1
|
Bai L, Gao Y, Li L, Liang Z, Qiao Y, Wang X, Yv L, Yang JJ, Xu JT. Poly-(ADP-ribose) polymerase 1-modulated production of CXCL1 in the dorsal root ganglion and spinal dorsal horn exacerbated inflammatory pain in rats. Int Immunopharmacol 2024; 143:113370. [PMID: 39405941 DOI: 10.1016/j.intimp.2024.113370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
Poly (ADP-ribose) polymerase 1 (PARP-1) serves as a transcriptional co-regulator and has been playing an important role in various inflammatory diseases. In the present study, we investigated the role and underlying mechanisms of action of PARP-1 in inflammatory pain. Intraplantar injection of complete Freund's adjuvant (CFA) was administered to the rats to induce inflammatory pain. Immunofluorescence, Western blotting, co-immunoprecipitation, and chromatin immunoprecipitation-quantitative polymerase chain reaction were performed to investigate the underlying mechanisms. Our results showed that CFA injection led to an increase in the production and activation of PARP-1 in both the L4/5 dorsal root ganglions (DRGs) and the spinal dorsal horn. Repeated intrathecal injections of Tiq-A or 5-AIQ, two specific inhibitors of PARP-1, and microinjections of AAV-PARP-1 shRNA into the L5 DRG or L5 spinal dorsal horn partially prevented the development of inflammatory pain. The established inflammatory pain was attenuated by a single bolus of intrathecal injection of Tiq-A or 5-AIQ on day 7 after the CFA injection. The CFA-induced mechanical allodynia and thermal hyperalgesia in female rats were alleviated by repeated intrathecal injections of Tiq-A. Moreover, repeated intrathecal injections of 5-AIQ inhibited the binding of NF-κB with CXCL1 promoter and reduced the production of CXCL1 in both the L4/5 DRGs and L4-6 spinal dorsal horns following CFA injection. Collectively, our results indicate that CFA-induced upregulation of PARP-1 by promoting CXCL1 expression in the DRG and probably in the spinal dorsal horn contributes to the pathogenesis of inflammatory pain. Thus, PARP-1 may be a potential pharmaceutical target for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Liying Bai
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Yan Gao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China; Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Lili Yv
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Xu C, Wang Y, Ni C, Xu M, Yin C, He Q, Ma B, Fu J, Zhao B, Chen L, Zhi T, Wei S, Cheng L, Xu H, Xiao J, Yang L, Xu Q, Kuang J, Liu B, Zhou Q, Lin X, Yao M, Ni H. Histone modifications and Sp1 promote GPR160 expression in bone cancer pain within rodent models. EMBO Rep 2024:10.1038/s44319-024-00292-6. [PMID: 39448865 DOI: 10.1038/s44319-024-00292-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/15/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
Bone cancer pain (BCP) affects ~70% of patients in advanced stages, primarily due to bone metastasis, presenting a substantial therapeutic challenge. Here, we profile orphan G protein-coupled receptors in the dorsal root ganglia (DRG) following tumor infiltration, and observe a notable increase in GPR160 expression. Elevated Gpr160 mRNA and protein levels persist from postoperative day 6 for over 18 days in the affected DRG, predominantly in small-diameter C-fiber type neurons specific to the tibia. Targeted interventions, including DRG microinjection of siRNA or AAV delivery, mitigate mechanical allodynia, cold, and heat hyperalgesia induced by the tumor. Tumor infiltration increases DRG neuron excitability in wild-type mice, but not in Gpr160 gene knockout mice. Tumor infiltration results in reduced H3K27me3 and increased H3K27ac modifications, enhanced binding of the transcription activator Sp1 to the Gpr160 gene promoter region, and induction of GPR160 expression. Modulating histone-modifying enzymes effectively alleviated pain behavior. Our study delineates a novel mechanism wherein elevated Sp1 levels facilitate Gpr160 gene transcription in nociceptive DRG neurons during BCP in rodents.
Collapse
Affiliation(s)
- Chengfei Xu
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
- Department of Anesthesiology, The Third People's Hospital of Bengbu, 38 Shengli Middle Road, 233000, Bengbu, China
| | - Yahui Wang
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Chaobo Ni
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Miao Xu
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Chengyu Yin
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Qiuli He
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Bing Ma
- Department of Anesthesiology, The Third People's Hospital of Bengbu, 38 Shengli Middle Road, 233000, Bengbu, China
| | - Jie Fu
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Baoxia Zhao
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Liping Chen
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Tong Zhi
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Shirong Wei
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Liang Cheng
- Department of Anesthesiology, The Third People's Hospital of Bengbu, 38 Shengli Middle Road, 233000, Bengbu, China
| | - Hui Xu
- Department of Anesthesiology, The First People's Hospital of Bengbu, 233000, Bengbu, China
| | - Jiajun Xiao
- Bengbu Hospital of Traditional Chinese Medicine, 4339 Huai-Shang Road, 233000, Bengbu, China
| | - Lei Yang
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Qingqing Xu
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Jiao Kuang
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Boyi Liu
- Department of Neurobiology and Acupuncture Research, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third Clinical Medical College, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Qinghe Zhou
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China
| | - Xuewu Lin
- Department of Pain Medicine, The First Affiliated Hospital of Bengbu Medical University, 233000, Bengbu, China.
| | - Ming Yao
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China.
| | - Huadong Ni
- Department of Anesthesiology and Pain Research Center, The Affiliated Hospital of Jiaxing University, 1882 Zhonghuan South Road, 314001, Jiaxing, China.
| |
Collapse
|
3
|
Li L, Ding Z, Ma F, Zhang K, Lu D, Wang H, Yang K. Spinal nerve transection-induced upregulation of KDM4A in the dorsal root ganglia contributes to the development and maintenance of neuropathic pain via promoting CCL2 expression in rats. Eur J Neurosci 2024; 60:5169-5188. [PMID: 39136140 DOI: 10.1111/ejn.16491] [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: 12/15/2023] [Accepted: 07/17/2024] [Indexed: 10/10/2024]
Abstract
Studies indicate that the lysine-specific demethylase 4A (KDM4A), acts as a key player in neuropathic pain, driving the process through its involvement in promoting neuroinflammation. Emerging evidence reveals that C-C Motif Chemokine Ligand 2 (CCL2) participates in neuroinflammation, which plays an important role in the development and maintenance of neuropathic pain. However, it remains unclear if KDM4A plays a role in regulating CCL2 in neuropathic pain. This study found that following spinal nerve transection (SNT) of the lumbar 5 nerve root in rats, the expression of KDM4A and CCL2 increased in the ipsilateral L4/5 dorsal root ganglia (DRG). Injecting KDM4A siRNA into the DRGs of rats post-SNT resulted in a higher paw withdrawal threshold (PWT) and paw-withdrawal latency (PWL) compared to the KDM4A scRNA group. In addition, prior microinjection of AAV-EGFP-KDM4A shRNA also alleviates the decrease in PWT and PWL caused by SNT. Correspondingly, microinjection of AAV-EGFP-KDM4A shRNA subsequent to SNT reduced the established mechanical and thermal hyperalgesia. Furthermore, AAV-EGFP-KDM4A shRNA injection decreased the expression of CCL2 in DRGs. ChIP-PCR analysis revealed that increased binding of p-STAT1 with the CCL2 promoter induced by SNT was inhibited by AAV-EGFP-KDM4A shRNA treatment. These findings suggest that KDM4A potentially influences neuropathic pain by regulating CCL2 expression in DRGs.
Collapse
Affiliation(s)
- Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zheng Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Ma
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai Zhang
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dan Lu
- Department of Respiratory Intensive Care, Zhengzhou Traditional Chinese Medicine Hospital, Zhengzhou, China
| | - Hongmin Wang
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kangli Yang
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Koyanagi M, Ogido R, Moriya A, Saigo M, Ihida S, Teranishi T, Kawada J, Katsuno T, Matsubara K, Terada T, Yamashita A, Imai S. Development of a 3-dimensional organotypic model with characteristics of peripheral sensory nerves. CELL REPORTS METHODS 2024; 4:100835. [PMID: 39116883 PMCID: PMC11384078 DOI: 10.1016/j.crmeth.2024.100835] [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: 01/24/2024] [Revised: 05/02/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
We developed a rat dorsal root ganglion (DRG)-derived sensory nerve organotypic model by culturing DRG explants on an organoid culture device. With this method, a large number of organotypic cultures can be produced simultaneously with high reproducibility simply by seeding DRG explants derived from rat embryos. Unlike previous DRG explant models, this organotypic model consists of a ganglion and an axon bundle with myelinated A fibers, unmyelinated C fibers, and stereo-myelin-forming nodes of Ranvier. The model also exhibits Ca2+ signaling in cell bodies in response to application of chemical stimuli to nerve terminals. Further, axonal transection increases the activating transcription factor 3 mRNA level in ganglia. Axons and myelin are shown to regenerate 14 days following transection. Our sensory organotypic model enables analysis of neuronal excitability in response to pain stimuli and tracking of morphological changes in the axon bundle over weeks.
Collapse
Affiliation(s)
- Madoka Koyanagi
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Ryosuke Ogido
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Akari Moriya
- Department of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Mamiko Saigo
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Satoshi Ihida
- New Business Promotion Division, Business Development Unit, Panel Semicon Laboratories, Sharp Corporation, Tenri, Nara 632-8567, Japan
| | - Tomoko Teranishi
- New Business Promotion Division, Business Development Unit, Panel Semicon Laboratories, Sharp Corporation, Tenri, Nara 632-8567, Japan
| | - Jiro Kawada
- Jiksak Bioengineering, Inc., Kawasaki, Kanagawa 210-0821, Japan
| | - Tatsuya Katsuno
- Division of Electron Microscopic Study, Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Kazuo Matsubara
- School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Tomohiro Terada
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Akira Yamashita
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan
| | - Satoshi Imai
- Department of Medical Neuropharmacology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan; Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto 606-8507, Japan.
| |
Collapse
|
5
|
Mitra S, Werner CT, Shwani T, Lopez AG, Federico D, Higdon K, Li X, Gobira PH, Thomas SA, Martin JA, An C, Chandra R, Maze I, Neve R, Lobo MK, Gancarz AM, Dietz DM. A Novel Role for the Histone Demethylase JMJD3 in Mediating Heroin-Induced Relapse-Like Behaviors. Biol Psychiatry 2024:S0006-3223(24)01452-5. [PMID: 39019389 DOI: 10.1016/j.biopsych.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 06/10/2024] [Accepted: 06/25/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Epigenetic changes that lead to long-term neuroadaptations following opioid exposure are not well understood. We examined how histone demethylase JMJD3 in the nucleus accumbens (NAc) influences heroin seeking after abstinence from self-administration. METHODS Male Sprague Dawley rats were trained to self-administer heroin. Western blotting and quantitative polymerase chain reaction were performed to quantify JMJD3 and bone morphogenetic protein (BMP) pathway expression in the NAc (n = 7-11/group). Pharmacological inhibitors or viral expression vectors were microinfused into the NAc to manipulate JMJD3 or the BMP pathway member SMAD1 (n = 9-11/group). The RiboTag capture method (n = 3-5/group) and viral vectors (n = 7-8/group) were used in male transgenic rats to identify the contributions of D1- and D2-expressing medium spiny neurons in the NAc. Drug seeking was tested by cue-induced response previously paired with drug infusion. RESULTS Levels of JMJD3 and phosphorylated SMAD1/5 in the NAc were increased after 14 days of abstinence from heroin self-administration. Pharmacological and virus-mediated inhibition of JMJD3 or the BMP pathway attenuated cue-induced seeking. Pharmacological inhibition of BMP signaling reduced JMJD3 expression and H3K27me3 levels. JMJD3 bidirectionally affected seeking: expression of the wild-type increased cue-induced seeking whereas expression of a catalytic dead mutant decreased it. JMJD3 expression was increased in D2+ but not D1+ medium spiny neurons. Expression of the mutant JMJD3 in D2+ neurons was sufficient to decrease cue-induced heroin seeking. CONCLUSIONS JMJD3 mediates persistent cellular and behavioral adaptations that underlie heroin relapse, and this activity is regulated by the BMP pathway.
Collapse
Affiliation(s)
- Swarup Mitra
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Craig T Werner
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Treefa Shwani
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Ana Garcia Lopez
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Dale Federico
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Kate Higdon
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Xiaofang Li
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Pedro H Gobira
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Shruthi A Thomas
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Jennifer A Martin
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Chunna An
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rachel Neve
- Gene Technology Core, Massachusetts General Hospital, Cambridge, Massachusetts
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Amy M Gancarz
- Department of Psychology, California State University, Bakersfield, Bakersfield, California
| | - David M Dietz
- Department of Pharmacology and Toxicology, Program in Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York.
| |
Collapse
|
6
|
Liang Z, Li L, Bai L, Gao Y, Qiao Y, Wang X, Yv L, Xu JT. Spinal nerve transection-induced upregulation of SAP97 via promoting membrane trafficking of GluA1-containing AMPA receptors in the dorsal horn contributes to the pathogenesis of neuropathic pain. Neurobiol Dis 2024; 194:106471. [PMID: 38461868 DOI: 10.1016/j.nbd.2024.106471] [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: 01/25/2024] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
Emerging evidence has implicated an important role of synapse-associated protein-97 (SAP97)-regulated GluA1-containing AMPARs membrane trafficking in cocaine restate and in contextual episodic memory of schizophrenia. Herein, we investigated the role of SAP97 in neuropathic pain following lumbar 5 spinal nerve transection (SNT) in rats. Our results showed that SNT led to upregulation of SAP97, enhanced the interaction between SAP97 and GluA1, and increased GluA1-containing AMPARs membrane trafficking in the dorsal horn. Microinjection of AAV-EGFP-SAP97 shRNA in lumbar 5 spinal dorsal horn inhibited SAP97 production, decreased SAP97-GluA1 interaction, reduced the membrane trafficking of GluA1-containing AMPARs, and partially attenuated neuropathic pain following SNT. Intrathecal injections of SAP97 siRNA or NASPM, an antagonist of GluA1-containing AMPARs, also partially reversed neuropathic pain on day 7, but not on day 14, after SNT. Spinal overexpression of SAP97 by AAV-EGFP-SAP97 enhanced SAP97-GluA1 interaction, increased the membrane insertion of GluA1-containing AMPARs, and induced abnormal pain in naïve rats. In addition, treatment with SAP97 siRNA or NASPM i.t. injection alleviated SNT-induced allodynia and hyperalgesia and exhibited a longer effect in female rats. Together, our results indicate that the SNT-induced upregulation of SAP97 via promoting GluA1-containing AMPARs membrane trafficking in the dorsal horn contributes to the pathogenesis of neuropathic pain. Targeting spinal SAP97 might be a promising therapeutic strategy to treatment of chronic pain.
Collapse
Affiliation(s)
- Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Lili Yv
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| |
Collapse
|
7
|
Pethő G, Kántás B, Horváth Á, Pintér E. The Epigenetics of Neuropathic Pain: A Systematic Update. Int J Mol Sci 2023; 24:17143. [PMID: 38138971 PMCID: PMC10743356 DOI: 10.3390/ijms242417143] [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: 10/10/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Epigenetics deals with alterations to the gene expression that occur without change in the nucleotide sequence in the DNA. Various covalent modifications of the DNA and/or the surrounding histone proteins have been revealed, including DNA methylation, histone acetylation, and methylation, which can either stimulate or inhibit protein expression at the transcriptional level. In the past decade, an exponentially increasing amount of data has been published on the association between epigenetic changes and the pathomechanism of pain, including its most challenging form, neuropathic pain. Epigenetic regulation of the chromatin by writer, reader, and eraser proteins has been revealed for diverse protein targets involved in the pathomechanism of neuropathic pain. They include receptors, ion channels, transporters, enzymes, cytokines, chemokines, growth factors, inflammasome proteins, etc. Most work has been invested in clarifying the epigenetic downregulation of mu opioid receptors and various K+ channels, two types of structures mediating neuronal inhibition. Conversely, epigenetic upregulation has been revealed for glutamate receptors, growth factors, and lymphokines involved in neuronal excitation. All these data cannot only help better understand the development of neuropathic pain but outline epigenetic writers, readers, and erasers whose pharmacological inhibition may represent a novel option in the treatment of pain.
Collapse
Affiliation(s)
- Gábor Pethő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti Str. 12., H-7624 Pécs, Hungary; (B.K.); (E.P.)
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2., H-7624 Pécs, Hungary;
| | - Boglárka Kántás
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti Str. 12., H-7624 Pécs, Hungary; (B.K.); (E.P.)
- Department of Obstetrics and Gynecology, University of Pécs, Édesanyák Str. 17., H-7624 Pécs, Hungary
| | - Ádám Horváth
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2., H-7624 Pécs, Hungary;
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti Str. 12., H-7624 Pécs, Hungary; (B.K.); (E.P.)
| |
Collapse
|
8
|
Sun C, Deng J, Ma Y, Meng F, Cui X, Li M, Li J, Li J, Yin P, Kong L, Zhang L, Tang P. The dual role of microglia in neuropathic pain after spinal cord injury: Detrimental and protective effects. Exp Neurol 2023; 370:114570. [PMID: 37852469 DOI: 10.1016/j.expneurol.2023.114570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/21/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition that is frequently accompanied by neuropathic pain, resulting in significant physical and psychological harm to a vast number of individuals globally. Despite the high prevalence of neuropathic pain following SCI, the precise underlying mechanism remains incompletely understood. Microglia are a type of innate immune cell that are present in the central nervous system (CNS). They have been observed to have a significant impact on neuropathic pain following SCI. This article presents a comprehensive overview of recent advances in understanding the role of microglia in the development of neuropathic pain following SCI. Specifically, the article delves into the detrimental and protective effects of microglia on neuropathic pain following SCI, as well as the mechanisms underlying their interconversion. Furthermore, the article provides a thorough overview of potential avenues for future research in this area.
Collapse
Affiliation(s)
- Chang Sun
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; Department of Orthopedics, Air Force Medical Center, PLA, Beijing, China
| | - Junhao Deng
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Yifei Ma
- School of Medicine, Nankai University, Tianjin, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiang Cui
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Ming Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jiantao Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jia Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Lingjie Kong
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| |
Collapse
|
9
|
Wang B, Liang Y, Bekker A, Hu H, Tao YX. Sensory neuron-specific long noncoding RNA in small non-peptidergic dorsal root ganglion neurons selectively impairs nerve injury-induced mechanical hypersensitivity. Life Sci 2023; 332:122120. [PMID: 37741322 PMCID: PMC10591916 DOI: 10.1016/j.lfs.2023.122120] [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: 08/05/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
AIMS Nerve injury-induced mechanical hypersensitivity is one of major clinical symptoms in neuropathic pain patients. Understanding molecular mechanisms underlying this symptom is crucial for developing effective therapies. The present study was to investigate whether sensory neuron-specific long noncoding RNA (SS-lncRNA) predominantly expressed in small non-peptidergic dorsal root ganglion (DRG) neurons repaired nerve injury-induced mechanical hypersensitivity. MATERIALS AND METHODS SS-lncRNA downregulation in the mas-related G protein-coupled receptor member D (Mrgprd)-expressed DRG neurons was rescued and mimicked by crossbreeding MrgprdCreERT2/+ lines with Rosa26SS-lncRNA knock-in mice and SS-lncRNAfl/fl mice, respectively, followed by tamoxifen injection. KEY FINDINGS Rescuing SS-lncRNA downregulation in the Mrgprd-expressed DRG neurons significantly reversed the spinal nerve ligation (SNL)-induced reduction of the calcium-activated potassium channel subfamily N member 1 (KCNN1) in these DRG neurons and alleviated the SNL-induced mechanical hypersensitivity, without affecting the SNL-induced heat and cold nociceptive hypersensitivities, on the ipsilateral side. Conversely, mimicking SS-lncRNA downregulation in the Mrgprd-expressed DRG neurons reduced basal KCNN1 expression in these DRG neurons and produced the enhanced response to mechanical stimulation, but not thermal and cold stimuli, on bilateral sides. Mechanistically, SS-lncRNA downregulation caused a reduction in its binding to lysine-specific demethylase 6B (KDM6B) and consequent recruitment of less KDM6B to Kcnn1 promoter and an increase of H3K27me3 enrichment in this promoter in injured DRG. SIGNIFICANCE Our findings suggest that SS-lncRNA downregulation in small non-peptidergic sensory neurons is required specifically for nerve injury-induced mechanical hypersensitivity likely through silencing KCNN1 expression caused by KDM6B-gated increase of H3K27me3 enrichment in Kcnn1 promoter in these neurons.
Collapse
Affiliation(s)
- Bing Wang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yingping Liang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Huijuan Hu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Physiology, Pharmacology & Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Physiology, Pharmacology & Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA.
| |
Collapse
|
10
|
Qiao Y, Li L, Bai L, Gao Y, Yang Y, Wang L, Wang X, Liang Z, Xu J. Upregulation of lysine-specific demethylase 6B aggravates inflammatory pain through H3K27me3 demethylation-dependent production of TNF-α in the dorsal root ganglia and spinal dorsal horn in rats. CNS Neurosci Ther 2023; 29:3479-3492. [PMID: 37287407 PMCID: PMC10580362 DOI: 10.1111/cns.14281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
AIMS Lysine-specific demethylase 6B (KDM6B) serves as a key mediator of gene transcription. It regulates expression of proinflammatory cytokines and chemokines in variety of diseases. Herein, the role and the underlying mechanisms of KDM6B in inflammatory pain were studied. METHODS The inflammatory pain was conducted by intraplantar injection of complete Freund's adjuvant (CFA) in rats. Immunofluorescence, Western blotting, qRT-PCR, and chromatin immunoprecipitation (ChIP)-PCR were performed to investigate the underlying mechanisms. RESULTS CFA injection led to upregulation of KDM6B and decrease in the level of H3K27me3 in the dorsal root ganglia (DRG) and spinal dorsal horn. The mechanical allodynia and thermal hyperalgesia following CFA were alleviated by the treatment of intrathecal injection of GSK-J4, and by microinjection of AAV-EGFP-KDM6B shRNA in the sciatic nerve or in lumbar 5 dorsal horn. The increased production of tumor necrosis factor-α (TNF-α) following CFA in the DRGs and dorsal horn was inhibited by these treatments. ChIP-PCR showed that CFA-induced increased binding of nuclear factor κB with TNF-α promoter was repressed by the treatment of microinjection of AAV-EGFP-KDM6B shRNA. CONCLUSIONS These results suggest that upregulated KDM6B via facilitating TNF-α expression in the DRG and spinal dorsal horn aggravates inflammatory pain.
Collapse
Affiliation(s)
- Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Li Wang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Ji‐Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
- Neuroscience Research InstituteZhengzhou UniversityZhengzhouChina
| |
Collapse
|
11
|
Gheorghe RO, Grosu AV, Magercu M, Ghenghea MS, Zbarcea CE, Tanase A, Negres S, Filippi A, Chiritoiu G, Gherghiceanu M, Dinescu S, Gaina G, Sapunar D, Ristoiu V. Switching Rat Resident Macrophages from M1 to M2 Phenotype by Iba1 Silencing Has Analgesic Effects in SNL-Induced Neuropathic Pain. Int J Mol Sci 2023; 24:15831. [PMID: 37958812 PMCID: PMC10648812 DOI: 10.3390/ijms242115831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Resident macrophages from dorsal root ganglia are important for the development of traumatic-induced neuropathic pain. In the first 5-7 days after a traumatic sciatic nerve injury (i.e., spinal nerve ligation (SNL), spared nerve injury (SNI), sciatic nerve transection or sciatic nerve ligation and transection), Ionized binding adapter protein 1 (Iba1) (+) resident macrophages cluster around dorsal root ganglia neurons, possibly contributing to nerve injury-induced hypersensitivity. Since infiltrating macrophages gradually recruited to the lesion site peak at about 7 days, the first few days post-lesion offer a window of opportunity when the contribution of Iba1 (+) resident macrophages to neuropathic pain pathogenesis could be investigated. Iba1 is an actin cross-linking cytoskeleton protein, specifically located only in macrophages and microglia. In this study, we explored the contribution of rat Iba1 (+) macrophages in SNL-induced neuropathic pain by using intra-ganglionic injections of naked Iba1-siRNA, delivered at the time the lesion occurred. The results show that 5 days after Iba1 silencing, Iba1 (+) resident macrophages are switched from an M1 (pro-inflammatory) phenotype to an M2 (anti-inflammatory) phenotype, which was confirmed by a significant decrease of M1 markers (CD32 and CD86), a significant increase of M2 markers (CD163 and Arginase-1), a reduced secretion of pro-inflammatory cytokines (IL-6, TNF-α and IL-1β) and an increased release of pro-regenerative factors (BDNF, NGF and NT-3) which initiated the regrowth of adult DRG neurites and reduced SNL-induced neuropathic pain. Our data show for the first time, that it is possible to induce macrophages towards an anti-inflammatory phenotype by interacting with their cytoskeleton.
Collapse
Affiliation(s)
- Roxana-Olimpia Gheorghe
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, District 5, 050095 Bucharest, Romania; (R.-O.G.)
| | - Andreea Violeta Grosu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, District 5, 050095 Bucharest, Romania; (R.-O.G.)
| | - Melania Magercu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, District 5, 050095 Bucharest, Romania; (R.-O.G.)
| | - Mihail-Sebastian Ghenghea
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, District 5, 050095 Bucharest, Romania; (R.-O.G.)
| | - Cristina Elena Zbarcea
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, 6 Traian Vuia Street, District 2, 02095 Bucharest, Romania
| | - Alexandra Tanase
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, 6 Traian Vuia Street, District 2, 02095 Bucharest, Romania
| | - Simona Negres
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, 6 Traian Vuia Street, District 2, 02095 Bucharest, Romania
| | - Alexandru Filippi
- Department of Biophysics, University of Medicine and Pharmacy “Carol Davila”, 8 Eroilor Sanitari Blvd., 050474 Bucharest, Romania
| | - Gabriela Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, 2996 Splaiul Independentei 296, District 6, 060031 Bucharest, Romania
| | - Mihaela Gherghiceanu
- Ultrastructural Pathology and Bioimaging Laboratory, Victor Babeș National Institute of Pathology Bucharest, 99-101 Splaiul Independentei, District 5, 050096 Bucharest, Romania
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei, District 5, 050095 Bucharest, Romania
| | - Gisela Gaina
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei, District 5, 050095 Bucharest, Romania
| | - Damir Sapunar
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| | - Violeta Ristoiu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, District 5, 050095 Bucharest, Romania; (R.-O.G.)
| |
Collapse
|
12
|
Zhang H, Li A, Liu YF, Sun ZM, Jin BX, Lin JP, Yang Y, Yao YX. Spinal TAOK2 contributes to neuropathic pain via cGAS-STING activation in rats. iScience 2023; 26:107792. [PMID: 37720090 PMCID: PMC10502416 DOI: 10.1016/j.isci.2023.107792] [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: 10/18/2022] [Revised: 05/25/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
Thousand and one amino acid kinase 2 (TAOK2) is a member of the mammalian sterile 20 kinase family and is implicated in neurodevelopmental disorders; however, its role in neuropathic pain remains unknown. Here, we found that TAOK2 was enriched and activated after chronic constriction injury (CCI) in the rat spinal dorsal horn. Meanwhile, cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling was also activated with hyperalgesia. Silencing TAOK2 reversed hyperalgesia and suppressed the activation of cGAS-STING signaling induced by CCI, while pharmacological activation of TAOK2 induced pain hypersensitivity and upregulation of cGAS-STING signaling in naive rats. Furthermore, pharmacological inhibition or gene silencing of cGAS-STING signaling attenuated CCI-induced hyperalgesia. Taken together, these data demonstrate that the activation of spinal TAOK2 contributes to CCI-induced hyperalgesia via cGAS-STING signaling activation, providing new molecular targets for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Ang Li
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
- Department of Anesthesia, People’s Hospital of Guizhou Province, Guiyang, Guizhou 550025, China
| | - Yu-Fan Liu
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Zhong-Ming Sun
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Bing-Xin Jin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jia-Piao Lin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yan Yang
- Department of Neurobiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
- Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yong-Xing Yao
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
13
|
Zhang J, Zhang X, Gao Y, Li L, Bai L, Wang L, Qiao Y, Wang X, Liang Z, Xu JT. Neuralized1-Mediated CPEB3 Ubiquitination in the Spinal Dorsal Horn Contributes to the Pathogenesis of Neuropathic Pain in Rats. ACS Chem Neurosci 2023; 14:3418-3430. [PMID: 37644621 DOI: 10.1021/acschemneuro.3c00313] [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] [Indexed: 08/31/2023] Open
Abstract
Compelling evidence has shown that Neuralized1 (Neurl1) facilitates hippocampal-dependent memory storage by modulating cytoplasmic polyadenylation element-binding protein 3 (CPEB3)-dependent protein synthesis. In the current study, we investigated the role of Neurl1 in the pathogenesis of neuropathic pain and the underlying mechanisms. The neuropathic pain was evaluated by lumbar 5 spinal nerve ligation (SNL) in rats. Immunofluorescence staining, Western blotting, qRT-PCR, and coimmunoprecipitation (Co-IP) were performed to investigate the underlying mechanisms. Our results showed that SNL led to an increase of Neurl1 in the spinal dorsal horn. Spinal microinjection of AAV-EGFP-Neurl1 shRNA alleviated mechanical allodynia; decreased the level of CPEB3 ubiquitination; inhibited the production of GluA1, GluA2, and PSD95; and reduced GluA1-containing AMPA receptors in the membrane of the dorsal horn following SNL. Knockdown of spinal CPEB3 decreased the production of GluA1, GluA2, and PSD95 in the dorsal horn and attenuated abnormal pain after SNL. Overexpression of Neurl1 in the dorsal horn resulted in pain-related hypersensitivity in naïve rats; raised the level of CPEB3 ubiquitination; increased the production of GluA1, GluA2, and PSD95; and augmented GluA1-containing AMPA receptors in the membrane in the dorsal horn. Moreover, spinal Neurl1 overexpression-induced mechanical allodynia in naïve rats was partially reversed by repeated intrathecal injections of CPEB3 siRNA. Collectively, our results suggest that SNL-induced upregulation of Neurl1 through CPEB3 ubiquitination-dependent production of GluA1, GluA2, and PSD95 in the dorsal horn contributes to the pathogenesis of neuropathic pain in rats. Targeting spinal Neurl1 might be a promising therapeutic strategy for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Li Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
- Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| |
Collapse
|
14
|
Chen Y, Han Y, Liu L, Liu M, Lin J, Tang Y, Guo S, He R, Wu Q. N 6-Methyladenosine methylase METTL3 contributes to neuropathic pain by epigenetic silencing of mu opioid receptor. Behav Brain Res 2023; 452:114592. [PMID: 37482304 DOI: 10.1016/j.bbr.2023.114592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/02/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
We aimed at exploring the role and mechanism of METTL3-mediated m6A modification in neuropathic pain. Male Sprague-Dawley rats were randomly divided into four groups: Sham operation group (Sham group), chronic constriction injury (CCI) of the sciatic nerve model group (NPP group), intrathecal injection of virus down-regulated METTL3 + CCI model group (M3 + NPP group) and intrathecal injection of negative control virus + CCI model group (Scr + NPP group). The M3 + NPP group and the Scr + NPP group were intrathecally injected with virus nineteen days before operation. The paw withdrawal mechanical thresholds and paw withdrawal latency were respectively recorded one day before operation, three days, five days and seven days after operation. The rats were sacrificed on the seventh day after operation, and their spinal cord tissues were taken. The frozen sections of rats were performed to observe the expression of green fluorescent protein of the virus. The methylation level of RNA, the protein expression of m6A-related enzyme (METTL3) and mu opioid receptor (MOR) in spinal cord tissues of the four groups were measured. Downregulation of METTL3 had no effect on the overall methylation level of the spinal cord, but it could regulate the methylation level of the OPRM1 gene RNA encoding MOR, partially restore the expression of MOR, and relieve pain in rats. In the process of NPP, METTL3 may inhibit the expression of MOR by regulating the methylation level of OPRM1 gene RNA encoding MOR, and ultimately promote the occurrence and development of NPP.
Collapse
Affiliation(s)
- Yao Chen
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China
| | - Yakun Han
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China
| | - Li Liu
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China
| | - Minqiang Liu
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China
| | - Jing Lin
- Central Sterile Supply Department, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China
| | - Yi Tang
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China
| | - Shanshan Guo
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China
| | - Renliang He
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China.
| | - Qiang Wu
- Department of Anesthsiology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong 518112, China.
| |
Collapse
|
15
|
Zhang J, Zhang X, Li L, Bai L, Gao Y, Yang Y, Wang L, Qiao Y, Wang X, Xu JT. Activation of Double-Stranded RNA-Activated Protein Kinase in the Dorsal Root Ganglia and Spinal Dorsal Horn Regulates Neuropathic Pain Following Peripheral Nerve Injury in Rats. Neurotherapeutics 2022; 19:1381-1400. [PMID: 35655111 PMCID: PMC9587175 DOI: 10.1007/s13311-022-01255-2] [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] [Accepted: 05/25/2022] [Indexed: 10/18/2022] Open
Abstract
Double-stranded RNA (dsRNA)-activated kinase (PKR) is an important component in inflammation and immune dysfunction. However, the role of PKR in neuropathic pain remains unclear. Here, we showed that lumbar 5 spinal nerve ligation (SNL) led to a significant increase in the level of phosphorylated PKR (p-PKR) in both the dorsal root ganglia (DRG) and spinal dorsal horn. Images of double immunofluorescence staining revealed that p-PKR was expressed in myelinated A-fibers, unmyelinated C-fibers, and satellite glial cells in the DRG. In the dorsal horn, p-PKR was located in neuronal cells, astrocytes, and microglia. Data from behavioral tests showed that intrathecal (i.t.) injection of 2-aminopurine (2-AP), a specific inhibitor of PKR activation, and PKR siRNA prevented the reductions in PWT and PWL following SNL. Established neuropathic pain was also attenuated by i.t. injection of 2-AP and PKR siRNA, which started on day 7 after SNL. Prior repeated i.t. injections of PKR siRNA prevented the SNL-induced degradation of IκBα and IκBβ in the cytosol and the nuclear translocation of nuclear factor κB (NF-κB) p65 in both the DRG and dorsal horn. Moreover, the SNL-induced increase in interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) production was diminished by this treatment. Collectively, these results suggest that peripheral nerve injury-induced PKR activation via NF-κB signaling-regulated expression of proinflammatory cytokines in the DRG and dorsal horn contributes to the pathogenesis of neuropathic pain. Our findings suggest that pharmacologically targeting PKR might be an effective therapeutic strategy for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Li Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China.
- Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, China.
| |
Collapse
|
16
|
Abstract
Neuropathic pain is a challenging clinical problem and remains difficult to treat. Altered gene expression in peripheral sensory nerves and neurons due to nerve injury is well documented and contributes critically to the synaptic plasticity in the spinal cord and the initiation and maintenance of chronic pain. However, our understanding of the epigenetic mechanisms regulating the transcription of pro-nociceptive (e.g., NMDA receptors and α2δ-1) and antinociceptive (e.g., potassium channels and opioid and cannabinoid receptors) genes are still limited. In this review, we summarize recent studies determining the roles of histone modifications (including methylation, acetylation, and ubiquitination), DNA methylation, and noncoding RNAs in neuropathic pain development. We review the epigenetic writer, reader, and eraser proteins that participate in the transcriptional control of the expression of key ion channels and neurotransmitter receptors in the dorsal root ganglion after traumatic nerve injury, which is commonly used as a preclinical model of neuropathic pain. A better understanding of epigenetic reprogramming involved in the transition from acute to chronic pain could lead to the development of new treatments for neuropathic pain.
Collapse
Affiliation(s)
- Krishna Ghosh
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|