51
|
Hanani M. How Do Peripheral Neurons and Glial Cells Participate in Pain Alleviation by Physical Activity? Cells 2025; 14:462. [PMID: 40136711 PMCID: PMC11941599 DOI: 10.3390/cells14060462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/21/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Chronic pain is a global health problem with major socioeconomic implications. Drug therapy for chronic pain is limited, prompting search for non-pharmacological treatments. One such approach is physical exercise, which has been found to be beneficial for numerous health issues. Research in recent years has yielded considerable evidence for the analgesic actions of exercise in humans and experimental animals, but the underlying mechanisms are far from clear. It was proposed that exercise influences the pain pathways by interacting with the immune system, mainly by reducing inflammatory responses, but the release of endogenous analgesic mediators is another possibility. Exercise acts on neurons and glial cells in both the central and peripheral nervous systems. This review focuses on the periphery, with emphasis on possible glia-neuron interactions. Key topics include interactions of Schwann cells with axons (myelinated and unmyelinated), satellite glial cells in sensory ganglia, enteric glial cells, and the sympathetic nervous system. An attempt is made to highlight several neurological diseases that are associated with pain and the roles that glial cells may play in exercise-induced pain alleviation. Among the diseases are fibromyalgia and Charcot-Marie-Tooth disease. The hypothesis that active skeletal muscles exert their effects on the nervous system by releasing myokines is discussed.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem 91240, Israel;
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
52
|
Li T, Zhou W, Ke J, Chen M, Wang Z, Hayashi L, Su X, Jia W, Huang W, Wang CS, Bengyella K, Yang Y, Hernandez R, Zhang Y, Song X, Xu T, Huang T, Liu Y. A pontine center in descending pain control. Neuron 2025:S0896-6273(25)00171-0. [PMID: 40132590 DOI: 10.1016/j.neuron.2025.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/17/2024] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
Pain sensation changes according to expectation, context, and mood, illustrating how top-down circuits affect somatosensory processing. Here, we used an intersectional strategy to identify anatomical and molecular-spatial features of supraspinal descending neurons activated by distinct noxious stimulation. This approach captured known descending pain pathways as well as spinal projecting neurons that are anatomically mapped to Barrington's nucleus in the dorsal pontine tegmentum. We determined that this population of neurons expresses corticotropin-releasing hormone in Barrington's nucleus (BarCrh) and exhibits time-locked firing in response to noxious stimulation. Chemogenetic activation of BarCrh neurons attenuated nocifensive responses as well as tactile neuropathic pain, while silencing these neurons resulted in thermal hyperalgesia and mechanical allodynia. Mechanistically, we demonstrated that pain-related input from the ventrolateral periaqueductal gray recruits BarCrh neurons, reduces ascending nociceptive transmission, and preferentially activates spinal dynorphin neurons to mediate analgesia. Our data expose a pontine inhibitory descending pathway that powerfully controls nocifensive sensory input to the brain.
Collapse
Affiliation(s)
- Tianming Li
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Wenjie Zhou
- Department of Cardiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.
| | - Jin Ke
- Shenzhen Key Laboratory of Neuropsychiatric Modulations, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, P.R. China
| | - Matthew Chen
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Zhen Wang
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Lauren Hayashi
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Xiaojing Su
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Wenbin Jia
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Wenxi Huang
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Chien-Sheng Wang
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Kapsa Bengyella
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Yang Yang
- Department of Neurology, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, P.R. China
| | - Rafael Hernandez
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Yan Zhang
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Xinglei Song
- Department of Anatomy and Physiology, Shanghai Jiao Tong University, School of Medicine, Shanghai, P.R. China
| | - Tianle Xu
- Department of Cardiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Tianwen Huang
- Shenzhen Key Laboratory of Neuropsychiatric Modulations, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, P.R. China.
| | - Yuanyuan Liu
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA.
| |
Collapse
|
53
|
Lan Q, Ouyang A, Chen Y, Li Y, Zhong B, Deng S. Pain, lactate, and anesthetics: intertwined regulators of tumor metabolism and immunity. Front Oncol 2025; 15:1534300. [PMID: 40165895 PMCID: PMC11955471 DOI: 10.3389/fonc.2025.1534300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Patients with advanced cancer frequently endure severe pain, which substantially diminishes their quality of life and can adversely impact survival. Analgesia, a critical modality for alleviating such pain, is now under scrutiny for its potential role in cancer progression, a relationship whose underlying mechanisms remain obscure. Emerging evidence suggests that lactate, once considered a metabolic byproduct, actively participates in the malignant progression of cancer by modulating both metabolic and immunological pathways within the tumor microenvironment. Furthermore, lactate is implicated in the modulation of cancer-related pain, exerting effects through direct and indirect mechanisms. This review synthesizes current understanding of lactate's production, transport, and functional roles in tumor cells, encompassing the regulation of tumor metabolism, immunity, and progression. Additionally, we dissect the complex, bidirectional relationship between lactate and pain, and assess the impact of anesthetics on pain relief, lactate homeostasis, and tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Simin Deng
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
54
|
Li X, Yang H, Qian M, Liu H, Zuo S, Liu JC, Ge WH, Zhou L. Intracellular metabotropic glutamate receptor 5 in spinal dorsal horn neurons contributes to pain in a mouse model of vincristine-induced neuropathic pain. Neurosci Lett 2025; 852:138193. [PMID: 40074023 DOI: 10.1016/j.neulet.2025.138193] [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: 11/05/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Vincristine (VCR) is a commonly used clinical anti-cancer drug, but it can also induce neurotoxicity and cause vincristine-induced neuropathic pain (VINP). The metabotropic glutamate receptor 5 (mGluR5) within spinal dorsal horn neurons regulates the transmission of pain mediated by glutamate. In this study, we investigated for the first time the role of mGluR5 in the transmission of noxious information in VINP. Expression of mGluR5 protein was significantly increased in the spinal cord from days 6 to 14 after VCR injection. Immunofluorescence double staining showed that mGluR5 colocalized with the neuron-specific marker NeuN. The intrathecal administration of MPEP (a specific antagonist of mGluR5) or DHPG (an agonist of mGluR5) influenced the pain threshold and mGluR5 protein expression in VINP mice. The expression of c-Fos protein was also affected by MPEP. Furthermore, simulated blockade of intracellular mGluR5 site by intrathecal injection of small interfering RNA (siRNA) of the excitatory amino acid transporter 3 (EAAT3) reduced mechanical allodynia and thermal hyperalgesia and suppressed the expression of mGluR5 and c-Fos proteins. The results collectively indicate that mGluR5 site in spinal dorsal horn neurons may be involved in the regulation of intracellular nociceptive signal transmission in VINP, and the expression of c-Fos largely depends on the intracellular mGluR5.
Collapse
Affiliation(s)
- Xiao Li
- Affiliated Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008 Jiangsu, China
| | - Hui Yang
- Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008 Jiangsu, China
| | - Ming Qian
- Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008 Jiangsu, China
| | - Hang Liu
- Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008 Jiangsu, China
| | - Shuang Zuo
- Affiliated Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008 Jiangsu, China
| | - Jin-Chun Liu
- Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008 Jiangsu, China.
| | - Wei-Hong Ge
- Affiliated Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008 Jiangsu, China; Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008 Jiangsu, China.
| | - Lin Zhou
- Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008 Jiangsu, China.
| |
Collapse
|
55
|
Chen TY, Yoshioka T, Hsu WL. NO Pain! No Cancer? The Crosstalk Between Nociception, ROS, and Cancer Development. FRONT BIOSCI-LANDMRK 2025; 30:31328. [PMID: 40152391 DOI: 10.31083/fbl31328] [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: 11/22/2024] [Revised: 01/02/2025] [Accepted: 01/31/2025] [Indexed: 03/29/2025]
Abstract
Transient receptor potential (TRP) channels, particularly those involved in nociception (nociceptive TRP channels), are implicated in both pain and cancer development. Activation of these channels by diverse stimuli triggers calcium influx, leading to mitochondrial oxidative stress and reactive oxygen species (ROS) accumulation. This ROS production contributes to both nociceptive signaling (causing pain) and aging processes, including genomic instability, a key driver of carcinogenesis. Although a direct causal link between pain and cancer onset remains elusive, the shared involvement of nociceptive TRP channels strongly suggests a correlation. This opinion article proposes targeting the crosstalk between nociceptive TRP channels and ROS as a promising therapeutic strategy to mitigate cancer and cancer-associated pain simultaneously. While further research is needed to definitively establish a causal relationship between pain and cancer risk, the available evidence suggests that inhibiting this pathway may offer significant benefits for both cancer prevention and treatment.
Collapse
Affiliation(s)
- Tzu-Yin Chen
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, 632007 Yunlin, Taiwan
| | - Tohru Yoshioka
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708 Kaohsiung, Taiwan
| | - Wen-Li Hsu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, 632007 Yunlin, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708 Kaohsiung, Taiwan
| |
Collapse
|
56
|
Kschonek J, Twele L, Deters K, Miller M, Reinmold J, Emmerich I, Hennig-Pauka I, Kemper N, Kreienbrock L, Wendt M, Kästner S, Grosse Beilage E. Part I: understanding pain in pigs-basic knowledge about pain assessment, measures and therapy. Porcine Health Manag 2025; 11:12. [PMID: 40069905 PMCID: PMC11895375 DOI: 10.1186/s40813-025-00421-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Pigs can suffer from pain due to spontaneously occurring diseases, wounds, injuries, trauma, and physiological conditions such as the farrowing process; however, this pain is often neglected. To increase knowledge and awareness about this phenomenon, the current article presents a scoping review of basic and new approaches for identifying, evaluating, and treating pain in pigs. METHODS A scoping review was conducted with results from a search of the electronic database VetSearch and CABI. With regard to eligibility criteria, 49 out of 725 publications between 2015 and the end of March 2023 were included. The findings are narratively synthesized and reported orienting on the PRISMA ScR guideline. RESULTS The results of this review showed that practitioners need to consider pain not only as a sign of a disease but also as a critical aspect of welfare. If both the symptoms of pain and the underlying reasons remain unassessed, the longevity and prosperity of pigs may be at risk. In this respect, veterinarians are obliged to know about intricacies of pain and pain mechanisms and to provide adequate treatment for their patients. CONCLUSION It is pivotal to increase knowledge about pain mechanisms, the reasons for heterogeneity in behavioural signs of pain, and methods for evaluating whether a pig is experiencing pain. This article will help practitioners update their knowledge of this topic and discuss the implications for everyday practice.
Collapse
Affiliation(s)
- Julia Kschonek
- Institute for Biometry, Epidemiology and Information Processing (IBEI), University of Veterinary Medicine, Foundation, Hannover, Bünteweg 2, 30559, Hannover, Germany.
| | - Lara Twele
- Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Bünteweg 9, 30559, Hannover, Germany
| | - Kathrin Deters
- Field Station for Epidemiology, University of Veterinary Medicine, Foundation, Hannover, Büscheler Str. 9, 49456, Bakum, Germany
| | - Moana Miller
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, University of Veterinary Medicine, Foundation, Hannover, Bischofsholer Damm 15, 30173, Hannover, Germany
| | - Jennifer Reinmold
- Field Station for Epidemiology, University of Veterinary Medicine, Foundation, Hannover, Büscheler Str. 9, 49456, Bakum, Germany
| | - Ilka Emmerich
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University Leipzig, An den Tierkliniken 39, 04103, Leipzig, Germany
| | - Isabel Hennig-Pauka
- Field Station for Epidemiology, University of Veterinary Medicine, Foundation, Hannover, Büscheler Str. 9, 49456, Bakum, Germany
| | - Nicole Kemper
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, University of Veterinary Medicine, Foundation, Hannover, Bischofsholer Damm 15, 30173, Hannover, Germany
| | - Lothar Kreienbrock
- Institute for Biometry, Epidemiology and Information Processing (IBEI), University of Veterinary Medicine, Foundation, Hannover, Bünteweg 2, 30559, Hannover, Germany
| | - Michael Wendt
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine, Foundation, Hannover, Bischofsholer Damm 15, 30173, Hannover, Germany
| | - Sabine Kästner
- Clinic for Small Animals, University of Veterinary Medicine, Foundation, Hannover, Bünteweg 2, 30559, Hannover, Germany
| | - Elisabeth Grosse Beilage
- Field Station for Epidemiology, University of Veterinary Medicine, Foundation, Hannover, Büscheler Str. 9, 49456, Bakum, Germany
| |
Collapse
|
57
|
Yagi H, Takao K, Hattori S, Minato Y, Kuwahara-Otani S, Maeda S, Noguchi K, Miyakawa T, Sato M. Deletion of filamin A-interacting protein (FILIP) results in a weak grip strength and abnormal responses to nociceptive stimulation. Neurosci Lett 2025; 851:138158. [PMID: 39961470 DOI: 10.1016/j.neulet.2025.138158] [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: 08/05/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
Filamin A-interacting protein (FILIP in mice, FILIP1 in humans) was first identified as a protein that negatively controls neuronal migration in rodents, and was subsequently demonstrated to be pivotal for the development of the neocortex. In the previous study, we generated FILIP knockout mice to investigate the in vivo functions of FILIP in cortical development. Since FILIP mRNA is widely expressed in the body, we systematically examined FILIP-knockout mice to determine the functions of FILIP throughout the body. Our results showed that FILIP-knockout mice exhibited weak grip strength and sensory abnormalities. Interestingly, we also found that FILIP was expressed in a subset of neurons in the dorsal root ganglion (DRG). Recent research has reported that FILIP1 mutations lead to severe neurological and musculoskeletal abnormalities, resulting in the proposal of a new disease entity, termed FILIP1opathy. It is expected that our FILIP-knockout mice could be used as a model for the pathological investigation of FILIP1opathy.
Collapse
Affiliation(s)
- Hideshi Yagi
- Department of Anatomy and Cell Biology, Hyogo Medical University, Hyogo, Japan; Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| | - Keizo Takao
- Frontier Technology Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Life Science Research Center, University of Toyama, Toyama, Japan
| | - Satoko Hattori
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Aichi, Japan; Research Creation Support Center, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yusuke Minato
- Department of Anatomy and Cell Biology, Hyogo Medical University, Hyogo, Japan
| | | | - Seishi Maeda
- Department of Anatomy and Cell Biology, Hyogo Medical University, Hyogo, Japan
| | - Koichi Noguchi
- Department of Anatomy and Neuroscience, Hyogo Medical University, Hyogo, Japan
| | - Tsuyoshi Miyakawa
- Frontier Technology Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Aichi, Japan
| | - Makoto Sato
- Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Research Center for Child Mental Development, University of Fukui, Fukui, Japan; United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui (UGSCD), Osaka, Japan; Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
58
|
Costa CJ, Prescott S, Fourie NH, Abey SK, Sherwin LB, Rahim-Williams B, Joseph PV, Posada-Quintero H, Hoffman RK, Henderson WA. Host Transcriptome and Microbial Variation in Relation to Visceral Hyperalgesia. Nutrients 2025; 17:921. [PMID: 40077792 PMCID: PMC11902232 DOI: 10.3390/nu17050921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Chronic visceral hypersensitivity is associated with an overstressed pain response to noxious stimuli (hyperalgesia). Microbiota are active modulators of host biology and are implicated in the etiology of visceral hypersensitivity. OBJECTIVES we studied the association between the circulating mRNA transcriptome, the intensity of induced visceral pain (IVP), and variation in the oral microbiome among participants with and without baseline visceral hypersensitivity. METHODS Transcriptomic profiles and microbial abundance were correlated with IVP intensity. Host mRNA and microbes associated with IVP were explored, linking variation in the microbiome to host RNA biology. RESULTS 259 OTUs were found to be associated with IVP through correlation to differential expression of 471 genes in molecular pathways related to inflammation and neural mechanisms, including Rho and PI3K/AKT pathways. The bacterial families Lachnospiraceae, Prevotellaceae, and Veillonellaceae showed the highest degree of association. Oral microbial profiles with reduced diversity were characteristic of participants with visceral hypersensitivity. CONCLUSIONS Our results suggest that the oral microbiome may be involved in systemic immune and inflammatory effects and play a role in nervous system and stem cell pathways. The interactions between visceral hypersensitivity, differentially expressed molecular pathways, and microbiota described here provide a framework for further work exploring the relationship between host and microbiome.
Collapse
Affiliation(s)
- Christopher J. Costa
- Department of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT 06030, USA;
| | - Stephanie Prescott
- Inova Health Services, L.J. Murphy Children’s Hospital, 3300 Gallows Rd, Falls Church, VA 22042, USA;
| | - Nicolaas H. Fourie
- National Institute of Nursing Research, National Institutes of Health, 31 Center Drive, Bethesda, MD 20892, USA
| | - Sarah K. Abey
- Laboratory of Neuroimaging, National Institute of Alcohol Abuse and Alcoholism, 10 Center Drive, Bethesda, MD 20814, USA
| | - LeeAnne B. Sherwin
- Sinclair School of Nursing, University of Missouri System, 915 Hitt Street, Columbia, MO 65203, USA;
| | - Bridgett Rahim-Williams
- Office of Research and Sponsored Programs, University of North Florida, 1 UNF Drive, Jacksonville, FL 32224, USA;
| | - Paule V. Joseph
- National Institute on Alcohol Abuse and Alcoholism, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 10 Center Drive, Bethesda, MD 20814, USA;
| | - Hugo Posada-Quintero
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Storrs, CT 06269, USA;
| | - Rebecca K. Hoffman
- Laboratory of Innovative and Translational Nursing Research, School of Nursing, University of PA, 418 Curie Blvd, Philadelphia, PA 19104, USA;
| | - Wendy A. Henderson
- Department of Biobehavioral Health Sciences, School of Nursing, University of PA, 418 Curie Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
59
|
Antal M. Molecular Anatomy of Synaptic and Extrasynaptic Neurotransmission Between Nociceptive Primary Afferents and Spinal Dorsal Horn Neurons. Int J Mol Sci 2025; 26:2356. [PMID: 40076973 PMCID: PMC11900602 DOI: 10.3390/ijms26052356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Sensory signals generated by peripheral nociceptors are transmitted by peptidergic and nonpeptidergic nociceptive primary afferents to the superficial spinal dorsal horn, where their central axon terminals establish synaptic contacts with secondary sensory spinal neurons. In the case of suprathreshold activation, the axon terminals release glutamate into the synaptic cleft and stimulate postsynaptic spinal neurons by activating glutamate receptors located on the postsynaptic membrane. When overexcitation is evoked by peripheral inflammation, neuropathy or pruritogens, peptidergic nociceptive axon terminals may corelease various neuropeptides, neurotrophins and endomorphin, together with glutamate. However, in contrast to glutamate, neuropeptides, neurotrophins and endomorphin are released extrasynaptically. They diffuse from the site of release and modulate the function of spinal neurons via volume transmission, activating specific extrasynaptic receptors. Thus, the released neuropeptides, neurotrophins and endomorphin may evoke excitation, disinhibition or inhibition in various spinal neuronal populations, and together with glutamate, induce overall overexcitation, called central sensitization. In addition, the synaptic and extrasynaptic release of neurotransmitters is subjected to strong retrograde control mediated by various retrogradely acting transmitters, messengers, and their presynaptic receptors. Moreover, the composition of this complex chemical apparatus is heavily dependent on the actual patterns of nociceptive primary afferent activation in the periphery. This review provides an overview of the complexity of this signaling apparatus, how nociceptive primary afferents can activate secondary sensory spinal neurons via synaptic and volume transmission in the superficial spinal dorsal horn, and how these events can be controlled by presynaptic mechanisms.
Collapse
Affiliation(s)
- Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
60
|
Reid P, Scherer K, Halasz D, Simal AL, Tang J, Zaheer F, Tuling J, Levine G, Michaud J, Clark AL, Descalzi G. Astrocyte neuronal metabolic coupling in the anterior cingulate cortex of mice with inflammatory pain. Brain Behav Immun 2025; 125:212-225. [PMID: 39694343 DOI: 10.1016/j.bbi.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/14/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024] Open
Abstract
Chronic pain is a major global concern, with at least 1 in 5 people suffering from chronic pain worldwide. Mounting evidence indicates that neuroplasticity of the anterior cingulate cortex (ACC) is a critical step in the development of chronic pain. Previously, we found that chronic pain and fear learning are both associated with enhanced neuronal excitability and cause similar neuroplasticity-related gene expression changes in the ACC of male mice. However, neuroplasticity, imposes large metabolic demands. In the brain, neurons have the highest energy needs and interact with astrocytes, which extract glucose from blood, mobilize glycogen, and release lactate in response to neuronal activity. Here, we use chronic and continuous inflammatory pain models in female and male mice to investigate the involvement of astrocyte-neuronal lactate shuttling (ANLS) in the ACC of female and male mice experiencing inflammatory pain. We found that ANLS in the mouse ACC promotes the development of chronic inflammatory pain, and expresses sex specific patterns of activation. Specifically, whereas both male and female mice show similar levels of chronic pain hypersensitivity, only male mice show sustained increases in lactate levels. Accordingly, chronic pain alters the expression levels of proteins involved in lactate metabolism and shuttling in a sexually dimorphic manner. We found that disrupting astrocyte-neuronal lactate shuttling in the ACC prior to inflammatory injury prevents the development of pain hypersensitivity in female and male mice, but only reduces temporary pain in male mice. Furthermore, using a transgenic mouse model (itga1-null mice) that displays a naturally occurring form of spontaneous osteoarthritis (OA), a painful inflammatory pain condition, we found that whereas both female and male mice develop OA, only male mice show increases in mechanisms involved in astrocyte-neuronal lactate shuttling. Our findings thus indicate that there are sex differences in astrocyte-neuronal metabolic coupling in the mouse ACC during chronic pain development.
Collapse
Affiliation(s)
- Paige Reid
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - Kaitlin Scherer
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - Danielle Halasz
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - Ana Leticia Simal
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - James Tang
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - Fariya Zaheer
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - Jaime Tuling
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - Gabriel Levine
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada
| | - Jana Michaud
- Department of Human Health & Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, ON, Canada
| | - Andrea L Clark
- Department of Human Health & Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, ON, Canada
| | - Giannina Descalzi
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph ON, Canada.
| |
Collapse
|
61
|
Meerschaert KA, Chiu IM. The gut-brain axis and pain signalling mechanisms in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2025; 22:206-221. [PMID: 39578592 DOI: 10.1038/s41575-024-01017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Visceral pain is a major clinical problem and one of the most common reasons patients with gastrointestinal disorders seek medical help. Peripheral sensory neurons that innervate the gut can detect noxious stimuli and send signals to the central nervous system that are perceived as pain. There is a bidirectional communication network between the gastrointestinal tract and the nervous system that mediates pain through the gut-brain axis. Sensory neurons detect mechanical and chemical stimuli within the intestinal tissues, and receive signals from immune cells, epithelial cells and the gut microbiota, which results in peripheral sensitization and visceral pain. This Review focuses on molecular communication between these non-neuronal cell types and neurons in visceral pain. These bidirectional interactions can be dysregulated during gastrointestinal diseases to exacerbate visceral pain. We outline the anatomical pathways involved in pain processing in the gut and how cell-cell communication is integrated into this gut-brain axis. Understanding how bidirectional communication between the gut and nervous system is altered during disease could provide new therapeutic targets for treating visceral pain.
Collapse
Affiliation(s)
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
62
|
Fatima SA, Akhtar B, Sharif A, Khan MI, Shahid M, Anjum F, Hussain F, Mobashar A, Ashraf M. Implications of nociceptor receptors and immune modulation: emerging therapeutic targets for autoimmune diseases. Inflammopharmacology 2025; 33:959-977. [PMID: 39955696 DOI: 10.1007/s10787-025-01653-w] [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: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 02/17/2025]
Abstract
Chronic painful autoimmune disorders such as multiple sclerosis (MS), inflammatory bowel disease (IBD), and rheumatoid arthritis (RA) induce significant discomfort. They are defined by persistent inflammation and immune-mediated tissue injury. The activation and sensitisation of nociceptors, mutated in various disorders, are fundamental components contributing to the pain experienced in these conditions. Recent discoveries indicate that immunological mediators and nociceptive receptors interact functionally within peripheral and central sensitisation pathways, amplifying chronic pain. This research examines the involvement of nociceptors in rheumatoid arthritis, multiple sclerosis, and inflammatory bowel disease. It explores how immune cells and pro-inflammatory cytokines induce, sensitise and regulate various nociceptive receptors (P2X, TRPA1 and TRPV1). Finally, we address possible future directions with respect to the treatment of long-lasting effects on immunity, and what new drug targets could be pursued as well, in order to counteract such either neuro-immune interactions in conditions involving the immunological system. By studying nociceptive mechanisms across autoimmune illnesses, we want to identify shared pathways and activation of nociceptors specific to individual diseases. This will shed insight on potential therapies for managing pain associated with autoimmune diseases.
Collapse
Affiliation(s)
- Syeda Asloob Fatima
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Bushra Akhtar
- Department of Pharmacy, Faculty of Health and Pharmaceutical Sciences, University of Agriculture, Faisalabad, Pakistan.
| | - Ali Sharif
- Department of Pharmacology, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore, Pakistan
| | - Muhammad Imran Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Pakistan
| | - Muhammad Shahid
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Fozia Anjum
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | - Fatma Hussain
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Aisha Mobashar
- Department of Pharmacology, Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Maham Ashraf
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, The University of Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
63
|
Abagnale S, Panico F, Sagliano L, Gosseries O, Trojano L. Pleasant touch: Behavioural and hemodynamic responses to a protocol for systematic assessment of tactile stimulation. Cortex 2025; 184:236-249. [PMID: 39908706 DOI: 10.1016/j.cortex.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025]
Abstract
Pleasant touch is a form of tactile stimulation mediated by tactile C afferent fibres. It involves the encoding of the emotional value associated with tactile stimulation and subserves important social functions. Although pleasant touch has gathered increased interest in recent years, no protocol has been proposed to assess it with a robust and reliable method. In the present study we adopted a rigorous protocol for evaluating the pleasantness or unpleasantness of 9 tactile (pleasant, unpleasant, or neutral) stimuli delivered on eight body areas in healthy individuals. We recorded participants' ratings on pleasantness and intensity of the stimulus, as well as their activity in the prefrontal cortex (PFC) by functional near-infrared spectroscopy (fNIRS). A questionnaire evaluated participants' subjective experience of touch in everyday life. The behavioural results confirmed the effectiveness of the protocol as the stimuli selected to evoke pleasantness were perceived as significantly more pleasant than unpleasant and neutral ones, whereas unpleasant stimuli were perceived as more intense than all other stimuli. The participants reported the palm of the hand, particularly the left one, as the most sensitive area to tactile stimulation. Judgements of pleasantness were positively correlated with subjective experience of touch in everyday life. fNIRS data showed increased activity in the prefrontal cortex particularly during stimulation with pleasant and unpleasant stimuli, consistent with behavioural findings. Overall, this study contributes to understand the processing of pleasant touch and its neural correlates, while introducing a rigorous protocol for investigating tactile stimulation. This protocol holds promise for future utilisation in both healthy and clinical populations.
Collapse
Affiliation(s)
- Simona Abagnale
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy; Fondazione Don Carlo Gnocchi ONLUS, Scientific Institute for Research and Health Care, Italy.
| | - Francesco Panico
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Laura Sagliano
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Olivia Gosseries
- Coma Science Group, GIGA-Consciousness, University of Liege, Belgium
| | - Luigi Trojano
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| |
Collapse
|
64
|
Das V, Lam AL, Smith MT. The angiotensin II type 2 receptor antagonists, PD123,319 ((S-( +)-1-[(4-(dimethylamino)-3-methylphenyl)methyl]-5-(diphenylacetyl)-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridine-6-carboxylic acid), EMA300 (5-(2,2-diphenylacetyl)-4-[(4-methoxy-3-methylphenyl)methyl]-1,4,6,7-tetrahydroimidazo[4,5-c]pyridine-6-carboxylic acid) and EMA401 ((3S)-5-(benzyloxy)-2-(2,2-diphenylacetyl)-6-methoxy-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid), evoke pain relief in a varicella zoster virus-induced rat model of neuropathic pain. Inflammopharmacology 2025; 33:1337-1348. [PMID: 39976667 PMCID: PMC11913958 DOI: 10.1007/s10787-025-01650-z] [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: 09/20/2024] [Accepted: 01/07/2025] [Indexed: 03/19/2025]
Abstract
Post-herpetic neuralgia (PHN) is a type of neuropathic (nerve) pain that persists for more than 3 months after crusting of the last shingles lesion. It is difficult to relieve with analgesic/adjuvant medications, and so novel analgesics are needed. Our aim was to use a rat model of varicella zoster virus (VZV)-induced neuropathic pain to assess the pain relief efficacy of several small molecule angiotensin II type 2 (AT2) receptor antagonists (PD123,319, EMA300, and EMA401) relative to clinically used analgesic/adjuvant agents from four different pharmacological classes. Male Wistar rats received a unilateral intraplantar injection of VZV-infected MRC-5 cells (2 × 104 infected cells) and paw withdrawal thresholds (PWTs) in the ipsilateral hindpaws were assessed using von Frey filaments. Animals with PWTs ≤ 8 g received single doses of PD123,319 (0.03-3 mg/kg), EMA300 (0.3-5 mg/kg), EMA401 (0.03-1 mg/kg), gabapentin (10-60 mg/kg), amitriptyline (5-30 mg/kg), morphine (0.1-3 mg/kg), meloxicam (5-20 mg/kg) or vehicle and PWT versus time curves were generated. Single doses of PD123,319, EMA300, EMA401, gabapentin and morphine-evoked dose-dependent anti-allodynia in the hindpaws of VZV-rats. The mean (95% confidence intervals) ED50s were 0.57 (0.04-1.7), 2.5 (1.0-3.7) and 0.41 (0.12-0.87) mg/kg for PD123,319, EMA300, and EMA401, respectively. The ED50s for gabapentin and morphine were 39.9 (25.1-64.8) and 0.04 (0.16-2.09) mg/kg, respectively. In conclusion, the anti-allodynic efficacy of EMA401 in a VZV-rat model of neuropathic pain is aligned with its analgesic efficacy in a Phase 2a clinical trial in patients with PHN. This model has utility for anti-allodynic efficacy assessment of novel AT2 receptor antagonists from drug discovery.
Collapse
Affiliation(s)
- V Das
- Centre for Integrated Preclinical Drug Development (CIPDD), School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - A L Lam
- Centre for Integrated Preclinical Drug Development (CIPDD), School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - M T Smith
- Centre for Integrated Preclinical Drug Development (CIPDD), School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
65
|
Tong SH, Liu DL, Liao P, Zhang SY, Zhou J, Zong Y, Zhang CQ, Huang YG, Gao JJ. Emerging role of macrophages in neuropathic pain. J Orthop Translat 2025; 51:227-241. [PMID: 40177638 PMCID: PMC11964759 DOI: 10.1016/j.jot.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 04/05/2025] Open
Abstract
Neuropathic pain is a complex syndrome caused by injury to the neurons, which causes persistent hypersensitivity and considerable inconvenience to the patient's whole life. Over the past two decades, the interaction between immune cells and neurons has been proven to play a crucial role in the development of neuropathic pain. Increasing studies have indicated the important role of macrophages for neuroinflammation and have shed light on the underlying molecular and cellular mechanisms. In addition, novel therapeutic methods targeting macrophages are springing up, which provide more options in our clinical treatment. Herein, we reviewed the characteristics of peripheral macrophages and their function in neuropathic pain, with the aim of better understanding how these cells contribute to pathological processes and paving the way for therapeutic approaches. Translational potential statement This review provides a comprehensive overview of the mechanisms underlying the interplay between the macrophages and nervous system during the progression of nerve injury. Additionally, it compiles existing intervention strategies targeting macrophages for the treatment of neuropathic pain. This information offers valuable insights for researchers seeking to address the challenge of this intractable pain.
Collapse
Affiliation(s)
- Si-Han Tong
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - De-Lin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Sen-Yao Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jian Zhou
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia, 6009, Australia
| | - Chang-Qing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yi-Gang Huang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jun-Jie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
66
|
Krishnamoorthy G, Narayana A, Balakrishnan D. The gut-masticatory muscles-temporomandibular joint pain axis-a scoping review. J Oral Facial Pain Headache 2025; 39:24-33. [PMID: 40129421 PMCID: PMC11934742 DOI: 10.22514/jofph.2025.021] [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: 10/04/2024] [Accepted: 11/29/2024] [Indexed: 03/26/2025]
Abstract
Orofacial pain has become the most common debilitating disease resulting in high healthcare costs, and compromising the quality of life, speech, aesthetics and masticatory function of those affected. As its aetiology is multifactorial and as the treatment involves a multidisciplinary holistic approach, arriving at a confirmative diagnosis is challenging. Numerous studies have been published that support the bidirectional link between gut health and other organs like the cardiovascular system, respiratory system, neurological and hormonal. Recent studies indicate a potential link between gut microbiota dysbiosis and chronic orofacial and temporomandibular joint (TMJ) pain. In this review, we enumerate the link between the metabolites released by the gut bacteria and how they regulate the pain mechanism of various types of orofacial pain like chronic, neuropathic and inflammatory in the orofacial and TMJ regions. We also discuss the potential link between pain and gender predisposition. Further, we review the recent non-invasive therapeutic options which can be put forth to use for treating orofacial and TMJ pain.
Collapse
Affiliation(s)
- Gayathri Krishnamoorthy
- Department of Prosthodontics and
Crown Bridge, Manipal College of Dental
Sciences, Manipal Academy of Higher
Education, 576104 Manipal, India
| | - Aparna Narayana
- Department of Prosthodontics and
Crown Bridge, Manipal College of Dental
Sciences, Manipal Academy of Higher
Education, 576104 Manipal, India
| | - Dhanasekar Balakrishnan
- Department of Prosthodontics and
Crown Bridge, Manipal College of Dental
Sciences, Manipal Academy of Higher
Education, 576104 Manipal, India
| |
Collapse
|
67
|
Losgott T, Kudlacek O, Yang JW, Schicker KW, Boehm S, Salzer I. The paracetamol metabolite N-acetyl-4-benzoquinoneimine (NAPQI) prevents modulation of K V7 channels via G-protein coupled receptors by interference with PIP 2 and Ca 2+ sensitivity. Br J Pharmacol 2025; 182:1341-1357. [PMID: 39627952 DOI: 10.1111/bph.17419] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Paracetamol has been found to alleviate inflammatory pain by modulating KV7 channels. Its metabolite N-acetyl-4-benzoquinoneimine (NAPQI) increases currents through these channels via a stretch of three cysteine residues in the channel S2-S3 linker. Through this effect, the excitability of neurons in the pain pathway is dampened. Inflammatory mediators, in turn, enhance the excitability of sensory neurons by inhibiting KV7 channels. Here, a specific interaction between NAPQI and the so-called inflammatory soup was investigated. EXPERIMENTAL APPROACH Currents through KV7 channels were measured in sensory neurons and after heterologous expression in tsA201 cells. In addition, changes in cytosolic Ca2+ and in the distribution of PIP2 (PI(4,5)P2) between membrane and cytosol were determined by fluorescence microscopy. KEY RESULTS NAPQI abolished Ca2+-mediated inhibitory effects of an 'inflammatory soup' containing ADP, ATP, bradykinin, histamine, 5-hydroxytryptamine, prostaglandin E2, substance P and a PAR2 agonist on KV7 channel currents in sensory neurons. Moreover, the increase of KV7.2 channel currents by quenching of cytosolic Ca2+ as well as the current decrease by depletion of membrane PIP2 was impaired by NAPQI. These effects were lost in mutant channels lacking the three cysteines in the S2-S3 linker. CONCLUSION AND IMPLICATION NAPQI targets the three-cysteine motif in the S2-S3 linker of KV7.2 channels to counteract the signalling cascades employed by inflammatory mediators that inhibit these channels. In sensory neurons, this abolishes the closure of KV7 channels by the inflammatory soup. This mechanism is likely involved in the alleviation of inflammatory pain by paracetamol.
Collapse
Affiliation(s)
- Thomas Losgott
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus W Schicker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Boehm
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
68
|
Chu CC, Hu YH, Li GZ, Chen J, Zhang NN, Gu YX, Wu SY, Zhang HF, Xu YY, Guo HL, Tian X, Chen F. Unveiling the significance of AKAP79/150 in the nervous system disorders: An emerging opportunity for future therapies? Neurobiol Dis 2025; 206:106812. [PMID: 39864527 DOI: 10.1016/j.nbd.2025.106812] [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: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025] Open
Abstract
A-kinase anchoring protein 79/150 (AKAP79/150) is a crucial scaffolding protein that positions various proteins at specific synaptic sites to modulate excitatory synaptic intensity. As our understanding of AKAP79/150's biology deepens, along with its significant role in the pathophysiology of various human disorders, there is growing evidence that reveals new opportunities for therapeutic interventions. In this review, we examine the fundamental structure and primary functions of AKAP79/150, emphasizing its pathophysiological mechanisms in different nervous system disorders, particularly inflammatory pain, epilepsy, depression, and Alzheimer's disease. We also discuss its potential therapeutic implications for patients suffering from these conditions.
Collapse
Affiliation(s)
- Chen-Chao Chu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Hu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Gui-Zhou Li
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jiang Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ning-Ning Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yi-Xue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Shi-Yu Wu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Feng Zhang
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang-Yang Xu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hong-Li Guo
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
| | - Feng Chen
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
69
|
Wu J, Kuang W, Zhu Z, Dou J, Yao J, Cao J, Zhang F, Xu G. Upregulation of NR2B Subunits of NMDA Receptors in the Lateral Parabrachial Nucleus Contributes to Chronic Pancreatitis Pain. CNS Neurosci Ther 2025; 31:e70313. [PMID: 40022510 PMCID: PMC11871393 DOI: 10.1111/cns.70313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
AIMS Chronic pancreatitis (CP) is a localized or diffuse chronic progressive inflammation of the pancreas that can be caused by a variety of factors and is characterized by abdominal pain. However, the underlying mechanisms are poorly understood. Increasing evidence suggests that central sensitization plays a crucial role in the development of visceral pain, but the precise mechanisms of central neural processing remain unclear. METHODS CP was induced using repeated intraperitoneal injections of caerulein in mice. Neurospecific anterograde tracing was achieved using herpes simplex virus type 1 (HSV-1). Fiber photometry was used to assess neuronal activity. Optogenetic, chemogenetic, or pharmacological approaches were applied to manipulate the lateral parabrachial nucleus (LPB) glutamatergic neurons. The abdominal withdrawal threshold (AWT) was measured to evaluate the CP pain. A glutamate sensor was used to detect glutamate release in the LPB. RESULTS In the present study, we demonstrated that glutamatergic neurons in the LPB are activated in CP mice, leading to the development of CP pain. Notably, glutamatergic release is increased in the LPB, and the increased release primarily mediates CP pain by binding to the N-methyl-D-aspartate (NMDA) receptor rather than α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Specifically, this process involves the binding of the N-Methyl-D-Aspartate Receptor Subunit 2B (NR2B) in the LPB, leading to the development of CP pain. CONCLUSIONS This study identified the NR2B subunits of NMDA receptors in the LPB as playing a critical role in the regulation of CP pain.
Collapse
Affiliation(s)
- Jing‐Lai Wu
- Clinical Research Center of Neurological DiseaseThe Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow UniversitySuzhouChina
| | - Wen‐Qiong Kuang
- Clinical Research Center of Neurological DiseaseThe Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow UniversitySuzhouChina
| | - Zheng‐Yan Zhu
- Clinical Research Center of Neurological DiseaseThe Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow UniversitySuzhouChina
| | - Jing‐Heng Dou
- Clinical Research Center of Neurological DiseaseThe Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow UniversitySuzhouChina
| | - Jia‐He Yao
- Clinical Research Center of Neurological DiseaseThe Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow UniversitySuzhouChina
| | - Jing Cao
- Department of AnatomySchool of Basic Medical Sciences, Zhengzhou UniversityZhengzhouHenanChina
| | - Fu‐Chao Zhang
- Clinical Research Center of Neurological DiseaseThe Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow UniversitySuzhouChina
| | - Guang‐Yin Xu
- Clinical Research Center of Neurological DiseaseThe Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow UniversitySuzhouChina
| |
Collapse
|
70
|
Jayathilake NJ, Phan TT, Kim J, Lee KP, Park JM. Modulating neuroplasticity for chronic pain relief: noninvasive neuromodulation as a promising approach. Exp Mol Med 2025; 57:501-514. [PMID: 40025172 PMCID: PMC11958754 DOI: 10.1038/s12276-025-01409-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/20/2024] [Accepted: 12/01/2024] [Indexed: 03/04/2025] Open
Abstract
Chronic neuropathic pain is a debilitating neuroplastic disorder that notably impacts the quality of life of millions of people worldwide. This complex condition, encompassing various manifestations, such as sciatica, diabetic neuropathy and postherpetic neuralgia, arises from nerve damage or malfunctions in pain processing pathways and involves various biological, physiological and psychological processes. Maladaptive neuroplasticity, known as central sensitization, plays a critical role in the persistence of chronic neuropathic pain. Current treatments for neuropathic pain include pharmacological interventions (for example, antidepressants and anticonvulsants), invasive procedures (for example, deep brain stimulation) and physical therapies. However, these approaches often have limitations and potential side effects. In light of these challenges, interest in noninvasive neuromodulation techniques as alternatives or complementary treatments for neuropathic pain is increasing. These methods aim to induce analgesia while reversing maladaptive plastic changes, offering potential advantages over conventional pharmacological practices and invasive methods. Recent technological advancements have spurred the exploration of noninvasive neuromodulation therapies, such as repetitive transcranial magnetic stimulation, transcranial direct current stimulation and transcranial ultrasound stimulation, as well as innovative transformations of invasive techniques into noninvasive methods at both the preclinical and clinical levels. Here this review aims to critically examine the mechanisms of maladaptive neuroplasticity in chronic neuropathic pain and evaluate the efficacy of noninvasive neuromodulation techniques in pain relief. By focusing on optimizing these techniques, we can better assess their short-term and long-term effects, refine treatment variables and ultimately improve the quality of neuropathic pain management.
Collapse
Affiliation(s)
- Nishani Jayanika Jayathilake
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Tien Thuy Phan
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jeongsook Kim
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kyu Pil Lee
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| | - Joo Min Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea.
- Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
71
|
Tanghetti EA, Sierra R, Estes M, Eck A, Intintoli A, Hofvander H, Cohen JL, Friedmann DP, Goldman MP, Pomerantz H, Wang JV, Geronemus RG, Anderson RR, Sakamoto FH. Treatment of Acne With a 1726 nm Laser, Air Cooling, and Real-Time Temperature Monitoring, Software-Assisted Power Adjustment to Achieve a Temperature Endpoint With Selective Sebaceous Gland Photothermolysis. Lasers Surg Med 2025; 57:236-251. [PMID: 39780333 DOI: 10.1002/lsm.23872] [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: 12/06/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVES This work highlights the methods used to develop a multi-pulse 1726 nm laser system combined with bulk air-cooling for selective sebaceous gland (SG) photothermolysis using thermal imaging and software algorithms. This approach enables treating to a desired tissue temperature and depth to provide a safe, effective, reproducible, and durable treatment of acne. METHODS We designed and built a 1726 nm laser system with a 40 W maximum power output, a highly controlled air-cooling device, and a thermal camera in the handpiece, which permits real-time temperature monitoring of the epidermis. IRB-approved safety and efficacy trials demonstrated SG damage at depth, resulting in safe, efficacious, and durable clinical outcomes. Bioheat transfer and light transport modeling confirmed that the pulsing protocols could produce therapeutic temperatures at various SG depths, while protecting the epidermis and dermis with bulk air-cooling. Similarly, we employed clinical observations and photothermal modeling to identify pain mitigation opportunities while maintaining therapeutic efficacy. Biopsies were subsequently taken for histological evaluation. RESULTS Clinical and histological data, confirmed with modeling, demonstrated that multi-pulse laser delivery with bulk air-cooling selectively increased SG temperature compared to surrounding dermis and at depths unachievable by a single pulse. Subjects showed an average 71% ILC reduction at 3 months posttreatment. We identified two different pulsing protocols with similar selective photothermolysis (SP) of the SG with very different pain responses. Thus, changing the pulsing protocols allowed for pain mitigation and eliminated the need for injectable anesthetic. Histology confirmed the selective damaging of the SG at depth and the preservation of the surrounding dermis and the epidermis. CONCLUSIONS The multi-pulse 1726 nm laser with bulk air-cooling, thermal monitoring, treat-to-temperature (and depth) control, and a unique pulsing protocol, is capable of selectively damaging SGs at depth without damage to the surrounding dermis or the epidermis. The system offers two different protocols that were developed with different levels of discomfort allowing for two different methods for pain mitigation (injectable vs. topical anesthesia).
Collapse
Affiliation(s)
- Emil A Tanghetti
- Center for Dermatology and Laser Surgery, Sacramento, California, USA
| | | | | | - Aubrey Eck
- Accure Acne Inc., Boulder, Colorado, USA
| | | | | | - Joel L Cohen
- AboutSkin Dermatology and Derm Surgery, Greenwood Village, Colorado, USA
| | - Daniel P Friedmann
- Westlake Dermatology Research Center, Westlake Dermatology & Cosmetic Surgery, Austin, Texas, USA
| | - Mitchel P Goldman
- Cosmetic Laser Dermatology, A Platinum Dermatology Partners Company, San Diego, California, USA
| | - Hyemin Pomerantz
- VivaSkin Dermatology and Aesthetics, Wellesley, Massachusetts, USA
| | - Jordan V Wang
- Laser & Skin Surgery Center of New York, New York, New York, USA
| | - Roy G Geronemus
- Laser & Skin Surgery Center of New York, New York, New York, USA
| | - R Rox Anderson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Fernanda H Sakamoto
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
72
|
Li H, Zhang Z, Zhu D, Zheng H, Zhu Z, Shen N, Guo Z, Wu X, Qi X, Li Q, Ma Q, Xiang H. A Dual-Responsive Fe₃O₄@ZIF-8 Nanoplatform Combining Magnetic Targeting and pH Sensitivity for Low Back Pain Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410874. [PMID: 39981971 DOI: 10.1002/smll.202410874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/06/2025] [Indexed: 02/22/2025]
Abstract
Low back pain (LBP) resulting from sciatic nerve compression presents major challenges in pain management, as traditional therapies provide only short-term relief and pose risks of systemic toxicity. In this study, an innovative Fe3O4@ZIF-8-RVC (FZR) dual-responsive nanoplatform is introduced that integrates magnetic targeting with pH-sensitive, sustained drug release to overcome these limitations. The FZR nanoplatform encapsulates ropivacaine (RVC) within the ZIF-8-coated Fe3O4 core, enabling precise and prolonged analgesia at the injury site through magnetic guidance and acid-triggered release. In vitro and in vivo assessments indicate that FZR achieves high drug loading, sustained release in acidic environments, and excellent biocompatibility, significantly extending analgesic effects in chronic nerve injury models while minimizing systemic exposure. Behavioral tests and molecular analyses in LBP rat models confirm that FZR effectively suppresses pain-related neuronal activity and central sensitization markers. This dual-responsive nanoplatform FZR offers a safe, long-lasting, and targeted therapeutic approach, holding strong potential for advancing pain relief in LBP and related neuropathic pain conditions.
Collapse
Affiliation(s)
- Hao Li
- Department of Orthopedics, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| | - Zhihao Zhang
- Department of Orthopedics, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| | - Dingding Zhu
- Faculty of Physics, Qingdao University, Qingdao, 266021, P. R. China
| | - Huiyuan Zheng
- School of Pharmacy, Qingdao University, Qingdao, 266021, P. R. China
| | - Zhongze Zhu
- Department of Orthopedics, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| | - Nana Shen
- The Department of Rehabiliation, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| | - Zhu Guo
- Department of Orthopedics, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| | - Xiaolin Wu
- Department of Orthopedics, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| | - Xiaoying Qi
- The Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| | - Qiang Li
- Faculty of Physics, Qingdao University, Qingdao, 266021, P. R. China
| | - Qingming Ma
- School of Pharmacy, Qingdao University, Qingdao, 266021, P. R. China
| | - Hongfei Xiang
- Department of Orthopedics, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, P. R. China
| |
Collapse
|
73
|
Palchaudhuri S, Lin BX, Osypenko D, Wu J, Kochubey O, Schneggenburger R. A posterior insula to lateral amygdala pathway transmits US-offset information with a limited role in fear learning. Cell Rep 2025; 44:115320. [PMID: 39954251 DOI: 10.1016/j.celrep.2025.115320] [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: 04/21/2023] [Revised: 03/21/2024] [Accepted: 01/27/2025] [Indexed: 02/17/2025] Open
Abstract
During fear learning, associations between a sensory cue (conditioned stimulus, CS) and an aversive stimulus (unconditioned stimulus, US) are formed in specific brain circuits. The lateral amygdala (LA) is involved in CS-US integration; however, US pathways to the LA remain understudied. Here, we investigated whether the posterior insular cortex (pInsCx), a hub for aversive state signaling, transmits US information to the LA during fear learning. We find that the pInsCx makes a robust, glutamatergic projection specifically targeting the anterior LA. In vivo Ca2+ imaging reveals that neurons in the pInsCx and anterior LA display US-onset and US-offset responses; imaging combined with axon silencing shows that the pInsCx selectively transmits US-offset information to the anterior LA. Optogenetic silencing, however, does not show a role for US-driven activity in the anterior LA or its pInsCx afferents in fear memory formation. Thus, we describe a cortical projection that carries US-offset information to the amygdala with a limited role in fear learning.
Collapse
Affiliation(s)
- Shriya Palchaudhuri
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Bei-Xuan Lin
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Denys Osypenko
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Jinyun Wu
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Olexiy Kochubey
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ralf Schneggenburger
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
74
|
Jariyasakulroj S, Shu Y, Lin Z, Chen J, Chang Q, Ko PF, Chen JF. Mapping cell diversity and dynamics in inflammatory temporomandibular joint osteoarthritis with pain at single-cell resolution. JCI Insight 2025; 10:e184379. [PMID: 39927459 PMCID: PMC11948589 DOI: 10.1172/jci.insight.184379] [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: 07/09/2024] [Accepted: 12/13/2024] [Indexed: 02/11/2025] Open
Abstract
Temporomandibular joint (TMJ) osteoarthritis with pain is a highly prevalent disorder affecting patients' quality of life. A comprehensive understanding of cell type diversity and its dynamics in painful TMJ osteoarthritis (TMJOA) is lacking. Here, we utilized an inflammatory TMJOA mouse model via intra-articular injection of CFA. TMJOA mice exhibited cartilage remodeling, bone loss, synovitis, increased osteoarthritis (OA) score, and orofacial pain, recapitulating hallmark symptoms in patients. Single-cell transcriptomic profiling of the TMJ was performed in conjunction with mouse genetic labeling, tissue clearing, light sheet and confocal 3D imaging, multiplex RNAscope, and immunodetection. We visualized, reconstructed, and analyzed the distribution and density of nociceptive innervation of TMJ at single-axon levels. We systematically mapped the heterogeneity and anatomical position of blood endothelial cells, synovial fibroblasts, and immune cells, including Cx3cr1-positive barrier macrophages. Importantly, TMJOA mice exhibited enhanced neurovascular coupling, sublining fibroblast hyperplasia, inflammatory immune cell expansion, disrupted signaling-dependent cell-cell interaction, and a breakdown of the sandwich-like organization consisting of synovial barrier macrophages and fibroblasts. By utilizing a mouse model with combined TMJ pain history and OA, we reveal the cellular diversity, anatomical structure, and cell dynamics of the TMJ at single-cell resolution, which facilitate our understanding and potential targeting of TMJOA.
Collapse
Affiliation(s)
- Supawadee Jariyasakulroj
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
- Department of Masticatory Science, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Yang Shu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Ziying Lin
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Jingyi Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Qing Chang
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Pao-Fen Ko
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
75
|
Jiang H, Fang W, Feng Y, Liu X, Zhao J, Xu L, Han G, Long X. Bafilomycin A1 mitigates subchondral bone degeneration and pain in TMJOA rats. Int Immunopharmacol 2025; 147:113947. [PMID: 39756167 DOI: 10.1016/j.intimp.2024.113947] [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: 11/12/2024] [Revised: 12/14/2024] [Accepted: 12/22/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Pain and disability are primary concerns for temporomandibular joint osteoarthritis (TMJOA) patients, and the efficacy of current treatments remains controversial. Overactive osteoclasts are associated with subchondral bone degeneration and pain in OA. The vacuolar H+-ATPase (V-ATPase) is crucial for differentiation and function in osteoclasts, but its role in TMJOA is not well defined. This study aims to evaluate the effects of the V-ATPase inhibitor, bafilomycin A1 (Baf A1) on the progression and pain of TMJOA. MATERIALS AND METHODS Pain behavior tests, histological staining, tartrate-resistant acid phosphatase (TRAP) staining, immunofluorescence staining, and micro-CT analysis were conducted to evaluate the therapeutic efficacy of Baf A1 in monosodium iodoacetate-induced TMJOA in rats. Additionally, TRAP staining, enzyme-linked immunosorbent assay and immunofluorescence staining were used to assess the inhibitory effects of Baf a1 on the osteoclastogenesis, secretion of netrin-1 and neurite growth of trigeminal ganglion (TG) neurons. RESULTS Baf A1 significantly mitigated subchondral bone degeneration by suppressing osteoclastogenesis and subsequently inhibited cartilage degradation in TMJOA rats. Baf A1 also effectively alleviated pain behavior by inhibiting expression of netrin-1 and innervation of sensory nerve in TMJOA rats. In vitro assays of osteoclast and TG further demonstrated the inhibitory effects of Baf A1 on osteoclastogenesis, secretion of netrin-1 and neurite outgrowth of TG. CONCLUSIONS This study demonstrates that Baf A1 inhibits V-ATPase to mitigate TMJOA degeneration and pain by suppressing osteoclastogenesis and secretion of netrin-1, thereby suggesting it as a potential clinical treatment option for degeneration and pain of TMJOA.
Collapse
Affiliation(s)
- Henghua Jiang
- Department of Orthodontic Division II, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Wei Fang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Yaping Feng
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Xin Liu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Jie Zhao
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Liqin Xu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Guangli Han
- Department of Orthodontic Division II, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| | - Xing Long
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
76
|
Shin DA, Chang MC. Consciousness Research Through Pain. Healthcare (Basel) 2025; 13:332. [PMID: 39942521 PMCID: PMC11816597 DOI: 10.3390/healthcare13030332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Consciousness is a complex and elusive phenomenon encompassing self-awareness, sensory perception, emotions, and cognition. Despite significant advances in neuroscience, understanding the neural mechanisms underlying consciousness remains challenging. Pain, as a subjective and multifaceted experience, offers a unique lens for exploring consciousness by integrating sensory inputs with emotional and cognitive dimensions. This study examines the relationship between consciousness and pain, highlighting the potential of pain as a model for understanding the interplay between subjective experience and neural activity. Methods: Literature review. Results: Key theories of consciousness, such as the Global Workspace Theory and the Integrated Information Theory, provide diverse frameworks for interpreting the emergence of consciousness. Similarly, pain research emphasizes the role of subjective interpretation and emotional context in shaping sensory experiences, reflecting broader challenges in consciousness studies. The limitations of current methodologies, particularly the difficulty of objectively measuring subjective phenomena, like pain and consciousness, are also addressed. This highlights the importance of neural correlates, with a particular focus on brain regions, such as the anterior cingulate cortex and the insula, which bridge sensory and emotional experiences. By analyzing the shared attributes of pain and consciousness, this study underscores the potential for pain to serve as a measurable proxy in consciousness research. Conclusions: Ultimately, it contributes to unraveling the neural and philosophical underpinnings of consciousness, offering implications for mental health treatment and advancements in artificial intelligence. This study fills a critical gap by leveraging pain as a measurable and reproducible model for exploring the neural and subjective mechanisms of consciousness. By combining theoretical frameworks with empirical evidence, it offers novel insights into how consciousness emerges from neural processes.
Collapse
Affiliation(s)
- Dong Ah Shin
- Department of Neurosurgery, Spine and Spinal Cord Institute, Severance Hospital, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea;
| | - Min Cheol Chang
- Department of Physical Medicine and Rehabilitation, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu 42415, Republic of Korea
| |
Collapse
|
77
|
Sun J, Wang D, Wei Y, Wang D, Ji Z, Sun W, Wang X, Wang P, Basmadji NP, Larrarte E, Pedraz JL, Ramalingam M, Xie S, Wang R. Capsaicin-induced Ca 2+ overload and ablation of TRPV1-expressing axonal terminals for comfortable tumor immunotherapy. NANOSCALE 2025; 17:3288-3305. [PMID: 39688368 DOI: 10.1039/d4nr04454a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
As a common malignancy symptom, cancer pain significantly affects patients' quality of life. Approximately 60%-90% of patients with advanced cancer experience debilitating pain. Therefore, a comprehensive treatment system that combines cancer pain suppression and tumor treatment could provide significant benefits for these patients. Here, we designed a manganese oxide (MnO2)/Bovine serum albumin (BSA)/polydopamine (PDA) composite nanoplatform internally loaded with capsaicin for cancer pain suppression and immunotherapy. MBD&C nanoparticles (NPs) can ablate tumor-innervated sensory nerve fibers via Transient receptor potential vanilloid 1 (TRPV1) channels, thereby reducing the pain caused by various inflammatory mediators. The ablation of TRPV1+ nerve terminals can also decrease the secretion of calcitonin gene-related peptide (CGRP) and substance P (SP) in sensory nerve fibers, thus reducing the tumor pain and inhibit tumor progression. MBD&C can promote calcium influx by activating overexpressed TRPV1 channels on the tumor membrane surface, thereby achieving cancer immunotherapy induced by endogenous Ca2+ overloading. In addition, MnO2 NPs can alleviate tumor hypoxia and mitigate the immunosuppressive tumor microenvironment (TME). Ultimately, this treatment system with dual capabilities of inhibiting tumor growth and relieving cancer pain makes comfortable tumor therapy feasible and paves the way for the development of patient-centered approaches to cancer treatment in the future.
Collapse
Affiliation(s)
- Jian Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China.
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai 264000, People's Republic of China.
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Deqiang Wang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Yiying Wei
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China.
| | - Danyang Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China.
| | - Zhengkun Ji
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Wanru Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China.
| | - Xin Wang
- Department of Rehabilitation Medicine, Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Pingyu Wang
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Nicola Paccione Basmadji
- TECNALIA, Basque Research & Technology Alliance (BRTA) Miñano, Spain
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology. Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain.
| | - Eider Larrarte
- TECNALIA, Basque Research & Technology Alliance (BRTA) Miñano, Spain
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology. Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain.
- Joint Research Laboratory (JRL) on Bioprinting and Advanced Pharma, Development, A Joint Venture of TECNALIA and University of the Basque Country (UPV/EHU), Centro de investigación Lascaray Ikergunea, Avenida Miguel de Unamuno, 01006 Vitoria-Gasteiz, Spain
- Bioaraba Health Research Institute, Jose Atxotegi, s/n, 01009 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Murugan Ramalingam
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology. Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain.
- Joint Research Laboratory (JRL) on Bioprinting and Advanced Pharma, Development, A Joint Venture of TECNALIA and University of the Basque Country (UPV/EHU), Centro de investigación Lascaray Ikergunea, Avenida Miguel de Unamuno, 01006 Vitoria-Gasteiz, Spain
- Bioaraba Health Research Institute, Jose Atxotegi, s/n, 01009 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
- School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Shuyang Xie
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai 264000, People's Republic of China.
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Ranran Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China.
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| |
Collapse
|
78
|
Tassou A, Richebe P, Rivat C. Mechanisms of chronic postsurgical pain. Reg Anesth Pain Med 2025; 50:77-85. [PMID: 39909543 DOI: 10.1136/rapm-2024-105964] [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: 08/14/2024] [Accepted: 10/25/2024] [Indexed: 02/07/2025]
Abstract
Chronic pain after surgery, also known as chronic postsurgical pain (CPSP), is recognized as a significant public health issue with serious medical and economic consequences. Current research on CPSP underscores the significant roles of both peripheral and central sensitization in pain development and maintenance. Peripheral sensitization occurs at the site of injury, through the hyperexcitability of nerve fibers due to surgical damage and the release of inflammatory mediators. This leads to increased expression of pronociceptive ion channels and receptors, such as transient receptor potential and acid-sensing ion channels (ASIC), enhancing pain signal transmission. Central sensitization involves long-term changes in the central nervous system, particularly in the spinal cord. In this context, sensitized spinal neurons become more responsive to pain signals, driven by continuous nociceptive input from the periphery, which results in an enhanced pain response characterized by hyperalgesia and/or allodynia. Key players in this process include N-methyl-D-aspartate receptor and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, along with proinflammatory cytokines and chemokines released by activated glia. These glial cells release substances that further increase neuronal excitability, maintaining the sensitized state and contributing to persistent pain. The activation of antinociceptive systems is required for the resolution of pain after surgery, and default in these systems may also be considered as an important component of CPSP. In this review, we will examine the clinical factors underlying CPSP in patients and the mechanisms previously established in preclinical models of CPSP that may explain how acute postoperative pain may transform into chronic pain in patients.
Collapse
Affiliation(s)
- Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Philippe Richebe
- Department of Anesthesiology and Pain Medicine, Polyclinique Bordeaux Nord Aquitaine (PBNA), Bordeaux, France
- Anesthesiology and Pain Medicine, Maisonneuve Rosemont Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Cyril Rivat
- University of Montpellier, Montpellier, France
- Institut des Neurosciences de Montpellier INSERM U1298, Montpellier, France
| |
Collapse
|
79
|
Tran NB, Lee SJ. Metallothionein-3-mediated intracellular zinc mediates antioxidant and anti-inflammatory responses in the complete Freund's adjuvant-induced inflammatory pain mouse model. Cell Death Discov 2025; 11:45. [PMID: 39904985 PMCID: PMC11794434 DOI: 10.1038/s41420-025-02322-1] [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: 10/22/2024] [Revised: 01/03/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
Chronic inflammatory pain is often caused by peripheral tissue damage and persistent inflammation. This disease substantially affects patients' physical and social well-being. We investigated the role of metallothionein-3 (MT3) in modulating complete Freund's adjuvant (CFA)-induced intracellular Zn2+ activity in an MT3 knockout mouse model of inflammatory pain in the hind paw. The results demonstrated that increasing intracellular Zn2+ levels ameliorate deficits in motor behavior, as well as inflammation in the paw, spleen, and thymus. Furthermore, intracellular Zn2+ was crucial in regulating oxidative stress markers (glutathione, superoxide dismutase, catalase, and malondialdehyde) and inflammatory cytokines, such as tumor necrosis factor-α and interleukin-6, in MT3 knockout mice induced with CFA. This study highlights the critical role of MT3 in coordinating the intracellular interaction with Zn2+, which is vital for the immune systems's protective functions. These interactions are fundamental for maintaining metal ion homeostasis and regulating the synthesis of various biomolecules in the body.
Collapse
Affiliation(s)
- Ngoc Buu Tran
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeonbuk-do, 54896, Republic of Korea
| | - Sook-Jeong Lee
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeonbuk-do, 54896, Republic of Korea.
| |
Collapse
|
80
|
Nakae A, Bu-Omer HM, Chang WC, Kishimoto C, Onishi Y, Sumioka H, Shiomi M. The Potential of a Robot Presence in Close Relationship to Influence Human Responses to Experimental Pain. Life (Basel) 2025; 15:229. [PMID: 40003638 PMCID: PMC11856449 DOI: 10.3390/life15020229] [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: 01/07/2025] [Revised: 01/29/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Pain management is a critical challenge in healthcare, often exacerbated by loneliness and emotional distress. This study investigated the potential of a communication robot, Moffuly, to reduce pain perception and influence hormonal responses in a controlled experimental setting. Nineteen healthy participants underwent heat pain stimulation under two conditions: with and without robotic interaction. Pain levels were assessed using the Short-form McGill Pain Questionnaire and the Visual Analogue Scale, while mood and mental states were evaluated through established questionnaires including the Profile of Mood States, Hospital Anxiety and Depression Scale, and Self-Rating Depression Scale. Hormonal changes, including cortisol, growth hormone, oxytocin, estradiol, and dehydroepiandrosterone-sulfate, were measured from blood samples collected at key time points. The results demonstrated significant reductions in subjective pain and improvements in mood following robotic interaction. These effects were accompanied by favorable hormonal changes, including increased oxytocin and decreased cortisol and growth hormone levels. The findings suggest that robotic interaction may serve as an innovative approach to pain management by addressing both physiological and psychological factors. This study highlights the potential of robotics to complement traditional therapies in alleviating pain and enhancing emotional well-being. By mitigating emotional distress and loneliness, robotic interventions may enhance existing pain therapies and offer innovative solutions for resource-limited healthcare systems.
Collapse
Affiliation(s)
- Aya Nakae
- Presence Media Research Group, Hiroshi Ishiguro Laboratories, Deep Interaction Laboratory Group, Advanced Telecommunications Research Institute International (ATR), 2-2-2 Hikaridai, Seika-Cho, Soraku-Gun, Kyoto 619-0288, Japan; (H.M.B.-O.); (W.-C.C.); (C.K.); (H.S.)
- Global Center for Medical Engineering and Informatics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Osaka 565-0871, Japan
- Laboratory of Science & Innovation for Pain, Graduate School of Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Osaka 565-0871, Japan
| | - Hani M. Bu-Omer
- Presence Media Research Group, Hiroshi Ishiguro Laboratories, Deep Interaction Laboratory Group, Advanced Telecommunications Research Institute International (ATR), 2-2-2 Hikaridai, Seika-Cho, Soraku-Gun, Kyoto 619-0288, Japan; (H.M.B.-O.); (W.-C.C.); (C.K.); (H.S.)
- Global Center for Medical Engineering and Informatics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Osaka 565-0871, Japan
- Laboratory of Science & Innovation for Pain, Graduate School of Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Osaka 565-0871, Japan
| | - Wei-Chuan Chang
- Presence Media Research Group, Hiroshi Ishiguro Laboratories, Deep Interaction Laboratory Group, Advanced Telecommunications Research Institute International (ATR), 2-2-2 Hikaridai, Seika-Cho, Soraku-Gun, Kyoto 619-0288, Japan; (H.M.B.-O.); (W.-C.C.); (C.K.); (H.S.)
- Laboratory of Science & Innovation for Pain, Graduate School of Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Osaka 565-0871, Japan
| | - Chie Kishimoto
- Presence Media Research Group, Hiroshi Ishiguro Laboratories, Deep Interaction Laboratory Group, Advanced Telecommunications Research Institute International (ATR), 2-2-2 Hikaridai, Seika-Cho, Soraku-Gun, Kyoto 619-0288, Japan; (H.M.B.-O.); (W.-C.C.); (C.K.); (H.S.)
- Global Center for Medical Engineering and Informatics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Osaka 565-0871, Japan
- Laboratory of Science & Innovation for Pain, Graduate School of Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Osaka 565-0871, Japan
| | - Yuya Onishi
- Department of Interaction Science Laboratories, Deep Interaction Laboratory Group, Advanced Telecommunications Research Institute International (ATR), 2-2-2 Hikaridai, Seika-Cho, Soraku-Gun, Kyoto 619-0288, Japan; (Y.O.); (M.S.)
| | - Hidenobu Sumioka
- Presence Media Research Group, Hiroshi Ishiguro Laboratories, Deep Interaction Laboratory Group, Advanced Telecommunications Research Institute International (ATR), 2-2-2 Hikaridai, Seika-Cho, Soraku-Gun, Kyoto 619-0288, Japan; (H.M.B.-O.); (W.-C.C.); (C.K.); (H.S.)
| | - Masahiro Shiomi
- Department of Interaction Science Laboratories, Deep Interaction Laboratory Group, Advanced Telecommunications Research Institute International (ATR), 2-2-2 Hikaridai, Seika-Cho, Soraku-Gun, Kyoto 619-0288, Japan; (Y.O.); (M.S.)
| |
Collapse
|
81
|
Wong C, Rodriguez-Hernandez LD, Lister KC, Gu N, Cai W, Hooshmandi M, Fan J, Brown N, Nguyen V, Ribeiro-da-Silva A, Bonin RP, Khoutorsky A. Targeting spinal mechanistic target of rapamycin complex 2 alleviates inflammatory and neuropathic pain. Brain 2025; 148:675-686. [PMID: 39167538 PMCID: PMC11788203 DOI: 10.1093/brain/awae275] [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: 01/15/2024] [Revised: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
The development and maintenance of chronic pain involve the reorganization of spinal nocioceptive circuits. The mechanistic target of rapamycin complex 2 (mTORC2), a central signalling hub that modulates both actin-dependent structural changes and mechanistic target of rapamycin complex 1 (mTORC1)-dependent mRNA translation, plays key roles in hippocampal synaptic plasticity and memory formation. However, its function in spinal plasticity and chronic pain is poorly understood. Here, we show that pharmacological activation of spinal mTORC2 induces pain hypersensitivity, whereas its inhibition, using downregulation of the mTORC2-defining component Rictor, alleviates both inflammatory and neuropathic pain. Cell type-specific deletion of Rictor showed that the selective inhibition of mTORC2 in a subset of excitatory neurons impairs spinal synaptic potentiation and alleviates inflammation-induced mechanical and thermal hypersensitivity and nerve injury-induced heat hyperalgesia. The ablation of mTORC2 in inhibitory interneurons strongly alleviated nerve injury-induced mechanical hypersensitivity. Our findings reveal the role of mTORC2 in chronic pain and highlight its cell type-specific functions in mediating pain hypersensitivity in response to peripheral inflammation and nerve injury.
Collapse
Affiliation(s)
- Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Luis David Rodriguez-Hernandez
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Kevin C Lister
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Ning Gu
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Weihua Cai
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Mehdi Hooshmandi
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Jonathan Fan
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Nicole Brown
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Vivienne Nguyen
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 0C7, Canada
- Alan Edwards Center for the Research on Pain, McGill University, Montreal, QC, H3A 2B4 Canada
| | - Robert P Bonin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
- University of Toronto Centre for the Study of Pain, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
- Alan Edwards Center for the Research on Pain, McGill University, Montreal, QC, H3A 2B4 Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, H3G 1Y6, Canada
| |
Collapse
|
82
|
Zhen G, Do N, Manh NV, Ha HJ, Kim H, Kim H, Choi K, Ann J, Lee J. Discovery of a Novel Multitarget Analgesic Through an In Vivo-Guided Approach. Pharmaceuticals (Basel) 2025; 18:205. [PMID: 40006019 PMCID: PMC11859264 DOI: 10.3390/ph18020205] [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: 12/26/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Pain is a complex condition influenced by peripheral, central, immune, and psychological factors. Multitarget approaches offer a more effective and safer alternative to single-target analgesics by enhancing efficacy, reducing side effects, and minimizing tolerance. This study aimed to identify a novel multitarget analgesic with improved pharmacological properties. Methods: An in vivo-guided screening approach was used to discover a new analgesic compound. Compound 29, derived from a novel scaffold inspired by opiranserin and vilazodone pharmacophores, was identified through analog screening in the formalin test. Its efficacy was further evaluated in the spinal nerve ligation (SNL) model of neuropathic pain. Mechanistic studies explored its interaction with neurotransmitter transporters and receptors, while pharmacokinetic and safety assessments were conducted to determine its stability, brain penetration, and potential toxicity. Results: Compound 29 demonstrated high potency in the formalin test, with an ED50 of 0.78 mg/kg in the second phase and a concentration-dependent effect in the first phase. In the SNL model, it produced dose-dependent analgesic effects, increasing withdrawal thresholds by 24% and 45% maximum possible effect (MPE) at 50 and 100 mg/kg, respectively. Mechanistic studies revealed strong triple uptake inhibition, particularly at dopamine (DAT) and serotonin (SERT) transporters, alongside high-affinity 5-HT2A receptor antagonism. Pharmacokinetic analysis indicated enhanced stability and blood-brain barrier permeability. In vitro studies confirmed its nontoxicity to HT-22 cells but revealed potential hERG inhibition and strong CYP3A4 inhibition. Conclusions: Compound 29 is a promising multitarget analgesic with potent efficacy and favorable pharmacokinetics. Ongoing optimization efforts aim to mitigate side effects and enhance its therapeutic profile for clinical application.
Collapse
Affiliation(s)
- Guo Zhen
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Nayeon Do
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Nguyen Van Manh
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Hee-Jin Ha
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Hee Kim
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Hyunsoo Kim
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Kwanghyun Choi
- Medifron DBT, Seoul 08502, Republic of Korea; (H.-J.H.); (H.K.); (H.K.); (K.C.)
| | - Jihyae Ann
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| | - Jeewoo Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; (G.Z.); (N.D.); (N.V.M.)
| |
Collapse
|
83
|
Du L, Zhu J, Liu S, Yang W, Hu X, Zhang W, Cui W, Yang Y, Wang C, Yang Y, Gao T, Zhang C, Zhang R, Lou M, Zhou H, Rao J, Maoying Q, Chu Y, Wang Y, Mi W. Transient receptor potential melastatin 8 contributes to the interleukin-33-mediated cold allodynia in a mouse model of neuropathic pain. Pain 2025; 166:347-359. [PMID: 39132923 DOI: 10.1097/j.pain.0000000000003346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/17/2024] [Indexed: 08/13/2024]
Abstract
ABSTRACT Cold allodynia is a common complaint of patients suffering from neuropathic pain initiated by peripheral nerve injury. However, the mechanisms that drive neuropathic cold pain remain elusive. In this study, we show that the interleukin (IL)-33/ST2 signaling in the dorsal root ganglion (DRG) is a critical contributor to neuropathic cold pain by interacting with the cold sensor transient receptor potential melastatin 8 (TRPM8). By using the St2-/- mice, we demonstrate that ST2 is required for the generation of nociceptor hyperexcitability and cold allodynia in a mouse model of spared nerve injury (SNI). Moreover, the selective elimination of ST2 function from the Nav1.8-expressing nociceptor markedly suppresses SNI-induced cold allodynia. Consistent with the loss-of-function studies, intraplantar injection of recombinant IL-33 (rIL-33) is sufficient to induce cold allodynia. Mechanistically, ST2 is co-expressed with TRPM8 in both mouse and human DRG neurons and rIL-33-induced Ca 2+ influx in mouse DRG neurons through TRPM8. Co-immunoprecipitation assays further reveal that ST2 interacts with TRPM8 in DRG neurons. Importantly, rIL-33-induced cold allodynia is abolished by pharmacological inhibition of TRPM8 and genetic ablation of the TRPM8-expressing neurons. Thus, our findings suggest that the IL-33/ST2 signaling mediates neuropathic cold pain through downstream cold-sensitive TRPM8 channels, thereby identifying a potential analgesic target for the treatment of neuropathic cold pain.
Collapse
Affiliation(s)
- Lixia Du
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Biochemistry, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianyu Zhu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shenbin Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xueming Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenwen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenqiang Cui
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yayue Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenghao Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yachen Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tianchi Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruofan Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengping Lou
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong Zhou
- Department of Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Jia Rao
- Department of Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Qiliang Maoying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Yuxia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| |
Collapse
|
84
|
Chen Y, Bajpai AK, Li N, Xiang J, Wang A, Gu Q, Ruan J, Zhang R, Chen G, Lu L. Discovery of Novel Pain Regulators Through Integration of Cross-Species High-Throughput Data. CNS Neurosci Ther 2025; 31:e70255. [PMID: 39924344 PMCID: PMC11807727 DOI: 10.1111/cns.70255] [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: 11/19/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/11/2025] Open
Abstract
AIMS Chronic pain is an impeding condition that affects day-to-day life and poses a substantial economic burden, surpassing many other health conditions. This study employs a cross-species integrated approach to uncover novel pain mediators/regulators. METHODS We used weighted gene coexpression network analysis to identify pain-enriched gene module. Functional analysis and protein-protein interaction (PPI) network analysis of the module genes were conducted. RNA sequencing compared pain model and control mice. PheWAS was performed to link genes to pain-related GWAS traits. Finally, candidates were prioritized based on node degree, differential expression, GWAS associations, and phenotype correlations. RESULTS A gene module significantly over-enriched with the pain reference set was identified (referred to as "pain module"). Analysis revealed 141 pain module genes interacting with 46 pain reference genes in the PPI network, which included 88 differentially expressed genes. PheWAS analysis linked 53 of these genes to pain-related GWAS traits. Expression correlation analysis identified Vdac1, Add2, Syt2, and Syt4 as significantly correlated with pain phenotypes across eight brain regions. NCAM1, VAMP2, SYT2, ADD2, and KCND3 were identified as top pain response/regulator genes. CONCLUSION The identified genes and molecular mechanisms may enhance understanding of pain pathways and contribute to better drug target identification.
Collapse
Affiliation(s)
- Ying Chen
- Department of Histology and Embryology, Medical CollegeNantong UniversityNantongJiangsuChina
| | - Akhilesh K. Bajpai
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Nan Li
- Department of Histology and Embryology, Medical CollegeNantong UniversityNantongJiangsuChina
| | - Jiahui Xiang
- Medical CollegeNantong UniversityNantongJiangsuChina
| | - Angelina Wang
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Qingqing Gu
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
- Department of CardiologyAffiliated Hospital of Nantong UniversityJiangsuChina
| | - Junpu Ruan
- Medical CollegeNantong UniversityNantongJiangsuChina
| | - Ran Zhang
- Medical CollegeNantong UniversityNantongJiangsuChina
| | - Gang Chen
- Department of Histology and Embryology, Medical CollegeNantong UniversityNantongJiangsuChina
- Department of AnesthesiologyAffiliated Hospital of Nantong UniversityJiangsu ProvinceChina
| | - Lu Lu
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| |
Collapse
|
85
|
Peterson IL, Scholpa NE, Bachtle KJ, Frye JB, Loppi SH, Thompson AD, Doyle K, Largent-Milnes TM, Schnellmann RG. Formoterol alters chemokine expression and ameliorates pain behaviors after moderate spinal cord injury in female mice. J Pharmacol Exp Ther 2025; 392:100015. [PMID: 40023611 DOI: 10.1124/jpet.124.002171] [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: 02/07/2024] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Secondary spinal cord injury (SCI) is characterized by increased cytokines and chemokines at the site of injury that have been associated with the development of neuropathic pain. Nearly 80% of SCI patients report suffering from chronic pain, which is poorly managed with available analgesics. While treatment with the US Food and Drug Administration-approved β2-adrenergic receptor agonist formoterol improves various aspects of recovery post-SCI in vivo, its effects on cytokines, chemokines, and neuropathic pain remain unknown. Female mice were subjected to moderate (60 kilodynes [kdyn]) or severe (80 kdyn) SCI followed by daily treatment with vehicle or formoterol (0.3 mg/kg, i.p.) beginning 8 hours after injury. The expression of proinflammatory cytokines/chemokines, such as interferon gamma-induced protein 10, macrophage inflammatory protein 1a, monocyte chemoattractant protein 1, B-cell attracting chemokine 1, and nuclear factor kappa-light-chain-enhancer of activated B-cells, was increased in the injury site of vehicle-treated mice 24 hours post-SCI, which was ameliorated with formoterol treatment, regardless of injury severity. Thermal hyperalgesia and mechanical allodynia, as measured by Hargreaves infrared apparatus and von Frey filaments, respectively, were assessed prior to SCI and then weekly beginning 21 days post-injury (DPI). While all injured mice exhibited decreased withdrawal latency following thermal stimulation compared with baseline, formoterol treatment reduced this response ∼15% by 35 DPI. Vehicle-treated mice displayed significant mechanical allodynia, as evidenced by a 55% decrease in withdrawal threshold from baseline. In contrast, mice treated with formoterol maintained a consistent withdrawal time at all times tested. These data indicate that formoterol reduces inflammation post-SCI, likely contributing to mitigation of neuropathic pain and further supporting the therapeutic potential of this treatment strategy. SIGNIFICANCE STATEMENT: Chronic pain is a detrimental consequence of spinal cord injury (SCI). We show that treatment with the US Food and Drug Administration-approved drug formoterol after SCI decreases injury site proinflammatory chemo-/cytokines and alters markers of glial cell activation and infiltration. Additionally, formoterol treatment improves locomotor function and body composition, and decreases lesion volume. Finally, formoterol treatment decreased mechanical allodynia and thermal hyperalgesia post-SCI. These data are suggestive of the mechanism of formoterol-induced recovery, and further indicate its potential as a therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ingrid L Peterson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona; Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Natalie E Scholpa
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona; Southern Arizona VA Health Care System, Tucson, Arizona.
| | - Kiara J Bachtle
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jennifer B Frye
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Sanna H Loppi
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Austin D Thompson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Kristian Doyle
- Southern Arizona VA Health Care System, Tucson, Arizona; Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | | | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona; Southern Arizona VA Health Care System, Tucson, Arizona; Department of Neuroscience, College of Medicine, University of Arizona, Tucson, Arizona; Southwest Environmental Health Science Center, University of Arizona, Tucson, Arizona; Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona.
| |
Collapse
|
86
|
Wani P, Anand R. Neuroplasticity and Pain Perception: Exploring the Complexities of Temporomandibular Disorders. Cureus 2025; 17:e79098. [PMID: 40104480 PMCID: PMC11918487 DOI: 10.7759/cureus.79098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2025] [Indexed: 03/20/2025] Open
Abstract
Temporomandibular disorders (TMDs) are prevalent conditions affecting the temporomandibular joint (TMJ), masticatory muscles, and associated structures, leading to pain, restricted movement, and joint noises. These disorders are multifactorial in origin, involving structural, functional, and psychological components. This review delves into the neurophysiological mechanisms of pain perception in TMDs, focusing on peripheral and central processes, including the role of neural plasticity in chronic pain. Peripheral mechanisms involve nociceptors in the TMJ, activated by inflammatory mediators, mechanical stress, and tissue damage, leading to pain. Peripheral sensitization, driven by factors such as cytokines and neuropeptides, enhances nociceptor sensitivity, contributing to chronic pain states. The trigeminal nerve is pivotal in transmitting nociceptive information to the central nervous system (CNS), with C-fibers and A-delta fibers involved in pain perception. Central sensitization, a hallmark of chronic pain in TMDs, involves neuroplastic changes in the CNS, including wind-up and long-term potentiation (LTP), enhancing pain perception and facilitating pain persistence. Neuroplasticity, both central and peripheral, plays a critical role in the development of chronic pain. Central plasticity includes synaptic changes and alterations in brain connectivity, which were observed in functional imaging studies of TMD patients. Peripheral plasticity involves the upregulation of ion channels and neurotransmitters, sustaining pain signals. Additionally, neuroimmune interactions between microglia, astrocytes, and pain pathways are integral to central sensitization. Understanding these mechanisms is crucial for developing effective treatments targeting both peripheral and central pain processes. Emerging therapies, including transient receptor potential (TRP) channel blockers and neuroimmune modulators, offer new avenues for managing TMD pain, emphasizing the need for a multifaceted treatment approach.
Collapse
Affiliation(s)
- Pinaki Wani
- Physiology, All India Institute of Medical Sciences, Raebareli, Raebareli, IND
| | | |
Collapse
|
87
|
Krishna AA, Abhirami BL, Kumaran A. Pain in rheumatoid arthritis: Emerging role of high mobility group box 1 protein-HMGB1. Life Sci 2025; 362:123361. [PMID: 39761742 DOI: 10.1016/j.lfs.2024.123361] [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: 10/14/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease where pain, driven by both inflammatory and non-inflammatory processes, is a major concern for patients. This pain can persist even after joint inflammation subsides. High mobility group box-1 (HMGB1) is a non-histone-DNA binding protein located in the nucleus that plays a key role in processes such as DNA transcription, recombination, and replication. HMGB1 can be released into the extracellular space through both passive and active mechanisms. Extracellular HMGB1 contributes to synovial inflammation, bone degradation, and the production of cytokines in RA by binding to toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE). It also forms complexes with molecules like lipopolysaccharide (LPS) and IL-1β, amplifying inflammatory responses. Due to its central role in these processes, HMGB1 is considered a promising therapeutic target in RA. It also acts as a nociceptive molecule in mediating pain in diseases such as diabetes and bone cancer. In this review, we explore how HMGB1 contributes to chronic pain in RA, supported by both in vitro and in vivo models. We begin by providing an overview of the mechanisms of pain in RA, the structure of HMGB1, its release mechanisms, and the therapeutic potential of targeting HMGB1 in RA. Following this, we highlight its role in peripheral and central pain sensitization through direct activation of the TLR4/MAPK/NF-κB pathway, as well as indirectly through downstream mediators, underscoring its potential as a target for managing RA pain.
Collapse
Affiliation(s)
- Anithakumari Aswathy Krishna
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Beena Levakumar Abhirami
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Alaganandam Kumaran
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
88
|
Ravuri HG, Sadowski P, Satake N, Mills PC. Efficacy of transdermal ketoprofen on surgical inflammation in dogs. Res Vet Sci 2025; 183:105512. [PMID: 39718306 DOI: 10.1016/j.rvsc.2024.105512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/18/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024]
Abstract
Ketoprofen is a non-steroidal anti-inflammatory drug (NSAID) used to treat pain and inflammation in dogs. Despite having effective analgesic efficacy, prolonged oral administration has been associated with adverse effects. Transdermal delivery of ketoprofen has reduced the incidence of adverse effects in humans and could potentially be used in veterinary clinical medicine. A transdermal (TD) ketoprofen formulation was recently developed for use in dogs and this study aimed to determine the pharmacodynamic activity of this formulation using surgical castration as an acute inflammatory model. Twelve dogs were randomly assigned to either a Control group (n = 6) or a TD group (n = 6). All dogs were castrated using standard surgical protocols, administered with pre-medication, consisting of acepromazine (0.055 mg/kg) and methadone (0.5 mg/kg) intramuscularly (IM) 30 min prior to induction of general anaesthesia. All dogs were then anaesthetised by injecting alfaxalone (2 mg/kg IV) via a 20 G 3 cm catheter in the left cephalic vein and subsequently maintained using isoflurane. Along with that TD group dogs also received TD ketoprofen (10 mg/kg) 1 h before pre-medication. Bloods were collected at 0 - hour (pre-surgery), and 1 and 2-h post-surgery and analysed for circulating eicosanoids using liquid chromatography mass spectrometry (LCMS) methods. Measured levels of Thromboxane B2 (TXB2) at both 1 and 2 h and Prostaglandin E2 (PGE2) at 2 h post-surgery were higher in the Control group compared to the TD group, suggesting pre-operative application of TD ketoprofen has a possible inhibitory effect on systemic inflammation and could be used to treat pain and inflammation in dogs.
Collapse
Affiliation(s)
- H G Ravuri
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - P Sadowski
- Central Analytical Research Facility, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - N Satake
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - P C Mills
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia.
| |
Collapse
|
89
|
Xu Q, Zheng Q, Cui X, Cleland A, Hincapie J, Raja SN, Dong X, Guan Y. Visualizing the modulation of neurokinin 1 receptor-positive neurons in the superficial dorsal horn by spinal cord stimulation in vivo. Pain 2025; 166:428-437. [PMID: 39140483 PMCID: PMC11723817 DOI: 10.1097/j.pain.0000000000003361] [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: 03/04/2024] [Accepted: 06/25/2024] [Indexed: 08/15/2024]
Abstract
ABSTRACT Spinal cord stimulation (SCS) is an effective modality for pain treatment, yet its underlying mechanisms remain elusive. Neurokinin 1 receptor-positive (NK1R + ) neurons in spinal lamina I play a pivotal role in pain transmission. To enhance our mechanistic understanding of SCS-induced analgesia, we investigated how different SCS paradigms modulate the activation of NK1R + neurons, by developing NK1R-Cre;GCaMP6s transgenic mice and using in vivo calcium imaging of superficial NK1R + neurons under anesthesia (1.5% isoflurane). Neurokinin 1 receptor-positive neurons in the lumbar spinal cord (L4-5) showed a greater activation by electrical test stimulation (TS, 3.0 mA, 1 Hz) at the hindpaw at 2 weeks after tibia-sparing nerve injury (SNI-t) than in naïve mice. Spinal cord stimulation was then delivered through a bipolar plate electrode placed epidurally at L1-2 level. The short-term 50-Hz high-intensity SCS (80% motor threshold [MoT], 10 minutes) induced robust and prolonged inhibition of NK1R + neuronal responses to TS in both naïve and SNI-t mice. The 30-minute 50-Hz and 900-Hz SCS applied at moderate intensity (50% MoT) also significantly inhibited neuronal responses in SNI-t mice. However, at low intensity (20% MoT), the 30-minute 900-Hz SCS only induced persistent neuronal inhibition in naïve mice, but not in SNI-t mice. In conclusion, both 10-minute high-intensity SCS and 30-minute SCS at moderate intensity inhibit the activation of superficial NK1R + neurons, potentially attenuating spinal nociceptive transmission. Furthermore, in vivo calcium imaging of NK1R + neurons provides a new approach for exploring the spinal neuronal mechanisms of pain inhibition by neuromodulation pain therapies.
Collapse
Affiliation(s)
- Qian Xu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
- Howard Hughes Medical Institute, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Qin Zheng
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Xiang Cui
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | | | | | - Srinivasa N. Raja
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
- Howard Hughes Medical Institute, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
- Department of Neurological Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, 21205, USA
- Department of Neurological Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
90
|
Rodrigues DCDN, Porto JCS, Dos Santos IL, Filho JIAB, Ferreira PMP. Repositioning anthelmintics for the treatment of inflammatory-based pathological conditions. Inflammopharmacology 2025; 33:551-571. [PMID: 39589670 DOI: 10.1007/s10787-024-01605-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/27/2024] [Indexed: 11/27/2024]
Abstract
Acute, uncontrolled and/or long-lasting inflammation causes a breakdown in immunological tolerance, leading to chronicity and contributing to a series of significant local or systemic tissue changes. Anti-inflammatory efficacy, fewer adverse effects, improved selectivity, and curative action are imminent issues for patients suffering from chronic inflammation-related pathologies. Then, we performed a complete and critical review about anthelmintics, discussing the main classes and the available preclinical evidence on repurposing to treat inflammation-based conditions. Despite low bioavailability, many benzimidazoles (albendazole and mebendazole), salicylanilides (niclosamide), macrocyclic lactones (avermectins), pyrazinoisoquinolones (praziquantel), thiazolides (nitazoxanide), piperazine derivatives, and imidazothiazoles (levamisole) indicate that repositioning is a promising strategy. They may represent a lower cost and time-saving course to expand anti-inflammatory options. Although mechanisms of action are not fully elucidated and well-delineated, in general, anthelmintics disrupt mitogen-activated protein kinases, the synthesis of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-8, IL-12, and IFN-γ), the migration and infiltration of leukocytes, and decrease COX-2 expression, which impacts negatively on the release of prostanoids and leukotrienes. Moreover, some of them reduce nuclear accumulation of NF-κB (niclosamide, albendazole, and ivermectin), levels of nitric oxide (nitazoxanide and albendazole), and mucus, cytokines, and bronchoconstriction in experimental inflammatory pulmonary diseases (ivermectin and niclosamide). Considering the linking between cytokines, bradykinin, histamine, and nociceptors with algesia, anthelmintics also stand out for treating inflammatory pain disorders (ivermectin, niclosamide, nitazoxanide, mebendazole, levamisole), including for cancer-related pain status. There are obstacles, including the low bioavailability and the first-pass metabolism.
Collapse
Affiliation(s)
- Débora Caroline do Nascimento Rodrigues
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí (UFPI), Universitaria Avenue, Teresina, Piauí, 64049-550, Brazil
| | - Jhonatas Cley Santos Porto
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí (UFPI), Universitaria Avenue, Teresina, Piauí, 64049-550, Brazil
| | - Ingredy Lopes Dos Santos
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí (UFPI), Universitaria Avenue, Teresina, Piauí, 64049-550, Brazil
| | - José Ivo Araújo Beserra Filho
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí (UFPI), Universitaria Avenue, Teresina, Piauí, 64049-550, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí (UFPI), Universitaria Avenue, Teresina, Piauí, 64049-550, Brazil.
| |
Collapse
|
91
|
García-Domínguez M. NGF in Neuropathic Pain: Understanding Its Role and Therapeutic Opportunities. Curr Issues Mol Biol 2025; 47:93. [PMID: 39996814 PMCID: PMC11854882 DOI: 10.3390/cimb47020093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Nerve growth factor (NGF) is one of the essential components that have been implicated in the pathophysiology of neuropathic pain, a condition that develops following nerve injury or dysfunction. This neurotrophin is critical for the survival and maintenance of sensory neurons, and its dysregulation has been implicated in the sensitization of pain pathways. NGF interacts with its receptor TrkA and p75NTR to activate intracellular signaling pathways associated with nociception and the emergence of allodynia and hyperalgesia. Therapeutic approaches employing neutralizing antibodies and molecule inhibitors have been highly effective at both preclinical and clinical levels, hence giving hope again for the use of NGF as an important biomarker and therapeutic target in the management of neuropathic pain. By exploiting the unique properties of NGF and its interactions within the nervous system, new therapeutic modalities could be designed to enhance efficacy while minimizing side effects. In conclusion, taking advantage of the multifaceted dynamics of NGF could provide effective pain management therapies to finally respond to the unmet needs of patients experiencing neuropathic pain.
Collapse
Affiliation(s)
- Mario García-Domínguez
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain;
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
92
|
Beignon F, Notais M, Diochot S, Baron A, Fajloun Z, Tricoire-Leignel H, Lenaers G, Mattei C. Neurotoxins Acting on TRPV1-Building a Molecular Template for the Study of Pain and Thermal Dysfunctions. Toxins (Basel) 2025; 17:64. [PMID: 39998081 PMCID: PMC11861614 DOI: 10.3390/toxins17020064] [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: 01/06/2025] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Transient Receptor Potential (TRP) channels are ubiquitous proteins involved in a wide range of physiological functions. Some of them are expressed in nociceptors and play a major role in the transduction of painful stimuli of mechanical, thermal, or chemical origin. They have been described in both human and rodent systems. Among them, TRPV1 is a polymodal channel permeable to cations, with a highly conserved sequence throughout species and a homotetrameric structure. It is sensitive to temperature above 43 °C and to pH below 6 and involved in various functions such as thermoregulation, metabolism, and inflammatory pain. Several TRPV1 mutations have been associated with human channelopathies related to pain sensitivity or thermoregulation. TRPV1 is expressed in a large part of the peripheral and central nervous system, most notably in sensory C and Aδ fibers innervating the skin and internal organs. In this review, we discuss how the transduction of nociceptive messages is activated or impaired by natural compounds and peptides targeting TRPV1. From a pharmacological point of view, capsaicin-the spicy ingredient of chilli pepper-was the first agonist described to activate TRPV1, followed by numerous other natural molecules such as neurotoxins present in plants, microorganisms, and venomous animals. Paralleling their adaptive protective benefit and allowing venomous species to cause acute pain to repel or neutralize opponents, these toxins are very useful for characterizing sensory functions. They also provide crucial tools for understanding TRPV1 functions from a structural and pharmacological point of view as this channel has emerged as a potential therapeutic target in pain management. Therefore, the pharmacological characterization of TRPV1 using natural toxins is of key importance in the field of pain physiology and thermal regulation.
Collapse
Affiliation(s)
- Florian Beignon
- University of Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France; (F.B.); (M.N.); (H.T.-L.); (G.L.)
| | - Margaux Notais
- University of Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France; (F.B.); (M.N.); (H.T.-L.); (G.L.)
| | - Sylvie Diochot
- Université Côte d’Azur, CNRS U7275, INSERM U1323, IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), 660 Route des Lucioles, Sophia-Antipolis, F-06560 Nice, France; (S.D.); (A.B.)
| | - Anne Baron
- Université Côte d’Azur, CNRS U7275, INSERM U1323, IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), 660 Route des Lucioles, Sophia-Antipolis, F-06560 Nice, France; (S.D.); (A.B.)
| | - Ziad Fajloun
- Laboratory of Applied Biotechnology (LBA3B), Department of Cell Culture, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon;
| | - Hélène Tricoire-Leignel
- University of Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France; (F.B.); (M.N.); (H.T.-L.); (G.L.)
| | - Guy Lenaers
- University of Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France; (F.B.); (M.N.); (H.T.-L.); (G.L.)
- Service de Neurologie, CHU d’Angers, F-49000 Angers, France
| | - César Mattei
- University of Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France; (F.B.); (M.N.); (H.T.-L.); (G.L.)
| |
Collapse
|
93
|
Zeng H, Zhang Z, Zhou D, Wang R, Verkhratsky A, Nie H. Investigation of the anti-inflammatory, anti-pruritic, and analgesic effects of sophocarpine inhibiting TRP channels in a mouse model of inflammatory itch and pain. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118882. [PMID: 39366497 DOI: 10.1016/j.jep.2024.118882] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sophocarpine is a bioactive compound extracted from the dried root of Sophorae Flavesentis Aiton, a plant that has been used for thousands of years for various conditions including skin itch and pain. Its antipruritic and analgesic effects are suggested in publications, while the molecular mechanisms underneath interacting with TRP channels are not understood. AIM OF THE STUDY We investigated the anti-inflammatory, antipruritic, and analgesic effects of sophocarpine in a murine inflammatory itch and pain model to elucidate the underlying mechanisms. MATERIALS AND METHODS We evaluated sophocarpine's anti-pruritic and analgesic effects by monitoring mice's scratching and wiping behaviors, and the anti-inflammatory effect by measuring psoriasis area and severity index (PASI) score. The mRNA and protein expression of TRPA1/TRPV1 was analyzed by real-time quantitative polymerase chain reaction and western blotting. We further investigated the relationship between sophocarpine and TRPA1/TRPV1 in mice administered allyl-isothiocyanate (AITC) or capsaicin and by molecular docking. RESULTS We found that sophocarpine decreased scratching bouts, wipes, and the PASI score, reduced the TNF-α and IL-1β in the skin and TRPA1 and TRPV1 in the trigeminal ganglion. Pretreatment of sophocarpine decreased AITC-induced scratching bouts and wipes and capsaicin-induced wipes. We also found potential competitive bindings between sophocarpine and AITC/capsaicin to TRPA1/TRPV1. CONCLUSIONS Sophocarpine is a potential competitive inhibitor of TRPA1 and TRPV1 channels eliciting strong anti-inflammatory, anti-pruritic, and analgesic effects, suggesting its significant therapeutic potential in treating diseases with inflammatory itch and pain.
Collapse
Affiliation(s)
- Hekun Zeng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Zhe Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Dan Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Ranjing Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania.
| | - Hong Nie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, China.
| |
Collapse
|
94
|
Shi S, Gong X. The Role of Microglia in Perioperative Pain and Pain Treatment: Recent Advances in Research. J Integr Neurosci 2025; 24:22675. [PMID: 40018770 DOI: 10.31083/jin22675] [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: 03/05/2024] [Revised: 06/29/2024] [Accepted: 08/12/2024] [Indexed: 03/01/2025] Open
Abstract
Microglia play a crucial role in monitoring the microenvironment of the central nervous system. Over the past decade, the role of microglia in the field of pain has gradually been unraveled. Microglia activation not only releases proinflammatory factors that enhance nociceptive signaling, but also participates in the resolving of pain. Opioids induce microglia activation, which enhances phagocytic activity and release of neurotoxic substances. Conversely, microglia activation reduces opioid efficacy and results in opioid tolerance. The application of microglia research to clinical pain management and drug development is a promising but challenging area. Microglia-targeted therapies may provide new avenues for pain management.
Collapse
Affiliation(s)
- Shengnan Shi
- Department of Anesthesiology, Institution of Neuroscience and Brain Disease, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, 441000 Xiangyang, Hubei, China
| | - Xingrui Gong
- Department of Anesthesiology, Institution of Neuroscience and Brain Disease, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, 441000 Xiangyang, Hubei, China
| |
Collapse
|
95
|
López-Carrillo J, Bernáldez-Sarabia J, Pawar TJ, Jiménez S, Dueñas S, Figueroa-Montiel A, Olivares-Romero JL, Granados-Soto V, Licea-Navarro AF, Caram-Salas NL. Systemic antihyperalgesic effect of a novel conotoxin from Californiconus californicus in an inflammatory pain model. FRONTIERS IN PAIN RESEARCH 2025; 5:1500789. [PMID: 39925365 PMCID: PMC11802583 DOI: 10.3389/fpain.2024.1500789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction This study explores the analgesic potential of the novel conotoxin O1_cal6.4b, derived from Californiconus californicus, as a candidate for pain management in a model of inflammatory pain. Methods O1_cal6.4b was systemically administered to Wistar rats, and its effects on thermal hyperalgesia and motor coordination were evaluated. Comparative analyses were conducted against O1_cal6.4d, ω-MVIIA, and standard analgesics (morphine, dexamethasone, and diclofenac). Structural differences between O1_cal6.4b and O1_cal6.4d were examined using in silico modeling and molecular dynamics simulations. Results Systemic administration of O1_cal6.4b significantly reduced thermal hyperalgesia in a dose-dependent manner without impairing motor coordination. The analgesic effect of O1_cal6.4b was superior to that of O1_cal6.4d, ω-MVIIA, and standard analgesics. Structural analyses revealed notable differences between O1_cal6.4b and O1_cal6.4d, suggesting unique functional properties. Discussion The findings indicate that O1_cal6.4b exhibits a promising analgesic profile with advantages over traditional opioid-based therapies. These results underscore the molecular diversity of conotoxins and highlight their potential as innovative analgesic treatments. Further research is needed to elucidate the mechanism of action of this novel conotoxin.
Collapse
Affiliation(s)
| | | | - Tushar J. Pawar
- Red de Estudios Moleculares Avanzados, Instituto de Ecología (INECOL), Xalapa, Mexico
| | - Samanta Jiménez
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
| | - Salvador Dueñas
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
| | | | | | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Alexei F. Licea-Navarro
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
- Dirección de Impulso a la Innovación y el Desarrollo (DIID), CICESE, Ensenada, Mexico
| | - Nadia L. Caram-Salas
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
- CONAHCYT. Av. Insurgentes Sur 1582, Col. Crédito Constructor, Deleg Benito Juárez, Mexico City, Mexico
| |
Collapse
|
96
|
Kumar S, Jin F, Park SJ, Choi W, Keuning SI, Massimino RP, Vu S, Lü W, Du J. Convergent Agonist and Heat Activation of Nociceptor TRPM3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634542. [PMID: 39896661 PMCID: PMC11785169 DOI: 10.1101/2025.01.23.634542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Detecting noxious heat is vital for survival, triggering pain responses that protect against harm1,2. The TRPM3 channel is a key nociceptor for sensing noxious heat and a promising therapeutic target for pain treatment and neurological disorders such as epilepsy3-11. Here, we functionally and structurally characterized TRPM3 in response to diverse stimuli: the synthetic superagonist CIM0216 Ref12, the anticonvulsant antagonist primidone13,14, and heat1,10,15. Our findings reveal that TRPM3 is intrinsically dynamic, with its intracellular domain (ICD) sampling both resting and activated states, though strongly favoring the resting state without stimulation. CIM0216 binds to the S1-S4 domain, inducing conformational changes in the ICD and shifting the equilibrium toward activation. Remarkably, heat induces similar ICD rearrangements, revealing a converged activation mechanism driven by chemical compounds and temperature. This mechanism is supported by functional data showing that mutations facilitating the ICD movement markedly increase the sensitivity of TRPM3 to both chemical and thermal signals. These findings establish a critical role of the ICD in temperature sensing in TRPM3, a mechanism likely conserved across the TRPM family. Finally, we show that primidone binds to the same site as CIM0216 but acts as an antagonist. This study provides a framework for understanding the thermal sensing mechanisms of temperature-sensitive ion channels and offers a structural foundation for developing TRPM3-target therapeutics for pain and neurological disorders.
Collapse
Affiliation(s)
- Sushant Kumar
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | - Sung Jin Park
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | - Sarah I. Keuning
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | | | - Wei Lü
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Juan Du
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
97
|
Dyson A, Gajjar G, Hoffman KC, Lewis D, Palega S, Rangel Silva E, Auwn J, Bellemer A. A nociceptor-specific RNAi screen in Drosophila larvae identifies RNA-binding proteins that regulate thermal nociception. PeerJ 2025; 13:e18857. [PMID: 39866556 PMCID: PMC11759608 DOI: 10.7717/peerj.18857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
Nociception is the process by which sensory neurons detect and encode potentially harmful environmental stimuli to generate behavioral responses. Nociceptor neurons exhibit plasticity in which their sensitivity to noxious stimuli and subsequent ability to drive behavior may be altered by environmental conditions, injury, infection, and inflammation. In some cases, nociceptor sensitization requires regulated changes in gene expression, and recent studies have indicated roles for post-transcriptional mechanisms in regulating these changes as an aspect of nociceptor plasticity. The larvae of Drosophila melanogaster have been developed as a powerful model for studying mechanisms of nociception, nociceptor plasticity, and nociceptor development. Diverse RNA-binding proteins regulate the development and morphology of larval nociceptors, implying important roles for post-transcriptional regulation of gene expression in these neurons, but the importance of these mechanisms for nociceptive behavior has not been investigated systematically. In this study, we conducted a nociceptor-specific RNAi screen of 112 candidate RNA-binding protein genes to identify those that are required for normal sensitivity to noxious thermal stimuli. The screen and subsequent validation experiments identified nine candidate genes (eIF2α, eIF4A, eIF4AIII, eIF4G2, mbl, SC35, snf, Larp4B and CG10445) that produce defects in nociceptive response latency when knocked down in larval nociceptors. Some of the genes identified have well-understood roles in the regulation of translation initiation and regulation of nociceptor sensitization in vertebrate and invertebrate animal models, suggesting an evolutionarily conserved role for these mechanisms in regulating nociceptor sensitivity. Other screen isolates have previously described roles in regulating nociceptor morphology and mRNA processing, but less clear roles in regulating nociceptor function. Further studies will be necessary to identify the mechanisms by which the identified RNA-binding proteins regulate sensory neuron function and the identities of the mRNAs that they target.
Collapse
Affiliation(s)
- Amber Dyson
- Department of Biology, Appalachian State University, Boone, North Carolina, United States
| | - Gita Gajjar
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, North Carolina, United States
| | - Katherine C. Hoffman
- Department of Biology, Appalachian State University, Boone, North Carolina, United States
| | - Dakota Lewis
- Department of Biology, Appalachian State University, Boone, North Carolina, United States
| | - Sara Palega
- Department of Biology, Appalachian State University, Boone, North Carolina, United States
| | - Erik Rangel Silva
- Department of Biology, Appalachian State University, Boone, North Carolina, United States
| | - James Auwn
- Department of Biology, Appalachian State University, Boone, North Carolina, United States
| | - Andrew Bellemer
- Department of Biology, Appalachian State University, Boone, North Carolina, United States
| |
Collapse
|
98
|
Peng BG. Fundamentals of intervertebral disc degeneration and related discogenic pain. World J Orthop 2025; 16:102119. [PMID: 39850042 PMCID: PMC11752479 DOI: 10.5312/wjo.v16.i1.102119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Lumbar intervertebral disc degeneration is thought to be the main cause of low back pain, although the mechanisms by which it occurs and leads to pain remain unclear. In healthy adult discs, vessels and nerves are present only in the outer layer of the annulus fibrosus and in the bony endplate. Animal models, and histological and biomechanical studies have shown that annulus tear or endplate injury is the initiating factor for painful disc degeneration. Injury to the disc triggers a local inflammatory repair response that activates nociceptors and promotes the synthesis of neuropeptides such as substance P and calcitonin gene-related peptide, by dorsal root ganglion neurons. These neuropeptides are transported to injured discs and act as pro-inflammatory molecules, promoting the production of an "inflammatory soup" by inducing vasodilatation and plasma extravasation as well as by promoting the release of chemical mediators from disc cells and infiltrating immune cells, causing neurogenic inflammation that leads to progressive disc degeneration and discogenic pain.
Collapse
Affiliation(s)
- Bao-Gan Peng
- Department of Orthopedics, The Third Medical Center, General Hospital of the Chinese People’s Liberation Army, Beijing 100039, China
| |
Collapse
|
99
|
Pușcașu C, Andrei C, Olaru OT, Zanfirescu A. Metabolite-Sensing Receptors: Emerging Targets for Modulating Chronic Pain Pathways. Curr Issues Mol Biol 2025; 47:63. [PMID: 39852178 PMCID: PMC11763455 DOI: 10.3390/cimb47010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Chronic pain is a debilitating condition affecting millions worldwide, often resulting from complex interactions between the nervous and immune systems. Recent advances highlight the critical role of metabolite-sensing G protein-coupled receptors (GPCRs) in various chronic pain types. These receptors link metabolic changes with cellular responses, influencing inflammatory and degenerative processes. Receptors such as free fatty acid receptor 1 (FFAR1/GPR40), free fatty acid receptor 4 (FFAR4/GPR120), free fatty acid receptor 2 (FFAR2/GPR43), and Takeda G protein-coupled receptor 5 (TGR5/GPR131/GPBAR1) are key modulators of nociceptive signaling. GPR40, activated by long-chain fatty acids, exhibits strong anti-inflammatory effects by reducing cytokine expression. Butyrate-activated GPR43 inhibits inflammatory mediators like nitric oxide synthase-2 and cyclooxygenase-2, mitigating inflammation. TGR5, activated by bile acids, regulates inflammation and cellular senescence through pathways like NF-κB and p38. These receptors are promising therapeutic targets in chronic pain, addressing the metabolic and inflammatory factors underlying nociceptive sensitization and tissue degeneration. This review explores the molecular mechanisms of metabolite-sensing receptors in chronic pain, their therapeutic potential, and challenges in clinical application. By uncovering these mechanisms, metabolite-sensing receptors could lead to safer, more effective pain management strategies.
Collapse
Affiliation(s)
| | - Corina Andrei
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (O.T.O.); (A.Z.)
| | | | | |
Collapse
|
100
|
Li G, Wang X, Wang Q, Han L, Bai J, Wang F, Yu B, Liu Z, Long X, Cheng Y. Coumarins rather than alkylamides evoke the numbing orosensation of pomelo peel. Food Chem 2025; 463:141502. [PMID: 39368197 DOI: 10.1016/j.foodchem.2024.141502] [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: 07/27/2024] [Revised: 09/20/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
Liangpingyou, a well-known Chinese pomelo (Citrus grandis L.) variety, elicits a unique and uncharacterized numbing aftertaste. To understand the molecular bases and characteristics of the pomelo-induced numbing sensation, we first determined that hydroxyl sanshools, the major Sichuan pepper chemosensates, were not responsible via silylation-GC-MS analysis. Pomelo peel juice was then subjected to solid-phase extraction to form 4 fractions, and key sensory-active substances were screened via taste dilution analysis. Three simple coumarins, meranzin hydrate, isomeranzin, and marmin, were identified to induce numbing, which has not been previously reported. Sensory studies via extensively modified half-tongue tests and verification steps revealed recognition thresholds within 0.49-1.78, 0.32-1.56, and 0.43-1.46 μmol/L for numbness, pungency, and astringency, respectively. The temporal dominance trends showed the following taste notes: Meranzin hydrate-numbing dominated, isomeranzin-numbing and pungent, and marmin-astringent and numbing. Molecular docking analysis suggested that coumarins target the receptors TRPV1, TPRA1, and KCNK3.
Collapse
Affiliation(s)
- Guijie Li
- Citrus Research Institute, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China.
| | - Xuting Wang
- Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Qundi Wang
- Citrus Research Institute, Southwest University, Chongqing, 400712, China.
| | - Leng Han
- Citrus Research Institute, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China.
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China.
| | - Fusheng Wang
- Citrus Research Institute, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China.
| | - Bo Yu
- Sichuan Dan Orange Modern Fruit Industry Co., Ltd, Danling, 620200, China
| | - Zhaojun Liu
- Chongqing Liangping District Agriculture and Rural Committee, Chongqing, 405200, China
| | - Xingyao Long
- Collaborative Innovation Center for Child Nutrition and Health Development, Chongqing University of Education, Chongqing, 400067, China.
| | - Yujiao Cheng
- Citrus Research Institute, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China.
| |
Collapse
|