Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhengqi Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liang Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kang Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weijian Chen
- Department of Orthopedics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Ye
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Burdette BE, Esparza AN, Zhu H, Wang S. Gasdermin D in pyroptosis. Acta Pharm Sin B 2021; 11:2768-2782. [PMID: 34589396 PMCID: PMC8463274 DOI: 10.1016/j.apsb.2021.02.006] [Citation(s) in RCA: 376] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Pyroptosis is the process of inflammatory cell death. The primary function of pyroptosis is to induce strong inflammatory responses that defend the host against microbe infection. Excessive pyroptosis, however, leads to several inflammatory diseases, including sepsis and autoimmune disorders. Pyroptosis can be canonical or noncanonical. Upon microbe infection, the canonical pathway responds to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), while the noncanonical pathway responds to intracellular lipopolysaccharides (LPS) of Gram-negative bacteria. The last step of pyroptosis requires the cleavage of gasdermin D (GsdmD) at D275 (numbering after human GSDMD) into N- and C-termini by caspase 1 in the canonical pathway and caspase 4/5/11 (caspase 4/5 in humans, caspase 11 in mice) in the noncanonical pathway. Upon cleavage, the N-terminus of GsdmD (GsdmD-N) forms a transmembrane pore that releases cytokines such as IL-1β and IL-18 and disturbs the regulation of ions and water, eventually resulting in strong inflammation and cell death. Since GsdmD is the effector of pyroptosis, promising inhibitors of GsdmD have been developed for inflammatory diseases. This review will focus on the roles of GsdmD during pyroptosis and in diseases.
Collapse
Key Words
- 7DG, 7-desacetoxy-6,7-dehydrogedunin
- ADRA2B, α-2B adrenergic receptor
- AIM, absent in melanoma
- ASC, associated speck-like protein
- Ac-FLTD-CMK, acetyl-FLTD-chloromethylketone
- BMDM, bone marrow-derived macrophages
- CARD, caspase activation
- CD, Crohn’s disease
- CTM, Chinese traditional medicine
- CTSG, cathepsin G
- Caspase
- DAMP, damage-associated molecular pattern
- DFNA5, deafness autosomal dominant 5
- DFNB59, deafness autosomal recessive type 59
- DKD, diabetic kidney disease
- DMF, dimethyl fumarate
- Damage-associated molecular patterns (DAMPs)
- ELANE, neutrophil expressed elastase
- ESCRT, endosomal sorting complexes required for transport
- FADD, FAS-associated death domain
- FDA, U.S. Food and Drug Administration
- FIIND, function to find domain
- FMF, familial Mediterranean fever
- GI, gastrointestinal
- GPX, glutathione peroxidase
- Gasdermin
- GsdmA/B/C/D/E, gasdermin A/B/C/D/E
- HAMP, homeostasis altering molecular pattern
- HIN, hematopoietic expression, interferon-inducible nature, and nuclear localization
- HIV, human immunodeficiency virus
- HMGB1, high mobility group protein B1
- IBD, inflammatory bowel disease
- IFN, interferon
- ITPR1, inositol 1,4,5-trisphosphate receptor type 1
- Inflammasome
- Inflammation
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAP3K7, mitogen-activated protein kinase kinase kinase 7
- MCC950, N-[[(1,2,3,5,6,7-hexahydro-s-indacen-4-yl)amino]carbonyl]-4-(1-hydroxy-1-methylethyl)-2-furansulfonamide
- NAIP, NLR family apoptosis inhibitory protein
- NBD, nucleotide-binding domain
- NEK7, NIMA-related kinase 7
- NET, neutrophil extracellular trap
- NIK, NF-κB inducing kinase
- NLR, NOD-like receptor
- NLRP, NLR family pyrin domain containing
- NSAID, non-steroidal anti-inflammatory drug
- NSCLC, non-small cell lung cancer
- NSP, neutrophil specific serine protease
- PAMP, pathogen-associated molecular pattern
- PKA, protein kinase A
- PKN1/2, protein kinase1/2
- PKR, protein kinase-R
- PRR, pattern recognition receptors
- PYD, pyrin domain
- Pathogen-associated molecular patterns (PAMPs)
- Pyroptosis
- ROS, reactive oxygen species
- STING, stimulator of interferon genes
- Sepsis
- TLR, Toll-like receptor
- UC, ulcerative colitis
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP–AMP synthase
- mtDNA, mitochondrial DNA
Collapse
Affiliation(s)
- Brandon E. Burdette
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Ashley N. Esparza
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Hua Zhu
- Department of Surgery, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Shanzhi Wang
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| |
Collapse
|
16
|
Zheng W, Liu B, Shi E. Perillaldehyde Alleviates Spinal Cord Ischemia-Reperfusion Injury Via Activating the Nrf2 Pathway. J Surg Res 2021; 268:308-317. [PMID: 34399353 DOI: 10.1016/j.jss.2021.06.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/31/2021] [Accepted: 06/11/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Spinal Cord ischemia-reperfusion injury (SCII) is one of the most destructive complications in thoracic-abdominal aortic surgery, which can cause physical abnormalities, paralysis and even brain death. Evidence has shown that perillaldehyde (PAH) can ameliorate rat's cerebra ischemia-reperfusion injury. However, the effect of PAH on SCII remains unknown. METHODS The current study established SCII rat models and oxygen and glucose deprivation/reoxygenation-induced BV2 microglia models to explore whether PAH could alleviate SCII symptoms and to investigate underlying mechanism. RESULTS SCII rats underwent severe neurologic motor dysfunction and histopathologic injury compared with the normal rats, which are exhibited by loss of motor neurons and decrease of nissl bodies. Treatment with PAH significantly ameliorated motor dysfunction and neuron damage. PAH downregulated the expression of NLR family pyrin domain containing 3, cleaved/pro caspase-1, interleukin-1β and interleukin-18 in spinal cord tissues of SCII rats. Besides, the contents of oxidative stress-related factors superoxide dismutase, manganese-dependent superoxide dismutase, catalase and glutathione peroxidase were significantly increased and malondialdehyde content was decreased after PAH treatment. PAH treatment upregulated the expression of nuclear factor-E2-related factor 2 and heme oxygenase-1 in spinal cord tissues of SCII rats. Our in vitro study confirmed that PAH inhibited microglial activation by activating the nuclear factor-E2-related factor 2/heme oxygenase-1 pathway, exhibited by alleviated inflammation and oxidative stress. CONCLUSIONS This study elucidates that PAH has the potential value for treating SCII, which provides an experimental basis for clinical trials in the future.
Collapse
Affiliation(s)
- Wenjun Zheng
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; Department of Cardiac Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Bing Liu
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; Department of Vascular Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Enyi Shi
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
17
|
Abstract
Significance: Kidney diseases remain a worldwide public health problem resulting in millions of deaths each year; they are characterized by progressive destruction of renal function by sustained inflammation. Pyroptosis is a lytic type of programmed cell death involved in inflammation, as well as a key fibrotic mechanism that is critical in the development of kidney pathology. Pyroptosis is induced by the cleavage of Gasdermins by various caspases and is executed by the insertion of the N-terminal fragment of cleaved Gasdermins into the plasma membrane, creating oligomeric pores and allowing the release of diverse proinflammatory products into the extracellular space. Inflammasomes are multiprotein complexes leading to the activation of caspase-1, which will cleave Gasdermin D, releasing several proinflammatory cytokines; this results in the initiation and amplification of the inflammatory response. Recent Advances: The efficacy of Gasdermin D cleavage is reduced by a change in the redox balance. Recently, several studies have shown that the attenuation of reactive oxygen species (ROS) production induced by antioxidant pathways results in a reduction of renal pyroptosis. In this review, we discuss the role of pyroptosis in the pathogenesis of chronic kidney disease (CKD) and acute kidney disease; summarize the clinical outcomes and different molecular mechanisms leading to Gasdermin activation; and examine studies about the capacity of antioxidants, particularly Nrf2 activators, to ameliorate Gasdermin activity. Future Directions: We illustrate the potential influence of the deregulation of redox balance on inflammasome activity and pyroptosis as a novel therapeutic approach for the treatment of kidney diseases. Antioxid. Redox Signal. 35, 40-60.
Collapse
Affiliation(s)
- Santiago Cuevas
- Molecular Inflammation Group, Biomedical Research Institute of Murcia, University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Pablo Pelegrín
- Molecular Inflammation Group, Biomedical Research Institute of Murcia, University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| |
Collapse
|
18
|
Corcoran SE, Halai R, Cooper MA. Pharmacological Inhibition of the Nod-Like Receptor Family Pyrin Domain Containing 3 Inflammasome with MCC950. Pharmacol Rev 2021; 73:968-1000. [PMID: 34117094 DOI: 10.1124/pharmrev.120.000171] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Activation of the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome drives release of the proinflammatory cytokines interleukin (IL)-1β and IL-18 and induces pyroptosis (lytic cell death). These events drive chronic inflammation, and as such, NLRP3 has been implicated in a large number of human diseases. These range from autoimmune conditions, the simplest of which is NLRP3 gain-of-function mutations leading to an orphan disease, cryopyrin-associated period syndrome, to large disease burden indications, such as atherosclerosis, heart failure, stroke, neurodegeneration, asthma, ulcerative colitis, and arthritis. The potential clinical utility of NLRP3 inhibitors is substantiated by an expanding list of indications in which NLRP3 activation has been shown to play a detrimental role. Studies of pharmacological inhibition of NLRP3 in nonclinical models of disease using MCC950 in combination with human genetics, epigenetics, and analyses of the efficacy of biologic inhibitors of IL-1β, such as anakinra and canakinumab, can help to prioritize clinical trials of NLRP3-directed therapeutics. Although MCC950 shows excellent (nanomolar) potency and high target selectivity, its pharmacokinetic and toxicokinetic properties limited its therapeutic development in the clinic. Several improved, next-generation inhibitors are now in clinical trials. Hence the body of research in a plethora of conditions reviewed herein may inform analysis of the potential translational value of NLRP3 inhibition in diseases with significant unmet medical need. SIGNIFICANCE STATEMENT: The nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is one of the most widely studied and best validated biological targets in innate immunity. Activation of NLRP3 can be inhibited with MCC950, resulting in efficacy in more than 100 nonclinical models of inflammatory diseases. As several next-generation NLRP3 inhibitors are entering proof-of-concept clinical trials in 2020, a review of the pharmacology of MCC950 is timely and significant.
Collapse
Affiliation(s)
- Sarah E Corcoran
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Reena Halai
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Matthew A Cooper
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| |
Collapse
|
19
|
Senol SP, Temiz-Resitoglu M, Guden DS, Sari AN, Sahan-Firat S, Tunctan B. Suppression of TLR4/MyD88/TAK1/NF-κB/COX-2 Signaling Pathway in the Central Nervous System by Bexarotene, a Selective RXR Agonist, Prevents Hyperalgesia in the Lipopolysaccharide-Induced Pain Mouse Model. Neurochem Res 2021; 46:624-637. [PMID: 33389386 DOI: 10.1007/s11064-020-03197-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/26/2022]
Abstract
A selective RXR agonist, bexarotene, has been shown to have anti-inflammatory, anti-nociceptive, and neuroprotective effects in several models of numerous neurological diseases characterized by systemic inflammation. The mechanisms underlying these effects remains unknown. To elucidate these mechanisms, we investigated whether the TLR4/MyD88/TAK1/NF-κB/COX-2 signaling pathway in the CNS mediates the effect of bexarotene to prevent hyperalgesia in the LPS-induced inflammatory pain mouse model. The reaction time to thermal stimuli within 30 s was evaluated by the hot plate test in male mice treated with saline, LPS (10 mg/kg), DMSO, and/or bexarotene (0.1, 1, 3, or 10 mg/kg) after 6 h. The latency to the thermal stimulus (18.11 ± 1.36 s) in the LPS-treated mice was significantly decreased by 30% compared with saline-treated mice (25.84 ± 1.99 s). Treatment with bexarotene only at a dose of 10 mg/kg showed a significant increase in the latency by 22.49 ± 1.00 s compared with LPS-treated mice. Bexarotene also prevented the reduction in RXRα protein expression associated with a rise in the expression of TLR4, MyD88, phosphorylated TAK1, NF-κB p65, phosphorylated NF-κB p65, COX-2, and IL-1β proteins, in addition to COX-2 activity and levels of PGE2 and IL-1β in the brains and spinal cords of the LPS-treated animals. Likely, decreased activity of TLR4/MyD88/TAK1/NF-κB/COX-2 signaling pathway in addition to increased pro-inflammatory cytokine formation in the CNS of mice participates in the protective effect of bexarotene against hyperalgesia induced by LPS.
Collapse
Affiliation(s)
- Sefika Pinar Senol
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Demet Sinem Guden
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Ayse Nihal Sari
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey.
| |
Collapse
|
20
|
Guden DS, Temiz-Resitoglu M, Senol SP, Kibar D, Yilmaz SN, Tunctan B, Malik KU, Sahan-Firat S. mTOR inhibition as a possible pharmacological target in the management of systemic inflammatory response and associated neuroinflammation by lipopolysaccharide challenge in rats. Can J Physiol Pharmacol 2021; 99:921-934. [PMID: 33641344 DOI: 10.1139/cjpp-2020-0487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuroinflammation plays a critical role during sepsis triggered by microglial activation. Mammalian target of rapamycin (mTOR) has gained attraction in neuroinflammation, however, the mechanism remains unclear. Our goal was to assess the effects of mTOR inhibition by rapamycin on inflammation, microglial activation, oxidative stress, and apoptosis associated with the changes in the inhibitor-κB (IκB)-α/nuclear factor-κB (NF-κB)/hypoxia-inducible factor-1α (HIF-1α) pathway activity following a systemic challenge with lipopolysaccharide (LPS). Rats received saline (10 mL/kg), LPS (10 mg/kg), and (or) rapamycin (1 mg/kg) intraperitoneally. Inhibition of mTOR by rapamycin blocked phosphorylated form of ribosomal protein S6, NF-κB p65 activity by increasing degradation of IκB-α in parallel with HIF-1α expression increased by LPS in the kidney, heart, lung, and brain tissues. Rapamycin attenuated the increment in the expression of tumor necrosis factor-α and interleukin-1β, the inducible nitric oxide synthase, gp91phox, and p47phox in addition to nitrite levels elicited by LPS in tissues or sera. Concomitantly, rapamycin treatment reduced microglial activation, brain expression of caspase-3, and Bcl-2-associated X protein while it increased expression of B cell lymphoma 2 induced by LPS. Overall, this study supports the hypothesis that mTOR contributes to the detrimental effect of LPS-induced systemic inflammatory response associated with neuroinflammation via IκB-α/NF-κB/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Demet Sinem Guden
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Sefika Pinar Senol
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Deniz Kibar
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Sakir Necat Yilmaz
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Department of Pharmacology, College of Medicine, Memphis, TN, USA
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| |
Collapse
|
21
|
Antituberculosis Drugs (Rifampicin and Isoniazid) Induce Liver Injury by Regulating NLRP3 Inflammasomes. Mediators Inflamm 2021; 2021:8086253. [PMID: 33688304 PMCID: PMC7914095 DOI: 10.1155/2021/8086253] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 01/13/2021] [Accepted: 01/23/2021] [Indexed: 02/07/2023] Open
Abstract
Patients being treated for pulmonary tuberculosis often suffer liver injury due to the effects of anti-TB drugs, and the underlying mechanisms for those injuries need to be clarified. In this study, rats and hepatic cells were administrated isoniazid (INH) and rifampin (RIF) and then treated with NLRP3-inflammasome inhibitors (INF39 and CP-456773) or NLRP3 siRNA. Histopathological changes that occurred in liver tissue were examined by H&E staining. Additionally, the levels IL-33, IL-18, IL-1β, NLRP3, ASC, and cleaved-caspase 1 expression in the liver tissues were also determined. NAT2 and CYP2E1 expression were identified by QRT-PCR analysis. Finally, in vitro assays were performed to examine the effects of siRNA targeting NLRP3. Treatment with the antituberculosis drugs caused significant liver injuries, induced inflammatory responses and oxidative stress (OS), activated NLRP3 inflammasomes, reduced the activity of drug-metabolizing enzymes, and altered the antioxidant defense system in rats and hepatic cells. The NLRP3 inflammasome was required for INH- and RIF-induced liver injuries that were produced by inflammatory responses, OS, the antioxidant defense system, and drug-metabolizing enzymes. This study indicated that the NLRP3 inflammasome is involved in antituberculosis drug-induced liver injuries (ATLIs) and suggests NLRP3 as a potential target for attenuating the inflammation response in ATLIs.
Collapse
|
22
|
Cagli A, Senol SP, Temiz-Resitoglu M, Guden DS, Sari AN, Sahan-Firat S, Tunctan B. Soluble epoxide hydrolase inhibitor trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl)urea prevents hyperalgesia through regulating NLRC4 inflammasome-related pro-inflammatory and anti-inflammatory signaling pathways in the lipopolysaccharide-induced pain mouse model. Drug Dev Res 2021; 82:815-825. [PMID: 33559150 DOI: 10.1002/ddr.21786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 01/22/2023]
Abstract
Epoxyeicosatrienoic acids (EETs) have anti-inflammatory effects and soluble epoxide hydrolase (sEH) inhibition might be a useful therapeutic approach to manage inflammatory disorders. The purpose of the study was to investigate whether nucleotide-binding and oligomerization domain-like receptor (NLR) C4 inflammasome-related pro-inflammatory and anti-inflammatory signaling pathways in the central nervous system (CNS) participates in the effect of trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl)urea (TPPU), a potent sEH inhibitor, to prevent hyperalgesia in the LPS-induced pain mouse model. The latency of pain within 30 s was measured by the hot plate test in male mice injected with saline, lipopolysaccharide (LPS) (10 mg/kg), and/or TPPU (0.3, 0.5, or 1 mg/kg) after 6 h. Hyperalgesia induced by LPS was associated with decreased 14,15-dihydroxyeicosatrienoic acid and interleukin (IL)-1β levels and enhanced expression of NLRC4, apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), caspase-1 p20, IL-1β, and caspase-11 p20 in the brains and spinal cords of the animals. Besides the increased expression of nicotinamide adenine dinucleotide phosphate oxidase (NOX) subunits (gp91phox and p47phox ) and nitrotyrosine, a decrease in NLRC3, inducible nitric oxide synthase (iNOS), and neuronal NOS (nNOS) expression was also observed in the tissues of LPS-treated mice. TPPU at 0.5 mg/kg dose prevented the changes induced by LPS. Likely, decreased activity of pro-inflammatory NLRC4/ASC/pro-caspase-1 and caspase-11 inflammasomes and NOX in addition to enhanced levels of anti-inflammatory EETs and expression of NLRC3, iNOS, and nNOS in the CNS of mice participates in the protective effect of TPPU against LPS-induced hyperalgesia.
Collapse
Affiliation(s)
- Ali Cagli
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Sefika Pinar Senol
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Demet Sinem Guden
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Ayse Nihal Sari
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| |
Collapse
|
23
|
Zhang L, Li J, Li Y, Wang Z, Wang G, Yu Y, Song C, Cui W. Spinal caspase-3 contributes to tibial fracture-associated postoperative allodynia via up-regulation of LRRTM1 expression in mice. Neurosci Lett 2020; 739:135429. [PMID: 33069813 DOI: 10.1016/j.neulet.2020.135429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/22/2020] [Accepted: 09/27/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bone fracture may subsequently cause chronic postoperative pain after orthopedic surgery, but mechanisms remain elusive. The necessity of caspase-3 in neuroinflammation and synaptic plasticity has been summarized in pathological pain. Leucine-rich repeat transmembrane protein 1 (LRRTM1) mediates synaptic delivery of AMPA receptor and synaptogenesis. This study evaluated whether caspase-3 and LRRTM1 are required for fracture-associated postoperative allodynia. METHODS A model of tibial fracture with intramedullary pinning in mice was established for the induction of postoperative pain, verified by measurement of mechanical paw withdrawal threshold and cold scores response to acetone. The caspase-3 specific inhibitor, recombinant caspase-3 and LRRTM1 knockdown by shRNA were utilized for the investigation of pathogenesis as well as the prevention of allodynia. Also, the activity of caspase-3 and the expression of LRRTM1 in the spinal dorsal horn were examined by Western blot and RT-qPCR. RESULTS This study reported that tibial fracture and orthopedic surgery produced long-lasting mechanical allodynia and cold allodynia, along with the up-modulation of spinal caspase-3 activity (but not caspase-3 expression) and LRRTM1 expression. Spinal caspase-3 inhibition prevented fracture-associated behavioral allodynia in a dose-dependent manner. Caspase-3 inhibitor also reduced the spinal increased LRRTM1 level after tibial fracture with pinning. Spinal LRRTM1 deficiency impaired fracture-caused postoperative pain. Intrathecal recombinant caspase-3 facilitated acute pain hypersensitivity and spinal LRRTM1 expression in naïve mice, reversing by LRRTM1 knockdown. CONCLUSION Our current results demonstrate the spinal up-regulation of LRRTM1 by caspase-3 activation in the development of tibial fracture-associated postoperative pain in mice.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Jing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Zhen Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Chengcheng Song
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| | - Wei Cui
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
24
|
Fu J, Zeng Z, Zhang L, Wang Y, Li P. 4'-O-β-D-glucosyl-5-O-methylvisamminol ameliorates imiquimod-induced psoriasis-like dermatitis and inhibits inflammatory cytokines production by suppressing the NF-κB and MAPK signaling pathways. ACTA ACUST UNITED AC 2020; 53:e10109. [PMID: 33146282 PMCID: PMC7643925 DOI: 10.1590/1414-431x202010109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/05/2020] [Indexed: 11/21/2022]
Abstract
Psoriasis is a chronic inflammatory skin disorder in humans, and the inflammatory reaction plays an important role in development and onset of psoriasis. 4'-O-β-D-glucosyl-5-O-methylvisamminol (4GMV) is one of the major active chromones isolated from Saposhnikoviae divaricata (Turcz.) Schischk, which has been reported to exhibit excellent anti-inflammatory activities. However, the possible therapeutic effect on psoriasis and underlying mechanism has not been reported. Thus, the aim of this study was to investigate the protective effect of 4GMV on the imiquimod (IMQ)-induced psoriasis-like lesions in BALB/c mice and the anti-inflammatory effect on the lipopolysaccharide (LPS)-induced inflammation in RAW264.7 macrophages. The results demonstrated that 4GMV decreased IMQ-induced keratinocyte proliferation and inflammatory cell infiltration. Moreover, 4GMV treatment significantly inhibited the production of NO, PEG 2, and cytokines such as interleukin (IL)-1β, IL-6, interferon (IFN)-γ, and IL-22 in LPS-stimulated RAW264.7 macrophages. 4GMV also suppressed the LPS-upregulated protein expressions of iNOS and COX-2 in a dose-dependent manner. Furthermore, qRT-PCR analysis showed that 4GMV down-regulated the mRNA level of IL-1β and IL-6 expression. Further studies by western blot indicated that 4GMV inhibited the activation of upstream mediator NF-κB by suppressing the expression of TLR4 and the phosphorylation of IκBα and p65. The phosphorylation of JNK, p38, and ERK were also markedly reversed by 4GMV in LPS-treated RAW264.7 macrophages. Taken together, these results demonstrated that 4GMV showed a protective effect in IMQ-induced psoriasis-like mice and inhibited inflammation through the NF-κB and MAPK signaling pathways, indicating that 4GMV might be a potential therapeutic drug for psoriasis.
Collapse
Affiliation(s)
- Jing Fu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, China
| | - Zuping Zeng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, China
| | - Lu Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, China
| |
Collapse
|
25
|
Li X, Yu Z, Zong W, Chen P, Li J, Wang M, Ding F, Xie M, Wang W, Luo X. Deficiency of the microglial Hv1 proton channel attenuates neuronal pyroptosis and inhibits inflammatory reaction after spinal cord injury. J Neuroinflammation 2020; 17:263. [PMID: 32891159 PMCID: PMC7487532 DOI: 10.1186/s12974-020-01942-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 08/25/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) causes neurological dysfunction with devastating consequences. SCI pathogenesis is accompanied by inflammasome activation and neuronal damage. But the spatial pattern and the time course of neuronal pyroptosis and apoptosis after SCI should be further elucidated. The microglial voltage-gated proton channel (Hv1) is implicated in reactive oxygen species (ROS)-induced neuronal damage following ischemic stroke. However, there is a lack of quantification on the neuronal pyroptosis and apoptosis associated with microglial Hv1 after SCI. METHODS We analyzed spatial and temporal characteristics of neuronal pyroptosis and apoptosis following SCI and investigated the effects of Hv1 deficiency on neuronal pyroptosis and the nod-like receptor 3 (NLRP3) inflammasome pathway by using a mouse model of SCI. We tested the effects of Hv1-deficient microglia on ROS production in vivo and examined the relationship between ROS and neuronal pyroptosis in vitro. RESULTS We observed that apoptosis was detected closer to the injury core than pyroptosis. The incidence of neuronal apoptosis peaked on day 1 after SCI and occurred before pyroptosis. Hv1 deficiency reduced neuronal apoptosis and NLRP3-inflammasome-mediated pyroptosis, improved axonal regeneration, and reduced motor deficits. SCI led to elevated ROS levels, whereas Hv1 deficiency downregulated microglial ROS generation. In vitro, ROS upregulated neuronal pyroptosis and activated the NLRP3 inflammasome pathway, both of which were reversed by addition of a ROS scavenger. Our results suggested that microglial Hv1 regulated neuronal apoptosis and NLRP3-induced neuronal pyroptosis after SCI by mediating ROS production. CONCLUSION Following SCI, neuronal pyroptosis lasted longer and occurred farther away from the injury core compared with that of neuronal apoptosis. Microglial Hv1 deficiency downregulated microglial ROS generation and reduced apoptosis and NLRP3-induced neuronal pyroptosis. Our findings may provide novel insights into Hv1-associated mechanisms underlying neuronal damage after SCI.
Collapse
Affiliation(s)
- Xuefei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weifeng Zong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peng Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengfei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
26
|
Huang R, Hou L, Ruan Z, Zhang D, Sun W, Wang Q. NLRP3 inflammasome mediates 2,5-hexanedione-induced neurotoxicity through regulation of macrophage infiltration in rats. Chem Biol Interact 2020; 330:109232. [PMID: 32860822 DOI: 10.1016/j.cbi.2020.109232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/13/2020] [Accepted: 08/21/2020] [Indexed: 01/20/2023]
Abstract
Currently, whether nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation contributes to neuropathy induced by 2,5-Hexanedione (HD), the toxic metabolite of n-hexane, remains unknown. In this study, we found that HD intoxication elevated NLRP3 expression, caspase-1 activation and interleukin-1β production in sciatic nerve of rats, indicating activation of NLRP3 inflammasome. The increased cleavage of gasdermin D (GSDMD) protein, an important mediator of pyroptosis, and axon degeneration were also observed in sciatic nerves of HD-intoxicated rats. Interestingly, glybenclamide, a widely used inhibitor of NLRP3 inflammasome, significantly reduced NLRP3 inflammasome activation, which was associated with decreased GSDMD cleavage and axon degeneration as well as improved motor performance of HD-intoxicated rats. Subsequently, we found that inhibition of NLRP3 inflammasome by glybenclamide attenuated macrophage infiltration, activation and M1 polarization in sciatic nerves of HD-intoxicated rats. Furthermore, decreased malondialdehyde (MDA) contents and increased glutathione (GSH) level and total anti-oxidative capacity were also observed in sciatic nerves of rats treated with combined glybenclamide and HD compared with HD alone group. Altogether, our findings suggest that NLRP3 inflammasome activation contributes to HD-induced neurotoxicity by enhancing macrophage infiltration and activation as well as oxidative stress, providing a novel mechanism of neuropathy induced by this neurotoxicant.
Collapse
Affiliation(s)
- Ruixue Huang
- School of Public Health, Dalian Medical University, Dalian, Liaoning Province, China
| | - Liyan Hou
- School of Public Health, Dalian Medical University, Dalian, Liaoning Province, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Zhengzheng Ruan
- School of Public Health, Dalian Medical University, Dalian, Liaoning Province, China
| | - Dongdong Zhang
- School of Public Health, Dalian Medical University, Dalian, Liaoning Province, China
| | - Wei Sun
- School of Public Health, Dalian Medical University, Dalian, Liaoning Province, China
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, Dalian, Liaoning Province, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
27
|
Faro J, Romero R, Schwenkel G, Garcia-Flores V, Arenas-Hernandez M, Leng Y, Xu Y, Miller D, Hassan SS, Gomez-Lopez N. Intra-amniotic inflammation induces preterm birth by activating the NLRP3 inflammasome†. Biol Reprod 2020; 100:1290-1305. [PMID: 30590393 DOI: 10.1093/biolre/ioy261] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/12/2018] [Accepted: 12/22/2018] [Indexed: 01/23/2023] Open
Abstract
Intra-amniotic inflammation is strongly associated with spontaneous preterm labor and birth, the leading cause of perinatal mortality and morbidity worldwide. Previous studies have suggested a role for the NLRP3 (NLR family pyrin domain-containing protein 3) inflammasome in the mechanisms that lead to preterm labor and birth. However, a causal link between the NLRP3 inflammasome and preterm labor/birth induced by intra-amniotic inflammation has not been established. Herein, using an animal model of lipopolysaccharide-induced intra-amniotic inflammation (IAI), we demonstrated that there was priming of the NLRP3 inflammasome (1) at the transcriptional level, indicated by enhanced mRNA expression of inflammasome-related genes (Nlrp3, Casp1, Il1b); and (2) at the protein level, indicated by greater protein concentrations of NLRP3, in both the fetal membranes and decidua basalis prior to preterm birth. Additionally, we showed that there was canonical activation of the NLRP3 inflammasome in the fetal membranes, but not in the decidua basalis, prior to IAI-induced preterm birth as evidenced by increased protein levels of active caspase-1. Protein concentrations of released IL1β were also increased in both the fetal membranes and decidua basalis, as well as in the amniotic fluid, prior to IAI-induced preterm birth. Finally, using the specific NLRP3 inhibitor, MCC950, we showed that in vivo inhibition of the NLRP3 inflammasome reduced IAI-induced preterm birth and neonatal mortality. Collectively, these results provide a causal link between NLRP3 inflammasome activation and spontaneous preterm labor and birth in the context of intra-amniotic inflammation. We also showed that, by targeting the NLRP3 inflammasome, adverse pregnancy and neonatal outcomes can be significantly reduced.
Collapse
Affiliation(s)
- Jonathan Faro
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - George Schwenkel
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yaozhu Leng
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sonia S Hassan
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
28
|
Rong J, Xu J, Liu Q, Xu J, Mou T, Zhang X, Chi H, Zhou H. Anti-inflammatory effect of up-regulated microRNA-221-3p on coronary heart disease via suppressing NLRP3/ASC/pro-caspase-1 inflammasome pathway activation. Cell Cycle 2020; 19:1478-1491. [PMID: 32372677 DOI: 10.1080/15384101.2020.1754562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE As some evidence has demonstrated the role of microRNA-221 (miR-221) on coronary heart disease (CHD), the aim of the present study was to investigate the effect of miR-221-3p on CHD via regulating NLRP3/ASC/pro-caspase-1 inflammasome pathway. METHODS Sixty CHD patients and 60 healthy controls were collected to detect the expression of miR-221-3p, NLRP3, ASC, pro-caspase-1 in peripheral blood and the contents of related factors in serum. The rats model of CHD was injected with miR-221-3p agomir or miR-221-3p antagomir to explore its functions in miR-221-3p, NLRP3, ASC and pro-caspase-1 expression, electrocardiogram data, cardiomyocytes apoptosis, myocardial injury, inflammatory reaction and oxidative stress of CHD rats. RESULTS MiR-221-3p declined and NLRP3, ASC and pro-caspase-1 raised in CHD. Up-regulated miR-221-3p reduced the change value of J-point and T-wave, decreased NLRP3, ASC and pro-caspase-1 expression, suppressed apoptosis in cardiomyocytes, as well as suppressed myocardial injury, inflammatory reaction and oxidative stress in CHD rats. CONCLUSION This study highlights that up-regulated miR-221-3p suppresses the overactivation of NLRP3/ASC/pro-caspase-1 inflammasome pathway and has an anti-inflammatory effect in CHD. Thus, miR-221-3p may serve as a potential target for the treatment of CHD.
Collapse
Affiliation(s)
- Jingfeng Rong
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Jijie Xu
- Cardiovascular Medicine Institute, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Qian Liu
- Cardiovascular Medicine Institute, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Jianjun Xu
- Cardiothoracic Surgery Department, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Ting Mou
- Department of Cardiovascular, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Xuhua Zhang
- Department of Cardiovascular, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Hao Chi
- Cardiothoracic Surgery Department, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Hua Zhou
- Department of Cardiovascular, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| |
Collapse
|
29
|
Biliktu M, Senol SP, Temiz-Resitoglu M, Guden DS, Horat MF, Sahan-Firat S, Sevim S, Tunctan B. Pharmacological inhibition of soluble epoxide hydrolase attenuates chronic experimental autoimmune encephalomyelitis by modulating inflammatory and anti-inflammatory pathways in an inflammasome-dependent and -independent manner. Inflammopharmacology 2020; 28:1509-1524. [PMID: 32128702 DOI: 10.1007/s10787-020-00691-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022]
Abstract
We aimed to determine the effect of soluble epoxide hydrolase (sEH) inhibition on chronic experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS), associated with changes in inflammasome-dependent and -independent inflammatory and anti-inflammatory pathways in the CNS of mice. C57BL/6 mice were used to induce chronic EAE by using an injection of MOG35-55 peptide/PT. Animals were observed daily and scored for EAE signs for 25 days after immunization. Following the induction of EAE, the scores were increased after 9 days and reached peak value as determined by ≥ 2 or ≤ 3 with 8% mortality rate on day 17. On day 17, mice were administered daily PBS, DMSO, or TPPU (a potent sEH inhibitor) (1, 3, or 10 mg/kg) until the end of the study. TPPU only at 3 mg/kg dose decreased the AUC values calculated from EAE scores obtained during the disease compared to EAE and vehicle control groups. On day 25, TPPU also caused an increase in the PPARα/β/γ and NLRC3 proteins and a decrease in the proteins of TLR4, MyD88, NF-κB p65, p-NF-κB p65, iNOS/nNOS, COX-2, NLRC4, ASC, caspase-1 p20, IL-1β, caspase-11 p20, NOX subunits (gp91phox and p47phox), and nitrotyrosine in addition to 14,15-DHET and IL-1β levels compared to EAE and vehicle control groups. Our findings suggest that pharmacological inhibition of sEH attenuates chronic EAE likely because of enhanced levels of anti-inflammatory EETs in addition to PPARα/β/γ and NLRC3 expression associated with suppressed inflammatory TLR4/MyD88/NF-κB signalling pathway, NLRC4/ASC/pro-caspase-1 inflammasome, caspase-11 inflammasome, and NOX activity that are responsible for inflammatory mediator formation in the CNS of mice.
Collapse
Affiliation(s)
- Merve Biliktu
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Sefika Pinar Senol
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Meryem Temiz-Resitoglu
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Demet Sinem Guden
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Mehmet Furkan Horat
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey
| | - Serhan Sevim
- Department of Neurology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33160, Yenisehir, Mersin, Turkey.
| |
Collapse
|
30
|
Spinal caspase-6 contributes to remifentanil-induced hyperalgesia via regulating CCL21/CXCR3 pathway in rats. Neurosci Lett 2020; 721:134802. [DOI: 10.1016/j.neulet.2020.134802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/22/2022]
|
31
|
Kanglexin, a novel anthraquinone compound, protects against myocardial ischemic injury in mice by suppressing NLRP3 and pyroptosis. Acta Pharmacol Sin 2020; 41:319-326. [PMID: 31645662 PMCID: PMC7468574 DOI: 10.1038/s41401-019-0307-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 09/06/2019] [Indexed: 01/02/2023]
Abstract
Pyroptosis is a form of inflammatory cell death that could be driven by the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation following myocardial infarction (MI). Emerging evidence suggests the therapeutic potential for ameliorating MI-induced myocardial damages by targeting NLRP3 and pyroptosis. In this study, we investigated the myocardial protection effect of a novel anthraquinone compound (4,5-dihydroxy-7-methyl-9,10-anthraquinone-2-ethyl succinate) named Kanglexin (KLX) in vivo and in vitro. Male C57BL/6 mice were pre-treated either with KLX (20, 40 mg· kg-1per day, intragastric gavage) or vehicle for 7 consecutive days prior to ligation of coronary artery to induce permanent MI. KLX administration dose-dependently reduced myocardial infarct size and lactate dehydrogenase release and improved cardiac function as compared to vehicle-treated mice 24 h after MI. We found that MI triggered NLRP3 inflammasome activation leading to conversion of interleukin-1β (IL-1β) and IL-18 into their active mature forms in the heart, which could expand the infarct size and drive cardiac dysfunction. We also showed that MI induced pyroptosis, as evidenced by increased DNA fragmentation, mitochondrial swelling, and cell membrane rupture, as well as increased levels of pyroptosis-related proteins, including gasdermin D, N-terminal GSDMD, and cleaved caspase-1. All these detrimental alterations were prevented by KLX. In hypoxia- or lipopolysaccharide (LPS)-treated neonatal mouse ventricular cardiomyocytes, we showed that KLX (10 μM) decreased the elevated levels of terminal deoxynucleotidyl transferase dUTP nick end labeling- and propidium iodide-positive cells, and pyroptosis-related proteins. We conclude that KLX prevents MI-induced cardiac damages and cardiac dysfunction at least partly through attenuating NLRP3 and subsequent cardiomyocyte pyroptosis, and it is worthy of more rigorous investigations for its potential for alleviating ischemic heart disease.
Collapse
|
32
|
Yin N, Gao Q, Tao W, Chen J, Bi J, Ding F, Wang Z. Paeoniflorin relieves LPS-induced inflammatory pain in mice by inhibiting NLRP3 inflammasome activation via transient receptor potential vanilloid 1. J Leukoc Biol 2020; 108:229-241. [PMID: 32083340 DOI: 10.1002/jlb.3ma0220-355r] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
LPS has been widely used to induce inflammatory pain, attributing to production of inflammatory cytokines and sensitization of nociceptors. Paeoniflorin (PF) possesses anti-nociceptive property, but its effect on LPS-induced inflammatory pain has not been investigated. In this study, we aimed to investigate the analgesic effect of PF on an inflammatory pain mouse model and explore the underlying mechanisms. LPS-induced inflammatory pain model was established in C57BL/6J mice after PF treatment. Then, thermal hyperalgesia, neutrophil infiltration, inflammatory cytokine production, intracellular Ca2+ levels, PKC activity, transient receptor potential vanilloid 1 (TRPV-1) expression, NF-κB transcription, and NLPR3 inflammasome activation were assessed by thermal withdrawal latency, histopathology, ELISA, intracellular Ca2+ concentration, immunohistochemistry, and Western blot, separately. PF significantly relieved inflammatory pain and paw edema in mice with LPS-induced inflammatory pain. Additionally, PF inhibited neutrophil infiltration, inflammatory cytokine production (IL-1β, TNF-α, and IL-6), intracellular Ca2+ levels, and PKC activity as well as suppressed TRPV-1 expression, NF-κB transcription, and NLPR3 inflammasome activation in the footpad tissue samples. Importantly, capsaicin (TRPV-1 agonists) obviously reversed the pain-relieving effect of PF, suggesting the involvement of TRPV-1 in the analgesic activity of PF. Our results indicated PF ameliorated LPS-induced inflammation and pain in mice by inhibiting TRPV-1-mediated NLRP3 inflammasome activation. These findings suggest that PF can be as a potential pharmacological agent for inflammatory pain and thus deserves more attention and further investigation.
Collapse
Affiliation(s)
- Nina Yin
- Department of Anatomy, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Qinghua Gao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Wenting Tao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Jiaojiao Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Jing Bi
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Fengmin Ding
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhigang Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
33
|
Chen F, Jiang G, Liu H, Li Z, Pei Y, Wang H, Pan H, Cui H, Long J, Wang J, Zheng Z. Melatonin alleviates intervertebral disc degeneration by disrupting the IL-1β/NF-κB-NLRP3 inflammasome positive feedback loop. Bone Res 2020; 8:10. [PMID: 32133213 PMCID: PMC7028926 DOI: 10.1038/s41413-020-0087-2] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 11/30/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response is induced by the overexpression of inflammatory cytokines, mainly interleukin (IL)-1β, and is one of the main causes of intervertebral disc degeneration (IVDD). NLR pyrin domain containing 3 (NLRP3) inflammasome activation is an important source of IL-1β. As an anti-inflammatory neuroendocrine hormone, melatonin plays various roles in different pathophysiological conditions. However, its roles in IVDD are still not well understood and require more examination. First, we demonstrated that melatonin delayed the progression of IVDD and relieved IVDD-related low back pain in a rat needle puncture IVDD model; moreover, NLRP3 inflammasome activation (NLRP3, p20, and IL-1β levels) was significantly upregulated in severely degenerated human discs and a rat IVDD model. Subsequently, an IL-1β/NF-κB-NLRP3 inflammasome activation positive feedback loop was found in nucleus pulposus (NP) cells that were treated with IL-1β. In these cells, expression of NLRP3 and p20 was significantly increased, NF-κB signaling was involved in this regulation, and mitochondrial reactive oxygen species (mtROS) production increased. Furthermore, we found that melatonin disrupted the IL-1β/NF-κB-NLRP3 inflammasome activation positive feedback loop in vitro and in vivo. Melatonin treatment decreased NLRP3, p20, and IL-1β levels by inhibiting NF-κB signaling and downregulating mtROS production. Finally, we showed that melatonin mediated the disruption of the positive feedback loop of IL-1β in vivo. In this study, we showed for the first time that IL-1β promotes its own expression by upregulating NLRP3 inflammasome activation. Furthermore, melatonin disrupts the IL-1β positive feedback loop and may be a potential therapeutic agent for IVDD.
Collapse
Affiliation(s)
- Fan Chen
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Guowei Jiang
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Hui Liu
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Zemin Li
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Yuxin Pei
- 2Department of Pediatric Intensive Care Unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Hua Wang
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Hehai Pan
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Haowen Cui
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Jun Long
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Jianru Wang
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Zhaomin Zheng
- 1Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
- 3Pain Research Center, Sun Yat-Sen University, Guangzhou, 510080 China
| |
Collapse
|
34
|
Pereira CA, Carlos D, Ferreira NS, Silva JF, Zanotto CZ, Zamboni DS, Garcia VD, Ventura DF, Silva JS, Tostes RC. Mitochondrial DNA Promotes NLRP3 Inflammasome Activation and Contributes to Endothelial Dysfunction and Inflammation in Type 1 Diabetes. Front Physiol 2020; 10:1557. [PMID: 32009974 PMCID: PMC6978691 DOI: 10.3389/fphys.2019.01557] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022] Open
Abstract
Background: NLRP3 inflammasome activation in response to several signals, including mitochondrial DNA (mDNA), regulates inflammatory responses by caspase-1 activation and interleukin-1β (IL-1β) release. Circulating mDNA is linked to micro and macrovascular complications in diabetes. However, a role for mDNA in endothelial dysfunction is not clear. We tested the hypothesis that mDNA contributes to diabetes-associated endothelial dysfunction and vascular inflammation via NLRP3 activation. Methods: Vascular reactivity, reactive oxygen species (ROS) generation, calcium (Ca2+) influx and caspase-1 and IL-1β activation were determined in mesenteric resistance arteries from normoglicemic and streptozotocin-induced diabetic C57BL/6 and NLRP3 knockout (Nlrp3-/- ) mice. Endothelial cells and mesenteric arteries were stimulated with mDNA from control (cmDNA) and diabetic (dmDNA) mice. Results: Diabetes reduced endothelium-dependent vasodilation and increased vascular ROS generation and caspase-1 and IL-1β activation in C57BL/6, but not in Nlrp3-/- mice. Diabetes increased pancreatic cytosolic mDNA. dmDNA decreased endothelium-dependent vasodilation. In endothelial cells, dmDNA activated NLRP3 via mitochondrial ROS and Ca2+ influx. Patients with type 1 diabetes exhibited increased circulating mDNA as well as caspase-1 and IL-1β activation. Conclusion: dmDNA activates endothelial NLRP3 inflammasome by mechanisms that involve Ca2+ influx and mitochondrial ROS generation. NLRP3 deficiency prevents diabetes-associated vascular inflammatory damage and endothelial dysfunction. Our study highlights the importance of NLRP3 inflammasome in diabetes-associated vascular dysfunction, which is key to diabetic complications.
Collapse
Affiliation(s)
- Camila A Pereira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Nathanne S Ferreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Josiane F Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Z Zanotto
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dario S Zamboni
- Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Valéria D Garcia
- Department of Experimental Psychology, Institute of Psychology, University of São Paulo, São Paulo, Brazil
| | - Dora Fix Ventura
- Department of Experimental Psychology, Institute of Psychology, University of São Paulo, São Paulo, Brazil
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
35
|
Cheng R, Wu Z, Li M, Shao M, Hu T. Interleukin-1β is a potential therapeutic target for periodontitis: a narrative review. Int J Oral Sci 2020; 12:2. [PMID: 31900383 PMCID: PMC6949296 DOI: 10.1038/s41368-019-0068-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/07/2019] [Accepted: 10/17/2019] [Indexed: 02/05/2023] Open
Abstract
Interleukin(IL)-1β, a pro-inflammatory cytokine, was elevated and participates in periodontitis. Not only the link between IL-1β and periodontitis was proved by clinical evidence, but also the increased IL-1β triggers a series of inflammatory reactions and promotes bone resorption. Currently, IL-1β blockage has been therapeutic strategies for autoimmune and autoinflammatory diseases such as rheumatoid arthritis, cryopyrin-associated periodic syndromes, gout and type II diabetes mellitus. It is speculated that IL-1β be a potential therapeutic target for periodontitis. The review focuses on the production, mechanism, present treatments and future potential strategies for IL-1β in periodontitis.
Collapse
Affiliation(s)
- Ran Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhiwu Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mingming Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Meiying Shao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tao Hu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
36
|
Ren J, Su D, Li L, Cai H, Zhang M, Zhai J, Li M, Wu X, Hu K. Anti-inflammatory effects of Aureusidin in LPS-stimulated RAW264.7 macrophages via suppressing NF-κB and activating ROS- and MAPKs-dependent Nrf2/HO-1 signaling pathways. Toxicol Appl Pharmacol 2019; 387:114846. [PMID: 31790703 DOI: 10.1016/j.taap.2019.114846] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 11/17/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022]
Abstract
Aureusidin, a naturally-occurring flavonoid, is found in various plants of Cyperaceae such as Heleocharis dulcis (Burm. f.) Trin., but its pharmacological effect and active mechanism are rarely reported. This study aimed to investigate the anti-inflammatory effect and action mechanism of Aureusidin in LPS-induced mouse macrophage RAW264.7 cells. The results suggested that lipopolysaccharide (LPS)-induced nitric oxide (NO), tumor necrosis factor-α (TNF-α) and prostaglandin E2 (PGE2) production were obviously inhibited by Aureusidin. Moreover, Aureusidin also significantly decreased the mRNA expression of various inflammatory factors in LPS-stimulated RAW264.7 cells. Furthermore, mechanistic studies showed that Aureusidin significantly inhibited nuclear transfer of nuclear factor-κB (NF-κB), while increasing the nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) as well as expression of Nrf2 target genes such as heme oxygenase (HO-1) and NAD(P)H:quinone oxidoreductase 1 (NQO1), but the addition of the HO-1 inhibitor Sn-protoporphyrin (Snpp) significantly abolished the anti-inflammatory effect of Aureusidin in LPS-stimulated RAW264.7 cells, confirming the view that HO-1 was involved in the anti-inflammatory effect. In addition, Aureusidin increased the levels of reactive oxygen species (ROS) and mitogen-activated protein kinase (MAPK) phosphorylation in RAW264.7 cells. Antioxidant N-acetylcysteine (NAC) or three MAPK inhibitors blocked the nuclear translocation of Nrf2 and HO-1 expression induced by Aureusidin, indicating that Aureusidin activated the Nrf2/HO-1 signaling pathway through ROS and MAPKs pathways. At the same time, co-treatment with the NAC blocked the phosphorylation of MAPKs. Results from molecular docking indicated that Aureusidin inhibited the NF-κB pathway by covalently binding to NF-κB. Thus, Aureusidin exerted the anti-inflammatory activity through blocking the NF-κB signaling pathways and activating the MAPKs and Nrf2/HO-1 signaling pathways. Based on the above results, Aureusidin may be an attractive therapeutic candidate for the inflammation-related diseases.
Collapse
Affiliation(s)
- Jie Ren
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China.
| | - Dan Su
- Changzhou No.2 People's Hospital, Changzhou, Jiangsu 213164, People's Republic of China.
| | - Lixia Li
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China
| | - Heng Cai
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China
| | - Meiju Zhang
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China
| | - Jingchen Zhai
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China
| | - Minyue Li
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China
| | - Xinyue Wu
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China
| | - Kun Hu
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, Jiangsu 213164, People's Republic of China
| |
Collapse
|
37
|
Hernandez A, Patil NK, Stothers CL, Luan L, McBride MA, Owen AM, Burelbach KR, Williams DL, Sherwood ER, Bohannon JK. Immunobiology and application of toll-like receptor 4 agonists to augment host resistance to infection. Pharmacol Res 2019; 150:104502. [PMID: 31689522 DOI: 10.1016/j.phrs.2019.104502] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/04/2019] [Accepted: 10/15/2019] [Indexed: 12/19/2022]
Abstract
Infectious diseases remain a threat to critically ill patients, particularly with the rise of antibiotic-resistant bacteria. Septic shock carries a mortality of up to ∼40% with no compelling evidence of promising therapy to reduce morbidity or mortality. Septic shock survivors are also prone to nosocomial infections. Treatment with toll-like receptor 4 (TLR4) agonists have demonstrated significant protection against common nosocomial pathogens in various clinically relevant models of infection and septic shock. TLR4 agonists are derived from a bacteria cell wall or synthesized de novo, and more recently novel small molecule TLR4 agonists have also been developed. TLR4 agonists augment innate immune functions including expansion and recruitment of innate leukocytes to the site of infection. Recent studies demonstrate TLR4-induced leukocyte metabolic reprogramming of cellular metabolism to improve antimicrobial function. Metabolic changes include sustained augmentation of macrophage glycolysis, mitochondrial function, and tricarboxylic acid cycle flux. These findings set the stage for the use of TLR4 agonists as standalone therapeutic agents or antimicrobial adjuncts in patient populations vulnerable to nosocomial infections.
Collapse
Affiliation(s)
- Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cody L Stothers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Margaret A McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine R Burelbach
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David L Williams
- Department of Surgery, East Tennessee State University, James H. Quillen College of Medicine, Johnson City, TN, USA
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
38
|
Pharmacological inhibition of the NLRP3 inflammasome as a potential target for cancer-induced bone pain. Pharmacol Res 2019; 147:104339. [DOI: 10.1016/j.phrs.2019.104339] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 12/27/2022]
|
39
|
Zhong X, Liu M, Yao W, Du K, He M, Jin X, Jiao L, Ma G, Wei B, Wei M. Epigallocatechin-3-Gallate Attenuates Microglial Inflammation and Neurotoxicity by Suppressing the Activation of Canonical and Noncanonical Inflammasome via TLR4/NF-κB Pathway. Mol Nutr Food Res 2019; 63:e1801230. [PMID: 31374144 DOI: 10.1002/mnfr.201801230] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 06/30/2019] [Indexed: 01/09/2023]
Abstract
SCOPE In this study, it has been investigated whether the neuroprotective efficacy of epigallocatechin-3-gallate (EGCG) is mediated by inhibition of canonical and noncanonical inflammasome activation via toll-like receptor 4 (TLR4)/NF-κB pathway both in LPS+Aβ-induced microglia in vitro and in APP/PS1 mice in vivo. METHODS AND RESULTS In BV2 cells, EGCG inhibits the expressions of Iba-1, cleaved IL-1β, and cleaved IL-18 induced by LPS+Aβ. Then, the supernatants are used to treat SH-SY5Y cells, and EGCG treatment significantly recovers the neurotoxicity from LPS+Aβ-induced microglial conditioned media. Subsequently, it has been found that EGCG reduces the microglial expressions of caspase-1 p20, NLRP3, and caspase-11 p26. Furthermore, the expression levels of Toll-like receptor 4 (TLR4), p-IKK/IKK, and p-NF-κB/NF-κB were decreased after EGCG treatment. As expected, when a caspase-1 specific inhibitor Z-YVAD-FMK, and an IKK and caspase-11 inhibitor wedelolactone are used for blocking, Z-YVAD-FMK and wedelolactone exacerbate the inhibitory efficacy than using EGCG alone. Finally, consistent with the results obtained in BV2 cells, EGCG treatment reduces microglial inflammation and neurotoxicity by suppressing the activation of canonical NLRP3 and noncanonical caspase-11-dependent inflammasome via TLR4/NF-κB pathway in LPS+Aβ-induced rat primary microglia and hippocampus of APP/PS1 mice. CONCLUSION EGCG attenuates microglial inflammation and neurotoxicity by inhibition of canonical NLRP3 and noncanonical caspase-11-dependent inflammasome activation via TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Xin Zhong
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Mingyan Liu
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Weifan Yao
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Ke Du
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Miao He
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Xin Jin
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Linchi Jiao
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Guowei Ma
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Binbin Wei
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| | - Minjie Wei
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang, 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, 110122, China
| |
Collapse
|
40
|
Zielinski MR, Systrom DM, Rose NR. Fatigue, Sleep, and Autoimmune and Related Disorders. Front Immunol 2019; 10:1827. [PMID: 31447842 PMCID: PMC6691096 DOI: 10.3389/fimmu.2019.01827] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022] Open
Abstract
Profound and debilitating fatigue is the most common complaint reported among individuals with autoimmune disease, such as systemic lupus erythematosus, multiple sclerosis, type 1 diabetes, celiac disease, chronic fatigue syndrome, and rheumatoid arthritis. Fatigue is multi-faceted and broadly defined, which makes understanding the cause of its manifestations especially difficult in conditions with diverse pathology including autoimmune diseases. In general, fatigue is defined by debilitating periods of exhaustion that interfere with normal activities. The severity and duration of fatigue episodes vary, but fatigue can cause difficulty for even simple tasks like climbing stairs or crossing the room. The exact mechanisms of fatigue are not well-understood, perhaps due to its broad definition. Nevertheless, physiological processes known to play a role in fatigue include oxygen/nutrient supply, metabolism, mood, motivation, and sleepiness-all which are affected by inflammation. Additionally, an important contributing element to fatigue is the central nervous system-a region impacted either directly or indirectly in numerous autoimmune and related disorders. This review describes how inflammation and the central nervous system contribute to fatigue and suggests potential mechanisms involved in fatigue that are likely exhibited in autoimmune and related diseases.
Collapse
Affiliation(s)
- Mark R Zielinski
- Veterans Affairs Boston Healthcare System, Boston, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - David M Systrom
- Department of Medicine, Harvard Medical School, Boston, MA, United States.,Department of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Noel R Rose
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
41
|
Ward R, Li W, Abdul Y, Jackson L, Dong G, Jamil S, Filosa J, Fagan SC, Ergul A. NLRP3 inflammasome inhibition with MCC950 improves diabetes-mediated cognitive impairment and vasoneuronal remodeling after ischemia. Pharmacol Res 2019; 142:237-250. [PMID: 30818045 PMCID: PMC6486792 DOI: 10.1016/j.phrs.2019.01.035] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/07/2019] [Accepted: 01/17/2019] [Indexed: 12/11/2022]
Abstract
Diabetes increases the risk and worsens the progression of cognitive impairment via the greater occurrence of small vessel disease and stroke. Yet, the underlying mechanisms are not fully understood. It is now accepted that cardiovascular health is critical for brain health and any neurorestorative approaches to prevent/delay cognitive deficits should target the conceptual neurovascular unit (NVU) rather than neurons alone. We have recently shown that there is augmented hippocampal NVU remodeling after a remote ischemic injury in diabetes. NLRP3 inflammasome signaling has been implicated in the development of diabetes and neurodegenerative diseases, but little is known about the impact of NLRP3 activation on functional and structural interaction within the NVU of hippocampus, a critical part of the brain that is involved in forming, organizing, and storing memories. Endothelial cells are at the center of the NVU and produce trophic factors such as brain derived neurotrophic factor (BDNF) contributing to neuronal survival, known as vasotrophic coupling. Therefore, the aims of this study focused on two hypotheses: 1) diabetes negatively impacts hippocampal NVU remodeling and worsens cognitive outcome after stroke, and 2) NLRP3 inhibition with MCC950 will improve NVU remodeling and cognitive outcome following stroke via vasotrophic (un)coupling between endothelial cells and hippocampal neurons. Stroke was induced through a 90-min transient middle cerebral artery occlusion (MCAO) in control and high-fat diet/streptozotocin-induced (HFD/STZ) diabetic male Wistar rats. Saline or MCC950 (3 mg/kg), an inhibitor of NLRP3, was injected at 1 and 3 h after reperfusion. Cognition was assessed over time and neuronal density, blood-brain barrier (BBB) permeability as well as NVU remodeling (aquaporin-4 [AQP4] polarity) was measured on day 14 after stroke. BDNF was measured in endothelial and hippocampal neuronal cultures under hypoxic and diabetes-mimicking condition with and without NLRP3 inhibition. Diabetes increased neuronal degeneration and BBB permeability, disrupted AQP4 polarity, impaired cognitive function and amplified NLRP3 activation after ischemia. Inhibition with MCC950 improved cognitive function and vascular integrity after stroke in diabetic animals and prevented hypoxia-mediated decrease in BDNF secretion. These results are the first to provide essential data showing MCC950 has the potential to become a therapeutic to prevent neurovascular remodeling and worsened cognitive decline in diabetic patients following stroke.
Collapse
Affiliation(s)
- Rebecca Ward
- Departments of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Weiguo Li
- Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, United States
| | - Yasir Abdul
- Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, United States
| | - LaDonya Jackson
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Guangkuo Dong
- Physiology, Augusta University, Augusta, GA, United States
| | - Sarah Jamil
- Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jessica Filosa
- Physiology, Augusta University, Augusta, GA, United States
| | - Susan C Fagan
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Adviye Ergul
- Physiology, Augusta University, Augusta, GA, United States; Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
42
|
Activation of Sphingosine-1-Phosphate Receptor 1 in the Spinal Cord Produces Mechanohypersensitivity Through the Activation of Inflammasome and IL-1β Pathway. THE JOURNAL OF PAIN 2019; 20:956-964. [PMID: 30802544 DOI: 10.1016/j.jpain.2019.02.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/29/2019] [Accepted: 02/16/2019] [Indexed: 12/20/2022]
Abstract
Sphingosine-1-phosphate (S1P) receptor 1 subtype (S1PR1) activation by its ligand S1P in the dorsal horn of the spinal cord causes mechanohypersensitivity. The cellular and molecular pathways remain poorly understood. We report that the activation of S1PR1 with an intrathecal injection of the highly selective S1PR1 agonist SEW2871 led to the development of mechanoallodynia by activating the nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome (increased expression of NLRP3, cleaved caspase 1 and mature IL-1β) in the dorsal horn of the spinal cord. The functional S1PR1 antagonist FTY720 blocked NLRP3 activation and IL-1β production. Moreover, inhibiting IL-10 signaling with an intrathecal injection of an anti-IL-10 antibody attenuated the beneficial effects exerted by FTY720. This finding suggests that disrupting S1PR1 signaling engages beneficial IL-10-dependent pathways. Notably, we found that mice with astrocyte-specific deletions of S1pr1 did not develop mechanoallodynia after intrathecal injection of SEW2871 and exhibited decreased levels of cleaved caspase 1, identifying astrocytes as a key cellular locus for S1PR1 activity. Our findings provide novel mechanistic insights on how S1PR1 activation in the spinal cord contributes to the development of nociception while identifying the cellular substrate for these activities. PERSPECTIVE: This study is the first to link the activation of NLRP3 and IL-1β signaling in the spinal cord and S1PR1 signaling in astrocytes to the development of S1PR1-evoked mechanoallodynia. These findings provide critical basic science insights to support the development of therapies targeted toward S1PR1.
Collapse
|
43
|
Zhang WJ, Fang ZM, Liu WQ. NLRP3 inflammasome activation from Kupffer cells is involved in liver fibrosis of Schistosoma japonicum-infected mice via NF-κB. Parasit Vectors 2019; 12:29. [PMID: 30635040 PMCID: PMC6330481 DOI: 10.1186/s13071-018-3223-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND NOD-like receptor protein 3 (NLRP3) inflammasome was reported as expressed in schistosomiasis-induced liver fibrosis (SSLF). We used an NLRP3 inflammasome inhibitor, MCC950, to investigate whether it inhibited liver fibrosis, and explored the preliminary molecular mechanism. METHODS BALB/c mice were infected with 15 cercariae through the abdominal skin. They received intraperitoneal injections of MCC950 on the day of infection and at day 22 post-infection. We examined their SSLF phenotype and the effect on liver fibrosis, primary Kupffer cells (KCs), and HSCs. Human hepatic stellate cell lines (human LX-2 cells) were treated with soluble egg antigen (SEA) released from the eggs. We then determined the expression of NLRP3 inflammasome and liver fibrosis-associated markers, liver granuloma and ALT/AST. RESULTS NLRP3 inflammasome expression in the liver was significantly increased, and eosinophilic granuloma and collagen deposition were found around the eggs in mice infected for 56 days. Additionally, IL-1β, ALT/AST in plasma, and NF-κB in liver tissue and in KCs were all greatly significantly increased. The above-mentioned indicators were largely reduced in mice treated with MCC950 on the day of infection. In vitro, lipopolysaccharide (LPS)/SEA could induce LX-2 cells to express NLRP3 and fibrosis markers, and the SEA-treated group was reversed by MCC950. Furthermore, NLRP3 inflammasome and liver fibrosis-associated markers were both increased in the primary KCs and HSCs isolated from infected mice. However, this effect was not observed in the same cells from the mice treated with MCC950 on the day of infection. Contrary to the aforementioned results, MCC950 treatment at day 22 post-infection aggravated this process. Surprisingly, NLRP3 inflammasome was involved in liver fibrosis mostly from KCs. CONCLUSIONS MCC950 acts dually on SSLF pathology and fibrosis in infected mice. Although MCC950 treatment improved SSLF on the day of infection, it exacerbated the pathological effects at day 22 post-infection. These dual effects were mediated via NF-κB. Moreover, NLRP3 inflammasome mainly came from KCs. Our results suggest that blocking NLRP3 on the day of infection may prove to be a promising direction in preventing SSLF.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, Hubei, People's Republic of China
| | - Zheng-Ming Fang
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, Hubei, People's Republic of China
| | - Wen-Qi Liu
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
44
|
Pan Z, Cui M, Dai G, Yuan T, Li Y, Ji T, Pan Y. Protective Effect of Anthocyanin on Neurovascular Unit in Cerebral Ischemia/Reperfusion Injury in Rats. Front Neurosci 2018; 12:947. [PMID: 30618576 PMCID: PMC6297832 DOI: 10.3389/fnins.2018.00947] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/29/2018] [Indexed: 12/17/2022] Open
Abstract
Treating cerebral ischemia continues to be a clinical challenge. Studies have shown that the neurovascular unit (NVU), as the central structural basis, plays a key role in cerebral ischemia. Here, we report that anthocyanin, a safe and natural antioxidant, could inhibit apoptosis and inflammation to protect NVU in rats impaired by middle cerebral artery occlusion/reperfusion (MCAO/R). Administration of anthocyanin significantly reduced infarct volume and neurological scores in MCAO/R rats. Anthocyanin could also markedly ameliorate cerebral edema and reduce the concentration of Evans blue (EB) by inhibiting MMP-9. Moreover, anthocyanin alleviated apoptotic injury resulting from MCAO/R through the regulation of Bcl-2 family proteins. The levels of inflammation-related molecules including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), which were over-expressed with MCAO/R, were decreased by anthocyanin. In addition, Nuclear factor-kappa B (NF-κB) and the NLRP3 inflammasome pathway might be involved in the anti-inflammatory effect of anthocyanin. In conclusion, anthocyanin could protect the NVU through multiple pathways, and play a protective role in cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Zihao Pan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengdi Cui
- Thyroid and Breast Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoliang Dai
- Department of Clinical Pharmacology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianjie Yuan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuhua Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tuo Ji
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Pan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
45
|
Pinellia pedatisecta lectin exerts a proinflammatory activity correlated with ROS-MAPKs/NF-κB pathways and the NLRP3 inflammasome in RAW264.7 cells accompanied by cell pyroptosis. Int Immunopharmacol 2018; 66:1-12. [PMID: 30415189 DOI: 10.1016/j.intimp.2018.11.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
Pinellia pedatisecta, a widely used herb in Chinese medicine, has proinflammatory toxicity related to its Pinellia pedatisecta lectin (PPL), but the mechanism is still unknown. However, for safer use, it is necessary to clarify its proinflammatory mechanism. Herein, we studied the mechanism in RAW264.7 cells. PPL decreased the mitochondrial membrane potential (MMP) and increased the outflow of calcium, accompanied by the overproduction of reactive oxygen species (ROS), which resulted in the activation of the MAPK and NF-κB pathways and the release of IL-1β. The maturation of IL-1β relied on caspase-1 p20, the active caspase-1, as demonstrated by adding caspase-1 inhibitor. While caspase-1 was associated with the activation of the NLRP3 inflammasome, we further found that the stimulation of PPL also contributed to the activation. In addition, TXNIP was downregulated, whereas NLRP3/caspase-1 p20/ASC was upregulated, and there was binding of TXNIP with NLRP3. There was also binding of NLRP3 with ASC and caspase-1. Further, we found that N-acetylcysteine (NAC), an ROS scavenger, could inhibit the PPL-stimulated activation of these pathways and the release of IL-1β. Moreover, PPL led to cell pyroptosis with pyknotic nuclei and plasma membrane rupture, which could be inhibited by NAC. All of these findings demonstrated an important role of ROS in the inflammation caused by PPL. Taken together, our data provide new mechanistic insights into the possible endogenous signaling pathways involved in the inflammation of RAW264.7 cells, stimulated by PPL.
Collapse
|
46
|
Recent Advances in the Molecular Mechanisms Underlying Pyroptosis in Sepsis. Mediators Inflamm 2018; 2018:5823823. [PMID: 29706799 PMCID: PMC5863298 DOI: 10.1155/2018/5823823] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/22/2018] [Indexed: 12/25/2022] Open
Abstract
Sepsis is recognized as a life-threatening organ dysfunctional disease that is caused by dysregulated host responses to infection. Up to now, sepsis still remains a dominant cause of multiple organ dysfunction syndrome (MODS) and death among severe condition patients. Pyroptosis, originally named after the Greek words “pyro” and “ptosis” in 2001, has been defined as a specific programmed cell death characterized by release of inflammatory cytokines. During sepsis, pyroptosis is required for defense against bacterial infection because appropriate pyroptosis can minimize tissue damage. Even so, pyroptosis when overactivated can result in septic shock, MODS, or increased risk of secondary infection. Proteolytic cleavage of gasdermin D (GSDMD) by caspase-1, caspase-4, caspase-5, and caspase-11 is an essential step for the execution of pyroptosis in activated innate immune cells and endothelial cells stimulated by cytosolic lipopolysaccharide (LPS). Cleaved GSDMD also triggers NACHT, LRR, and PYD domain-containing protein (NLRP) 3-mediated activation of caspase-1 via an intrinsic pathway, while the precise mechanism underlying GSDMD-induced NLRP 3 activation remains unclear. Hence, this study provides an overview of the recent advances in the molecular mechanisms underlying pyroptosis in sepsis.
Collapse
|
47
|
Del Campo JA, Gallego P, Grande L. Role of inflammatory response in liver diseases: Therapeutic strategies. World J Hepatol 2018; 10:1-7. [PMID: 29399273 PMCID: PMC5787673 DOI: 10.4254/wjh.v10.i1.1] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/21/2017] [Accepted: 01/15/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammation and tumorigenesis are tightly linked pathways impacting cancer development. Inflammasomes are key signalling platforms that detect pathogenic microorganisms, including hepatitis C virus (HCV) infection, and sterile stressors (oxidative stress, insulin resistance, lipotoxicity) able to activate pro-inflammatory cytokines interleukin-1β and IL-18. Most of the inflammasome complexes that have been described to date contain a NOD-like receptor sensor molecule. Redox state and autophagy can regulate inflammasome complex and, depending on the conditions, can be either pro- or anti-apoptotic. Acute and chronic liver diseases are cytokine-driven diseases as several proinflammatory cytokines (IL-1α, IL-1β, tumor necrosis factor-alpha, and IL-6) are critically involved in inflammation, steatosis, fibrosis, and cancer development. NLRP3 inflammasome gain of function aggravates liver disease, resulting in severe liver fibrosis and highlighting this pathway in the pathogenesis of non-alcoholic fatty liver disease. On the other hand, HCV infection is the primary catalyst for progressive liver disease and development of liver cancer. It is well established that HCV-induced IL-1β production by hepatic macrophages plays a critical and central process that promotes liver inflammation and disease. In this review, we aim to clarify the role of the inflammasome in the aggravation of liver disease, and how selective blockade of this main pathway may be a useful strategy to delay fibrosis progression in liver diseases.
Collapse
Affiliation(s)
- José A Del Campo
- Department of Digestive Diseases, Valme University Hospital and CIBERehd, Sevilla 41014, Spain
| | - Paloma Gallego
- Department of Digestive Diseases, Valme University Hospital and CIBERehd, Sevilla 41014, Spain
| | - Lourdes Grande
- Department of Digestive Diseases, Valme University Hospital and CIBERehd, Sevilla 41014, Spain
| |
Collapse
|
48
|
Che H, Li Q, Zhang T, Ding L, Zhang L, Shi H, Yanagita T, Xue C, Chang Y, Wang Y. A comparative study of EPA-enriched ethanolamine plasmalogen and EPA-enriched phosphatidylethanolamine on Aβ42 induced cognitive deficiency in a rat model of Alzheimer's disease. Food Funct 2018; 9:3008-3017. [DOI: 10.1039/c8fo00643a] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The possible molecular mechanism of EPA-pPE and EPA-PE on AD.
Collapse
Affiliation(s)
- Hongxia Che
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Qian Li
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Tiantian Zhang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Lin Ding
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Lingyu Zhang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Haohao Shi
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Teruyoshi Yanagita
- Department of Health and Nutrition Science
- Nishikyushu University
- Kanzaki
- Japan
| | - Changhu Xue
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
- Qingdao National Laboratory for Marine Science and Technology
| | - Yaoguang Chang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Yuming Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
- Qingdao National Laboratory for Marine Science and Technology
| |
Collapse
|
49
|
3-(Naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride attenuates NLRP3 inflammasome-mediated signaling pathway in lipopolysaccharide-stimulated BV2 microglial cells. Biochem Biophys Res Commun 2018; 495:151-156. [DOI: 10.1016/j.bbrc.2017.10.131] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
|
50
|
Du SH, Qiao DF, Chen CX, Chen S, Liu C, Lin Z, Wang H, Xie WB. Toll-Like Receptor 4 Mediates Methamphetamine-Induced Neuroinflammation through Caspase-11 Signaling Pathway in Astrocytes. Front Mol Neurosci 2017; 10:409. [PMID: 29311802 PMCID: PMC5733023 DOI: 10.3389/fnmol.2017.00409] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/27/2017] [Indexed: 02/02/2023] Open
Abstract
Methamphetamine (METH) is an amphetamine-typed stimulant drug that is increasingly being abused worldwide. Previous studies have shown that METH toxicity is systemic, especially targeting dopaminergic neurons in the central nervous system (CNS). However, the role of neuroinflammation in METH neurotoxicity remains unclear. We hypothesized that Toll-like receptor 4 (TLR4) and Caspase-11 are involved in METH-induced astrocyte-related neuroinflammation. We tested our hypothesis by examining the changes of TLR4 and Caspase-11 protein expression in primary cultured C57BL/6 mouse astrocytes and in the midbrain and striatum of mice exposed to METH with western blot and double immunofluorescence labeling. We also determined the effects of blocking Caspase-11 expression with wedelolactone (a specific inhibitor of Caspase-11) or siRNA on METH-induced neuroinflammation in astrocytes. Furthermore, we determined the effects of blocking TLR4 expression with TAK-242 (a specific inhibitor of TLR4) or siRNA on METH-induced neuroinflammation in astrocytes. METH exposure increased Caspase-11 and TLR4 expression both in vitro and in vivo, with the effects in vitro being dose-dependent. Inhibition of Caspase-11 expression with either wedelolactone or siRNAs reduced the expression of inflammasome NLRP3 and pro-inflammatory cytokines. In addition, blocking TLR4 expression inhibited METH-induced activation of NF-κB and Caspase-11 in vitro and in vivo, suggesting that TLR4-Caspase-11 pathway is involved in METH-induced neuroinflammation. These results indicate that Caspase-11 and TLR4 play an important role in METH-induced neuroinflammation and may be potential gene targets for therapeutics in METH-caused neurotoxicity.
Collapse
Affiliation(s)
- Si-Hao Du
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Dong-Fang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chuan-Xiang Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Si Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou, China
| | - Zhoumeng Lin
- Department of Anatomy and Physiology, Institute of Computational Comparative Medicine (ICCM), College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|