1
|
Jiang C, Zhao J, Zhang Y, Zhu X. Role of EPAC1 in chronic pain. Biochem Biophys Rep 2024; 37:101645. [PMID: 38304575 PMCID: PMC10832381 DOI: 10.1016/j.bbrep.2024.101645] [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: 11/18/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
Chronic pain usually lasts over three months and commonly occurs in chronic diseases (cancer, arthritis, and diabetes), injuries (herniated discs, torn ligaments), and many major pain disorders (neuropathic pain, fibromyalgia, chronic headaches). Unfortunately, there is currently a lack of effective treatments to help people with chronic pain to achieve complete relief. Therefore,it is particularly important to understand the mechanism of chronic pain and find new therapeutic targets. The exchange protein directly activated by cyclic adenosine monophosphate(cAMP) (EPAC) has been recognized for its functions in nerve regeneration, stimulating insulin release, controlling vascular pressure, and controlling other metabolic activities. In recent years, many studies have found that the subtype of EPAC, EPAC1 is involved in the regulation of neuroinflammation and plays a crucial role in the regulation of pain, which is expected to become a new therapeutic target for chronic pain. This article reviews the major contributions of EPAC1 in chronic pain.
Collapse
Affiliation(s)
- Chenlu Jiang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong, 226001, China
| | - Jiacheng Zhao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong, 226001, China
| | - Yihang Zhang
- Medical School of Nantong University, Nantong, 226001, China
| | - Xiang Zhu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| |
Collapse
|
2
|
Jin Y, Zhou J, Fang Y, Song H, Lin S, Pan B, Liu L, Xiong B. Electroacupuncture prevents the development or establishment of chronic pain via IL-33/ST2 signaling in hyperalgesic priming model rats. Neurosci Lett 2024; 820:137611. [PMID: 38142925 DOI: 10.1016/j.neulet.2023.137611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Chronic pain is acomplexhealth issue. Compared to acute pain, which has a protective value, chronic pain is defined as persistent pain after tissue injury. Few clinical advances have been made to prevent the transition from acute to chronic pain. Electroacupuncture (EA), the most common form of acupuncture, is widely used in clinical practice to relieve pain. METHODS The hyperalgesic priming model, established via a carrageenan injection followed by a prostaglandin E2 injection, was used to investigate the development or establishment of chronic pain. We observed the hyperalgesic effect of EA on rats and investigated the expression p38 mitogen-activated protein kinase, interleukin-33 (IL-33), and its receptor ST2 in astrocytes in the L4-L6 spinal cord dorsal horns (SDHs) after EA. The IL-33/ST2 signaling pathway in SDH is associated with the development of chronic pain. RESULTS EA can reverse the pain threshold in hyperalgesic priming model rats and regulates the expression of phosphorylated p38, IL-33, and ST2 in astrocytes in the L4-L6 SDHs. We discovered that EA raises the pain threshold. This suggests that EA can prevent the development or establishment of chronic pain by inhibiting IL-33/ST2 signaling in the lower central nervous system. CONCLUSIONS EA can alleviate the development or establishment of chronic pain by modulating IL-33/ST2 signaling in SDHs. Our findings will help clinicians understand the mechanisms of EA analgesia.
Collapse
Affiliation(s)
- Ying Jin
- Department of Rehabilitation in Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou City, Zhejiang Province 310009, China; Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, 155 Hanzhong Road, Nanjing City, Jiangsu 210029, China
| | - Jie Zhou
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, 219 Moganshan Road, Xihu District, Hangzhou City, Zhejiang Province 310005, China
| | - Yinfeng Fang
- The School of Communication Engineering, Hangzhou Dianzi University, Hangzhou City, Zhejiang Province 310018, China
| | - Hongyun Song
- Department of Rehabilitation in Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou City, Zhejiang Province 310009, China
| | - Shiming Lin
- Department of Rehabilitation in Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou City, Zhejiang Province 310009, China
| | - Bowen Pan
- Department of Traumatology, Affiliated Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Lanying Liu
- Department of Acupuncture and Rehabilitation, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, 155 Hanzhong Road, Nanjing City, Jiangsu 210029, China.
| | - Bing Xiong
- Department of Rehabilitation in Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou City, Zhejiang Province 310009, China.
| |
Collapse
|
3
|
Silnitsky S, Rubin SJS, Zerihun M, Qvit N. An Update on Protein Kinases as Therapeutic Targets-Part I: Protein Kinase C Activation and Its Role in Cancer and Cardiovascular Diseases. Int J Mol Sci 2023; 24:17600. [PMID: 38139428 PMCID: PMC10743896 DOI: 10.3390/ijms242417600] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Protein kinases are one of the most significant drug targets in the human proteome, historically harnessed for the treatment of cancer, cardiovascular disease, and a growing number of other conditions, including autoimmune and inflammatory processes. Since the approval of the first kinase inhibitors in the late 1990s and early 2000s, the field has grown exponentially, comprising 98 approved therapeutics to date, 37 of which were approved between 2016 and 2021. While many of these small-molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP binding pocket have been massively successful for oncological indications, their poor selectively for protein kinase isozymes have limited them due to toxicities in their application to other disease spaces. Thus, recent attention has turned to the use of alternative allosteric binding mechanisms and improved drug platforms such as modified peptides to design protein kinase modulators with enhanced selectivity and other pharmacological properties. Herein we review the role of different protein kinase C (PKC) isoforms in cancer and cardiovascular disease, with particular attention to PKC-family inhibitors. We discuss translational examples and carefully consider the advantages and limitations of each compound (Part I). We also discuss the recent advances in the field of protein kinase modulators, leverage molecular docking to model inhibitor-kinase interactions, and propose mechanisms of action that will aid in the design of next-generation protein kinase modulators (Part II).
Collapse
Affiliation(s)
- Shmuel Silnitsky
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Samuel J. S. Rubin
- Department of Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA;
| | - Mulate Zerihun
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| |
Collapse
|
4
|
Nagaraja S, Tewari SG, Reifman J. Predictive analytics identifies key factors driving hyperalgesic priming of muscle sensory neurons. Front Neurosci 2023; 17:1254154. [PMID: 37942142 PMCID: PMC10629345 DOI: 10.3389/fnins.2023.1254154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/25/2023] [Indexed: 11/10/2023] Open
Abstract
Hyperalgesic priming, a form of neuroplasticity induced by inflammatory mediators, in peripheral nociceptors enhances the magnitude and duration of action potential (AP) firing to future inflammatory events and can potentially lead to pain chronification. The mechanisms underlying the development of hyperalgesic priming are not well understood, limiting the identification of novel therapeutic strategies to combat chronic pain. In this study, we used a computational model to identify key proteins whose modifications caused priming of muscle nociceptors and made them hyperexcitable to a subsequent inflammatory event. First, we extended a previously validated model of mouse muscle nociceptor sensitization to incorporate Epac-mediated interaction between two G protein-coupled receptor signaling pathways commonly activated by inflammatory mediators. Next, we calibrated and validated the model simulations of the nociceptor's AP response to both innocuous and noxious levels of mechanical force after two subsequent inflammatory events using literature data. Then, by performing global sensitivity analyses that simulated thousands of nociceptor-priming scenarios, we identified five ion channels and two molecular processes (from the 18 modeled transmembrane proteins and 29 intracellular signaling components) as potential regulators of the increase in AP firing in response to mechanical forces. Finally, when we simulated specific neuroplastic modifications in Kv1.1 and Nav1.7 alone as well as with simultaneous modifications in Nav1.7, Nav1.8, TRPA1, and Kv7.2, we observed a considerable increase in the fold change in the number of triggered APs in primed nociceptors. These results suggest that altering the expression of Kv1.1 and Nav1.7 might regulate the neuronal hyperexcitability in primed mechanosensitive muscle nociceptors.
Collapse
Affiliation(s)
- Sridevi Nagaraja
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Development Command, Fort Detrick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Shivendra G. Tewari
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Development Command, Fort Detrick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Jaques Reifman
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, US Army Medical Research and Development Command, Fort Detrick, MD, United States
| |
Collapse
|
5
|
Bohic M, Pattison LA, Jhumka ZA, Rossi H, Thackray JK, Ricci M, Mossazghi N, Foster W, Ogundare S, Twomey CR, Hilton H, Arnold J, Tischfield MA, Yttri EA, St John Smith E, Abdus-Saboor I, Abraira VE. Mapping the neuroethological signatures of pain, analgesia, and recovery in mice. Neuron 2023; 111:2811-2830.e8. [PMID: 37442132 PMCID: PMC10697150 DOI: 10.1016/j.neuron.2023.06.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 12/16/2022] [Accepted: 06/16/2023] [Indexed: 07/15/2023]
Abstract
Ongoing pain is driven by the activation and modulation of pain-sensing neurons, affecting physiology, motor function, and motivation to engage in certain behaviors. The complexity of the pain state has evaded a comprehensive definition, especially in non-verbal animals. Here, in mice, we used site-specific electrophysiology to define key time points corresponding to peripheral sensitivity in acute paw inflammation and chronic knee pain models. Using supervised and unsupervised machine learning tools, we uncovered sensory-evoked coping postures unique to each model. Through 3D pose analytics, we identified movement sequences that robustly represent different pain states and found that commonly used analgesics do not return an animal's behavior to a pre-injury state. Instead, these analgesics induce a novel set of spontaneous behaviors that are maintained even after resolution of evoked pain behaviors. Together, these findings reveal previously unidentified neuroethological signatures of pain and analgesia at heightened pain states and during recovery.
Collapse
Affiliation(s)
- Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA
| | - Luke A Pattison
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Z Anissa Jhumka
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Heather Rossi
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Joshua K Thackray
- Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA; Tourette International Collaborative Genetics Study (TIC Genetics), Piscataway, NJ, USA
| | - Matthew Ricci
- Data Science Initiative, Brown University, Providence, RI, USA; School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nahom Mossazghi
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - William Foster
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Simon Ogundare
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Colin R Twomey
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Justin Arnold
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Max A Tischfield
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA; Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA; Tourette International Collaborative Genetics Study (TIC Genetics), Piscataway, NJ, USA
| | - Eric A Yttri
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Victoria E Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
6
|
Du J, Yi M, Xi D, Wang S, Liu B, Shao X, Liang Y, He X, Fang J, Fang J. Satellite glial cells drive the transition from acute to chronic pain in a rat model of hyperalgesic priming. Front Mol Neurosci 2023; 16:1089162. [PMID: 36818653 PMCID: PMC9931746 DOI: 10.3389/fnmol.2023.1089162] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Chronic pain is one of the most common clinical syndromes affecting patients' quality of life. Regulating the transition from acute to chronic pain is a novel therapeutic strategy for chronic pain that presents a major clinical challenge. However, the mechanism underlying pain transitions remains poorly understood. A rat hyperalgesic priming (HP) model, which mimics pain transition, was established decades ago. Here, this HP model and RNA sequencing (RNA-seq) were used to study the potential role of neuroinflammation in pain transition. In this study, HP model rats developed prolonged hyperalgesia in the hind paw after carrageenan (Car) and PGE2 injection, accompanied by obvious satellite glial cell (SGC) activation in the dorsal root ganglion (DRG), as indicated by upregulation of GFAP. RNA-Seq identified a total of differentially expressed genes in the ipsilateral DRG in HP model rats. The expression of several representative genes was confirmed by real-time quantitative PCR (qPCR). Functional analysis of the differentially expressed genes indicated that genes related to the inflammatory and neuroinflammatory response showed the most significant changes in expression. We further found that the expression of the chemokine CXCL1 was significantly upregulated in the rat DRG. Pharmacological blockade of CXCL1 reduced protein kinase C epsilon overproduction as well as hyperalgesia in HP rats but did not prevent the upregulation of GFAP in the DRG. These results reveal that neuroinflammatory responses are involved in pain transition and may be the source of chronic pain. The chemokine CXCL1 in the DRG is a pivotal contributor to chronic pain and pain transition in HP model rats. Thus, our study provides a putative novel target for the development of effective therapeutics to prevent pain transition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Junfan Fang
- *Correspondence: Jianqiao Fang, ; Junfan Fang,
| |
Collapse
|
7
|
Shi M, Zhou J, Hu R, Xu H, Chen Y, Wu X, Chen B, Ma R. EA participates in pain transition through regulating KCC2 expression by BDNF-TrkB in the spinal cord dorsal horn of male rats. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100115. [PMID: 36875547 PMCID: PMC9982673 DOI: 10.1016/j.ynpai.2023.100115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 06/18/2023]
Abstract
The pathogenesis of chronic pain is complex and poorly treated, seriously affecting the quality of life of patients. Electroacupuncture (EA) relieves pain by preventing the transition of acute pain into chronic pain, but its mechanism of action is still unclear. Here, we aimed to investigate whether EA can inhibit pain transition by increasing KCC2 expression via BDNF-TrkB. We used hyperalgesic priming (HP) model to investigate the potential central mechanisms of EA intervention on pain transition. HP model male rats showed significant and persistent mechanically abnormal pain. Brain derived neurotrophic factor (BDNF) expression and Tropomyosin receptor kinase B (TrkB) phosphorylation were upregulated in the affected spinal cord dorsal horn (SCDH) of HP model rats, accompanied by K+-Cl-- Cotransporter-2 (KCC2) expression was down-regulated. EA significantly increased the mechanical pain threshold in HP model male rats and decreased BDNF and p-TrkB overexpression and upregulated KCC2 expression. Blockade of BDNF with BDNF neutralizing antibody attenuated mechanical abnormal pain in HP rats. Finally, administration of exogenous BDNF by pharmacological methods reversed the EA-induced resistance to abnormal pain. In all, these results suggest that BDNF-TrkB contributes to mechanical abnormal pain in HP model rats and that EA ameliorates mechanical abnormal pain through upregulation of KCC2 by BDNF-TrkB in SCDH. Our study further supports EA as an effective treatment to prevent the transition of acute pain into chronic pain.
Collapse
Affiliation(s)
- Mengting Shi
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jie Zhou
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Acupuncture and Moxibustion, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Rong Hu
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Haipeng Xu
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yi Chen
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xingying Wu
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Bowen Chen
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Ruijie Ma
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Acupuncture and Moxibustion, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Chen L, Wang H, Xing J, Shi X, Huang H, Huang J, Xu C. Silencing P2X7R Alleviates Diabetic Neuropathic Pain Involving TRPV1 via PKCε/P38MAPK/NF-κB Signaling Pathway in Rats. Int J Mol Sci 2022; 23:ijms232214141. [PMID: 36430617 PMCID: PMC9696864 DOI: 10.3390/ijms232214141] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 11/19/2022] Open
Abstract
Transient receptor potential vanillic acid 1 (TRPV1) is an ion channel activated by heat and inflammatory factors involved in the development of various types of pain. The P2X7 receptor is in the P2X family and is associated with pain mediated by satellite glial cells. There might be some connection between the P2X7 receptor and TRPV1 in neuropathic pain in diabetic rats. A type 2 diabetic neuropathic pain rat model was induced using high glucose and high-fat diet for 4 weeks and low-dose streptozocin (35 mg/kg) intraperitoneal injection to destroy islet B cells. Male Sprague Dawley rats were administrated by intrathecal injection of P2X7 shRNA and p38 inhibitor, and we recorded abnormal mechanical and thermal pain and nociceptive hyperalgesia. One week later, the dorsal root ganglia from the L4-L6 segment of the spinal cord were harvested for subsequent experiments. We measured pro-inflammatory cytokines, examined the relationship between TRPV1 on neurons and P2X7 receptor on satellite glial cells by measuring protein and transcription levels of P2X7 receptor and TRPV1, and measured protein expression in the PKCε/P38 MAPK/NF-κB signaling pathway after intrathecal injection. P2X7 shRNA and p38 inhibitor relieved hyperalgesia in diabetic neuropathic pain rats and modulated inflammatory factors in vivo. P2X7 shRNA and P38 inhibitors significantly reduced TRPV1 expression by downregulating the PKCε/P38 MAPK/NF-κB signaling pathway and inflammatory factors in dorsal root ganglia. Intrathecal injection of P2X7 shRNA alleviates nociceptive reactions in rats with diabetic neuropathic pain involving TRPV1 via PKCε/P38 MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lisha Chen
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | - Hongji Wang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | - Juping Xing
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | - Xiangchao Shi
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | - Huan Huang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | - Jiabao Huang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
| | - Changshui Xu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, China
- Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang 330006, China
- The Clinical Medical School, Jiangxi Medical College, Shangrao 334000, China
- The First Affiliated Hospital, Jiangxi Medical College, Shangrao 334000, China
- Correspondence: ; Tel.: +86-791-86360556
| |
Collapse
|
9
|
Li W, Liu J, Chen A, Dai D, Zhao T, Liu Q, Song J, Xiong L, Gao XF. Shared nociceptive dorsal root ganglion neurons participating in acupoint sensitization. Front Mol Neurosci 2022; 15:974007. [PMID: 36106140 PMCID: PMC9465389 DOI: 10.3389/fnmol.2022.974007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
When the body is under pathological stress (injury or disease), the status of associated acupoints changes, including decreased pain threshold. Such changes in acupoint from a “silent” to an “active” state are considered “acupoint sensitization,” which has become an important indicator of acupoint selection. However, the mechanism of acupoint sensitization remains unclear. In this study, by retrograde tracing, morphological, chemogenetic, and behavioral methods, we found there are some dorsal root ganglion (DRG) neurons innervating the ST36 acupoint and ipsilateral hind paw (IHP) plantar simultaneously. Inhibition of these shared neurons induced analgesia in the complete Freund’s adjuvant (CFA) pain model and obstruction of nociceptive sensation in normal mice, and elevated the mechanical pain threshold (MPT) of ST36 acupoint in the CFA model. Excitation of shared neurons induced pain and declined the MPT of ST36 acupoint. Furthermore, most of the shared DRG neurons express TRPV1, a marker of nociceptive neurons. These results indicate that the shared nociceptive DRG neurons participate in ST36 acupoint sensitization in CFA-induced chronic pain. This raised a neural mechanism of acupoint sensitization at the level of primary sensory transmission.
Collapse
Affiliation(s)
- Wanrong Li
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Jia Liu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Aiwen Chen
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Danqing Dai
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Tiantian Zhao
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Qiong Liu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Jianren Song
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lize Xiong
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
- *Correspondence: Lize Xiong,
| | - Xiao-Fei Gao
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
- Xiao-Fei Gao,
| |
Collapse
|