1
|
Zhang Q, Zhang X, He Q, Tian Y, Liu Z. Cimifugin Alleviates Chronic Constriction Injury of the Sciatic Nerve by Suppressing Inflammatory Response and Schwann Cell Apoptosis. Cell Biochem Biophys 2025; 83:823-836. [PMID: 39392551 DOI: 10.1007/s12013-024-01513-4] [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] [Accepted: 08/28/2024] [Indexed: 10/12/2024]
Abstract
Inflammation and Schwann cell apoptosis play critical roles in neuropathic pain after sciatic nerve injury. This study aimed to explore the function and mechanism of cimifugin in lipopolysaccharide (LPS)-stimulated rat Schwann cells and sciatic nerves of rats treated with chronic constriction injury (CCI). Thermal, mechanical and cold hyperalgesia of rats in response to cimifugin or mecobalamin (the positive drug control) treatment were evaluated through behavioral tests. H&E staining of sciatic nerves was performed for pathological observation. ELISA was conducted to assess concentrations of inflammatory cytokines in rat serum and sciatic nerves. The intensity of S100β in sciatic nerves was determined using immunohistochemistry. Flow cytometry analysis was conducted for detection of Schwann cell apoptosis. RT-qPCR was performed to measure mRNA levels of inflammatory factors in Schwann cells. Immunofluorescence staining was performed to detect cellular p65/NF-κB activity. Western blotting was performed to quantify protein levels of apoptotic markers and factors associated with the NF-κB and MAPK pathways in rat nerves and Schwann cells. As shown by experimental data, cimifugin mitigated thermal, mechanical and cold hyperalgesia of CCI rats. Cimifugin repressed inflammatory cell infiltration, reduced proinflammatory cytokine levels while increasing anti-inflammatory factor (IL-10) level in serum or sciatic nerves of CCI rats. Cimifugin enhanced S100β expression and downregulated apoptotic markers in vivo. The anti-inflammatory and anti-apoptotic properties of cimifugin were verified in the LPS-stimulated Schwann cells. Moreover, cimifugin suppressed nuclear translocation of p65 NF-κB in vitro and repressed the phosphorylation of IκB, p65 NF-κB, p38 MAPK, ERK1/2, as well as JNK in CCI rats. In conclusion, cimifugin alleviates neuropathic pain after sciatica by suppressing inflammatory response and Schwann cell apoptosis via inactivation of NF-κB and MAPK pathways.
Collapse
Affiliation(s)
- Qijuan Zhang
- Department of rehabilitation medicine, Wuhan Orthopaedic Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Wuhan Sports University), Wuhan, 430070, China.
| | - Xiaoli Zhang
- Wuhan Fiberhome technical service Co. Ltd, Wuhan, 430000, China
| | - Qing He
- Department of rehabilitation medicine, Wuhan Orthopaedic Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Wuhan Sports University), Wuhan, 430070, China
| | - Yu Tian
- Department of rehabilitation medicine, Wuhan Orthopaedic Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Wuhan Sports University), Wuhan, 430070, China
| | - Zhengmao Liu
- Department of rehabilitation medicine, Wuhan Orthopaedic Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Wuhan Sports University), Wuhan, 430070, China
| |
Collapse
|
2
|
Yang S, Guan Y, Zheng C, Xia X, Ma X, Jiang J. FOXO3-induced microRNA-128-3p promotes the progression of spinal cord injury in mice via regulating NLRP3 inflammasome-mediated pyroptosis. Front Immunol 2025; 16:1526721. [PMID: 40061945 PMCID: PMC11885150 DOI: 10.3389/fimmu.2025.1526721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/03/2025] [Indexed: 05/13/2025] Open
Abstract
Background Spinal cord injury (SCI) remains a severe condition with an extremely high disability rate and complex pathophysiologic mechanisms. Pyroptosis, an inflammatory form of cell death triggered by certain inflammasomes, has a key role in a variety of inflammatory diseases, including SCI. However, it is unclear whether microRNAs (miRNAs), novel regulators in the SCI, are involved in SCI-induced pyroptosis. Methods Two GEO miRNA expression profiles (GSE158195 and GSE90452) were downloaded, and the differentially expressed miRNAs were analyzed by bioinformatics methods. An in vivo animal model and an in vitro cellular model of SCI were constructed in female C57BL/6 mice and BV-2 cells for studying the possible roles of FOXO3, miR-128-3p and NLRP3-mediated pyroptosis in SCI. Markers of ROS, cell pyroptosis and inflammation were measured by RT-qPCR, Western blotting, immunofluorescence, flow cytometry, and enzyme-linked immunosorbent assays. Histopathological changes in spinal cord tissue were detected using hematoxylin and eosin and immunohistochemical. The Basso-Beattie-Bresnahan (BBB) score was used to evaluate the motor function of mice in each group. Results Bioinformatics analysis of GSE158195 and GSE90452 datasets revealed a significant downregulation of miR-128-3p, a phenomenon that was consistently observed in the SCI mice model. Functionally, miR-128-3p upregulation improved functional behavioral recovery, relieved pathological injury, repressed oxidative stress, and alleviated pyroptosis and inflammation in the mouse SCI models. We also confirmed that Thioredoxin-interacting protein (TXNIP) was the target gene of miR-128-3p, and overexpression of TXNIP can effectively reverse the improvement of miR-128-3p in SCI cell model. Moreover, we found that transcription factor FOXO3 facilitated miR-128-3p expression, and its overexpression resulted in similar effects of miR-128-3p in the SCI cell model. Conclusion To the best of our knowledge, this is the first report demonstrating miR-128-3p improved secondary injury in SCI through the modulation of cell pyroptosis pathway. Our results suggest that FOXO3/miR-128-3p/TXNIP/NLRP3-mediated pyroptosis axis may be a potential therapeutic target for SCI.
Collapse
Affiliation(s)
| | | | - Chaojun Zheng
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinlei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | | | | |
Collapse
|
3
|
Ji S, Chen D, Ding F, Gu X, Xue Q, Zhou C, Cao M, Yu S. Salidroside exerts neuroprotective effects on retrograde neuronal death following neonatal axotomy via activation of PI3K/Akt pathway and deactivation of p38 MAPK pathway. Toxicol Appl Pharmacol 2025; 494:117178. [PMID: 39617258 DOI: 10.1016/j.taap.2024.117178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
Salidroside, a glucoside of tyrosol, is a powerful active ingredient extracted from the Chinese herb medicine Rhodiola rosea L.. As a neuroprotective agent, the application of salidroside in combination with neural tissue engineering has recently attracted much attention in peripheral nerve repair and reconstruction. However, the cellular and molecular mechanisms by which salidroside promotes nerve regeneration remain to be elucidated. We aim to evaluate the long-term neuroprotective potential of salidroside in an experimental rat model of neonatal sciatic nerve crush injury, with a focus on target-deprived neuronal death and the mechanisms involved. Behavioral analysis showed that salidroside dose-dependently improved voluntary hindlimb behavior and rod rotation ability following neonatal axotomy during an 8-week observation period. According to electrophysiology, Fluoro-Gold retrograde tracing, histological and immunohistochemical analyses, salidroside significantly improved nerve regeneration and reinnervation. Nissle and TUNEL staining, as well as caspase-3 activation assay indicated a beneficial effect of salidroside on retrograde loss and apoptosis of motoneurons within 2 weeks after axotomy. qPCR, ELISA and oxidative stress experiments revealed that salidroside improved the imbalance of spinal microenvironment, including oxidative stress and down-regulation of neurotrophic factors. Western blotting analysis showed that salidroside enhanced the activation of PI3K/Akt and inhibited the p38 MAPK signaling pathway following axotomy. The oxidative stress and axonal disconnection/regeneration models of primary motoneurons in vitro further confirmed the involvement of these two pathways in the neuroprotective effects of salidroside. These data provide a theoretical basis for the application of salidroside in peripheral nerve repair and reconstruction.
Collapse
Affiliation(s)
- Shengtao Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Daiyue Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Qiu Xue
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China; Department of General Surgery, Nantong Tumor Hospital, Nantong Fifth People's Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Chun Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China.
| | - Maohong Cao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China.
| | - Shu Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China.
| |
Collapse
|
4
|
Dhir S, Derue H, Ribeiro-da-Silva A. Temporal changes of spinal microglia in murine models of neuropathic pain: a scoping review. Front Immunol 2024; 15:1460072. [PMID: 39735541 PMCID: PMC11671780 DOI: 10.3389/fimmu.2024.1460072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/18/2024] [Indexed: 12/31/2024] Open
Abstract
Neuropathic pain (NP) is an ineffectively treated, debilitating chronic pain disorder that is associated with maladaptive changes in the central nervous system, particularly in the spinal cord. Murine models of NP looking at the mechanisms underlying these changes suggest an important role of microglia, the resident immune cells of the central nervous system, in various stages of disease progression. However, given the number of different NP models and the resource limitations that come with tracking longitudinal changes in NP animals, many studies fail to truly recapitulate the patterns that exist between pain conditions and temporal microglial changes. This review integrates how NP studies are being carried out in murine models and how microglia changes over time can affect pain behavior in order to inform better study design and highlight knowledge gaps in the field. 258 peer-reviewed, primary source articles looking at spinal microglia in murine models of NP were selected using Covidence. Trends in the type of mice, statistical tests, pain models, interventions, microglial markers and temporal pain behavior and microglia changes were recorded and analyzed. Studies were primarily conducted in inbred, young adult, male mice having peripheral nerve injury which highlights the lack of generalizability in the data currently being collected. Changes in microglia and pain behavior, which were both increased, were tested most commonly up to 2 weeks after pain initiation despite aberrant microglia activity also being recorded at later time points in NP conditions. Studies using treatments that decrease microglia show decreased pain behavior primarily at the 1- and 2-week time point with many studies not recording pain behavior despite the involvement of spinal microglia dysfunction in their development. These results show the need for not only studying spinal microglia dynamics in a variety of NP conditions at longer time points but also for better clinically relevant study design considerations.
Collapse
Affiliation(s)
- Simran Dhir
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Hannah Derue
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
5
|
Luo F, Zhang J, Miao Y, Wu D, Shen H, Lu M. Paeoniflorin regulates microglia-astrocyte crosstalk, inhibits inflammatory response, and alleviates neuropathic pain through HSP90AA1/HMGB1 signaling pathway. Int J Biochem Cell Biol 2024; 176:106675. [PMID: 39395636 DOI: 10.1016/j.biocel.2024.106675] [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: 05/29/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Given the unclear, complex pathogenesis of neuropathic pain and the potential of paeoniflorin in relieving neuropathic pain, this study aimed to further clarify the therapeutic effect of paeoniflorin on neuropathic pain and to preliminarily explore the possible protective mechanisms of paeoniflorin. Chronic constrictive injury-induced Sprague Dawley rats and lipopolysaccharide-induced BV-2 cells were used for in vivo and in vitro experiments, respectively. The exosome uptake assay of mouse astrocytes (PKH-67 fluorescent labeling) and the mechanical nociceptive assay (the von Frey fibrous filaments) were performed. The effects of paeoniflorin and its downstream mechanisms on microglial and astrocyte activation, inflammation-associated proteins and exosome marker were determined. Paeoniflorin alleviated mechanical abnormal pain, decreased levels of ionized calcium binding adapter molecule-1 (Iba-1), glial fibrillary acidic protein, Heat Shock Protein 90 Alpha Family Class A Member 1 (HSP90AA1, inflammatory factor) and High Mobility Group Box 1 (HMGB1, inflammation-related protein), and inhibited neuronal apoptosis in chronic constrictive injury rats or lipopolysaccharide-induced BV-2 cells. However, these effects were offset by HSP90AA1 overexpression in lipopolysaccharide-induced BV-2 cells. Exosomes of BV-2 cells could be absorbed by mouse astrocytes. In addition, HSP90AA1 overexpression reversed the effects of paeoniflorin on HMGB1 expression and inflammatory factors and proteins in mouse astrocytes co-cultured with exosome. Collectively, paeoniflorin alleviates neuropathic pain and inhibits inflammatory responses in chronic constrictive injury by modulating microglia-astrocyte crosstalk through HSP90AA1/HMGB1 pathways, which further evidences the potential of paeoniflorin in the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Fengqin Luo
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), China
| | - Juan Zhang
- Department of Pain, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), China
| | - Yunfei Miao
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), China
| | - Danhong Wu
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), China
| | - Hongxia Shen
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), China
| | - Man Lu
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), China.
| |
Collapse
|
6
|
Zhao Y, Chen Y, Liu Z, Zhou L, Huang J, Luo X, Luo Y, Li J, Lin Y, Lai J, Liu J. TXNIP knockdown protects rats against bupivacaine-induced spinal neurotoxicity via the inhibition of oxidative stress and apoptosis. Free Radic Biol Med 2024; 219:1-16. [PMID: 38614227 DOI: 10.1016/j.freeradbiomed.2024.04.223] [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: 01/07/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Bupivacaine (BUP) is an anesthetic commonly used in clinical practice that when used for spinal anesthesia, might exert neurotoxic effects. Thioredoxin-interacting protein (TXNIP) is a member of the α-arrestin protein superfamily that binds covalently to thioredoxin (TRX) to inhibit its function, leading to increased oxidative stress and activation of apoptosis. The role of TXNIP in BUP-induced oxidative stress and apoptosis remains to be elucidated. In this context, the present study aimed to explore the effects of TXNIP knockdown on BUP-induced oxidative stress and apoptosis in the spinal cord of rats and in PC12 cells through the transfection of adeno-associated virus-TXNIP short hairpin RNA (AAV-TXNIP shRNA) and siRNA-TXNIP, respectively. In vivo, a rat model of spinal neurotoxicity was established by intrathecally injecting rats with BUP. The BUP + TXNIP shRNA and the BUP + Control shRNA groups of rats were injected with an AAV carrying the TXNIP shRNA and the Control shRNA, respectively, into the subarachnoid space four weeks prior to BUP treatment. The Basso, Beattie & Bresnahan (BBB) locomotor rating score, % MPE of TFL, H&E staining, and Nissl staining analyses were conducted. In vitro, 0.8 mM BUP was determined by CCK-8 assay to establish a cytotoxicity model in PC12 cells. Transfection with siRNA-TXNIP was carried out to suppress TXNIP expression prior to exposing PC12 cells to BUP. The results revealed that BUP effectively induced neurological behavioral dysfunction and neuronal damage and death in the spinal cord of the rats. Similarly, BUP triggered cytotoxicity and apoptosis in PC12 cells. In addition, treated with BUP both in vitro and in vivo exhibited upregulated TXNIP expression and increased oxidative stress and apoptosis. Interestingly, TXNIP knockdown in the spinal cord of rats through transfection of AAV-TXNIP shRNA exerted a protective effect against BUP-induced spinal neurotoxicity by ameliorating behavioral and histological outcomes and promoting the survival of spinal cord neurons. Similarly, transfection with siRNA-TXNIP mitigated BUP-induced cytotoxicity in PC12 cells. In addition, TXNIP knockdown mitigated the upregulation of ROS, MDA, Bax, and cleaved caspase-3 and restored the downregulation of GSH, SOD, CAT, GPX4, and Bcl2 induced upon BUP exposure. These findings suggested that TXNIP knockdown protected against BUP-induced spinal neurotoxicity by suppressing oxidative stress and apoptosis. In summary, TXNIP could be a central signaling hub that positively regulates oxidative stress and apoptosis during neuronal damage, which renders TXNIP a promising target for treatment strategies against BUP-induced spinal neurotoxicity.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China; Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Road, Nanchong, 637000, Sichuan, China
| | - Yuanyuan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Ziru Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Lei Zhou
- Department of Anesthesiology, Meishan People's Hospital, No. 288 South Fourth Section of Dongpo Avenue, 620020, Sichuan, China
| | - Jiao Huang
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Xi Luo
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yunpeng Luo
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, 557300, Guizhou, China
| | - Jia Li
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China; Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Yunan Lin
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Jian Lai
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| | - Jingchen Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
7
|
Basu P, Maier C, Averitt DL, Basu A. NLR family pyrin domain containing 3 (NLRP3) inflammasomes and peripheral neuropathic pain - Emphasis on microRNAs (miRNAs) as important regulators. Eur J Pharmacol 2023; 955:175901. [PMID: 37451423 DOI: 10.1016/j.ejphar.2023.175901] [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: 02/18/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Neuropathic pain is caused by the lesion or disease of the somatosensory system and can be initiated and/or maintained by both central and peripheral mechanisms. Nerve injury leads to neuronal damage and apoptosis associated with the release of an array of pathogen- or damage-associated molecular patterns to activate inflammasomes. The activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome contributes to neuropathic pain and may represent a novel target for pain therapeutic development. In the current review, we provide an up-to-date summary of the recent findings on the involvement of NLRP3 inflammasome in modulating neuropathic pain development and maintenance, focusing on peripheral neuropathic conditions. Here we provide a detailed review of the mechanisms whereby NLRP3 inflammasomes contribute to neuropathic pain via (1) neuroinflammation, (2) apoptosis, (3) pyroptosis, (4) proinflammatory cytokine release, (5) mitochondrial dysfunction, and (6) oxidative stress. We then present the current research literature reporting on the antinociceptive effects of several natural products and pharmacological interventions that target activation, expression, and/or regulation of NLRP3 inflammasome. Furthermore, we emphasize the effects of microRNAs as another regulator of NLRP3 inflammasome. In conclusion, we summarize the possible caveats and future perspectives that might provide successful therapeutic approaches against NLRP3 inflammasome for treating or preventing neuropathic pain conditions.
Collapse
Affiliation(s)
- Paramita Basu
- Pittsburgh Center for Pain Research, The Pittsburgh Project to End Opioid Misuse, Department of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Camelia Maier
- Division of Biology, School of the Sciences, Texas Woman's University, Denton, TX, 76204-5799, USA.
| | - Dayna L Averitt
- Division of Biology, School of the Sciences, Texas Woman's University, Denton, TX, 76204-5799, USA.
| | - Arpita Basu
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV, 89154, USA.
| |
Collapse
|
8
|
Chen W, Wang X, Sun Q, Zhang Y, Liu J, Hu T, Wu W, Wei C, Liu M, Ding Y, Liu D, Chong Y, Wang P, Zhu H, Cui W, Zhang J, Li Q, Yang F. The upregulation of NLRP3 inflammasome in dorsal root ganglion by ten-eleven translocation methylcytosine dioxygenase 2 (TET2) contributed to diabetic neuropathic pain in mice. J Neuroinflammation 2022; 19:302. [PMID: 36527131 PMCID: PMC9756585 DOI: 10.1186/s12974-022-02669-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The nucleotide oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) in dorsal root ganglion (DRG) contributes to pain hypersensitivity in multiple neuropathic pain models, but the function of the NLRP3 in diabetic neuropathic pain (DNP) and the regulation mechanism are still largely unknown. Epigenetic regulation plays a vital role in the controlling of gene expression. Ten-eleven translocation methylcytosine dioxygenase 2 (TET2) is a DNA demethylase that contributes to transcriptional activation. TET2 is also involved in high glucose (HG)-induced pathology. METHODS DNP was induced in mice via the intraperitoneal injection of streptozotocin (STZ) for five consecutive days and the mechanical threshold was evaluated in STZ-diabetic mice by using von Frey hairs. The expression level of the NLRP3 pathway and TET2 in DRG were determined through molecular biology experiments. The regulation of the NLRP3 pathway by TET2 was examined in in vitro and in vivo conditions. RESULTS In the present research, we first established the DNP model and found that NLRP3 pathway was activated in DRG. The treatment of NLRP3 inhibitor MCC950 alleviated the mechanical allodynia of DNP mice. Then we revealed that in STZ-diabetic mice DRG, the genomic DNA was demethylated, and the expression of DNA demethylase TET2 was increased evidently. Using RNA-sequencing analysis, we found that the expression of Txnip, a gene that encodes a thioredoxin-interacting protein (TXNIP) which mediates NLRP3 activation, was elevated in the DRG after STZ treatment. In addition, knocking down of TET2 expression in DRG using TET2-siRNA suppressed the mRNA expression of Txnip and subsequently inhibited the expression/activation of NLRP3 inflammasome in vitro and in vivo as well as relieved the pain sensitivity of DNP animals. CONCLUSION The results suggested that the upregulation of the TXNIP/NLRP3 pathway by TET2 in DRG was involved in the pain hypersensitivity of the DNP model.
Collapse
Affiliation(s)
- Wen Chen
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China ,grid.24695.3c0000 0001 1431 9176International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Xiaotong Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Qingyu Sun
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Yurui Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Jing Liu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Tingting Hu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Weihua Wu
- grid.24696.3f0000 0004 0369 153XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Chao Wei
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Meng Liu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Yumeng Ding
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Dianxin Liu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Yingzi Chong
- grid.24696.3f0000 0004 0369 153XDepartment of Anesthesiology Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070 China
| | - Peipei Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Hongwei Zhu
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| | - Weihua Cui
- grid.24696.3f0000 0004 0369 153XDepartment of Anesthesiology Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070 China
| | - Jiannan Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Qian Li
- grid.24696.3f0000 0004 0369 153XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China ,grid.24696.3f0000 0004 0369 153XAdvanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069 China ,grid.24696.3f0000 0004 0369 153XKey Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, 100069 China
| | - Fei Yang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China ,grid.24696.3f0000 0004 0369 153XAdvanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069 China
| |
Collapse
|
9
|
Sun Q, Hu T, Zhang Y, Wang X, Liu J, Chen W, Wei C, Liu D, Wu W, Lan T, Ding Y, Luo Z, Liu M, Shen D, Xiao Z, Hu L, Pang M, Ma Y, Shi L, Wang P, Zhang J, Li Q, Yang F. IRG1/itaconate increases IL-10 release to alleviate mechanical and thermal hypersensitivity in mice after nerve injury. Front Immunol 2022; 13:1012442. [PMID: 36311727 PMCID: PMC9612919 DOI: 10.3389/fimmu.2022.1012442] [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: 08/05/2022] [Accepted: 09/28/2022] [Indexed: 09/19/2023] Open
Abstract
Inflammation plays an important role in the occurrence and development of neuropathic pain. Immune-responsive gene 1 (IRG1) decarboxylates cis-aconitate to produce itaconate in the mitochondria. Itaconate serves as an immunomodulator of macrophages and represses inflammation in infectious diseases. Recently, a study showed that an itaconate derivative inhibits neuroinflammation and reduces chronic pain in mice. However, the function and molecular mechanisms of endogenous itaconate in neuropathic pain have not been fullyelucidated. In this study, the content of itaconate in the ipsilateral spinal cord after nerve-injured mice was detected with mass spectrometry. The Irg1-/- mouse was constructed to determine the role of endogenous itaconate in the chronic constriction nerve injury (CCI) model. The analgesic effect of exogenous itaconate was assessed with intraperitoneal and intrathecal administration in both male and female CCI mice. The spinal application of 4-OI also reduced the evoked responses of wide dynamic range neurons in CCI mice. The potential analgesic mechanism of itaconate was explored through molecular biology experiments and verified in Interleukin (IL)-10-/- mice. We found the levels of itaconate and IRG1 in the spinal cord significantly increased after CCI. Irg1 deficiency aggravated the mechanical and heat hypersensitivity, while the exogenous administration of the itaconate derivative 4-OI alleviated the neuropathic pain in male and female CCI mice. Mechanistically, the treatment of 4-OI increased the level of IL-10 and activates STAT3/β-endorphin pathway in the spinal cord, and the analgesia effect of itaconate was impaired in IL-10-/- mice. Finally, we showed that the upregulation of IL-10 induced by 4-OI was mainly from spinal neurons through Nrf2 pathway. This study demonstrated the analgesic effect of endogenous and exogenous itaconate in the neuropathic pain model, suggesting that the spinal IL-10/STAT3/β-endorphin pathway might mediate the analgesia effect of itaconate.
Collapse
Affiliation(s)
- Qingyu Sun
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Anesthesiology, Chang Hai Hospital, Naval Military Medical University, Shanghai, China
| | - Tingting Hu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yurui Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaotong Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wen Chen
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chao Wei
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dianxin Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Weihua Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ting Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yumeng Ding
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhaoli Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Meng Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Danmin Shen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhongnan Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Liye Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Miaoyi Pang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yiran Ma
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Peipei Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jiannan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Liu P, Li X, Liu J, Zhang H, You Z, Zhang J. TXNIP Participated in NLRP3-Mediated Inflammation in a Rat Model of Cervical Spondylotic Myelopathy. J Inflamm Res 2022; 15:4547-4559. [PMID: 35971339 PMCID: PMC9375583 DOI: 10.2147/jir.s373614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/30/2022] [Indexed: 12/02/2022] Open
Abstract
Background Cervical spondylotic myelopathy (CSM) is a spinal cord disease caused by cervical disc degeneration and related pathological changes. Cervical spondylotic myelopathy may result from inflammation responses and neuronal damage. Thioredoxin-interacting protein (TXNIP)/NOD-like receptor protein 3 (NLRP3) signaling promotes inflammation. However, the effects of TXNIP/NLRP3 on the pathogenesis of CSM have not been reported. Methods A rat model of chronic cervical cord compression was established to observe changes in the levels of of TNXIP/NeuN and NLRP3/NeuN expression in the damaged anterior horn of the spinal cord following progression of CSM. Rats were injected with TXNIP small interfering RNA (siRNA) and scrambled control to determine the effects of TXNIP inhibition on NLRP3-mediated inflammation in rats with CSM. Behaviors effects and the expression of NLRP3 and pro-caspase-1 in the damaged spinal cord were evaluated. Results The expression levels of TXNIP and NLRP3 were significantly increased in the damaged anterior horn of the spinal cord following CSM. Injection of TXNIP siRNA significantly improved behavioral measures and decreased apoptosis in the damaged anterior horn of spinal cord. Furthermore, the levels of NLRP3 and pro-caspase-1 in the lesioned area were reduced by the TXNIP siRNA injection. Conclusion Thioredoxin-interacting protein participated in NLRP3 mediated inflammation in a rat model of CSM, which indicated that TXNIP may be a potential therapeutic target in improving CSM.
Collapse
Affiliation(s)
- Peisheng Liu
- Department of Spinal Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Xiaofeng Li
- Department of Spinal Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Jing Liu
- Basic Department, Yantai Vocational College, Yantai, People's Republic of China
| | - Hengjia Zhang
- Department of Spinal Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Zhitao You
- Department of Spinal Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Jianfeng Zhang
- Department of Spinal Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| |
Collapse
|
11
|
RGS6 Drives Spinal Cord Injury by Inhibiting AMPK Pathway in Mice. DISEASE MARKERS 2022; 2022:4535652. [PMID: 35510037 PMCID: PMC9061016 DOI: 10.1155/2022/4535652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 11/18/2022]
Abstract
Objective. Oxidative stress and inflammation play critical roles in the pathogenesis of spinal cord injury (SCI). Regulator of G protein signaling 6 (RGS6) is involved in controlling ROS generation and inflammatory response under different contexts. This study is aimed at investigating its role and underlying mechanism in SCI. Methods. Contusive SCI mouse models were generated, and lentiviral vectors were injected to silence or overexpress RGS6 in the spinal cord. To inhibit AMP-activated protein kinase (AMPK) activity, SCI mice were intraperitoneally injected with compound C (20 mg/kg) every two days. Oxidative and inflammatory markers were detected. Results. Spinal RGS6 expression was elevated upon SCI stimulation. RGS6 knockdown suppressed, while RGS6 overexpression aggravated oxidative stress, inflammation, and SCI in mice. Mechanistically, RGS6 elevation during SCI deactivated AMPK pathway, thereby exacerbating oxidative stress and inflammation in SCI mice. Conclusion. RGS6 is required for the initiation and progression of SCI, and knocking down RGS6 may provide promising therapeutic strategies for SCI patients.
Collapse
|