1
|
Neyama H, Dozono N, Sahara Y, Nishikawa K, Ueda H. A novel lysophosphatidic acid receptor 1 antagonist with high brain penetrability has a curative effect in the empathic pain-related fibromyalgia model. J Pharmacol Sci 2025; 158:139-142. [PMID: 40288824 DOI: 10.1016/j.jphs.2025.03.012] [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: 02/06/2025] [Revised: 03/15/2025] [Accepted: 03/29/2025] [Indexed: 04/29/2025] Open
Abstract
Lysophosphatidic acid receptor 1 (LPA1) plays a pivotal role in the pathophysiology of various diseases, especially chronic pain. In this study, we assessed the biochemical properties of Compound A, a novel LPA1 antagonist and its beneficial effects in the fibromyalgia (FM)-like pain model. Compound A was found to be a high-affinity and selective LPA1 antagonist and have high brain penetrability. Repeated oral administrations of Compound A reversed the hyperalgesia as late as 9 days after the treatments, suggesting this compound has a curative effect in the FM model.
Collapse
Affiliation(s)
- Hiroyuki Neyama
- Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8521, Japan; Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Naoki Dozono
- Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8521, Japan
| | - Yasuka Sahara
- Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8521, Japan
| | - Kenji Nishikawa
- Pharmaceutical Research Laboratory, Pharmaceutical Division, UBE Corporation, Yamaguchi, 755-8633, Japan
| | - Hiroshi Ueda
- Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8521, Japan; Graduate Institute of Pharmacology, National Defense Medical Center, Taiwan, 114201, Taipei, China; Laboratory for the Study of Pain, Research Institute for Production Development, Kyoto, 606-0805, Japan.
| |
Collapse
|
2
|
Chen N, Tu Y, Liu DQ, Zhang Y, Tian YK, Zhou YQ, Yang SB. Exploring the Role of RhoA/ROCK Signaling in Pain: A Narrative Review. Aging Dis 2025:AD.2024.1539. [PMID: 40249935 DOI: 10.14336/ad.2024.1539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/13/2025] [Indexed: 04/20/2025] Open
Abstract
Despite significant progress in understanding the mechanisms of pain and developing therapeutic agents, pain remains a challenging and unresolved clinical issue. The Ras homolog gene family member A (RhoA), a member of the small guanosine triphosphate hydrolases (GTPases) of the Ras homolog family, is involved in transmitting signals that regulate various cellular processes. RhoA exerts its effects through a range of downstream effectors, with Rho-associated kinase (ROCK) being the most extensively studied. Emerging evidence suggests that the RhoA/ROCK signaling pathway plays a crucial role in pain transmission and sensitization. Our work indicates that targeting the RhoA/ROCK signaling pathway may offer a promising therapeutic avenue for alleviating pain.
Collapse
|
3
|
Yosipovitch G, Kim B, Luger T, Lerner E, Metz M, Adiri R, Canosa JM, Cha A, Ständer S. Similarities and differences in peripheral itch and pain pathways in atopic dermatitis. J Allergy Clin Immunol 2024; 153:904-912. [PMID: 38103700 DOI: 10.1016/j.jaci.2023.10.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/22/2023] [Accepted: 10/12/2023] [Indexed: 12/19/2023]
Abstract
Atopic dermatitis (AD) is predominantly characterized by intense itching, but concomitant skin pain is experienced by more than 40% of patients. Patients with AD display considerable somatosensory aberrations, including increased nerve sensitivity to itch stimuli (hyperknesis), perception of itch from innocuous stimuli (alloknesis), or perception of pain from innocuous stimuli (allodynia). This review summarizes the current understanding of the similarities and differences in the peripheral mechanisms underlying itch and pain in AD. These distinct yet reciprocal sensations share many similarities in the peripheral nervous system, including common mediators (such as serotonin, endothelin-1, IL-33, and thymic stromal lymphopoietin), receptors (such as members of the G protein-coupled receptor family and Toll-like receptors), and ion channels for signal transduction (such as certain members of the transient receptor potential [TRP] cation channels). Itch-responding neurons are also sensitive to pain stimuli. However, there are distinct differences between itch and pain signaling. For example, specific immune responses are associated with pain (type 1 and/or type 3 cytokines and certain chemokine C-C [CCL2, CCL5] and C-X-C [CXCL] motif ligands) and itch (type 2 cytokines, including IL-31, and periostin). The TRP melastatin channels TRPM2 and TRPM3 have a role in pain but no known role in itch. Activation of μ-opioid receptors is known to alleviate pain but exacerbate itch. Understanding the connection between itch and pain mechanisms may offer new insights into the treatment of chronic pain and itch in AD.
Collapse
Affiliation(s)
- Gil Yosipovitch
- Miami Itch Center, Miller School of Medicine, University of Miami, Miami, Fla.
| | - Brian Kim
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St Louis, Mo
| | | | - Ethan Lerner
- Massachusetts General Hospital, Charlestown, Mass
| | - Martin Metz
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Roni Adiri
- Pfizer Pharmaceuticals Israel Ltd, Herzliya Pituach, Israel
| | | | | | - Sonja Ständer
- Center for Chronic Pruritus, Münster University Hospital, Münster, Germany
| |
Collapse
|
4
|
Sumitani M, Kimura A, Mochizuki T, Akiyama T, Uranbileg B, Takahashi T, Hirai T, Hayakawa K, Chikuda H, Kurano M. Cerebrospinal Fluid Lysophosphatidylcholine Species for Distinguishing Narrowing of the Lumbar Spine. World Neurosurg 2024; 183:e571-e575. [PMID: 38181872 DOI: 10.1016/j.wneu.2023.12.148] [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: 12/25/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Reoperation, sometimes multiple, is common with progressively worse outcomes in patients with degenerative lumbar spine diseases. Lysophosphatidylcholine (LPC), a precursor of lysophosphatidic acid, in the cerebrospinal fluid (CSF) is a possible biomarker for neuropathic pain and discriminating neuropathic pain caused by lumbar spinal canal stenosis (LSCS) from other etiologies. This study aimed to explore this possible use of LPC species in the CSF. METHODS Patients with LSCS (n = 137) and persistent spinal pain syndrome (n = 22) were subjected in this multi-site observational study. The CSF was collected by lumbar puncture. Using liquid chromatography-tandem mass spectrometry, we measured 6 LPC species, (16:0), (18:0), (18:1), (18:2), (20:4), and (22:6), in the CSF. We compared the LPC values between the groups and determined the cutoff levels that could efficiently discriminate the groups with high accuracy. RESULTS The levels of all measured LPC species were significantly higher in the LSCS group than the persistent spinal pain syndrome group. Four LPC species demonstrated more than 0.80 area under the curve obtained from the receiver operating characteristic curve analysis. Although the specificity of cutoff levels for the 6 LPC species was low to moderate, their sensitivity was consistently high. CONCLUSIONS The existing diagnostic protocols combining physical examinations and morphological imaging studies for lumbar spinal pain have limited sensitivity. Measuring LPC species in the CSF is a promising objective laboratory test and could be suitable for detecting the presence of lumbar spinal stenosis and can help indications for surgery.
Collapse
Affiliation(s)
- Masahiko Sumitani
- Department of Pain and Palliative Medicine, The University of Tokyo Hospital, Tokyo, Japan.
| | - Atsushi Kimura
- Department of Orthopaedic Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Takao Mochizuki
- Department of Orthopaedic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Toru Akiyama
- Department of Orthopaedic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Takuya Takahashi
- Department of Orthopaedic and Spinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takashi Hirai
- Department of Orthopaedic and Spinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kentaro Hayakawa
- Department of Orthopaedics and Spine Surgery, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Hirotaka Chikuda
- Department of Orthopaedic Surgery, Gunma University, Maebashi, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
5
|
Ueda H, Neyama H. Fibromyalgia Animal Models Using Intermittent Cold and Psychological Stress. Biomedicines 2023; 12:56. [PMID: 38255163 PMCID: PMC10813244 DOI: 10.3390/biomedicines12010056] [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: 11/14/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Fibromyalgia (FM) is a chronic pain condition characterized by widespread musculoskeletal pain and other frequent symptoms such as fatigue, sleep disturbance, cognitive impairment, and mood disorder. Based on the view that intermittent stress would be the most probable etiology for FM, intermittent cold- and intermittent psychological stress-induced generalized pain (ICGP and IPGP) models in mice have been developed and validated as FM-like pain models in terms of the patho-physiological and pharmacotherapeutic features that are shared with clinical versions. Both models show long-lasting and generalized pain and female-predominant sex differences after gonadectomy. Like many other neuropathic pain models, ICGP and IPGP were abolished in lysophosphatidic acid receptor 1 (LPAR1) knock-out mice or by LPAR1 antagonist treatments, although deciding the clinical importance of this mechanism depends on waiting for the development of a clinically available LPAR1 antagonist. On the other hand, the nonsteroidal anti-inflammatory drug diclofenac with morphine did not suppress hyperalgesia in these models, and this is consistent with the clinical findings. Pharmacological studies suggest that the lack of morphine analgesia is associated with opioid tolerance upon the stress-induced release of endorphins and subsequent counterbalance through anti-opioid NMDA receptor mechanisms. Regarding pharmacotherapy, hyperalgesia in both models was suppressed by pregabalin and duloxetine, which have been approved for FM treatment in clinic. Notably, repeated treatments with mirtazapine, an α2 adrenergic receptor antagonist-type antidepressant, and donepezil, a drug for treating Alzheimer's disease, showed potent therapeutic actions in these models. However, the pharmacotherapeutic treatment should be carried out 3 months after stress, which is stated in the FM guideline, and many preclinical studies, such as those analyzing molecular and cellular mechanisms, as well as additional evidence using different animal models, are required. Thus, the ICGP and IPGP models have the potential to help discover and characterize new therapeutic medicines that might be used for the radical treatment of FM, although there are several limitations to be overcome.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan;
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114201, Taiwan
| | - Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan;
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
6
|
Ono T, Yamashita T, Kano R, Inoue M, Okada S, Kano K, Koizumi S, Iwabuchi K, Hirabayashi Y, Matsuo I, Nakashima Y, Kamiguchi H, Kohro Y, Tsuda M. GPR55 contributes to neutrophil recruitment and mechanical pain induction after spinal cord compression in mice. Brain Behav Immun 2023; 110:276-287. [PMID: 36898418 DOI: 10.1016/j.bbi.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Pain transmission and processing in the nervous system are modulated by various biologically active substances, including lysophospholipids, through direct and indirect actions on the somatosensory pathway. Lysophosphatidylglucoside (LysoPtdGlc) was recently identified as a structurally unique lysophospholipid that exerts biological actions via the G protein-coupled receptor GPR55. Here, we demonstrated that GPR55-knockout (KO) mice show impaired induction of mechanical pain hypersensitivity in a model of spinal cord compression (SCC) without the same change in the models of peripheral tissue inflammation and peripheral nerve injury. Among these models, only SCC recruited peripheral inflammatory cells (neutrophils, monocytes/macrophages, and CD3+ T-cells) in the spinal dorsal horn (SDH), and GPR55-KO blunted these recruitments. Neutrophils were the first cells recruited to the SDH, and their depletion suppressed the induction of SCC-induced mechanical hypersensitivity and inflammatory responses in compressed SDH. Furthermore, we found that PtdGlc was present in the SDH and that intrathecal administration of an inhibitor of secretory phospholipase A2 (an enzyme required for producing LysoPtdGlc from PtdGlc) reduced neutrophil recruitment to compressed SDH and suppressed pain induction. Finally, by screening compounds from a chemical library, we identified auranofin as a clinically used drug with an inhibitory effect on mouse and human GPR55. Systemically administered auranofin to mice with SCC effectively suppressed spinal neutrophil infiltration and pain hypersensitivity. These results suggest that GPR55 signaling contributes to the induction of inflammatory responses and chronic pain after SCC via the recruitment of neutrophils and may provide a new target for reducing pain induction after spinal cord compression, such as spinal canal stenosis.
Collapse
Affiliation(s)
- Teruaki Ono
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomohiro Yamashita
- Department of Drug Discovery Structural Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryota Kano
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mariko Inoue
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shota Okada
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Koki Kano
- Graduate School of Science and Technology, Gunma University, 1-5-1, Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kazuhisa Iwabuchi
- Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan
| | - Yoshio Hirabayashi
- Cellular Informatics Laboratory, RIKEN, 2-1, Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ichiro Matsuo
- Graduate School of Science and Technology, Gunma University, 1-5-1, Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hiroyuki Kamiguchi
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yuta Kohro
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
7
|
Zhang D, Decker AM, Woodhouse K, Snyder R, Patel P, Harris DL, Tao YX, Li JX, Zhang Y. Isoquinolone derivatives as lysophosphatidic acid receptor 5 (LPA5) antagonists: Investigation of structure-activity relationships, ADME properties and analgesic effects. Eur J Med Chem 2022; 243:114741. [PMID: 36126387 PMCID: PMC10155261 DOI: 10.1016/j.ejmech.2022.114741] [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: 07/07/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 11/18/2022]
Abstract
Blockade of lysophosphatidic acid receptor 5 (LPA5) by a recently reported antagonist AS2717638 (2) attenuated inflammatory and neuropathic pains, although it showed moderate in vivo efficacy and its structure-activity relationships and the ADME properties are little studied. We therefore designed and synthesized a series of isoquinolone derivatives and evaluated their potency in LPA5 calcium mobilization and cAMP assays. Our results show that substituted phenyl groups or bicyclic aromatic rings such as benzothiophenes or benzofurans are tolerated at the 2-position, 4-substituted piperidines are favored at the 4-position, and methoxy groups at the 6- and 7-positions are essential for activity. Compounds 65 and 66 showed comparable in vitro potency, excellent selectivity against LPA1-LPA4 and >50 other GPCRs, moderate metabolic stability, and high aqueous solubility and brain permeability. Both 65 and 66 significantly attenuated nociceptive hypersensitivity at lower doses than 2 and had longer-lasting effects in an inflammatory pain model, and 66 also dose-dependently reduced mechanical allodynia in the chronic constriction injury model and opioid-induced hyperalgesia at doses that had no effect on the locomotion in rats. These results suggest that these isoquinolone derivatives as LPA5 antagonists are of promise as potential analgesics.
Collapse
Affiliation(s)
- Dehui Zhang
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Ann M Decker
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Kristen Woodhouse
- Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, Buffalo, NY, 14203, USA
| | - Rodney Snyder
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Purvi Patel
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Danni L Harris
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, Buffalo, NY, 14203, USA
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
8
|
Langedijk J, Araya EI, Barroso AR, Tolenaars D, Nazaré M, Belabed H, Schoene J, Chichorro JG, Oude Elferink R. An LPAR5-antagonist that reduces nociception and increases pruriception. FRONTIERS IN PAIN RESEARCH 2022; 3:963174. [PMID: 35959236 PMCID: PMC9360597 DOI: 10.3389/fpain.2022.963174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction The G-protein coupled receptor LPAR5 plays a prominent role in LPA-mediated pain and itch signaling. In this study we focus on the LPAR5-antagonist compound 3 (cpd3) and its ability to affect pain and itch signaling, both in vitro and in vivo. Methods Nociceptive behavior in wild type mice was induced by formalin, carrageenan or prostaglandin E2 (PGE2) injection in the hind paw, and the effect of oral cpd3 administration was measured. Scratch activity was measured after oral administration of cpd3, in mice overexpressing phospholipase A2 (sPLA2tg), in wild type mice (WT) and in TRPA1-deficient mice (Trpa1 KO). In vitro effects of cpd3 were assessed by measuring intracellular calcium release in HMC-1 and HEK-TRPA1 cells. Results As expected, nociceptive behavior (induced by formalin, carrageenan or PGE2) was reduced after treatment with cpd3. Unexpectedly, cpd3 induced scratch activity in mice. In vitro addition of cpd3 to HEK-TRPA1 cells induced an intracellular calcium wave that could be inhibited by the TRPA1-antagonist A-967079. In Trpa1 KO mice, however, the increase in scratch activity after cpd3 administration was not reduced. Conclusions Cpd3 has in vivo antinociceptive effects but induces scratch activity in mice, probably by activation of multiple pruriceptors, including TRPA1. These results urge screening of antinociceptive candidate drugs for activity with pruriceptors.
Collapse
Affiliation(s)
- Jacqueline Langedijk
- Amsterdam University Medical Centers (UMC), Tytgat Institute for Liver and Intestinal Research, University of Amsterdam, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism (AG&M), Amsterdam, Netherlands
| | - Erika Ivanna Araya
- Department of Pharmacology, Biological Sciences Sector, Federal University of Parana, Curitiba, Brazil
| | - Amanda Ribeiro Barroso
- Department of Pharmacology, Biological Sciences Sector, Federal University of Parana, Curitiba, Brazil
| | - Dagmar Tolenaars
- Amsterdam University Medical Centers (UMC), Tytgat Institute for Liver and Intestinal Research, University of Amsterdam, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism (AG&M), Amsterdam, Netherlands
| | - Marc Nazaré
- Departments of Chemical Biology and Structural Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Hassane Belabed
- Departments of Chemical Biology and Structural Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Jens Schoene
- Departments of Chemical Biology and Structural Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Ronald Oude Elferink
- Amsterdam University Medical Centers (UMC), Tytgat Institute for Liver and Intestinal Research, University of Amsterdam, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism (AG&M), Amsterdam, Netherlands
- *Correspondence: Ronald Oude Elferink
| |
Collapse
|
9
|
Neyama H, Nishiyori M, Cui Y, Watanabe Y, Ueda H. Lysophosphatidic acid receptor type-1 mediates brain activation in micro-Positron Emission Tomography analysis in a fibromyalgia-like mouse model. Eur J Neurosci 2022; 56:4224-4233. [PMID: 35666711 DOI: 10.1111/ejn.15729] [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: 11/15/2021] [Accepted: 05/30/2022] [Indexed: 11/26/2022]
Abstract
The intermittent cold stress-induced generalized pain response mimics the pathophysiological and pharmacotherapeutic features reported for fibromyalgia patients, including the presence of chronic generalized pain and female dominance. In addition, the intermittent cold stress-induced generalized pain is abolished in lysophosphatidic acid receptor type-1 knockout mice, as reported in many cases of neuropathic pain models. This study aimed to identify the brain loci involved in the intermittent cold stress generalized pain response and test their dependence on the lysophosphatidic acid receptor type-1. Positron emission tomography analyses using 2-deoxy-2-[18 F]fluoro-D-glucose in the presence of a pain stimulus showed that intermittent cold stress causes a significant increase in uptake in the ipsilateral regions, including the salience networking-related anterior cingulate cortex and insular cortex and the cognition-related hippocampus. A significant decrease was observed in the default mode network-related posterior cingulate cortex. Almost these intermittent cold stress-induced changes were abolished in lysophosphatidic acid receptor type-1 knockout mice. There results suggest that the intermittent cold stress-induced generalized pain response is mediated by the lysophosphatidic acid receptor type-1 in specific brain loci related to salience networking and cognition, which may lead to further developments in the treatment of fibromyalgia.
Collapse
Affiliation(s)
- Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Michiko Nishiyori
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Yilong Cui
- Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Laboratory for the Study of Pain, Research Institute for Production Development, Kyoto, Japan
| |
Collapse
|
10
|
Su J, Krock E, Barde S, Delaney A, Ribeiro J, Kato J, Agalave N, Wigerblad G, Matteo R, Sabbadini R, Josephson A, Chun J, Kultima K, Peyruchaud O, Hökfelt T, Svensson CI. Pain-like behavior in the collagen antibody-induced arthritis model is regulated by lysophosphatidic acid and activation of satellite glia cells. Brain Behav Immun 2022; 101:214-230. [PMID: 35026421 DOI: 10.1016/j.bbi.2022.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/14/2021] [Accepted: 01/07/2022] [Indexed: 12/30/2022] Open
Abstract
Inflammatory and neuropathic-like components underlie rheumatoid arthritis (RA)-associated pain, and lysophosphatidic acid (LPA) is linked to both joint inflammation in RA patients and to neuropathic pain. Thus, we investigated a role for LPA signalling using the collagen antibody-induced arthritis (CAIA) model. Pain-like behavior during the inflammatory phase and the late, neuropathic-like phase of CAIA was reversed by a neutralizing antibody generated against LPA and by an LPA1/3 receptor inhibitor, but joint inflammation was not affected. Autotaxin, an LPA synthesizing enzyme was upregulated in dorsal root ganglia (DRG) neurons during both CAIA phases, but not in joints or spinal cord. Late-phase pronociceptive neurochemical changes in the DRG were blocked in Lpar1 receptor deficient mice and reversed by LPA neutralization. In vitro and in vivo studies indicated that LPA regulates pain-like behavior via the LPA1 receptor on satellite glia cells (SGCs), which is expressed by both human and mouse SGCs in the DRG. Furthermore, CAIA-induced SGC activity is reversed by phospholipid neutralization and blocked in Lpar1 deficient mice. Our findings suggest that the regulation of CAIA-induced pain-like behavior by LPA signalling is a peripheral event, associated with the DRGs and involving increased pronociceptive activity of SGCs, which in turn act on sensory neurons.
Collapse
Affiliation(s)
- Jie Su
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Emerson Krock
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ada Delaney
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Jungo Kato
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Nilesh Agalave
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Gustaf Wigerblad
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Roger Sabbadini
- LPath Inc, San Diego, United States; Department of Biology, San Diego State University, 92182, United States
| | - Anna Josephson
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Jerold Chun
- Translational Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States
| | - Kim Kultima
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Medical Sciences, Uppsala University, 75185 Uppsala, Sweden
| | | | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
11
|
Durán AM, Beeson WL, Firek A, Cordero-MacIntyre Z, De León M. Dietary Omega-3 Polyunsaturated Fatty-Acid Supplementation Upregulates Protective Cellular Pathways in Patients with Type 2 Diabetes Exhibiting Improvement in Painful Diabetic Neuropathy. Nutrients 2022; 14:nu14040761. [PMID: 35215418 PMCID: PMC8876723 DOI: 10.3390/nu14040761] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Omega-3 polyunsaturated fatty acids (PUFAs) have been proposed to improve chronic neuroinflammatory diseases in peripheral and central nervous systems. For instance, docosahexaenoic acid (DHA) protects nerve cells from noxious stimuli in vitro and in vivo. Recent reports link PUFA supplementation to improving painful diabetic neuropathy (pDN) symptoms, but cellular mechanisms responsible for this therapeutic effect are not well understood. The objective of this study is to identify distinct cellular pathways elicited by dietary omega-3 PUFA supplementation in patients with type 2 diabetes mellitus (T2DM) affected by pDN. Methods: Forty volunteers diagnosed with type 2 diabetes were enrolled in the “En Balance-PLUS” diabetes education study. The volunteers participated in weekly lifestyle/nutrition education and daily supplementation with 1000 mg DHA and 200 mg eicosapentaenoic acid. The Short-Form McGill Pain Questionnaire validated clinical determination of baseline and post-intervention pain complaints. Laboratory and untargeted metabolomics analyses were conducted using blood plasma collected at baseline and after three months of participation in the dietary regimen. The metabolomics data were analyzed using random forest, hierarchical clustering, ingenuity pathway analysis, and metabolic pathway mapping. Results: The data show that metabolites involved in oxidative stress and glutathione production shifted significantly to a more anti-inflammatory state post supplementation. Example of these metabolites include cystathionine (+90%), S-methylmethionine (+9%), glycine cysteine-glutathione disulfide (+157%) cysteinylglycine (+19%), glutamate (−11%), glycine (+11%), and arginine (+13.4%). In addition, the levels of phospholipids associated with improved membrane fluidity such as linoleoyl-docosahexaenoyl-glycerol (18:2/22:6) (+253%) were significantly increased. Ingenuity pathway analysis suggested several key bio functions associated with omega-3 PUFA supplementation such as formation of reactive oxygen species (p = 4.38 × 10−4, z-score = −1.96), peroxidation of lipids (p = 2.24 × 10−5, z-score = −1.944), Ca2+ transport (p = 1.55 × 10−4, z-score = −1.969), excitation of neurons (p = 1.07 ×10−4, z-score = −1.091), and concentration of glutathione (p = 3.06 × 10−4, z-score = 1.974). Conclusion: The reduction of pro-inflammatory and oxidative stress pathways following dietary omega-3 PUFA supplementation is consistent with the promising role of these fatty acids in reducing adverse symptoms associated with neuroinflammatory diseases and painful neuropathy.
Collapse
Affiliation(s)
- Alfonso M. Durán
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
| | - W. Lawrence Beeson
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA 92350, USA
| | - Anthony Firek
- Comparative Effectiveness and Clinical Outcomes Research Center, Riverside University Health System Medical Center, Moreno Valley, CA 92555, USA;
| | - Zaida Cordero-MacIntyre
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA 92350, USA
| | - Marino De León
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Correspondence: ; Tel.: +1-909-558-9474
| |
Collapse
|
12
|
Ding YQ, Qi JG. Sensory root demyelination: Transforming touch into pain. Glia 2021; 70:397-413. [PMID: 34549463 DOI: 10.1002/glia.24097] [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: 05/24/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 11/12/2022]
Abstract
The normal feeling of touch is vital for nearly every aspect of our daily life. However, touching is not always felt as touch, but also abnormally as pain under numerous diseased conditions. For either mechanistic understanding of the faithful feeling of touch or clinical management of chronic pain, there is an essential need to thoroughly dissect the neuropathological changes that lead to painful touch or tactile allodynia and their corresponding cellular and molecular underpinnings. In recent years, we have seen remarkable progress in our understanding of the neural circuits for painful touch, with an increasing emphasis on the upstream roles of non-neuronal cells. As a highly specialized form of axon ensheathment by glial cells in jawed vertebrates, myelin sheaths not only mediate their outstanding neural functions via saltatory impulse propagation of temporal and spatial precision, but also support long-term neuronal/axonal integrity via metabolic and neurotrophic coupling. Therefore, myelinopathies have been implicated in diverse neuropsychiatric diseases, which are traditionally recognized as a result of the dysfunctions of neural circuits. However, whether myelinopathies can transform touch into pain remains a long-standing question. By summarizing and reframing the fragmentary but accumulating evidence so far, the present review indicates that sensory root demyelination represents a hitherto underappreciated neuropathological change for most neuropathic conditions of painful touch and offers an insightful window into faithful tactile sensation as well as a potential therapeutic target for intractable painful touch.
Collapse
Affiliation(s)
- You-Quan Ding
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jian-Guo Qi
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Tanaka K, Dozono N, Neyama H, Nagai J, Tsukahara R, Nagayasu K, Kaneko S, Ueda H. Secreted PLA 2-III is a possible therapeutic target to treat neuropathic pain. Biochem Biophys Res Commun 2021; 568:167-173. [PMID: 34237486 DOI: 10.1016/j.bbrc.2021.06.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/15/2021] [Indexed: 10/20/2022]
Abstract
Lysophosphatidic acid (LPA) plays a critical role in developing and maintaining chronic pain in various animal models. Previous studies have reported that cytosolic and calcium-independent phospholipase A2 (PLA2) is involved in the LPA receptor-mediated amplification of LPA production in the spinal dorsal horn (SDH) after nerve injury, while the involvement of secreted PLA2 (sPLA2) remains unclear. The present study revealed that only sPLA2 -III among 11 species of PLA2 showed a significant upregulation of gene expression in the SDH. Intraspinal injection of adeno-associated virus-miRNA targeting sPLA2-III prevented hyperalgesia and unique hypoalgesia in mice treated with partial sciatic nerve ligation. In addition, intrathecal treatment with antisense oligodeoxynucleotide or siRNA targeting sPLA2-III significantly reversed the established thermal hyperalgesia. In the high-throughput screening of sPLA2-III inhibitors from the chemical library, we identified two hit compounds. Through in vitro characterization of PLA2 inhibitor profiles and in vivo assessment of the anti-hyperalgesic effects of known PLA2 inhibitors as well as hit compounds, sPLA2-III was found to be a novel therapeutic target molecule for the treatment of Neuropathic pain.
Collapse
Affiliation(s)
- Keigo Tanaka
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, 606-8501, Japan
| | - Naoki Dozono
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, 606-8501, Japan; Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, 852-8521, Japan
| | - Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, 852-8521, Japan; RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Jun Nagai
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, 852-8521, Japan
| | - Ryoko Tsukahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, 852-8521, Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, 606-8501, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, 606-8501, Japan
| | - Hiroshi Ueda
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, 606-8501, Japan; Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, 852-8521, Japan; Laboratory for the Study of Pain, Research Institute for Production Development, Kyoto, 606-0805, Japan.
| |
Collapse
|
14
|
Geraldo LHM, Spohr TCLDS, Amaral RFD, Fonseca ACCD, Garcia C, Mendes FDA, Freitas C, dosSantos MF, Lima FRS. Role of lysophosphatidic acid and its receptors in health and disease: novel therapeutic strategies. Signal Transduct Target Ther 2021; 6:45. [PMID: 33526777 PMCID: PMC7851145 DOI: 10.1038/s41392-020-00367-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidic acid (LPA) is an abundant bioactive phospholipid, with multiple functions both in development and in pathological conditions. Here, we review the literature about the differential signaling of LPA through its specific receptors, which makes this lipid a versatile signaling molecule. This differential signaling is important for understanding how this molecule can have such diverse effects during central nervous system development and angiogenesis; and also, how it can act as a powerful mediator of pathological conditions, such as neuropathic pain, neurodegenerative diseases, and cancer progression. Ultimately, we review the preclinical and clinical uses of Autotaxin, LPA, and its receptors as therapeutic targets, approaching the most recent data of promising molecules modulating both LPA production and signaling. This review aims to summarize the most update knowledge about the mechanisms of LPA production and signaling in order to understand its biological functions in the central nervous system both in health and disease.
Collapse
Affiliation(s)
- Luiz Henrique Medeiros Geraldo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | | | | | | | - Celina Garcia
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio de Almeida Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Catarina Freitas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Fabio dosSantos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavia Regina Souza Lima
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Ueda H, Dozono N, Tanaka K, Kaneko S, Neyama H, Uchida H. Allodynia by Splenocytes From Mice With Acid-Induced Fibromyalgia-Like Generalized Pain and Its Sexual Dimorphic Regulation by Brain Microglia. Front Neurosci 2021; 14:600166. [PMID: 33424538 PMCID: PMC7785978 DOI: 10.3389/fnins.2020.600166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/24/2020] [Indexed: 12/23/2022] Open
Abstract
Fibromyalgia (FM), a disease of unknown etiology characterized by chronic generalized pain, is partly recapitulated in an animal model induced by repeated acid saline injections into the gastrocnemius muscle. Here, we attempted to investigate the sex difference in pain hypersensitivity (mechanical allodynia and hypersensitivity to electrical stimulation) in the repeated acid saline-induced FM-like generalized pain (AcGP) model. The first unilateral acid injection into gastrocnemius muscle at day 0/D0 and second injection at D5 (post day 0, P0) induced transient and long-lasting mechanical allodynia, respectively, on both sides of male and female mice. The pretreatment with gonadectomy did not affect the first injection-induced allodynia in both sexes, but gradually reversed the second injection-induced allodynia in male but not female mice. Moreover, the AcGP in male mice was abolished by intracerebroventricular minocycline treatments during D4–P4 or P5–P11, but not by early treatments during D0–D5 in male but not female mice, suggesting that brain microglia are required for AcGP in late-onset and sex-dependent manners. We also found that the intravenous treatments of splenocytes derived from male but not female mice treated with AcGP caused allodynia in naive mice. In addition, the purified CD4+ T cells derived from splenocytes of acid-treated male mice retained the ability to cause allodynia in naive mice. These findings suggest that FM-like AcGP has multiple sexual dimorphic mechanisms.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Naoki Dozono
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Keigo Tanaka
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hitoshi Uchida
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
16
|
Pathogenic mechanisms of lipid mediator lysophosphatidic acid in chronic pain. Prog Lipid Res 2020; 81:101079. [PMID: 33259854 DOI: 10.1016/j.plipres.2020.101079] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
A number of membrane lipid-derived mediators play pivotal roles in the initiation, maintenance, and regulation of various types of acute and chronic pain. Acute pain, comprising nociceptive and inflammatory pain warns us about the presence of damage or harmful stimuli. However, it can be efficiently reversed by opioid analgesics and anti-inflammatory drugs. Prostaglandin E2 and I2, the representative lipid mediators, are well-known causes of acute pain. However, some lipid mediators such as lipoxins, resolvins or endocannabinoids suppress acute pain. Various types of peripheral and central neuropathic pain (NeuP) as well as fibromyalgia (FM) are representatives of chronic pain and refractory owing to abnormal pain processing distinct from acute pain. Accumulating evidence demonstrated that lipid mediators represented by lysophosphatidic acid (LPA) are involved in the initiation and maintenance of both NeuP and FM in experimental animal models. The LPAR1-mediated peripheral mechanisms including dorsal root demyelination, Cavα2δ1 expression in dorsal root ganglion, and LPAR3-mediated amplification of central LPA production via glial cells are involved in the series of molecular mechanisms underlying NeuP. This review also discusses the involvement of lipid mediators in emerging research directives, including itch-sensing, sexual dimorphism, and the peripheral immune system.
Collapse
|
17
|
Neyama H, Dozono N, Uchida H, Ueda H. Mirtazapine, an α2 Antagonist-Type Antidepressant, Reverses Pain and Lack of Morphine Analgesia in Fibromyalgia-Like Mouse Models. J Pharmacol Exp Ther 2020; 375:1-9. [PMID: 32665319 DOI: 10.1124/jpet.120.265942] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Treatment of fibromyalgia is an unmet medical need; however, its pathogenesis is still poorly understood. In a series of studies, we have demonstrated that some pharmacological treatments reverse generalized chronic pain but do not affect the lack of morphine analgesia in the intermittent cold stress (ICS)-induced fibromyalgia-like pain model in mice. Here we report that repeated intraperitoneal treatments with mirtazapine, which is presumed to disinhibit 5-hydroxytriptamine (5-HT) release and activate 5-HT1 receptor through mechanisms of blocking presynaptic adrenergic α2 and postsynaptic 5-HT2 and 5-HT3 receptors, completely reversed the chronic pain for more than 4 to 5 days after the cessation of treatments. The repeated mirtazapine treatments also recovered the morphine analgesia after the return of nociceptive threshold to the normal level. The microinjection of small interfering RNA (siRNA) adrenergic α2a receptor (ADRA2A) into the habenula, which showed a selective upregulation of α2 receptor gene expression after ICS, reversed the hyperalgesia but did not recover the morphine analgesia. However, both reversal of hyperalgesia and recovery of morphine analgesia were observed when siRNA ADRA2A was administered intracerebroventricularly. As the habenular is reported to be involved in the emotion/reward-related pain and hypoalgesia, these results suggest that mirtazapine could attenuate pain and/or augment hypoalgesia by blocking the habenular α2 receptor after ICS. The recovery of morphine analgesia in the ICS model, on the other hand, seems to be mediated through a blockade of α2 receptor in unidentified brain regions. SIGNIFICANCE STATEMENT: This study reports possible mechanisms underlying the complete reversal of hyperalgesia and recovery of morphine analgesia by mirtazapine, a unique antidepressant with adrenergic α2 and serotonergic receptor antagonist properties, in a type of intermittently repeated stress (ICS)-induced fibromyalgia-like pain model. Habenula, a brain region which is related to the control of emotional pain, was found to play key roles in the antihyperalgesia, whereas other brain regions appeared to be involved in the recovery of morphine analgesia in the ICS model.
Collapse
Affiliation(s)
- Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Dozono
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hitoshi Uchida
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
18
|
Lysophosphatidic Acid Receptor 1- and 3-Mediated Hyperalgesia and Hypoalgesia in Diabetic Neuropathic Pain Models in Mice. Cells 2020; 9:cells9081906. [PMID: 32824296 PMCID: PMC7465054 DOI: 10.3390/cells9081906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 12/28/2022] Open
Abstract
Lysophosphatidic acid (LPA) signaling is known to play key roles in the initiation and maintenance of various chronic pain models. Here we examined whether LPA signaling is also involved in diabetes-induced abnormal pain behaviors. The high-fat diet (HFD) showing elevation of blood glucose levels and body weight caused thermal, mechanical hyperalgesia, hypersensitivity to 2000 or 250 Hz electrical-stimulation and hyposensitivity to 5 Hz stimulation to the paw in wild-type (WT) mice. These HFD-induced abnormal pain behaviors and body weight increase, but not elevated glucose levels were abolished in LPA1−/− and LPA3−/− mice. Repeated daily intrathecal (i.t.) treatments with LPA1/3 antagonist AM966 reversed these abnormal pain behaviors. Similar abnormal pain behaviors and their blockade by daily AM966 (i.t.) or twice daily Ki16425, another LPA1/3 antagonist was also observed in db/db mice which show high glucose levels and body weight. Furthermore, streptozotocin-induced similar abnormal pain behaviors, but not elevated glucose levels or body weight loss were abolished in LPA1−/− and LPA3−/− mice. These results suggest that LPA1 and LPA3 play key roles in the development of both type I and type II diabetic neuropathic pain.
Collapse
|
19
|
Rivera RR, Lin M, Bornhop EC, Chun J. Conditional Lpar1 gene targeting identifies cell types mediating neuropathic pain. FASEB J 2020; 34:8833-8842. [PMID: 32929779 PMCID: PMC7383719 DOI: 10.1096/fj.202000317r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/26/2023]
Abstract
LPA1 is one of six known receptors (LPA1-6) for lysophosphatidic acid (LPA). Constitutive Lpar1 null mutant mice have been instrumental in identifying roles for LPA-LPA1 signaling in neurobiological processes, brain development, and behavior, as well as modeling human neurological diseases like neuropathic pain. Constitutive Lpar1 null mutant mice are protected from partial sciatic nerve ligation (PSNL)-induced neuropathic pain, however, the cell types that are functionally responsible for mediating this protective effect are unknown. Here, we report the generation of an Lpar1flox/flox conditional null mutant mouse that allows for cre-mediated conditional deletion, combined with a PSNL pain model. Lpar1flox/flox mice were crossed with cre transgenic lines driven by neural gene promoters for nestin (all neural cells), synapsin (neurons), or P0 (Schwann cells). CD11b-cre transgenic mice were also used to delete Lpar1 in microglia. PSNL-initiated pain responses were reduced following cre-mediated Lpar1 deletion with all three neural promoters as well as the CD11b promoter, supporting involvement of Schwann cells, central and/or peripheral neurons, and microglia in mediating pain. Interestingly, rescue responses were nonidentical, implicating distinct roles for Lpar1-expressing cell types. Our results with a new Lpar1 conditional mouse mutant expand an understanding of LPA1 signaling in the PSNL model of neuropathic pain.
Collapse
Affiliation(s)
- Richard R. Rivera
- Degenerative Disease ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCAUSA
| | - Mu‐En Lin
- Molecular Biology Department, Dorris Neuroscience CenterThe Scripps Research InstituteLa JollaCAUSA
- Biomedical Sciences Graduate ProgramUniversity of California San DiegoLa JollaCAUSA
- Present address:
RevMAb BiosciencesSouth San FranciscoCAUSA
| | - Emily C. Bornhop
- Degenerative Disease ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCAUSA
| | - Jerold Chun
- Degenerative Disease ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCAUSA
| |
Collapse
|
20
|
Plastira I, Bernhart E, Joshi L, Koyani CN, Strohmaier H, Reicher H, Malle E, Sattler W. MAPK signaling determines lysophosphatidic acid (LPA)-induced inflammation in microglia. J Neuroinflammation 2020; 17:127. [PMID: 32326963 PMCID: PMC7178949 DOI: 10.1186/s12974-020-01809-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
Background In the extracellular environment, lysophosphatidic acid (LPA) species are generated via autotaxin (ATX)-mediated hydrolysis of lysophospholipid precursors. Members of the LPA family are potent lipid mediators transmitting signals via six different G protein-coupled LPA receptors (LPAR1-6). The LPA signaling axis is indispensable for brain development and function of the nervous system; however, during damage of the central nervous system, LPA levels can increase and aberrant signaling events counteract brain function. Here, we investigated regulation of the ATX/LPA/LPAR axis in response to lipopolysaccharide-induced systemic inflammation in mice and potential neurotoxic polarization programs in LPA-activated primary murine microglia. Methods In vivo, LPAR1-6 expression was established by qPCR in whole murine brain homogenates and in FACS-sorted microglia. ELISAs were used to quantitate LPA concentrations in the brain and cyto-/chemokine secretion from primary microglia in vitro. Transcription factor phosphorylation was analyzed by immunoblotting, and plasma membrane markers were analyzed by flow cytometry. We used MAPK inhibitors to study signal integration by the JNK, p38, and ERK1/2 branches in response to LPA-mediated activation of primary microglia. Results Under acute and chronic inflammatory conditions, we observed a significant increase in LPA concentrations and differential regulation of LPAR, ATX (encoded by ENPP2), and cytosolic phospholipase A2 (encoded by PLA2G4A) gene expression in the brain and FACS-sorted microglia. During pathway analyses in vitro, the use of specific MAPK antagonists (SP600125, SB203580, and PD98059) revealed that JNK and p38 inhibition most efficiently attenuated LPA-induced phosphorylation of proinflammatory transcription factors (STAT1 and -3, p65, and c-Jun) and secretion of IL-6 and TNFα. All three inhibitors decreased LPA-mediated secretion of IL-1β, CXCL10, CXCL2, and CCL5. The plasma membrane marker CD40 was solely inhibited by SP600125 while all three inhibitors affected expression of CD86 and CD206. All MAPK antagonists reduced intracellular COX-2 and Arg1 as well as ROS and NO formation, and neurotoxicity of microglia-conditioned media. Conclusion In the present study, we show that systemic inflammation induces aberrant ATX/LPA/LPAR homeostasis in the murine brain. LPA-mediated polarization of primary microglia via MAPK-dependent pathways induces features reminiscent of a neurotoxic phenotype.
Collapse
Affiliation(s)
- Ioanna Plastira
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Eva Bernhart
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Lisha Joshi
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Chintan N Koyani
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria.,Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Heimo Strohmaier
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Helga Reicher
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Wolfgang Sattler
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria. .,Center for Explorative Lipidomics, BioTechMed, Graz, Austria.
| |
Collapse
|
21
|
Neyama H, Dozono N, Ueda H. NR2A-NMDA Receptor Blockade Reverses the Lack of Morphine Analgesia Without Affecting Chronic Pain Status in a Fibromyalgia-Like Mouse Model. J Pharmacol Exp Ther 2020; 373:103-112. [PMID: 31941720 DOI: 10.1124/jpet.119.262642] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/13/2020] [Indexed: 03/08/2025] Open
Abstract
We have developed an experimental fibromyalgia-like mouse model using intermittent cold stress (ICS), where chronic pain is generalized, female predominant, and abolished in type 1 lysophosphatidic acid receptor-knockout (LPA1 -/-) mice but is not reversed by systemic or brain treatment with morphine. We investigated two issues in the present study: (1) whether chronic pain mechanisms and lack of brain morphine analgesia are associated in the ICS model and (2) what mechanisms are involved in the lack of morphine analgesia. ICS-induced hyperalgesia was not affected in μ-opioid receptor-knockout (MOPr -/-) mice, whereas the lack of brain morphine analgesia remained unchanged in LPA1 -/- mice, which completely abolished the hyperalgesia in the ICS model. In contrast, the lack of morphine analgesia was abolished in NR2A-NMDA receptor-knockout (NR2A -/- ) mice and blocked by intracerebroventricular (i.c.v.) injection of (R)-CPP, an NR2A antagonist, or by microinjection of siRNA NR2A into the periaqueductal gray matter region, whereas no change was observed with Ro 04-5595, an NMDA receptor subtype 2B antagonist (i.c.v.). The lack of morphine analgesia was also reversed by concomitant treatment with 1 mg/kg intraperitoneal (i.p.) of dextromethorphan, which possesses NMDA receptor antagonist activity but no analgesic activity. Finally, the hyperalgesia was completely reversed by methadone, which possesses both MOPr agonist and NMDA receptor antagonist activity. Indeed, methadone analgesia was abolished in MOPr -/- mice. These results suggest that chronic pain status and lack of morphine analgesia are independent of each other, and that lack of morphine analgesia is mediated by activation of the NR2A-NMDA receptor system. SIGNIFICANCE STATEMENT: This study reports that a type of intermittently repeated stress causes widespread pain that does not respond to morphine. Because this lack of morphine analgesia is not affected in mice, in which chronic pain is abolished, the mechanisms underlying chronic pain and lack of morphine analgesia are independent of each other. Through speculation that a lack of morphine analgesia may be a secondary event to endogenous opioid analgesic tolerance, the authors demonstrate that an antiopioid N-methyl-D-aspartate receptor system counterbalances the μ-opioid receptor-mediated analgesic mechanisms in the intermittent cold stress model.
Collapse
Affiliation(s)
- Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Dozono
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
22
|
Ueda H. LPA receptor signaling as a therapeutic target for radical treatment of neuropathic pain and fibromyalgia. Pain Manag 2019; 10:43-53. [PMID: 31852400 DOI: 10.2217/pmt-2019-0036] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Since the first discovery that the bioactive lipid, lysophosphatidic acid (LPA) and LPA1 receptor signaling play a role in the initiation of neuropathic pain (NeuP), accumulated reports have supported the original findings and extended the study toward possible therapeutic applications. The present review describes beneficial roles of LPA receptor signaling in a variety of chronic pain, such as peripheral NeuP induced by nerve injury, chemotherapy and diabetes, central NeuP induced by cerebral ischemia with hemorrhage and spinal cord injury, and fibromyalgia-like wide spread pain induced by repeated cold, psychological and muscular acidic stress. Emerging mechanistic findings are the feed-forward amplification of LPA production through LPA1, LPA3 and microglia and the evidence for maintenance of chronic pain by LPA receptor signaling.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Molecular Pharmacology, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
23
|
González-Gil I, Zian D, Vázquez-Villa H, Hernández-Torres G, Martínez RF, Khiar-Fernández N, Rivera R, Kihara Y, Devesa I, Mathivanan S, Del Valle CR, Zambrana-Infantes E, Puigdomenech M, Cincilla G, Sanchez-Martinez M, Rodríguez de Fonseca F, Ferrer-Montiel AV, Chun J, López-Vales R, López-Rodríguez ML, Ortega-Gutiérrez S. A Novel Agonist of the Type 1 Lysophosphatidic Acid Receptor (LPA 1), UCM-05194, Shows Efficacy in Neuropathic Pain Amelioration. J Med Chem 2019; 63:2372-2390. [PMID: 31790581 DOI: 10.1021/acs.jmedchem.9b01287] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neuropathic pain (NP) is a complex chronic pain state with a prevalence of almost 10% in the general population. Pharmacological options for NP are limited and weakly effective, so there is a need to develop more efficacious NP attenuating drugs. Activation of the type 1 lysophosphatidic acid (LPA1) receptor is a crucial factor in the initiation of NP. Hence, it is conceivable that a functional antagonism strategy could lead to NP mitigation. Here we describe a new series of LPA1 agonists among which derivative (S)-17 (UCM-05194) stands out as the most potent and selective LPA1 receptor agonist described so far (Emax = 118%, EC50 = 0.24 μM, KD = 19.6 nM; inactive at autotaxin and LPA2-6 receptors). This compound induces characteristic LPA1-mediated cellular effects and prompts the internalization of the receptor leading to its functional inactivation in primary sensory neurons and to an efficacious attenuation of the pain perception in an in vivo model of NP.
Collapse
Affiliation(s)
- Inés González-Gil
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Debora Zian
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Henar Vázquez-Villa
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Gloria Hernández-Torres
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - R Fernando Martínez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Nora Khiar-Fernández
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Richard Rivera
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Isabel Devesa
- IDiBE, Universidad Miguel Hernández de Elche, E-03202 Alicante, Spain
| | | | - Cristina Rosell Del Valle
- Instituto de Investigación Biomédica de Málaga, UGC Salud Mental, Universidad de Málaga, Hospital Universitario Regional de Málaga, E-29010 Málaga, Spain
| | - Emma Zambrana-Infantes
- Instituto de Investigación Biomédica de Málaga, UGC Salud Mental, Universidad de Málaga, Hospital Universitario Regional de Málaga, E-29010 Málaga, Spain
| | - María Puigdomenech
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, E-08193 Barcelona, Spain
| | - Giovanni Cincilla
- Molomics, Barcelona Science Park, Baldiri i Reixac 4-8, E-08028 Barcelona, Spain
| | | | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga, UGC Salud Mental, Universidad de Málaga, Hospital Universitario Regional de Málaga, E-29010 Málaga, Spain.,Departamento de Psicobiología, Facultad de Psicología, Universidad Complutense de Madrid, Pozuelo de Alarcón, E-28223 Madrid, Spain
| | | | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Rubén López-Vales
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, E-08193 Barcelona, Spain
| | - María L López-Rodríguez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Silvia Ortega-Gutiérrez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| |
Collapse
|
24
|
Uranbileg B, Ito N, Kurano M, Saigusa D, Saito R, Uruno A, Kano K, Ikeda H, Yamada Y, Sumitani M, Sekiguchi M, Aoki J, Yatomi Y. Alteration of the lysophosphatidic acid and its precursor lysophosphatidylcholine levels in spinal cord stenosis: A study using a rat cauda equina compression model. Sci Rep 2019; 9:16578. [PMID: 31719574 PMCID: PMC6851136 DOI: 10.1038/s41598-019-52999-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Cauda equina compression (CEC) is a major cause of neurogenic claudication and progresses to neuropathic pain (NP). A lipid mediator, lysophosphatidic acid (LPA), is known to induce NP via the LPA1 receptor. To know a possible mechanism of LPA production in neurogenic claudication, we determined the levels of LPA, lysophosphatidylcholine (LPC) and LPA-producing enzyme autotaxin (ATX), in the cerebrospinal fluid (CSF) and spinal cord (SC) using a CEC as a possible model of neurogenic claudication. Using silicon blocks within the lumbar epidural space, we developed a CEC model in rats with motor dysfunction. LPC and LPA levels in the CSF were significantly increased from day 1. Importantly, specific LPA species (16:0, 18:2, 20:4) were upregulated, which have been shown to produce by ATX detected in the CSF, without changes on its level. In SC, the LPC and LPA levels did not change, but mass spectrometry imaging analysis revealed that LPC was present in a region where the silicon blocks were inserted. These results propose a model for LPA production in SC and CSF upon neurogenic claudication that LPC produced locally by tissue damages is converted to LPA by ATX, which then leak out into the CSF.
Collapse
Affiliation(s)
- Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuko Ito
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo, Tokyo, Japan.
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Ritsumi Saito
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Miyagi, Japan.,Medical Biochemistry, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Kuniyuki Kano
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Hitoshi Ikeda
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshitsugu Yamada
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo, Tokyo, Japan
| | - Masahiko Sumitani
- Department of Pain and Palliative Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Miho Sekiguchi
- Department of Orthopaedic Surgery, Fukushima Medical University, School of Medicine, Fukushima, Japan
| | - Junken Aoki
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Ueda H. [Lysophosphatidic Acid Receptor Signaling Underlying Chronic Pain and Neuroprotective Mechanisms through Prothymosin α]. YAKUGAKU ZASSHI 2019; 139:1403-1415. [PMID: 31685737 DOI: 10.1248/yakushi.19-00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For my Ph.D. research topic, I isolated endogenous morphine-like analgesic dipeptide, kyotorphin, which mediates Met-enkephalin release, and discovered kyotorphin synthetase, a putative receptor and antagonist. Furthermore, I succeeded in purifying μ-opioid receptor and functional reconstitution with purified G proteins. After receiving my full professor position at Nagasaki University in 1996, I worked on two topics of research, molecular mechanisms of chronic pain through lysophosphatidic acid (LPA) and identification and characterization of neuroprotective protein, prothymosin α. In a series of studies, we have shown that LPA signaling defines the molecular mechanisms of neuropathic pain and fibromyalgia in terms of development and maintenance. Above all, the discovery of feed-forward system in LPA production and pain memory may contribute to better understanding of chronic pain and future analgesic drug discovery. Regarding prothymosin α, we first discovered it as neuronal necrosis-inhibitory molecule through two independent mechanisms, such as toll-like receptor and F0/F1 ATPase, both which protect neurons through indirect mechanisms. Prothymosin α is released by non-classical and non-vesicular mechanisms on various stresses, such as ischemia, starvation, and heat-shock. Thus it may be called a new type of neuroprotective damage-associated molecular patterns (DAMPs)/Alarmins. Heterozygotic mice showed a defect in memory-learning and neurogenesis as well as anxiogenic behaviors. Small peptide, P6Q derived from prothymosin α retains neuroprotective actions, which include blockade of cerebral hemorrhage caused by late treatment with tissue plasminogen activator in the stroke model in mice.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University, Institute of Biomedical Sciences
| |
Collapse
|
26
|
Affiliation(s)
- Hiroshi Ueda
- Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
27
|
Lind AL, Just D, Mikus M, Fredolini C, Ioannou M, Gerdle B, Ghafouri B, Bäckryd E, Tanum L, Gordh T, Månberg A. CSF levels of apolipoprotein C1 and autotaxin found to associate with neuropathic pain and fibromyalgia. J Pain Res 2019; 12:2875-2889. [PMID: 31686904 PMCID: PMC6800548 DOI: 10.2147/jpr.s215348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Objective Neuropathic pain and fibromyalgia are two common and poorly understood chronic pain conditions that lack satisfactory treatments, cause substantial suffering and societal costs. Today, there are no biological markers on which to base chronic pain diagnoses, treatment choices or to understand the pathophysiology of pain for the individual patient. This study aimed to investigate cerebrospinal fluid (CSF) protein profiles potentially associated with fibromyalgia and neuropathic pain. Methods CSF samples were collected from 25 patients with neuropathic pain (two independent sets, n=14 patients for discovery, and n=11 for verification), 40 patients with fibromyalgia and 134 controls without neurological disease from two different populations. CSF protein profiling of 55 proteins was performed using antibody suspension bead array technology. Results We found increased levels of apolipoprotein C1 (APOC1) in CSF of neuropathic pain patients compared to controls and there was a trend for increased levels also in fibromyalgia patients. In addition, levels of ectonucleotide pyrophosphatase family member 2 (ENPP2, also referred to as autotaxin) were increased in the CSF of fibromyalgia patients compared to all other groups including patients with neuropathic pain. Conclusion The increased levels of APOC1 and ENPP2 found in neuropathic pain and fibromyalgia patients may shed light on the underlying mechanisms of these conditions. Further investigation is required to elucidate their role in maintaining pain and other main symptoms of these disorders.
Collapse
Affiliation(s)
- Anne-Li Lind
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - David Just
- Division of Affinity Proteomics, SciLifeLab, Deptartment of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Maria Mikus
- Division of Affinity Proteomics, SciLifeLab, Deptartment of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Claudia Fredolini
- Division of Affinity Proteomics, SciLifeLab, Deptartment of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Marina Ioannou
- Division of Affinity Proteomics, SciLifeLab, Deptartment of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Gerdle
- Pain and Rehabilitation Center, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Bijar Ghafouri
- Pain and Rehabilitation Center, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Emmanuel Bäckryd
- Pain and Rehabilitation Center, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Lars Tanum
- Department of R&D in Mental Health, Akershus University Hospital, Lørenskog, Norway
| | - Torsten Gordh
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anna Månberg
- Division of Affinity Proteomics, SciLifeLab, Deptartment of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
28
|
Targeting the Nav1.8 ion channel engenders sex-specific responses in lysophosphatidic acid-induced joint neuropathy. Pain 2019; 160:269-278. [PMID: 30211781 DOI: 10.1097/j.pain.0000000000001399] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Joint neuropathic pain occurs in a subset of arthritis patients, and lysophosphatidic acid (LPA) has been implicated as a mediator of joint neuropathy. The mechanism by which LPA promotes neuropathic pain is unknown but may be related to altered signalling of the voltage-gated sodium channel Nav1.8 located on nociceptors. Because arthritis and neuropathic pain are more prevalent in females, this study aimed to explore potential sex differences in the development of LPA-induced joint neuropathy and whether Nav1.8 played a role in the associated neuropathic pain. Joint neuropathy was induced in male and female Wistar rats (179-284 g) by intra-articular injection of 50-µg LPA. Pain behaviour was assessed over 21 days using von Frey hair algesiometry. On day 21, electrophysiological recordings of joint primary afferents were conducted to measure peripheral sensitisation. Saphenous nerve morphology and expression of the nerve-damage marker ATF3 and Nav1.8 in ipsilateral dorsal root ganglions were compared on the basis of sex. The analgesic properties of the selective Nav1.8 antagonist A-803467 was determined in pain behaviour and electrophysiology experiments. Females developed more severe mechanical allodynia than males after LPA treatment. Lysophosphatidic acid caused more pronounced demyelination of the saphenous nerve in females, but no sex differences were observed in the expression of ATF3 or Nav1.8 in dorsal root ganglion neurones. Blockade of Nav1.8 channels with A-803467 resulted in a decrease in joint mechanosensitivity and secondary allodynia with females exhibiting a greater response. These findings suggest that LPA has sex-specific effects on joint neuropathy and Nav1.8 gating, which should be considered when treating neuropathic arthritis patients.
Collapse
|
29
|
Lysophosphatidic acids and their substrate lysophospholipids in cerebrospinal fluid as objective biomarkers for evaluating the severity of lumbar spinal stenosis. Sci Rep 2019; 9:9144. [PMID: 31235770 PMCID: PMC6591408 DOI: 10.1038/s41598-019-45742-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 06/14/2019] [Indexed: 12/28/2022] Open
Abstract
Lysophospholipids (LPLs) are known to have potentially important roles in the initiation and maintenance of neuropathic pain in animal models. This study investigated the association between the clinical severity of lumbar spinal stenosis (LSS) and the cerebrospinal fluid (CSF) levels of LPLs, using human samples. We prospectively identified twenty-eight patients with LSS and fifteen controls with idiopathic scoliosis or bladder cancer without neurological symptoms. We quantified LPLs from CSF using liquid chromatography-tandem mass spectrometry. We assessed clinical outcome measures of LSS (Neuropathic Pain Symptom Inventory (NPSI) and Zurich Claudication Questionnaire (ZCQ)) and categorized patients into two groups according to their severity. Five species of lysophosphatidic acid (LPA), nine species of lysophosphatidylcholine (LPC), and one species of lysophosphatidylinositol (LPI) were detected. The CSF levels of all species of LPLs were significantly higher in LSS patients than controls. Patients in the severe NPSI group had significantly higher LPL levels (three species of LPA and nine species of LPC) than the mild group. Patients in the severe ZCQ group also had significantly higher LPL levels (four species of LPA and nine species of LPC). This investigation demonstrates a positive correlation between the CSF levels of LPLs and the clinical severity of LSS. LPLs are potential biomarkers for evaluating the severity of LSS.
Collapse
|
30
|
|
31
|
Suckau O, Gross I, Schrötter S, Yang F, Luo J, Wree A, Chun J, Baska D, Baumgart J, Kano K, Aoki J, Bräuer AU. LPA 1 , LPA 2 , LPA 4 , and LPA 6 receptor expression during mouse brain development. Dev Dyn 2019; 248:375-395. [PMID: 30847983 PMCID: PMC6593976 DOI: 10.1002/dvdy.23] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/17/2022] Open
Abstract
Background LPA is a small bioactive phospholipid that acts as an extracellular signaling molecule and is involved in cellular processes, including cell proliferation, migration, and differentiation. LPA acts by binding and activating at least six known G protein–coupled receptors: LPA1–6. In recent years, LPA has been suggested to play an important role both in normal neuronal development and under pathological conditions in the nervous system. Results We show the expression pattern of LPA receptors during mouse brain development by using qRT‐PCR, in situ hybridization, and immunocytochemistry. Only LPA1, LPA2,LPA4, and LPA6 mRNA transcripts were detected throughout development stages from embryonic day 16 until postnatal day 30 of hippocampus, neocortex, cerebellum, and bulbus olfactorius in our experiments, while expression of LPA3 and LPA5 genes was below detection level. In addition to our qRT‐PCR results, we also analyzed the cellular protein expression of endogenous LPA receptors, with focus on LPA1 and LPA2 within postnatal brain slices and primary neuron differentiation with and without cytoskeleton stabilization and destabilization. Conclusions The expression of LPA receptors changes depends on the developmental stage in mouse brain and in cultured hippocampal primary neurons. Interestingly, we found that commercially available antibodies for LPA receptors are largely unspecific. LPA1, ‐2, ‐4, and ‐6 genes are dynamically expressed during postnatal brain development. LPA1, ‐2, ‐4, and ‐6 genes are differently expressed in the hippocampus, neocortex, cerebellum, and bulbus olfactorius. LPA1 and ‐2 gene expression alters during neuronal differentiation. LPA1, ‐2, ‐3, ‐4, and ‐6 genes are expressed in glia cells, but differed in gene expression levels.
Collapse
Affiliation(s)
- Olga Suckau
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Isabel Gross
- Institute of Anatomy, Universitätsmedizin Rostock, Rostock, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Sandra Schrötter
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fan Yang
- Albrecht Kossel Institute for Neuroregeneration, Rostock University Medical Center, Rostock, Germany
| | - Jiankai Luo
- Albrecht Kossel Institute for Neuroregeneration, Rostock University Medical Center, Rostock, Germany
| | - Andreas Wree
- Institute of Anatomy, Universitätsmedizin Rostock, Rostock, Germany
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California
| | - David Baska
- Translational Animal Research Center, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jan Baumgart
- Translational Animal Research Center, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| | - Anja U Bräuer
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute of Anatomy, Universitätsmedizin Rostock, Rostock, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
32
|
Involvement of lysophosphatidic acid-induced astrocyte activation underlying the maintenance of partial sciatic nerve injury-induced neuropathic pain. Pain 2019; 159:2170-2178. [PMID: 29939962 DOI: 10.1097/j.pain.0000000000001316] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have previously demonstrated that lysophosphatidic acid (LPA) plays key roles in the initial mechanisms for neuropathic pain (NeuP) development. Here, we examined whether LPA receptor mechanisms and LPA production are related to the glial activation at a late stage after partial sciatic nerve ligation (pSNL) by use of microglial inhibitor, Mac1-saporin or astrocyte inhibitor, and L-α-aminoadipate (L-AA). Although single intrathecal injection of LPA1/3 antagonist, Ki-16425 did not affect the pain threshold at day 7 after the spinal cord injury, repeated treatments of each compound gradually reversed the basal pain threshold to the control level. The intrathecal administration of a microglia inhibitor, Mac-1-saporin reversed the late hyperalgesia and LPA production at day 14 after the pSNL, whereas L-AA inhibited the hyperalgesia, but had no effect on LPA production. The involvement of LPA receptors in astrocyte activation in vivo was evidenced by the findings that Ki-16425 treatments abolished the upregulation of CXCL1 in activated astrocytes in the spinal dorsal horn of mice at day 14 after the pSNL, and that Ki-16425 reversed the LPA-induced upregulation of several chemokine gene expressions in primary cultured astrocytes. Finally, we found that significant hyperalgesia was observed with intrathecal administration of primary cultured astrocytes, which had been stimulated by LPA in a Ki-16425-reversible manner. All these findings suggest that LPA production and LPA1/3 receptor activation through differential glial mechanisms play key roles in the maintenance as well as initiation mechanisms in NeuP.
Collapse
|
33
|
Ueda H, Neyama H, Sasaki K, Miyama C, Iwamoto R. Lysophosphatidic acid LPA 1 and LPA 3 receptors play roles in the maintenance of late tissue plasminogen activator-induced central poststroke pain in mice. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 5:100020. [PMID: 31194070 PMCID: PMC6550111 DOI: 10.1016/j.ynpai.2018.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 11/17/2022]
Abstract
We developed a mouse model for central post-stroke pain (CPSP), a centrally-originated neuropathic pain (NeuP). In this mode, mice were first injected with Rose Bengal, followed by photo-irradiation of left middle cerebral artery (MCA) to generate thrombosis. Although the MCA thrombosis was soon dissolved, the reduced blood flow remained for more than 24 h due to subsequent occlusion of microvessels. This photochemically induced thrombosis (PIT) model showed a hypersensitivity to the electrical stimulation of both sides of paw, but did not show any abnormal pain in popular thermal or mechanical nociception tests. When tissue-type plasminogen activator (tPA) was injected 6 h after the PIT stress, tPA-dependent hypersensitivity to the electrical paw stimulation and stable thermal and mechanical hyperalgesia on both sides for more than 17 or 18 days after the PIT treatment. These hyperalgesic effects were abolished in lysophosphatidic acid receptor 1 (LPA1)- and lysophosphatidic acid receptor 3 (LPA3)-deficient mice. When Ki-16425, an LPA1 and LPA3 antagonist was treated twice daily for 6 days consecutively, the thermal and mechanical hyperalgesia at day 17 and 18 were significantly reversed. The liquid chromatography-mass spectrometry (LC-MS/MS) analysis revealed that there is a significant increase in several species of LPA molecules in somatosensory S-I and medial dorsal thalamus (MD), but not in striatum or ventroposterior thalamus. All these results suggest that LPA1 and LPA3 signaling play key roles in the development and maintenance of CPSP.
Collapse
Key Words
- CPSP, central post-stroke pain
- Central poststroke pain
- DMSO, dimethyl sulfoxide
- EPW, electrical stimulation-induced paw withdrawal
- HE, Hematoxylin and Eosin
- LC–MS/MS
- LC–MS/MS, liquid chromatography–mass spectrometry
- LPA1, lysophosphatidic acid receptor 1
- LPA1-KO, LPA1-deficient
- LPA3, lysophosphatidic acid receptor 3
- Lysophosphatidic acid
- MCA, middle cerebral artery
- MD, medial dorsal thalamus
- MRM, multiple reaction monitoring
- NeuP, neuropathic pain
- PFA, paraformaldehyde
- PIT, photochemically induced thrombosis
- PWL, paw withdrawal latency
- Photochemically induced thrombosis
- RB, Rose Bengal
- S-I, sensory cortex
- TTC, 2,3,5-triphenyltetrazolium chloride
- i.v., intravenously
- pSNL, partial sciatic nerve ligation
- tMCAO, transient middle cerebral artery occlusion
- tPA
- tPA, tissue-type plasminogen activator
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University, Institute of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | | | | | | | | |
Collapse
|
34
|
Kuwajima K, Sumitani M, Kurano M, Kano K, Nishikawa M, Uranbileg B, Tsuchida R, Ogata T, Aoki J, Yatomi Y, Yamada Y. Lysophosphatidic acid is associated with neuropathic pain intensity in humans: An exploratory study. PLoS One 2018; 13:e0207310. [PMID: 30408112 PMCID: PMC6224112 DOI: 10.1371/journal.pone.0207310] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/29/2018] [Indexed: 12/15/2022] Open
Abstract
The underlying mechanisms of neuropathic pain remain to be elucidated. Basic animal research has suggested that lysophosphatidic acids, which are bioactive lipids produced by autotaxin from lysophosphatidylcholine, may play key roles in the initiation and maintenance of neuropathic pain. Here, we investigated the clinical relevance of lysophosphatidic acids signaling on neuropathic pain in humans. Eighteen patients who had been diagnosed with neuropathic pain with varied etiologies participated in the study. Cerebrospinal fluid samples were obtained by lumbar puncture and the concentrations of 12 species of lysophosphatidic acids and lysophosphatidylcholine, autotaxin, and the phosphorylated neurofilament heavy subunit were measured. Pain symptoms were assessed using an 11-point numeric rating scale and the Neuropathic Pain Symptom Inventory regarding intensity and descriptive dimensions of neuropathic pain. The total lysophosphatidic acids were significantly associated with both pain intensity and symptoms. 18:1 and 20:4 lysophosphatidic acids in particular demonstrated the most correlations with dimensions of pain symptoms. Autotaxin and the phosphorylated neurofilament heavy subunit showed no association with pain symptoms. In conclusions, lysophosphatidic acids were significantly associated with pain symptoms in neuropathic pain patients. These results suggest that lysophosphatidic acids signaling might be a potential therapeutic target for neuropathic pain.
Collapse
Affiliation(s)
- Ken Kuwajima
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Masahiko Sumitani
- Department of Pain and Palliative Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Kuniyuki Kano
- Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Masako Nishikawa
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Rikuhei Tsuchida
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Toru Ogata
- Department of Rehabilitation for Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | - Junken Aoki
- Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yoshitsugu Yamada
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
35
|
LPA5 signaling is involved in multiple sclerosis-mediated neuropathic pain in the cuprizone mouse model. J Pharmacol Sci 2018; 136:93-96. [DOI: 10.1016/j.jphs.2018.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/29/2017] [Accepted: 01/09/2018] [Indexed: 11/21/2022] Open
|
36
|
Ueda H, Neyama H. LPA1 receptor involvement in fibromyalgia-like pain induced by intermittent psychological stress, empathy. NEUROBIOLOGY OF PAIN 2017; 1:16-25. [PMID: 31194005 PMCID: PMC6550118 DOI: 10.1016/j.ynpai.2017.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/22/2022]
Abstract
Treatment for fibromyalgia is an unmet medical need and its pathogenesis is still poorly understood. The present study demonstrated that intermittent psychological stress (IPS), or empathy causes generalized chronic abnormal pain with female predominance. The persistence of the pain phenotype was dependent on the unpredictability of the stressor. The pain was reversed by pregabalin (PGB), duloxetine (DLX) or mirtazapine (Mir), but not by diclofenac or morphine. Differential administration of these existing medicines revealed that the sites of PGB and Mir actions exist in the brain, but not in the spinal cord, while that of DLX is preferentially in the spinal cord. It is interesting to note that the intracerebroventricular injection of PGB or Mir showed potent analgesia for 24 h or longer, though systemic injection of these medicines shows anti-hyperalgesia just for several hours. These results indicate that initial intense actions in the target brain may prevent the forthcoming development of pain memory. IPS-induced abnormal pain was prevented in mice deficient of lysophosphatidic acid receptor 1 (LPA1) gene, and completely cured by the repeated intrathecal treatments with LPA1 antagonist, AM966, which did not show acute action. All these results suggest that IPS model is an experimental animal model, which mimics the pathophysiology and pharmacotherapy in fibromyalgia in clinic, and LPA1 signaling plays crucial roles in the IPS-induced fibromyalgia-like abnormal pain.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Corresponding author at: Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| | | |
Collapse
|