1
|
Menezes J, Jakobsson JE, Bersellini Farinotti A, Krock E, Hunt MA, Simon N, Venckute Larsson S, Tanum L, Kultima K, Kosek E, Svensson CI. Comparative Analysis of Lysophosphatidic Acid Levels in Fibromyalgia and Other Painful Conditions in Female Patients. Eur J Pain 2025; 29:e70022. [PMID: 40269628 PMCID: PMC12018871 DOI: 10.1002/ejp.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/09/2025] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Previous work found a decrease in lysophosphatidylcholines (LPCs) in fibromyalgia (FM) serum, prompting the hypothesis that this decrease could be due to increased conversion of LPC to lysophosphatidic acid (LPA) through autotaxin (ATX). LPA has pronociceptive functions, and increased LPA levels could modulate FM pain. METHODS This study quantified LPA levels in serum and lumbar cerebrospinal fluid (CSF) and serum ATX levels in FM patients, comparing with healthy controls (HCs), osteoarthritis (OA), degenerative disc disease (DDD) and lumbar disc herniation (LDH) patients. RESULTS We found increased serum LPA levels in FM and OA patients, with no changes in FM lumbar CSF. Unexpectedly, a positive correlation between serum LPA and conditioned pain modulation was observed in FM patients, while LPA levels were correlated with pain intensity and Knee Injury and Osteoarthritis Outcome Scores in OA. Serum ATX levels in FM patients were comparable to those in HC but correlated significantly with FM LPA levels (in one cohort), as well as with pain duration and the maximal weekly pain intensity. CONCLUSIONS This study suggests that increased LPA levels play distinct roles in FM and OA patients. In FM, LPA levels were linked to less impaired inhibitory pain pathways, while LPA levels in OA correlated with pain intensity and knee-related impairment. ATX levels in FM serum are associated with pain intensity and duration. These findings underscore the complex role of LPA and ATX in FM pathophysiology. Future studies are essential to clarify LPA's specific roles and to develop therapies. SIGNIFICANCE STATEMENT This study provides novel insights into the role of LPA in FM and other chronic pain conditions. Although ATX levels were unchanged in FM, a positive correlation between serum ATX and LPA supports the role of ATX in LPA conversion. These findings suggest complex lipid dysregulation in FM, with LPA potentially modulating pain pathways. Further research is needed to clarify LPA's role and its potential as a biomarker or therapeutic target.
Collapse
Affiliation(s)
- Joana Menezes
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Jenny E. Jakobsson
- Department of Medical SciencesUppsala UniversityUppsalaSweden
- Clinical Pain Research, Department of Surgical SciencesUppsala UniversityUppsalaSweden
| | - Alex Bersellini Farinotti
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Emerson Krock
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
- Faculty of Dental Medicine and Oral Health SciencesAlan Edwards Centre for Research on Pain, McGill UniversityMontrealCanada
| | - Matthew A. Hunt
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Nils Simon
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Sigita Venckute Larsson
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| | - Lars Tanum
- Department of R&D in Mental HealthAkershus University HospitalLørenskogNorway
| | - Kim Kultima
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Eva Kosek
- Clinical Pain Research, Department of Surgical SciencesUppsala UniversityUppsalaSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Camilla I. Svensson
- Department of Physiology and PharmacologyCenter for Molecular Medicine, Karolinska InstitutetSolnaSweden
| |
Collapse
|
2
|
Jakobsson JE, Menezes J, Krock E, Hunt MA, Carlsson H, Vaivade A, Emami Khoonsari P, Agalave NM, Sandström A, Kadetoff D, Tour Sohlin J, Erngren I, Al-Grety A, Freyhult E, Sandor K, Kosek E, Svensson CI, Kultima K. Fibromyalgia patients have altered lipid concentrations associated with disease symptom severity and anti-satellite glial cell IgG antibodies. THE JOURNAL OF PAIN 2025; 29:105331. [PMID: 39922554 DOI: 10.1016/j.jpain.2025.105331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/02/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Autoimmunity and immunoglobulin G (IgG) autoantibodies may contribute to pain in a subset of fibromyalgia (FM) patients. Previously, IgG from FM patients was found to induce pain-like behavior in mice and bind to satellite glial cells (anti-SGC IgG). The anti-SGC IgG levels were also associated with more severe symptomatology. Lipid metabolism in FM subjects is altered with lysophosphatidylcholines (LPCs) acting as pain mediators. The relationship between autoantibodies, lipid metabolism, and FM symptomatology remains unclear. Serum lipidomics with liquid chromatography mass spectrometry, anti-SGC IgG levels, and clinical measures were examined in 35 female FM subjects and 33 age- and body mass index-balanced healthy controls (HC). Fibromyalgia subjects with higher anti-SGC IgG levels experienced more intense pain than those with lower levels. Sixty-three lipids were significantly altered between FM subjects and HC or between FM subjects with severe (FM severe) and mild symptoms (FM mild). Compared to HC, FM subjects had lower concentrations of lipid species belonging to the classes LPC (n = 10), lysophosphatidylethanolamine (n = 7), phosphatidylcholine (n = 4), and triglyceride (n = 5), but higher concentrations of diglyceride (n = 3). Additionally, FM severe had higher LPC 19:0, 22:0, and 24:1 and lower sphingomyelin (n = 9) concentrations compared to FM mild. Positive associations were seen for LPC 22:0 and 24:1 with pain intensity and anti-SGC IgG levels in FM subjects. Taken together, these results suggest an association between altered lipid metabolism and autoimmune mechanisms in FM.
Collapse
Affiliation(s)
- Jenny E Jakobsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Joana Menezes
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emerson Krock
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matthew A Hunt
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Carlsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Aina Vaivade
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Nilesh M Agalave
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Angelica Sandström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Diana Kadetoff
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Ida Erngren
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Asma Al-Grety
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Eva Freyhult
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eva Kosek
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Ren J, Yu L, Lin J, Liu Y, Ma L, Huang Y, Sun N, Deng Y, Zhong D, Zhou B, Jiang B, Yan M. Elevated 18:1 lysophosphatidylcholine contributes to neuropathic pain in peripheral nerve injury. Reg Anesth Pain Med 2025:rapm-2024-106195. [PMID: 40016127 DOI: 10.1136/rapm-2024-106195] [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: 11/01/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Neuropathic pain is a maladaptive and chronic condition with limited effective treatments. Although recent studies have suggested that certain lipid metabolites, like lysophosphatidylcholine (LPC), may contribute to chronic pain, their specific roles and mechanisms remain unclear. OBJECTIVE This study investigated the role and mechanism of LPC(18:1), a lipid subtype, in neuropathic pain caused by nerve injury. METHODS Using a mouse model of spinal nerve ligation, LPC(18:1) levels were measured in serum, dorsal root ganglion (DRG), spinal cord (SC) and cerebrospinal fluid (CSF). Nociception was assessed using von Frey and Hargreaves' methods, while molecular analyses explored inflammatory pathways and oxidative stress. RESULTS LPC(18:1) levels significantly increased in the serum, DRG and CSF after nerve injury. Administration of LPC(18:1) induced heightened pain responses and activated inflammatory pathways, including protein kinase C (PKC) and extracellular regulated protein kinase (ERK) in the DRG, as well as glial cells in the SC. The findings suggested that oxidative stress played a role in LPC(18:1) production, and its effects were mediated by G protein-coupled receptor 132 (GPR132). CONCLUSION LPC(18:1) may serve as a potential biomarker and therapeutic target for managing neuropathic pain.
Collapse
Affiliation(s)
- Jinxuan Ren
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Lina Yu
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jiaqi Lin
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, Shanghai, China
| | - Ying Liu
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Longfei Ma
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Yangyuxin Huang
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Na Sun
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Yutao Deng
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Da Zhong
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Binglin Zhou
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Baochun Jiang
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Min Yan
- Department of Anesthesiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Steinmeyer J. Phospholipids and Sphingolipids in Osteoarthritis. Biomolecules 2025; 15:250. [PMID: 40001553 PMCID: PMC11853253 DOI: 10.3390/biom15020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Many studies now emphasize the intricate relationship between lipid metabolism and osteoarthritis (OA), a leading cause of disability. This narrative review examines alterations in the levels of phospholipids (PLs) and sphingolipids (SLs) in synovial fluid (SF), plasma, serum, and articular tissues; discusses their role in joint lubrication, inflammation, and cartilage degradation; and describes their potential as diagnostic markers and therapeutic targets. Key findings include stage-dependent elevated levels of specific PLs and SLs in the SF, blood, and tissue of OA patients, implicating them as possible biomarkers of disease severity and progression. Studies suggest that beyond the involvement of these lipids in joint lubrication, individual species, such as lysophosphatidylcholine (LPC) 16:0, lysophosphatidic acid (LPA), ceramide-1-phosphate (C1P), and sphingosine-1-phosphate (S1P), contribute to pain, inflammation, and degradation of joints through various signaling pathways. Cross-species comparisons suggest that dogs and mice experience similar lipidomic changes during OA as humans, rendering them valuable models for studying lipid-related mechanisms. PLs and SLs in SF appear to originate primarily from the synovial blood capillaries through diffusion. In addition, lipids that are produced locally by fibroblast-like synoviocytes (FLSs) are influenced by cytokines and growth factors that regulate the biosynthesis of PLs for joint lubrication. Emerging research has identified genes such as UGCG and ESYT1 as regulators of lipid metabolism in OA. Further, we examine the suitability of lipids as biomarkers of OA and the potential of targeting the PL and SL pathways to treat OA, emphasizing the need for further research to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Juergen Steinmeyer
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics and Orthopaedic Surgery, Justus Liebig University, 35392 Giessen, Germany
| |
Collapse
|
5
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.23.563601. [PMID: 37961151 PMCID: PMC10634739 DOI: 10.1101/2023.10.23.563601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
|
6
|
Hamed Hamed D, Rodríguez-Pérez C, Pruimboom L, Navarro-Ledesma S. Relationship Between Metabolic Profile, Pain, and Functionality in Patients with Frozen Shoulder: A Cross-Sectional Study. Healthcare (Basel) 2024; 12:2444. [PMID: 39685066 DOI: 10.3390/healthcare12232444] [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: 10/28/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Frozen shoulder (FS), or adhesive capsulitis, is a disabling condition characterized by pain and restricted shoulder mobility. AIMS This study investigates the relationship between metabolic biomarkers-liver enzymes and thyroid function-and pain and shoulder functionality in patients with FS. METHODS A total of 32 patients (22 women and 10 men) were included in this cross-sectional study. Participants underwent clinical evaluations and blood tests to assess metabolic biomarkers, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma-glutamyl transferase (GGT), and thyroid-stimulating hormone (TSH). Pain and functionality were measured using the Shoulder Pain and Disability Index (SPADI). Correlation and multiple regression analyses were performed to assess the associations between biomarkers, pain, and functionality. RESULTS Significant negative correlations were found between AST (r = -0.528, p = 0.029), ALT (r = -0.533, p = 0.027), GGT (r = -0.602, p = 0.011), and TSH (r = -0.556, p = 0.017) with total pain scores. A significant negative correlation was also observed between TSH and SPADI scores (r = -0.511, p = 0.039). Multiple regression analysis showed that GGT (β = -0.335, p = 0.008) and TSH (β = -0.298, p = 0.014) were the strongest predictors of pain. These findings suggest that metabolic biomarkers, particularly liver enzymes and thyroid function, play a significant role in the pathophysiology of frozen shoulder. The results highlight the importance of assessing these biomarkers for better understanding and managing pain and functionality in patients with FS. CONCLUSIONS Further research is needed to explore the underlying mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Dina Hamed Hamed
- Clinical Medicine and Public Health Ph.D. Program, Faculty of Health Sciences, University of Granada, 18001 Granada, Spain
- Department of Physiotherapy, University Hospital of Melilla, C. Luis de Ostáriz 12, 52005 Melilla, Spain
| | - Celia Rodríguez-Pérez
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology "Jose Mataix" (INYTA-UGR), Biomedical Research Centre (CIMB), University of Granada, Avda. del Conocimiento s/n, 18100 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Leo Pruimboom
- Clinical Psychoneuroimmunology, University of Granada and PNI Europe, 52004 Melilla, Spain
| | - Santiago Navarro-Ledesma
- Clinical Psychoneuroimmunology, University of Granada and PNI Europe, 52004 Melilla, Spain
- Department of Physiotherapy, Faculty of Health Sciences, Campus of Melilla, University of Granada, Querol Street, 5, 52004 Melilla, Spain
| |
Collapse
|
7
|
Zhou R, Fu W, Vasylyev D, Waxman SG, Liu CJ. Ion channels in osteoarthritis: emerging roles and potential targets. Nat Rev Rheumatol 2024; 20:545-564. [PMID: 39122910 DOI: 10.1038/s41584-024-01146-0] [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] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Osteoarthritis (OA) is a highly prevalent joint disease that causes substantial disability, yet effective approaches to disease prevention or to the delay of OA progression are lacking. Emerging evidence has pinpointed ion channels as pivotal mediators in OA pathogenesis and as promising targets for disease-modifying treatments. Preclinical studies have assessed the potential of a variety of ion channel modulators to modify disease pathways involved in cartilage degeneration, synovial inflammation, bone hyperplasia and pain, and to provide symptomatic relief in models of OA. Some of these modulators are currently being evaluated in clinical trials. This review explores the structures and functions of ion channels, including transient receptor potential channels, Piezo channels, voltage-gated sodium channels, voltage-dependent calcium channels, potassium channels, acid-sensing ion channels, chloride channels and the ATP-dependent P2XR channels in the osteoarthritic joint. The discussion spans channel-targeting drug discovery and potential clinical applications, emphasizing opportunities for further research, and underscoring the growing clinical impact of ion channel biology in OA.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Wenyu Fu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Dmytro Vasylyev
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Huang C, Sun PY, Jiang Y, Liu Y, Liu Z, Han SL, Wang BS, Huang YX, Ren AR, Lu JF, Jiang Q, Li Y, Zhu MX, Yao Z, Tian Y, Qi X, Li WG, Xu TL. Sensory ASIC3 channel exacerbates psoriatic inflammation via a neurogenic pathway in female mice. Nat Commun 2024; 15:5288. [PMID: 38902277 PMCID: PMC11190258 DOI: 10.1038/s41467-024-49577-3] [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: 06/01/2023] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Psoriasis is an immune-mediated skin disease associated with neurogenic inflammation, but the underlying molecular mechanism remains unclear. We demonstrate here that acid-sensing ion channel 3 (ASIC3) exacerbates psoriatic inflammation through a sensory neurogenic pathway. Global or nociceptor-specific Asic3 knockout (KO) in female mice alleviates imiquimod-induced psoriatic acanthosis and type 17 inflammation to the same extent as nociceptor ablation. However, ASIC3 is dispensable for IL-23-induced psoriatic inflammation that bypasses the need for nociceptors. Mechanistically, ASIC3 activation induces the activity-dependent release of calcitonin gene-related peptide (CGRP) from sensory neurons to promote neurogenic inflammation. Botulinum neurotoxin A and CGRP antagonists prevent sensory neuron-mediated exacerbation of psoriatic inflammation to similar extents as Asic3 KO. In contrast, replenishing CGRP in the skin of Asic3 KO mice restores the inflammatory response. These findings establish sensory ASIC3 as a critical constituent in psoriatic inflammation, and a promising target for neurogenic inflammation management.
Collapse
Affiliation(s)
- Chen Huang
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Basic Medicine Experimental Teaching Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Pei-Yi Sun
- Department of Dermatology, Xinhua Hospital, Institute of Dermatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yiming Jiang
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Otorhinolaryngology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yuandong Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Zhichao Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Shao-Ling Han
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bao-Shan Wang
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yong-Xin Huang
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - An-Ran Ren
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian-Fei Lu
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qin Jiang
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Li
- Basic Medicine Experimental Teaching Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhirong Yao
- Department of Dermatology, Xinhua Hospital, Institute of Dermatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Xin Qi
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wei-Guang Li
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
- Ministry of Education-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| | - Tian-Le Xu
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
9
|
Negm A, Stobbe K, Ben Fradj S, Sanchez C, Landra-Willm A, Richter M, Fleuriot L, Debayle D, Deval E, Lingueglia E, Rovere C, Noel J. Acid-sensing ion channel 3 mediates pain hypersensitivity associated with high-fat diet consumption in mice. Pain 2024; 165:470-486. [PMID: 37733484 DOI: 10.1097/j.pain.0000000000003030] [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: 09/19/2022] [Accepted: 07/07/2023] [Indexed: 09/23/2023]
Abstract
ABSTRACT Lipid-rich diet is the major cause of obesity, affecting 13% of the worldwide adult population. Obesity is a major risk factor for metabolic syndrome that includes hyperlipidemia and diabetes mellitus. The early phases of metabolic syndrome are often associated with hyperexcitability of peripheral small diameter sensory fibers and painful diabetic neuropathy. Here, we investigated the effect of high-fat diet-induced obesity on the activity of dorsal root ganglion (DRG) sensory neurons and pain perception. We deciphered the underlying cellular mechanisms involving the acid-sensing ion channel 3 (ASIC3). We show that mice made obese through consuming high-fat diet developed the metabolic syndrome and prediabetes that was associated with heat pain hypersensitivity, whereas mechanical sensitivity was not affected. Concurrently, the slow conducting C fibers in the skin of obese mice showed increased activity on heating, whereas their mechanosensitivity was not altered. Although ASIC3 knockout mice fed with high-fat diet became obese, and showed signs of metabolic syndrome and prediabetes, genetic deletion, and in vivo pharmacological inhibition of ASIC3, protected mice from obesity-induced thermal hypersensitivity. We then deciphered the mechanisms involved in the heat hypersensitivity of mice and found that serum from high-fat diet-fed mice was enriched in lysophosphatidylcholine (LPC16:0, LPC18:0, and LPC18:1). These enriched lipid species directly increased the activity of DRG neurons through activating the lipid sensitive ASIC3 channel. Our results identify ASIC3 channel in DRG neurons and circulating lipid species as a mechanism contributing to the hyperexcitability of nociceptive neurons that can cause pain associated with lipid-rich diet consumption and obesity.
Collapse
Affiliation(s)
- Ahmed Negm
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Katharina Stobbe
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Selma Ben Fradj
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Clara Sanchez
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Arnaud Landra-Willm
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Margaux Richter
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | | | | | - Emmanuel Deval
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Eric Lingueglia
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Carole Rovere
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Jacques Noel
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| |
Collapse
|
10
|
Qiao C, He M, Wang S, Jiang X, Wang F, Li X, Tan S, Chao Z, Xin W, Gao S, Yuan J, Li Q, Xu Z, Zheng X, Zhao J, Liu G. Multi-omics analysis reveals substantial linkages between the oral-gut microbiomes and inflamm-aging molecules in elderly pigs. Front Microbiol 2023; 14:1250891. [PMID: 37789859 PMCID: PMC10542583 DOI: 10.3389/fmicb.2023.1250891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/17/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction The accelerated aging of the global population has emerged as a critical public health concern, with increasing recognition of the influential role played by the microbiome in shaping host well-being. Nonetheless, there remains a dearth of understanding regarding the functional alterations occurring within the microbiota and their intricate interactions with metabolic pathways across various stages of aging. Methods This study employed a comprehensive metagenomic analysis encompassing saliva and stool samples obtained from 45 pigs representing three distinct age groups, alongside serum metabolomics and lipidomics profiling. Results Our findings unveiled discernible modifications in the gut and oral microbiomes, serum metabolome, and lipidome at each age stage. Specifically, we identified 87 microbial species in stool samples and 68 in saliva samples that demonstrated significant age-related changes. Notably, 13 species in stool, including Clostridiales bacterium, Lactobacillus johnsonii, and Oscillibacter spp., exhibited age-dependent alterations, while 15 salivary species, such as Corynebacterium xerosis, Staphylococcus sciuri, and Prevotella intermedia, displayed an increase with senescence, accompanied by a notable enrichment of pathogenic organisms. Concomitant with these gut-oral microbiota changes were functional modifications observed in pathways such as cell growth and death (necroptosis), bacterial infection disease, and aging (longevity regulating pathway) throughout the aging process. Moreover, our metabolomics and lipidomics analyses unveiled the accumulation of inflammatory metabolites or the depletion of beneficial metabolites and lipids as aging progressed. Furthermore, we unraveled a complex interplay linking the oral-gut microbiota with serum metabolites and lipids. Discussion Collectively, our findings illuminate novel insights into the potential contributions of the oral-gut microbiome and systemic circulating metabolites and lipids to host lifespan and healthy aging.
Collapse
Affiliation(s)
- Chuanmin Qiao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
- Hainan Yazhou Bay Seed Laboratory, Sanya, China
| | - Maozhang He
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Shumei Wang
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinjie Jiang
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Feng Wang
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Xinjian Li
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Shuyi Tan
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Zhe Chao
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Wenshui Xin
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Shuai Gao
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Jingli Yuan
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Qiang Li
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Zichun Xu
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Xinli Zheng
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| | - Jianguo Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guangliang Liu
- Institute of Animal Science and Veterinary Medicine, Academy of Agricultural Sciences, Haikou, China
| |
Collapse
|
11
|
Timm T, Hild C, Liebisch G, Rickert M, Lochnit G, Steinmeyer J. Functional Characterization of Lysophospholipids by Proteomic and Lipidomic Analysis of Fibroblast-like Synoviocytes. Cells 2023; 12:1743. [PMID: 37443777 PMCID: PMC10340184 DOI: 10.3390/cells12131743] [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: 06/01/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Synovial fluid (SF) from human knee joints with osteoarthritis (OA) has elevated levels of lysophosphatidylcholine (LPC) species, but their functional role is not well understood. This in vitro study was designed to test the hypothesis that various LPCs found elevated in OA SF and their metabolites, lysophosphatidic acids (LPAs), modulate the abundance of proteins and phospholipids (PLs) in human fibroblast-like synoviocytes (FLSs), with even minute chemical variations in lysophospholipids determining the extent of regulation. Cultured FLSs (n = 5-7) were treated with one of the LPC species, LPA species, IL-1β, or a vehicle. Tandem mass tag peptide labeling coupled with LC-MS/MS/MS was performed to quantify proteins. The expression of mRNA from regulated proteins was analyzed using RT-PCR. PL synthesis was determined via ESI-MS/MS, and the release of radiolabeled PLs was determined by means of liquid scintillation counting. In total, 3960 proteins were quantified using multiplexed MS, of which 119, 8, and 3 were significantly and reproducibly regulated by IL-1β, LPC 16:0, and LPC 18:0, respectively. LPC 16:0 significantly inhibited the release of PLs and the synthesis of phosphatidylcholine, LPC, and sphingomyelin. Neither LPC metabolite-LPA 16:0 nor LPA 18:0-had any reproducible effect on the levels of each protein. In conclusion, small chemical variations in LPC species can result in the significantly altered expression and secretion of proteins and PLs from FLSs. IL-1β influenced all proteins that were reproducibly regulated by LPC 16:0. LPC species are likely to modulate FLS protein expression only in more advanced OA stages with low IL-1β levels. None of the eight proteins being significantly regulated by LPC 16:0 have been previously reported in OA. However, our in vitro findings show that the CD81 antigen, calumenin, and B4E2C1 are promising candidates for further study, focusing in particular on their potential ability to modulate inflammatory and catabolic mechanisms.
Collapse
Affiliation(s)
- Thomas Timm
- Protein Analytics Group, Institute of Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Christiane Hild
- Laboratory for Experimental Orthopedics, Department of Orthopedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Gerhard Liebisch
- Department for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Markus Rickert
- Laboratory for Experimental Orthopedics, Department of Orthopedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Guenter Lochnit
- Protein Analytics Group, Institute of Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Juergen Steinmeyer
- Laboratory for Experimental Orthopedics, Department of Orthopedics, Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
12
|
Hung CH, Chin Y, Fong YO, Lee CH, Han DS, Lin JH, Sun WH, Chen CC. Acidosis-related pain and its receptors as targets for chronic pain. Pharmacol Ther 2023; 247:108444. [PMID: 37210007 DOI: 10.1016/j.pharmthera.2023.108444] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Sensing acidosis is an important somatosensory function in responses to ischemia, inflammation, and metabolic alteration. Accumulating evidence has shown that acidosis is an effective factor for pain induction and that many intractable chronic pain diseases are associated with acidosis signaling. Various receptors have been known to detect extracellular acidosis and all express in the somatosensory neurons, such as acid sensing ion channels (ASIC), transient receptor potential (TRP) channels and proton-sensing G-protein coupled receptors. In addition to sense noxious acidic stimulation, these proton-sensing receptors also play a vital role in pain processing. For example, ASICs and TRPs are involved in not only nociceptive activation but also anti-nociceptive effects as well as some other non-nociceptive pathways. Herein, we review recent progress in probing the roles of proton-sensing receptors in preclinical pain research and their clinical relevance. We also propose a new concept of sngception to address the specific somatosensory function of acid sensation. This review aims to connect these acid-sensing receptors with basic pain research and clinical pain diseases, thus helping with better understanding the acid-related pain pathogenesis and their potential therapeutic roles via the mechanism of acid-mediated antinociception.
Collapse
Affiliation(s)
- Chih-Hsien Hung
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin Chin
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-On Fong
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Der-Shen Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Jiann-Her Lin
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
13
|
Khoury S, Colas J, Breuil V, Kosek E, Ahmed AS, Svensson CI, Marchand F, Deval E, Ferreira T. Identification of Lipid Biomarkers for Chronic Joint Pain Associated with Different Joint Diseases. Biomolecules 2023; 13:biom13020342. [PMID: 36830710 PMCID: PMC9953120 DOI: 10.3390/biom13020342] [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: 01/02/2023] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Lipids, especially lysophosphatidylcholine LPC16:0, have been shown to be involved in chronic joint pain through the activation of acid-sensing ion channels (ASIC3). The aim of the present study was to investigate the lipid contents of the synovial fluids from controls and patients suffering from chronic joint pain in order to identify characteristic lipid signatures associated with specific joint diseases. For this purpose, lipids were extracted from the synovial fluids and analyzed by mass spectrometry. Lipidomic analyses identified certain choline-containing lipid classes and molecular species as biomarkers of chronic joint pain, regardless of the pathology, with significantly higher levels detected in the patient samples. Moreover, correlations were observed between certain lipid levels and the type of joint pathologies. Interestingly, LPC16:0 levels appeared to correlate with the metabolic status of patients while other choline-containing lipids were more specifically associated with the inflammatory state. Overall, these data point at selective lipid species in synovial fluid as being strong predictors of specific joint pathologies which could help in the selection of the most adapted treatment.
Collapse
Affiliation(s)
- Spiro Khoury
- Université de Poitiers, Laboratoire Lipotoxicity and Channelopathies (LiTch)—ConicMeds, 86073 Poitiers, France
- Correspondence:
| | - Jenny Colas
- Université de Poitiers, Laboratoire Lipotoxicity and Channelopathies (LiTch)—ConicMeds, 86073 Poitiers, France
- Université de Poitiers, Laboratoire PRéTI, 86073 Poitiers, France
| | - Véronique Breuil
- Université Côte d’Azur (UCA), UMR E-4320 MATOs CEA/iBEB/SBTN, Faculté de Médecine, CEDEX 2, 06107 Nice, France
- Service de Rhumatologie, Hôpital Pasteur, CHU de Nice, 06000 Nice, France
| | - Eva Kosek
- Department of Clinical Neuroscience, Karolinska Institutet, 17165 Solna, Sweden
- Department of Surgical Sciences, Uppsala University, 75185 Uppsala, Sweden
| | - Aisha S. Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17165 Solna, Sweden
| | - Fabien Marchand
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, 63001 Clermont-Ferrand, France
| | - Emmanuel Deval
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, 06560 Valbonne, France
| | - Thierry Ferreira
- Université de Poitiers, Laboratoire Lipotoxicity and Channelopathies (LiTch)—ConicMeds, 86073 Poitiers, France
- Université de Poitiers, Laboratoire PRéTI, 86073 Poitiers, France
| |
Collapse
|
14
|
Klipp RC, Bankston JR. A loosely coordinated interaction site for arachidonic acid on ASICs. J Gen Physiol 2023; 155:213849. [PMID: 36723670 PMCID: PMC9929933 DOI: 10.1085/jgp.202213307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Molecular dynamics simulations reveal a putative interaction surface for PUFAs on TM1 of ASICs that is not tightly conserved between isoforms.
Collapse
Affiliation(s)
- Robert C. Klipp
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John R. Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA,Correspondence to John R. Bankston:
| |
Collapse
|
15
|
Zhang Z, Zhang C, Li Y, Wang C, Yu Q. Lipid and metabolic alteration involvement in physiotherapy for chronic nonspecific low back pain. Lipids Health Dis 2022; 21:125. [PMID: 36434687 PMCID: PMC9700977 DOI: 10.1186/s12944-022-01737-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Chronic nonspecific low back pain (cNLBP) is a common health problem worldwide, affecting 65-80% of the population and greatly affecting people's quality of life and productivity. It also causes huge economic losses. Manual therapy (MT) and therapeutic exercise (TE) are effective treatment options for cNLBP physiotherapy-based treatment. However, the underlying mechanisms that promote cNLBP amelioration by MT or TE are incompletely understood. METHODS Seventeen recruited subjects were randomly divided into an MT group and a TE group. Subjects in the MT group performed muscular relaxation, myofascial release, and mobilization for 20 min during each treatment session. The treatment lasted for a total of six sessions, once every two days. Subjects in the TE group completed motor control and core stability exercises for 30 min during each treatment session. The motor control exercise included stretching of the trunk and extremity muscles through trunk and hip rotation and flexion training. Stabilization exercises consisted of the (1) bridge exercise, (2) single-leg-lift bridge exercise, (3) side bridge exercise, (4) two-point bird-dog position with an elevated contralateral leg and arm, (5) bear crawl exercise, and (6) dead bug exercise. The treatment lasted for a total of six sessions, with one session every two days. Serum samples were collected from subjects before and after physiotherapy-based treatment for lipidomic and metabolomic measurements. RESULTS Through lipidomic analysis, we found that the phosphatidylcholine/phosphatidylethanolamine (PC/PE) ratio decreased and the sphingomyelin/ceramide (SM/Cer) ratio increased in cNLBP patients after MT or TE treatment. In addition, eight metabolites enriched in pyrimidine and purine differed significantly in cNLBP patients who received MT treatment. A total of nine metabolites enriched in pyrimidine, tyrosine, and galactose pathways differed significantly in cNLBP patients after TE treatment during metabolomics analysis. CONCLUSION Our study was the first to elucidate the alterations in the lipidomics and metabolomics of cNLBP physiotherapy-based treatment and can expand our knowledge of cNLBP physiotherapy-based treatment.
Collapse
Affiliation(s)
- Zhou Zhang
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510275 Guangzhou, P. R. China
| | - Chanjuan Zhang
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510275 Guangzhou, P. R. China
| | - Yuelong Li
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510275 Guangzhou, P. R. China
| | - Chuhuai Wang
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510275 Guangzhou, P. R. China
| | - Qiuhua Yu
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510275 Guangzhou, P. R. China
| |
Collapse
|
16
|
Verkest C, Salinas M, Diochot S, Deval E, Lingueglia E, Baron A. Mechanisms of Action of the Peptide Toxins Targeting Human and Rodent Acid-Sensing Ion Channels and Relevance to Their In Vivo Analgesic Effects. Toxins (Basel) 2022; 14:toxins14100709. [PMID: 36287977 PMCID: PMC9612379 DOI: 10.3390/toxins14100709] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/16/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are voltage-independent H+-gated cation channels largely expressed in the nervous system of rodents and humans. At least six isoforms (ASIC1a, 1b, 2a, 2b, 3 and 4) associate into homotrimers or heterotrimers to form functional channels with highly pH-dependent gating properties. This review provides an update on the pharmacological profiles of animal peptide toxins targeting ASICs, including PcTx1 from tarantula and related spider toxins, APETx2 and APETx-like peptides from sea anemone, and mambalgin from snake, as well as the dimeric protein snake toxin MitTx that have all been instrumental to understanding the structure and the pH-dependent gating of rodent and human cloned ASICs and to study the physiological and pathological roles of native ASICs in vitro and in vivo. ASICs are expressed all along the pain pathways and the pharmacological data clearly support a role for these channels in pain. ASIC-targeting peptide toxins interfere with ASIC gating by complex and pH-dependent mechanisms sometimes leading to opposite effects. However, these dual pH-dependent effects of ASIC-inhibiting toxins (PcTx1, mambalgin and APETx2) are fully compatible with, and even support, their analgesic effects in vivo, both in the central and the peripheral nervous system, as well as potential effects in humans.
Collapse
Affiliation(s)
- Clément Verkest
- CNRS (Centre National de la Recherche Scientifique), 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”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Miguel Salinas
- CNRS (Centre National de la Recherche Scientifique), 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”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Sylvie Diochot
- CNRS (Centre National de la Recherche Scientifique), 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”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Emmanuel Deval
- CNRS (Centre National de la Recherche Scientifique), 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”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Eric Lingueglia
- CNRS (Centre National de la Recherche Scientifique), 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”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Anne Baron
- CNRS (Centre National de la Recherche Scientifique), 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”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
- Correspondence:
| |
Collapse
|
17
|
Lysophosphatidylcholine: Potential Target for the Treatment of Chronic Pain. Int J Mol Sci 2022; 23:ijms23158274. [PMID: 35955410 PMCID: PMC9368269 DOI: 10.3390/ijms23158274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/26/2022] Open
Abstract
The bioactive lipid lysophosphatidylcholine (LPC), a major phospholipid component of oxidized low-density lipoprotein (Ox-LDL), originates from the cleavage of phosphatidylcholine by phospholipase A2 (PLA2) and is catabolized to other substances by different enzymatic pathways. LPC exerts pleiotropic effects mediated by its receptors, G protein-coupled signaling receptors, Toll-like receptors, and ion channels to activate several second messengers. Lysophosphatidylcholine (LPC) is increasingly considered a key marker/factor positively in pathological states, especially inflammation and atherosclerosis development. Current studies have indicated that the injury of nervous tissues promotes oxidative stress and lipid peroxidation, as well as excessive accumulation of LPC, enhancing the membrane hyperexcitability to induce chronic pain, which may be recognized as one of the hallmarks of chronic pain. However, findings from lipidomic studies of LPC have been lacking in the context of chronic pain. In this review, we focus in some detail on LPC sources, biochemical pathways, and the signal-transduction system. Moreover, we outline the detection methods of LPC for accurate analysis of each individual LPC species and reveal the pathophysiological implication of LPC in chronic pain, which makes it an interesting target for biomarkers and the development of medicine regarding chronic pain.
Collapse
|
18
|
Pidoux L, Delanoe K, Barbier J, Marchand F, Lingueglia E, Deval E. Single Subcutaneous Injection of Lysophosphatidyl-Choline Evokes ASIC3-Dependent Increases of Spinal Dorsal Horn Neuron Activity. Front Mol Neurosci 2022; 15:880651. [PMID: 35774865 PMCID: PMC9239072 DOI: 10.3389/fnmol.2022.880651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022] Open
Abstract
Lysophosphatidyl-choline (LPC), a member of the phospholipid family, is an emerging player in pain. It is known to modulate different pain-related ion channels, including Acid-Sensing Ion Channel 3 (ASIC3), a cationic channel mainly expressed in peripheral sensory neurons. LPC potentiates ASIC3 current evoked by mild acidifications, but can also activate the channel at physiological pH. Very recently, LPC has been associated to chronic pain in patients suffering from fibromyalgia or osteoarthritis. Accordingly, repetitive injections of LPC within mouse muscle or joint generate both persistent pain-like and anxiety-like behaviors in an ASIC3-dependent manner. LPC has also been reported to generate acute pain behaviors when injected intraplantarly in rodents. Here, we explore the mechanism of action of a single cutaneous injection of LPC by studying its effects on spinal dorsal horn neurons. We combine pharmacological, molecular and functional approaches including in vitro patch clamp recordings and in vivo recordings of spinal neuronal activity. We show that a single cutaneous injection of LPC exclusively affects the nociceptive pathway, inducing an ASIC3-dependent sensitization of nociceptive fibers that leads to hyperexcitabilities of both high threshold (HT) and wide dynamic range (WDR) spinal neurons. ASIC3 is involved in LPC-induced increase of WDR neuron’s windup as well as in WDR and HT neuron’s mechanical hypersensitivity, and it participates, together with TRPV1, to HT neuron’s thermal hypersensitivity. The nociceptive input induced by a single LPC cutaneous rather induces short-term sensitization, contrary to previously described injections in muscle and joint. If the effects of peripheral LPC on nociceptive pathways appear to mainly depend on peripheral ASIC3 channels, their consequences on pain may also depend on the tissue injected. Our findings contribute to a better understanding of the nociceptive signaling pathway activated by peripheral LPC via ASIC3 channels, which is an important step regarding the ASIC3-dependent roles of this phospholipid in acute and chronic pain conditions.
Collapse
Affiliation(s)
- Ludivine Pidoux
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
| | - Kevin Delanoe
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
| | - Julie Barbier
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Fabien Marchand
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Eric Lingueglia
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
| | - Emmanuel Deval
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
- *Correspondence: Emmanuel Deval,
| |
Collapse
|