1
|
Liu R, Zhu T, Chu X, Xu Y, Wang L, Wan Q, Li T. Transcranial direct current stimulation alleviates chronic pain in knee osteoarthritis by modulating microglial and astrocytic polarization and neuroinflammation. Life Sci 2025:123753. [PMID: 40409582 DOI: 10.1016/j.lfs.2025.123753] [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: 12/15/2024] [Revised: 05/06/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
AIMS Knee osteoarthritis (KOA) chronic pain is linked to neuroinflammation mediated by reactive astrocytes in the primary somatosensory cortex (S1). Reactive astrocytes are classified into neurotoxic A1 and neuroprotective A2 phenotypes, with activated microglia promoting A1 astrocyte formation during chronic pain progression. This study aimed to investigate whether transcranial direct current stimulation (tDCS) can alleviate KOA chronic pain by modulating glial phenotype conversion and neuroinflammatory processes. MAIN METHODS Rats received an intra-articular injection of monosodium iodoacetate (MIA) in the left knee to model KOA pain. Additionally, rats received intraperitoneal injections of the NF-κB inhibitor BAY 11-7082 and underwent tDCS. Pain thresholds were assessed using von Frey filaments and a hot plate. Changes in microglia, astrocytes, and inflammatory factor expression were analyzed with Western blotting, immunofluorescence, and reverse transcription-quantitative PCR. KEY FINDINGS At 4 and 7 days after MIA injection, microglia exhibited a proinflammatory M1 phenotype, accompanied by increased expression of IL-1α, TNF-α, and C1q. From day 7 to 21 post-injection, astrocytes displayed a neurotoxic A1 phenotype. The NF-κB/NLRP3/IL-18 signaling pathway was significantly upregulated in KOA rats. Treatment with BAY 11-7082 or tDCS significantly alleviated mechanical allodynia and thermal hyperalgesia, shifting microglia from M1 to M2 and astrocytes from A1 to A2 polarization, while suppressing the NF-κB/NLRP3/IL-18 pathway and reducing neuroinflammation. SIGNIFICANCE These findings suggest that tDCS may alleviate KOA chronic pain through modulation of glial activation states and suppression of central neuroinflammation, highlighting its potential as a non-invasive therapeutic approach.
Collapse
Affiliation(s)
- Rujuan Liu
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China; Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Ting Zhu
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Xiao Chu
- Department of Pharmacy of Qingdao Municipal Hospital, Qingdao, China
| | - Yifan Xu
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lin Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Qi Wan
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China.
| | - Tieshan Li
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
2
|
Zhang J, Jiang Y, Zhang Z, Li S, Fan H, Gu J, Mao R, Xu X. Repulsive guidance molecules b (RGMb): molecular mechanism, function and role in diseases. Expert Rev Mol Med 2024; 26:e24. [PMID: 39375839 PMCID: PMC11488336 DOI: 10.1017/erm.2024.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/23/2023] [Accepted: 06/11/2024] [Indexed: 10/09/2024]
Abstract
Repulsive guidance molecule b (RGMb), a glycosylphosphatidylinositol-anchored member of the RGM family, is initially identified as a co-receptor of bone morphogenetic protein (BMP) in the nervous system. The expression of RGMb is transcriptionally regulated by dorsal root ganglion 11 (DRG11), which is a transcription factor expressed in embryonic DRG and dorsal horn neurons and plays an important role in the development of sensory circuits. RGMb is involved in important physiological processes such as embryonic development, immune response, intercellular adhesion and tumorigenesis. Furthermore, RGMb is mainly involved in the regulation of RGMb-neogenin-Rho and BMP signalling pathways. The recent discovery of programmed death-ligand 2 (PD-L2)-RGMb binding reveals that the cell signalling network and functional regulation centred on RGMb are extremely complex. The latest report suggests that down-regulation of the PD-L2-RGMb pathway in the gut microbiota promotes an anti-tumour immune response, which defines a potentially effective immune strategy. However, the biological function of RGMb in a variety of human diseases has not been fully determined, and will remain an active research field. This article reviews the properties and functions of RGMb, focusing on its role under various physiological and pathological conditions.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Zijian Zhang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Shilin Li
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Haowen Fan
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jinhua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
3
|
Li D, Liu C, Wang H, Li Y, Wang Y, An S, Sun S. The Role of Neuromodulation and Potential Mechanism in Regulating Heterotopic Ossification. Neurochem Res 2024; 49:1628-1642. [PMID: 38416374 DOI: 10.1007/s11064-024-04118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
Heterotopic ossification (HO) is a pathological process characterized by the aberrant formation of bone in muscles and soft tissues. It is commonly triggered by traumatic brain injury, spinal cord injury, and burns. Despite a wide range of evidence underscoring the significance of neurogenic signals in proper bone remodeling, a clear understanding of HO induced by nerve injury remains rudimentary. Recent studies suggest that injury to the nervous system can activate various signaling pathways, such as TGF-β, leading to neurogenic HO through the release of neurotrophins. These pathophysiological changes lay a robust groundwork for the prevention and treatment of HO. In this review, we collected evidence to elucidate the mechanisms underlying the pathogenesis of HO related to nerve injury, aiming to enhance our understanding of how neurological repair processes can culminate in HO.
Collapse
Affiliation(s)
- Dengju Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong First Medical University, Jinan, Shandong, China
| | - Changxing Liu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Haojue Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yunfeng Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yaqi Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Senbo An
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
| | - Shui Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
4
|
Li Y, Yin C, Jiang J, Yang H, Zhang F, Xing Y, Wang W, Lu C. Tumor necrosis factor α-induced protein 8-like-2 controls microglia phenotype via metabolic reprogramming in BV2 microglial cells and responses to neuropathic pain. Int J Biochem Cell Biol 2024; 169:106541. [PMID: 38309648 DOI: 10.1016/j.biocel.2024.106541] [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: 06/16/2023] [Revised: 01/07/2024] [Accepted: 01/28/2024] [Indexed: 02/05/2024]
Abstract
Microglial are major players in neuroinflammation that have recently emerged as potential therapeutic targets for neuropathic pain. Glucose metabolic programming has been linked to differential activation state and function in microglia. Tumor necrosis factor α-induced protein 8-like-2 (TNFAIP8L2) is an important component in regulating the anti-inflammatory response. However, the role of TNFAIP8L2 in microglia differential state during neuropathic pain and its interplay with glucose metabolic reprogramming in microglia has not yet been determined. Thus, we aimed to investigate the role of TNFAIP8L2 in the status of microglia in vitro and in vivo. BV2 microglial cells were treated with lipopolysaccharides plus interferon-gamma (LPS/IFNγ) or interleukin-4 (IL-4) to induce the two different phenotypes of microglia in vitro. In vivo experiments were conducted by chronic constriction injury of the sciatic nerve (CCI). We investigated whether TNFAIP8L2 regulates glucose metabolic programming in BV2 microglial cells. The data in vitro showed that TNFAIP8L2 lowers glycolysis and increases mitochondrial oxidative phosphorylation (OXPHOS) in inflammatory microglia. Blockade of glycolytic pathway abolished TNFAIP8L2-mediated differential activation of microglia. TNFAIP8L2 suppresses inflammatory microglial activation and promotes restorative microglial activation in BV2 microglial cells and in spinal cord microglia after neuropathic pain. Furthermore, TNFAIP8L2 controls differential activation of microglia and glucose metabolic reprogramming through the MAPK/mTOR/HIF-1α signaling axis. This study reveals that TNFAIP8L2 plays a critical role in neuropathic pain, providing important insights into glucose metabolic reprogramming and microglial phenotypic transition, which indicates that TNFAIP8L2 may be used as a potential drug target for the prevention of neuropathic pain.
Collapse
Affiliation(s)
- Yeqi Li
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cui Yin
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinhong Jiang
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huan Yang
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Feifei Zhang
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanhong Xing
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wuyang Wang
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chen Lu
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Xu L, Pan F, Guo Z. TIPE2: A Candidate for Targeting Antitumor Immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:755-763. [PMID: 38377476 DOI: 10.4049/jimmunol.2300433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/18/2023] [Indexed: 02/22/2024]
Abstract
TNF-α-induced protein 8-like 2 (TIPE2 or TNFAIP8L2) is a recently discovered negative regulator of innate and adaptive immunity. TIPE2 is expressed in a wide range of tissues, both immune and nonimmune, and is implicated in the maintenance of immune homeostasis within the immune system. Furthermore, TIPE2 has been shown to play a pivotal role in the regulation of inflammation and the development of tumor. This review focuses on the structural characteristics, expression patterns, and functional roles of TIPE proteins, with a particular emphasis on the role and underlying mechanisms of TIPE2 in immune regulation and its involvement in different diseases. However, the current body of evidence is still limited in providing a comprehensive understanding of the complex role of TIPE2 in the human body, warranting further investigation to elucidate the possible mechanisms and functions of TIPE2 in diverse disease contexts.
Collapse
Affiliation(s)
- Luxia Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
6
|
Tian S, Zheng H, Wu W, Wu L. Predicting Diagnostic Biomarkers Associated with Pyroptosis in Neuropathic Pain Based on Machine Learning and Experimental Validation. J Inflamm Res 2024; 17:1121-1145. [PMID: 38406324 PMCID: PMC10893895 DOI: 10.2147/jir.s445382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Previous studies have shown that pyroptosis plays a vital role in the progress of neuropathic pain (NP), but the molecular mechanisms have not been fully elucidated. The aim of this study was to identify crucial pyroptosis-related genes (PRGs) in NP. Methods We identified pyroptosis-related differentially expressed genes (PRDEGs) in NP by machine learning analysis of the GSE24982 and GSE60670 datasets. Furthermore, these PRDEGs were subjected to Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, Gene Set Enrichment Analysis (GSEA) and Friends analysis, respectively. Meanwhile, receiver operator characteristic (ROC) analysis was performed to assess the diagnostic value of PRDEGs in NP. Finally, we performed immune infiltration analysis of key PRDEGs using CIBERSORTR R package. Results We found that 5 PRDEGs by least absolute shrinkage and selection operator (LASSO) regression and random forest and verified by RT-qPCR. GO, KEGG and GSEA revealed that these PRDEGs were mainly enriched in regulation of neuron death, IL-4 signaling, IL-23 pathway, and NF-κB pathway. ROC analysis revealed that most of the PRDEGs performed well in diagnosing NP. We also revealed transcription factors, miRNA regulatory networks and drug interaction networks of PRDEGs. For immune infiltration analysis, PRDEGs were mainly correlated with dendritic cells, monocytes and follicular T helper cells, suggested that it might be involved in the regulation of neuroimmune-related signaling. Conclusion A total of five PRDEGs were can be employed as NP biomarkers, particularly Tlr4, Il1b and Casp8, and provide additional evidence for a vital role of pyroptosis in NP.
Collapse
Affiliation(s)
- Sheng Tian
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Heqing Zheng
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Wei Wu
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Lanxiang Wu
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| |
Collapse
|
7
|
Tang J, Lin M, Ou C. High-throughput transcriptome sequencing reveals the critical role of long non-coding RNA Gm14376 in the occurrence of neuropathic pain. Neurosci Lett 2023; 810:137312. [PMID: 37236343 DOI: 10.1016/j.neulet.2023.137312] [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: 01/30/2023] [Revised: 04/02/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023]
Abstract
Long noncoding RNAs (lncRNAs) have been suggested as important regulators in neuropathic pain. Our study aims to explore the possible molecular mechanism underlying the role of long non-coding RNA (lncRNA) Gm14376 in neuropathic pain in mice by high-throughput transcriptome sequencing. A mouse model of spared nerve injury (SNI) was constructed for mechanical, thermal and spontaneous pain testing. Transcriptomic changes in lncRNAs and mRNAs in the dorsal root ganglion (DRG) of SNI mice were analyzed using RNA-sequencing techniques in conjunction with public data analysis. AAV5 viral vector was constructed to assess the effect of Gm14376 on SNI-induced pain hypersensitivity and inflammatory response. Cis-target genes of Gm14376 were obtained and the functions of Gm14376 were analyzed by GO and KEGG pathway enrichment analyses. Results from bioinformatic analysis identified a conserved Gm14376, which was up-regulated in the DRG of SNI mice, specifically in response to nerve injury. Overexpression of Gm14376 in DRG induced neuropathic pain-like symptoms in mice. Furthermore, the functions of Gm14376 were related to the phosphatidylinositol 3-kinase (PI3K)/Akt pathway and fibroblast growth factor 3 (Fgf3) was identified as the cis-target gene of Gm14376. Gm14376 could directly up-regulate Fgf3 expression to activate the PI3K/Akt pathway, which alleviated pain hypersensitivity to mechanical and thermal stimuli and reduced the release of inflammatory factors in SNI mice. From our data, we conclude that SNI-induced up-regulation of Gm14376 expression in DRG activates the PI3K/Akt pathway through up-regulation of Fgf3 expression, thereby promoting the development of neuropathic pain in mice.
Collapse
Affiliation(s)
- Jian Tang
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Department of Anesthesiology, Southwest Medical University, Luzhou, Sichuan Province 64600, 0, China
| | - Min Lin
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Department of Anesthesiology, Southwest Medical University, Luzhou, Sichuan Province 64600, 0, China
| | - Cehua Ou
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
8
|
Yang Z, Zhang F, Abdul M, Jiang J, Li Y, Li Y, Yin C, Xing Y, Liu S, Lu C. Tumor necrosis factor-α-induced protein 8-like 2 alleviates morphine antinociceptive tolerance through reduction of ROS-mediated apoptosis and MAPK/NF-κB signaling pathways. Neuropharmacology 2023:109667. [PMID: 37451333 DOI: 10.1016/j.neuropharm.2023.109667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Chronic morphine tolerance is a repulsive barrier to the clinical treatment of pain. Whereas the underlying molecular mechanisms of morphine tolerance remain unknown. Here, we proposed that tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) is an essential control point regarding the progression of chronic morphine tolerance. We found that TIPE2 levels in the lumbar spinal cord were significantly downregulated in the morphine tolerance mouse model. Specifically, decreased TIPE2 by morphine tolerance was primarily expressed in spinal neurons, while increased expression of spinal TIPE2 distinctly attenuated the chronic morphine antinociceptive tolerance and tolerance-associated hyperalgesia. We also observed that increased expression of spinal TIPE2 significantly reduced morphine tolerance-induced neuronal ROS production and apoptosis, along with the activation of MAPKs and NF-κB signaling pathways. Moreover, the increased TIPE2 expression inhibited neuronal activation and glial reactivity in the spinal dorsal horn after chronic morphine exposure. Additionally, TIPE2 overexpression in cultured SH-SY5Y cells significantly suppressed ROS production and apoptosis in response to morphine challenge. Therefore, we can conclude that the upregulation of spinal TIPE2 may attenuate the morphine antinociceptive tolerance via TIPE2-dependent downregulation of neuronal ROS, inhibition of neuronal apoptosis, suppression of MAPKs and NF-κB activation. TIPE2 may be a potential strategy for preventing morphine tolerance in the future studies and clinical settings. Schematic diagram for the proposed mechanisms of TIPE2 regulates morphine antinociceptive tolerance. TIPE2 may alleviate morphine antinociceptive tolerance by regulating MAPK/NF-κB signaling pathways and apoptosis, which might be associated with ROS production.
Collapse
Affiliation(s)
- Zhong Yang
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Feifei Zhang
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mannan Abdul
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China; School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinhong Jiang
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanqiang Li
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yeqi Li
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cui Yin
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanhong Xing
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Su Liu
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chen Lu
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
9
|
Zaninelli TH, Mizokami SS, Bertozzi MM, Saraiva-Santos T, Pinho-Ribeiro FA, de Oliveira GI, Streck R, Araújo EJA, Arakawa NS, Borghi SM, Casagrande R, Verri WA. Kaurenoic Acid Reduces Ongoing Chronic Constriction Injury-Induced Neuropathic Pain: Nitric Oxide Silencing of Dorsal Root Ganglia Neurons. Pharmaceuticals (Basel) 2023; 16:ph16030343. [PMID: 36986443 PMCID: PMC10051686 DOI: 10.3390/ph16030343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Kaurenoic acid (KA) is a diterpene extracted from Sphagneticola trilobata (L.) Pruski. KA presents analgesic properties. However, the analgesic activity and mechanisms of action of KA in neuropathic pain have not been investigated so far; thus, we addressed these points in the present study. A mouse model of neuropathic pain was induced by chronic constriction injury (CCI) of the sciatic nerve. Acute (at the 7th-day post-CCI surgery) and prolonged (from 7–14th days post-CCI surgery) KA post-treatment inhibited CCI-induced mechanical hyperalgesia at all evaluated time points, as per the electronic version of von Frey filaments. The underlying mechanism of KA was dependent on activating the NO/cGMP/PKG/ATP-sensitive potassium channel signaling pathway since L-NAME, ODQ, KT5823, and glibenclamide abolished KA analgesia. KA reduced the activation of primary afferent sensory neurons, as observed by a reduction in CCI-triggered colocalization of pNF-κB and NeuN in DRG neurons. KA treatment also increased the expression of neuronal nitric oxide synthase (nNOS) at the protein level as well as the intracellular levels of NO in DRG neurons. Therefore, our results provide evidence that KA inhibits CCI neuropathic pain by activating a neuronal analgesic mechanism that depends on nNOS production of NO to silence the nociceptive signaling that generates analgesia.
Collapse
Affiliation(s)
- Tiago H. Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Sandra S. Mizokami
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Mariana M. Bertozzi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Telma Saraiva-Santos
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Felipe A. Pinho-Ribeiro
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Gabriele Inácio de Oliveira
- Department of Pharmaceutical Sciences, Center of Health Sciences, Londrina State University, Londrina 86039-440, Paraná, Brazil
| | - Renata Streck
- Department of Histology, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Eduardo J. A. Araújo
- Department of Histology, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Nilton S. Arakawa
- Department of Pharmaceutical Sciences, Center of Health Sciences, Londrina State University, Londrina 86039-440, Paraná, Brazil
| | - Sergio M. Borghi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Sciences, Londrina State University, Londrina 86039-440, Paraná, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
- Correspondence:
| |
Collapse
|
10
|
Soluble Epoxide Hydrolase Inhibitor TPPU Alleviates Nab-Paclitaxel-Induced Peripheral Neuropathic Pain via Suppressing NF- κB Signalling in the Spinal Cord of a Rat. Pain Res Manag 2023; 2023:9058774. [PMID: 36819745 PMCID: PMC9931472 DOI: 10.1155/2023/9058774] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/31/2022] [Accepted: 01/24/2023] [Indexed: 02/10/2023]
Abstract
Objective Paclitaxel-induced peripheral neuropathy (PIPN) is a debilitating and difficult-to-treat side effect of paclitaxel. Soluble epoxide hydrolase (sEH) can rapidly metabolize the endogenous anti-inflammatory mediators' epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids. This study aimed to assess whether the sEH inhibitor N-(1-(1-oxopropy)-4-piperidinyl]-N'-(trifluoromethoxy) phenyl)-urea (TPPU) plays a critical role in PIPN of rats and provides a new target for treatment. Methods A Sprague-Dawley male rat model of PIPN induced by nab-paclitaxel was established. Rats were randomly divided into a control group, nab-paclitaxel group, and nab-paclitaxel + TPPU (sEH inhibitor) group, with 36 rats in each group. The effects of the sEH inhibitor TPPU on behavioural assays, apoptosis, glial activation, axonal injury, microstructure, and permeability of the blood-spinal cord barrier were detected, and the underlying mechanisms were explored by examining the expression of NF-κB signalling pathways, inflammatory cytokines, and oxidative stress. Results The results showed that the mechanical and thermal pain thresholds of rats were decreased after nab-paclitaxel treatment, accompanied by an increased expression of axonal injury-related proteins, enhanced cell apoptosis, aggravated destruction of vascular permeability, intense glial responses, and elevated inflammatory cytokines and oxidative stress in the L4-L6 spinal cord. TPPU restored the mechanical and thermal thresholds, decreased cell apoptosis, alleviated axonal injury and glial responses, and protected vascular permeability by increasing the expression of tight junction proteins. TPPU relieved PIPN by inhibiting the activation of the sEH and NF-κB signalling pathways by decreasing the levels of inflammatory cytokines and oxidative stress. Conclusion These findings support a role for sEH in PIPN and suggest that the inhibition of sEH represents a potential new therapeutic target for PIPN.
Collapse
|
11
|
Lu P, Fang K, Cheng W, Yu B. High-frequency electrical stimulation reduced hyperalgesia and the activation of the Myd88 and NFκB pathways in chronic constriction injury of sciatic nerve-induced neuropathic pain mice. Neurosci Lett 2023; 796:137064. [PMID: 36638955 DOI: 10.1016/j.neulet.2023.137064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/05/2023] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Neuropathic pain has become a global public problem and health burden. Pharmacological interventions are the primary treatment, but the drug cure rate is low with side effects. There is an urgent need to develop novel treatment approaches. High frequency electrical stimulation (KHES) has been widely applied in clinical analgesia. However, its mechanism is poorly understood. In this study, datasets related to neuropathic pain were obtained from the GEO database. The differentially expressed genes (DEGs) and key genes were analyzed through functional enrichment analysis, showing that most of the pathways involve the inflammation. The MyD88 and NFκB pathways were further studied. KHES significantly alleviated mechanical and thermal allodynia in chronic constriction injury of the sciatic nerve mice. KHES also inhibited the increase in Myd88 and p-NFκB expression. The administration of NFκB pathway activator partly reversed the antinociceptive effects of KHES, and NFκB pathway inhibitor achieved analgesic effects similar to those of KHES. Therefore, KHES might be a novel intervention for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Peixin Lu
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| | - Kexin Fang
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| | - Wen Cheng
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| | - Bin Yu
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|