1
|
You C, Zhou W, Ye P, Zhang L, Sun W, Tian L, Peng B, Hu M, Xu B. LncRNA 4933431K23Rik modulate microglial phenotype via inhibiting miR-10a-5p in spinal cord injury induced neuropathic pain. Sci Rep 2025; 15:11620. [PMID: 40185764 PMCID: PMC11971278 DOI: 10.1038/s41598-025-91021-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 02/18/2025] [Indexed: 04/07/2025] Open
Abstract
Neuropathic pain (NP) is caused by primary damage and dysfunction of nervous system, in which spinal cord injury (SCI) is a common cause of NP. Evidence shows that neuroinflammation and oxidative stress are related to the pathophysiology of NP, in which the activation of microglia and astrocytes in spinal is significant. Therefore, understanding the molecular mechanism of NP after SCI is of great significance. The rat model of SCI was established and BV2 cell was treated with LPS. The exosomes derived from astrocytes were extracted by centrifugation. The morphology of the exosomes was observed by electron microscope and the surface markers were detected by Western blot. LncRNA in astrocytes and astrocyte-derived exosomes were detected by qRT-PCR. The expression of microglia activation markers CD68 and Iba-1 was detected by immunohistochemistry. The von Frey test was applied to assess mechanical hypersensitivity. The heat plate analgesia instrument was used to evaluate Paw withdrawal latency (PWL). QRT-PCR used to detect expression of LncRNA49rik and miR-10a-5p. Western blot was used to detect MAPK/PI3K/AKT / mTOR signal pathway and COX2, iNOS. The content of MDA and the activity of SOD were detected by oxidative stress kit. The concentrations of IL-6, IL-1β, IL-18 and IFN-αwere detected by ELISA. The targeting relationship between LncRNA49rik and miR-10a-5p was analyzed by bioinformatics and double luciferase activity, Rip and FISH experiments. LncRNA49rik was highly expressed in astrocytes and its derived exosomes. SCI stimulated astrocytes to release exosome containing LncRNA49rik and promote microglia activation to increase inflammatory response. At the same time, overexpression of LncRNA49rik increased the incidence of NP and aggravated the level of inflammation and oxidative stress in rats with SCI. MiR-10a-5p is the target of LncRNA4933431K23Rik. Overexpression of LncRNA49rik significantly inhibited the up-regulation of miR-10a-5p. Overexpression of miR-10a-5p inhibited hyperalgesia and inflammation in SCI rats. In addition, transfection of miR-10a-5p mimics significantly inhibited the expression of MAPK/PI3K/AKT and up-regulated the expression of mTOR. Mechanism studies have shown that overexpression of miR-10a-5p weakens the phenotypic induction of microglia induced by LncRNA4933431K23Rik. LncRNA4933431K23Rik regulates microglial phenotype through inhibiting miR-10a-5p, which is responsible for NP induced by SCI.
Collapse
Affiliation(s)
- Changhui You
- Department of Anesthesiology, The First School of Clinical Medicine, General Hospital of Southern Theatre Command of PLA, Southern Medical University, Guangzhou, China
- Pain Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Waiping Zhou
- Pain Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Ye
- Radiology Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhang
- Pain Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenchao Sun
- Pain Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Tian
- Pain Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bocheng Peng
- Pain Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengying Hu
- Pain Department, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Xu
- Department of Anesthesiology, The First School of Clinical Medicine, General Hospital of Southern Theatre Command of PLA, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Ke X, Cai H, Luo F, Zheng X, Hu Q, Zhou Y, Wang Y, Zhang X, Chen Y, Chen G. TRPC4 Mediates Trigeminal Neuropathic Pain via Ca 2+-ERK/P38-ATF2 Pathway in the Trigeminal Ganglion of Mice. CNS Neurosci Ther 2025; 31:e70368. [PMID: 40202077 PMCID: PMC11979714 DOI: 10.1111/cns.70368] [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: 11/29/2024] [Revised: 02/19/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Trigeminal neuropathic pain (TNP) is a debilitating condition characterized by chronic facial pain, yet its underlying mechanisms remain incompletely understood. Transient Receptor Potential Canonical 4 (TRPC4) has been reported to promote the development of abnormal pain or pain hypersensitivity in neuropathic pain. However, the specific contribution of TRPC4 to TNP pathogenesis remains unclear. AIM This study aimed to investigate the role of TRPC4 in a mouse model of trigeminal neuropathic pain induced by chronic constriction of the unilateral infraorbital nerve (CION). METHODS Adult male/female mice were subjected to either CION surgery or sham surgery. Behavioral assays were conducted to assess facial pain-like responses over a 28-day period. TRPC4 distribution in the trigeminal ganglion (TG) was evaluated using Immunofluorescence. TRPC4 inhibitor ML204 and agonist Englerin A were employed to evaluate the impact of TRPC4 on facial pain-like behaviors. A TRPC4-overexpressing HEK293 cell model was conducted via plasmid transfection. To assess the function of TRPC4, we employed cellular calcium imaging technology to investigate the effects of modulating TRPC4 function by analyzing dynamic changes in intracellular calcium ion concentrations in primary trigeminal ganglion neurons and HEK293 cells. Trpc4 shRNA was used to specifically knock down TRPC4 in the trigeminal ganglion. Western blot analysis was used to assess the activation of ERK, P38, and ATF2 signaling pathways. RESULTS Mice subjected to CION exhibited persistent facial pain-like behaviors and a significant increase in TRPC4 expression in TG neurons. Trpc4 shRNA or pharmacological inhibition with ML204 attenuated CION-induced pain behaviors, while activation of TRPC4 with Englerin A induced pain-like responses in naive mice. Calcium imaging revealed that both Englerin A and TRPC4 overexpression elevated intracellular Ca²2+ levels in TG neurons and HEK293 cells. This Ca²2+ influx triggered the activation of ERK and P38, leading to enhanced ATF2 activation. Downregulation of TRPC4 in the TG reduced ERK/P38 phosphorylation and ATF2 expression and activation. CONCLUSION This study provides the first evidence that TRPC4 plays a critical role in CION-induced trigeminal neuropathic pain by promoting the activation of the downstream transcription factor ATF2 via the Ca²2+-ERK/P38 pathway.
Collapse
Affiliation(s)
- Xinlong Ke
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Huajing Cai
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Fangla Luo
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Xing Zheng
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Qian Hu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Youfa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityZhejiangHangzhouChina
| | - Xiangnan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityZhejiangHangzhouChina
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| |
Collapse
|
3
|
Lee KH, Kim UJ, Cha M, Lee BH. Inhibiting Nav1.7 channels in pulpitis: An in vivo study on neuronal hyperexcitability. Biochem Biophys Res Commun 2024; 717:150044. [PMID: 38718567 DOI: 10.1016/j.bbrc.2024.150044] [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: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024]
Abstract
Pulpitis constitutes a significant challenge in clinical management due to its impact on peripheral nerve tissue and the persistence of chronic pain. Despite its clinical importance, the correlation between neuronal activity and the expression of voltage-gated sodium channel 1.7 (Nav1.7) in the trigeminal ganglion (TG) during pulpitis is less investigated. The aim of this study was to examine the relationship between experimentally induced pulpitis and Nav1.7 expression in the TG and to investigate the potential of selective Nav1.7 modulation to attenuate TG abnormal activity associated with pulpitis. Acute pulpitis was induced at the maxillary molar (M1) using allyl isothiocyanate (AITC). The mice were divided into three groups: control, pulpitis model, and pulpitis model treated with ProTx-II, a selective Nav1.7 channel inhibitor. After three days following the surgery, we conducted a recording and comparative analysis of the neural activity of the TG utilizing in vivo optical imaging. Then immunohistochemistry and Western blot were performed to assess changes in the expression levels of extracellular signal-regulated kinase (ERK), c-Fos, collapsin response mediator protein-2 (CRMP2), and Nav1.7 channels. The optical imaging result showed significant neurological excitation in pulpitis TGs. Nav1.7 expressions exhibited upregulation, accompanied by signaling molecular changes suggestive of inflammation and neuroplasticity. In addition, inhibition of Nav1.7 led to reduced neural activity and subsequent decreases in ERK, c-Fos, and CRMP2 levels. These findings suggest the potential for targeting overexpressed Nav1.7 channels to alleviate pain associated with pulpitis, providing practical pain management strategies.
Collapse
Affiliation(s)
- Kyung Hee Lee
- Department of Dental Hygiene, Division of Health Science, Dongseo University, Busan, 47011, Republic of Korea
| | - Un Jeng Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
4
|
Asiedu K. Neurophysiology of corneal neuropathic pain and emerging pharmacotherapeutics. J Neurosci Res 2024; 102:e25285. [PMID: 38284865 DOI: 10.1002/jnr.25285] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/12/2023] [Accepted: 12/02/2023] [Indexed: 01/30/2024]
Abstract
The altered activity generated by corneal neuronal injury can result in morphological and physiological changes in the architecture of synaptic connections in the nervous system. These changes can alter the sensitivity of neurons (both second-order and higher-order projection) projecting pain signals. A complex process involving different cell types, molecules, nerves, dendritic cells, neurokines, neuropeptides, and axon guidance molecules causes a high level of sensory rearrangement, which is germane to all the phases in the pathomechanism of corneal neuropathic pain. Immune cells migrating to the region of nerve injury assist in pain generation by secreting neurokines that ensure nerve depolarization. Furthermore, excitability in the central pain pathway is perpetuated by local activation of microglia in the trigeminal ganglion and alterations of the descending inhibitory modulation for corneal pain arriving from central nervous system. Corneal neuropathic pain may be facilitated by dysfunctional structures in the central somatosensory nervous system due to a lesion, altered synaptogenesis, or genetic abnormality. Understanding these important pathways will provide novel therapeutic insight.
Collapse
Affiliation(s)
- Kofi Asiedu
- School of Optometry & Vision Science, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Wu L, Tang H. The role of N6-methyladenosine modification in rodent models of neuropathic pain: from the mechanism to therapeutic potential. Biomed Pharmacother 2023; 166:115398. [PMID: 37647691 DOI: 10.1016/j.biopha.2023.115398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
Neuropathic pain (NP) is a common chronic pain condition resulted from lesions or diseases of somatosensory nervous system, but the pathogenesis remains unclear. A growing body of evidence supports the relationship between pathogenesis and N6-methyladenosine (m6A) modifications of RNA. However, studies on the role of m6A modifications in NP are still at an early stage. Elucidating different etiologies is important for understanding the specific pathogenesis of NP. This article provides a comprehensive review on the role of m6A methylation modifications including methyltransferases ("writers"), demethylases ("erasers"), and m6A binding proteins ("readers") in NP models. Further analysis of the pathogenic mechanism relationship between m6A and NP provided novel theoretical and practical significance for clinical treatment of NP.
Collapse
Affiliation(s)
- Liping Wu
- Guangxi University of Traditional Chinese Medicine, Nanning, China; The First Clinical Medical College of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Hongliang Tang
- Guangxi Traditional Chinese Medicine University Affiliated Fangchenggang Hospital.
| |
Collapse
|
6
|
Shao J, Yu W, Wei W, Wang S, Zheng Z, Li L, Sun Y, Zhang J, Li Z, Ren X, Zang W, Cao J. MAPK-ERK-CREB signaling pathway upregulates Nav1.6 in oxaliplatin-induced neuropathic pain in the rat. Toxicol Lett 2023; 384:149-160. [PMID: 37453670 DOI: 10.1016/j.toxlet.2023.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
The voltage-gated sodium channel subtype Nav1.6 is involved in the electrophysiological changes of primary sensory neurons that occur in oxaliplatin-induced neuropathic pain, but its regulatory mechanism remains unclear. In this study, Western blot, RT-qPCR, immunofluorescence staining, chromatin immunoprecipitation were used to prove the mechanism of MAPK-ERK-CREB signaling pathway participating in oxaliplatin-induced neuropathic pain by regulating Nav1.6. The results showed that p-Raf1 and p-ERK, key molecules in MAPK/ERK pathway, and Nav1.6 were significantly increased in DRGs of oxaliplatin-induced neuropathic pain rats. Inhibition of p-Raf1 and p-ERK respectively not only reduced the expression of Nav1.6 protein in DRGs of OXA rats, but also caused a decrease in Nav1.6 mRNA, which led us to further explore the transcription factor CREB regulated by MAPK/ERK pathway. Results showed that CREB was co-distributed with Nav1.6. Inhibition of CREB resulted in decreased mRNA and protein expression of Nav1.6, and alleviated oxaliplatin-induced neuropathic pain. A chromatin immunoprecipitation experiment proved that OXA caused p-CREB to directly bind to the promoter region of Scn8A, which is the encoding gene for Nav1.6, and promote the transcription of Scn8A. In summary, in this study, we found that oxaliplatin can activate the MAPK/ERK pathway, which promotes the expression and activation of CREB and leads to an increase in Scn8A transcription, and then leads to an increase in Nav1.6 protein expression to enhance neuronal excitability and cause pain. This study provides an experimental basis for the molecular mechanism of sodium channel regulation in oxaliplatin-induced neuropathic pain.
Collapse
Affiliation(s)
- Jinping Shao
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Wenli Yu
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China; Xinyang Central Hospital, Xinyang, China
| | - Wei Wei
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Suifeng Wang
- Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China; The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenli Zheng
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Lei Li
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Yanyan Sun
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Jingjing Zhang
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Zhihua Li
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Xiuhua Ren
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Weidong Zang
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China.
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Zhengzhou 450001, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|