1
|
Cen J, Hu N, Shen J, Gao Y, Lu H. Pathological Functions of Lysosomal Ion Channels in the Central Nervous System. Int J Mol Sci 2024; 25:6565. [PMID: 38928271 PMCID: PMC11203704 DOI: 10.3390/ijms25126565] [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: 04/03/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Lysosomes are highly dynamic organelles that maintain cellular homeostasis and regulate fundamental cellular processes by integrating multiple metabolic pathways. Lysosomal ion channels such as TRPML1-3, TPC1/2, ClC6/7, CLN7, and TMEM175 mediate the flux of Ca2+, Cl-, Na+, H+, and K+ across lysosomal membranes in response to osmotic stimulus, nutrient-dependent signals, and cellular stresses. These ion channels serve as the crucial transducers of cell signals and are essential for the regulation of lysosomal biogenesis, motility, membrane contact site formation, and lysosomal homeostasis. In terms of pathophysiology, genetic variations in these channel genes have been associated with the development of lysosomal storage diseases, neurodegenerative diseases, inflammation, and cancer. This review aims to discuss the current understanding of the role of these ion channels in the central nervous system and to assess their potential as drug targets.
Collapse
Affiliation(s)
| | | | | | - Yongjing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| | - Huanjun Lu
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| |
Collapse
|
2
|
Ronan EA, Nagel M, Emrick JJ. The anatomy, neurophysiology, and cellular mechanisms of intradental sensation. FRONTIERS IN PAIN RESEARCH 2024; 5:1376564. [PMID: 38590718 PMCID: PMC11000636 DOI: 10.3389/fpain.2024.1376564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Somatosensory innervation of the oral cavity enables the detection of a range of environmental stimuli including minute and noxious mechanical forces. The trigeminal sensory neurons underlie sensation originating from the tooth. Prior work has provided important physiological and molecular characterization of dental pulp sensory innervation. Clinical dental experiences have informed our conception of the consequence of activating these neurons. However, the biological role of sensory innervation within the tooth is yet to be defined. Recent transcriptomic data, combined with mouse genetic tools, have the capacity to provide important cell-type resolution for the physiological and behavioral function of pulp-innervating sensory neurons. Importantly, these tools can be applied to determine the neuronal origin of acute dental pain that coincides with tooth damage as well as pain stemming from tissue inflammation (i.e., pulpitis) toward developing treatment strategies aimed at relieving these distinct forms of pain.
Collapse
Affiliation(s)
- Elizabeth A. Ronan
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| | - Maximilian Nagel
- Sensory Cells and Circuits Section, National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | - Joshua J. Emrick
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
3
|
Sessle BJ. Modulatory Processes in Craniofacial Pain States. ADVANCES IN NEUROBIOLOGY 2024; 35:107-124. [PMID: 38874720 DOI: 10.1007/978-3-031-45493-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Pain is a common symptom associated with many disorders affecting the craniofacial tissues that include the teeth and their supporting structures, the jaw, face and tongue muscles, and the temporomandibular joint. Most acute craniofacial pain states are easily recognized and readily treated, but chronic craniofacial pain states (e.g., temporomandibular disorders [TMD], trigeminal neuropathies, and some headaches) may be especially challenging to manage successfully. This chapter provides an overview of the processes that underlie craniofacial pain, with a focus on the pain-modulatory mechanisms operating in craniofacial tissues and in the central nervous system (CNS), including the role of endogenous chemical processes such as those involving opioids. The chapter outlines in particular findings from preclinical studies that have provided substantial information about the neural as well as nonneural (e.g., glial) processes involved in the initiation, transmission, and modulation of nociceptive signals in the trigeminal system, and also draws attention to their clinical correlates. The increased understanding gained from these preclinical studies of how nociceptive signals can be modulated will contribute to improvements in presently available therapeutic approaches to manage craniofacial pain as well as to the development of novel analgesic approaches.
Collapse
Affiliation(s)
- Barry J Sessle
- Department of Oral Physiology Faculty of Dentistry, Department of Physiology Faculty of Medicine, Centre for the Study of Pain, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Wedel S, Hahnefeld L, Schreiber Y, Namendorf C, Heymann T, Uhr M, Schmidt MV, de Bruin N, Hausch F, Thomas D, Geisslinger G, Sisignano M. SAFit2 ameliorates paclitaxel-induced neuropathic pain by reducing spinal gliosis and elevating pro-resolving lipid mediators. J Neuroinflammation 2023; 20:149. [PMID: 37355700 DOI: 10.1186/s12974-023-02835-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Chemotherapy-induced neuropathic pain (CIPN) describes a pathological pain state that occurs dose-dependently as a side effect and can limit or even impede an effective cancer therapy. Unfortunately, current treatment possibilities for CIPN are remarkably confined and mostly inadequate as CIPN therapeutics themselves consist of low effectiveness and may induce severe side effects, pointing out CIPN as pathological entity with an emerging need for novel treatment targets. Here, we investigated whether the novel and highly specific FKBP51 inhibitor SAFit2 reduces paclitaxel-induced neuropathic pain. METHODS In this study, we used a well-established multiple low-dose paclitaxel model to investigate analgesic and anti-inflammatory properties of SAFit2. For this purpose, the behavior of the mice was recorded over 14 days and the mouse tissue was then analyzed using biochemical methods. RESULTS Here, we show that SAFit2 is capable to reduce paclitaxel-induced mechanical hypersensitivity in mice. In addition, we detected that SAFit2 shifts lipid levels in nervous tissue toward an anti-inflammatory and pro-resolving lipid profile that counteracts peripheral sensitization after paclitaxel treatment. Furthermore, SAFit2 reduced the activation of astrocytes and microglia in the spinal cord as well as the levels of pain-mediating chemokines. Its treatment also increased anti-inflammatory cytokines levels in neuronal tissues, ultimately leading to a resolution of neuroinflammation. CONCLUSIONS In summary, SAFit2 shows antihyperalgesic properties as it ameliorates paclitaxel-induced neuropathic pain by reducing peripheral sensitization and resolving neuroinflammation. Therefore, we consider SAFit2 as a potential novel drug candidate for the treatment of paclitaxel-induced neuropathic pain.
Collapse
Affiliation(s)
- Saskia Wedel
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Christian Namendorf
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Tim Heymann
- Department of Biochemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Manfred Uhr
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Mathias V Schmidt
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Natasja de Bruin
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Felix Hausch
- Department of Biochemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany.
| |
Collapse
|
5
|
Siddiqui YD, Nie X, Wang S, Abbasi Y, Park L, Fan X, Thumbigere-Math V, Chung MK. Substance P aggravates ligature-induced periodontitis in mice. Front Immunol 2023; 14:1099017. [PMID: 37122730 PMCID: PMC10140340 DOI: 10.3389/fimmu.2023.1099017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Periodontitis is one of the most common oral diseases in humans, affecting over 40% of adult Americans. Pain-sensing nerves, or nociceptors, sense local environmental changes and often contain neuropeptides. Recent studies have suggested that nociceptors magnify host response and regulate bone loss in the periodontium. A subset of nociceptors projected to periodontium contains neuropeptides, such as calcitonin gene-related peptide (CGRP) or substance P (SP). However, the specific roles of neuropeptides from nociceptive neural terminals in periodontitis remain to be determined. In this study, we investigated the roles of neuropeptides on host responses and bone loss in ligature-induced periodontitis. Deletion of tachykinin precursor 1 (Tac1), a gene that encodes SP, or treatment of gingiva with SP antagonist significantly reduced bone loss in ligature-induced periodontitis, whereas deletion of calcitonin related polypeptide alpha (Calca), a gene that encodes CGRP, showed a marginal role on bone loss. Ligature-induced recruitment of leukocytes, including neutrophils, and increase in cytokines leading to bone loss in periodontium was significantly less in Tac1 knockout mice. Furthermore, intra-gingival injection of SP, but not neurokinin A, induced a vigorous inflammatory response and osteoclast activation in alveolar bone and facilitated bone loss in ligature-induced periodontitis. Altogether, our data suggest that SP plays significant roles in regulating host responses and bone resorption in ligature-induced periodontitis.
Collapse
Affiliation(s)
- Yasir Dilshad Siddiqui
- Program in Neuroscience, Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, United States
- Department of Preventive Dentistry, College of Dentistry, Jouf University, Sakaka, Saudi Arabia
| | - Xuguang Nie
- Program in Neuroscience, Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Sheng Wang
- Program in Neuroscience, Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Yasaman Abbasi
- Program in Neuroscience, Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Lauren Park
- Program in Neuroscience, Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Xiaoxuan Fan
- Department of Microbiology and Immunology, Flow Cytometry Shared Service, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Vivek Thumbigere-Math
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Man-Kyo Chung
- Program in Neuroscience, Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
6
|
Fang XX, Zhai MN, Zhu M, He C, Wang H, Wang J, Zhang ZJ. Inflammation in pathogenesis of chronic pain: Foe and friend. Mol Pain 2023; 19:17448069231178176. [PMID: 37220667 DOI: 10.1177/17448069231178176] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023] Open
Abstract
Chronic pain is a refractory health disease worldwide causing an enormous economic burden on individuals and society. Accumulating evidence suggests that inflammation in the peripheral nervous system (PNS) and central nervous system (CNS) is the major factor in the pathogenesis of chronic pain. The inflammation in the early- and late phase may have distinctive effects on the initiation and resolution of pain, which can be viewed as friend or foe. On the one hand, painful injuries lead to the activation of glial cells and immune cells in the PNS, releasing pro-inflammatory mediators, which contribute to the sensitization of nociceptors, leading to chronic pain; neuroinflammation in the CNS drives central sensitization and promotes the development of chronic pain. On the other hand, macrophages and glial cells of PNS and CNS promote pain resolution via anti-inflammatory mediators and specialized pro-resolving mediators (SPMs). In this review, we provide an overview of the current understanding of inflammation in the deterioration and resolution of pain. Further, we summarize a number of novel strategies that can be used to prevent and treat chronic pain by controlling inflammation. This comprehensive view of the relationship between inflammation and chronic pain and its specific mechanism will provide novel targets for the treatment of chronic pain.
Collapse
Affiliation(s)
- Xiao-Xia Fang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Meng-Nan Zhai
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Meixuan Zhu
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Cheng He
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Heng Wang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Juan Wang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Zhi-Jun Zhang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
7
|
Domenichiello AF, Wilhite BC, Nara P, Pitcher MH, Keyes GS, Mannes AJ, Bushnell MC, Ramsden CE. Biochemical and behavioral effects of decreasing dietary linoleic acid and increasing eicosapentaenoic acid and docosahexaenoic acid in a rat chronic monoarthrits model. Prostaglandins Leukot Essent Fatty Acids 2022; 187:102512. [PMID: 36347090 PMCID: PMC9729441 DOI: 10.1016/j.plefa.2022.102512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Clinical studies have demonstrated that decreasing linoleic acid (LA) while increasing eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in diets evokes an analgesic effect in headache sufferers. We utilized a rat chronic monoarthritis model to determine if these analgesic effects can be reproduced in rats and to and further probe potential analgesic mechanisms. We fed 8 rats a control diet (with fatty acid levels similar to standard US diets) and 8 rats a low LA diet with added EPA and DHA (H3L6 diet) and after 10 weeks, performed a unilateral intraarticular injection of Complete's Freund Adjuvant (CFA). We evaluated thermal and mechanical sensitivity as well as hind paw weight bearing prior to and at 4 and 20 days post CFA injection. At 28 days post CFA injection rats were euthanized and tissue collected. H3L6 diet fed rats had higher concentrations of EPA and DHA, as well as higher concentrations of oxidized lipids derived from these fatty acids, in hind paw and plasma, compared to control fed rats. LA and oxidized LA metabolites were lower in the plasma and hind paw of H3L6 compared to control fed rats. Diet did not affect thermal or mechanical sensitivity, nor did it affect hind paw weight bearing. In conclusion, the H3L6 diet evoked biochemical changes in rats but did not impact pain related behavioral measures in this chronic monoarthritis model.
Collapse
Affiliation(s)
- Anthony F Domenichiello
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), 10 Center Drive, 3D57, Bethesda, Baltimore, MD 20892, USA.
| | - Breanne C Wilhite
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - Pranavi Nara
- Department of Perioperative Medicine, NIH Clinical Center, NIH, Bethesda, MD, USA
| | - Mark H Pitcher
- National Center for Complimentary and Integrative Health, NIH, Bethesda, MD, USA
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), 10 Center Drive, 3D57, Bethesda, Baltimore, MD 20892, USA
| | - Andrew J Mannes
- Department of Perioperative Medicine, NIH Clinical Center, NIH, Bethesda, MD, USA
| | - M Catherine Bushnell
- National Center for Complimentary and Integrative Health, NIH, Bethesda, MD, USA
| | - Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), 10 Center Drive, 3D57, Bethesda, Baltimore, MD 20892, USA; National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| |
Collapse
|
8
|
Lai Y, Bäumer W, Meneses C, Roback DM, Robertson JB, Mishra SK, Lascelles BDX, Nolan MW. Irradiation of the Normal Murine Tongue Causes Upregulation and Activation of Transient Receptor Potential (TRP) Ion Channels. Radiat Res 2021; 196:331-344. [PMID: 34324688 DOI: 10.1667/rade-21-000103.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/15/2021] [Indexed: 11/03/2022]
Abstract
Signal transduction at sensory neurons occurs via transmembrane flux of cations, which is largely governed by the transient receptor potential (TRP) family of ion channels. It is unknown whether TRP channel activation contributes to the pain that accompanies radiation-induced oral mucositis. This study sought to characterize changes in TRP channel expression and function that occur in the locally irradiated tissues and afferent neurons of mice. Female CD-1 mice received single high-dose (27 Gy) tongue irradiation, or sham irradiation. Animals were euthanized either before overt glossitis developed (days 1 and 5 postirradiation), when glossitis was severe (day 11), or after mice had recovered (days 21 and 45). Tongue irradiation caused upregulation of the Trpv1 gene in trigeminal ganglia (TG) neurons. Other TRP genes (Trpv2, Trpv4, Trpa1, Trpm8) and Gfrα3 (which acts upstream of several TRP channels) were also upregulated in TGs and/or tongue tissue, in response to radiation. Ex vivo calcium imaging experiments demonstrated that the proportions of TG neurons responding to histamine (an activator of TRPV1, TRPV4 and TRPA1), TNF-α (an activator of TRPV1, TRPV2 and TRPV4), and capsaicin (a TRPV1 agonist), were increased as early as one day after tongue irradiation; these changes persisted for at least 21 days. In a subsequent experiment, we found that genetic deletion of TRPV1 mitigated weight loss (a surrogate marker of pain severity) in mice with severe glossitis. The results intimate that various TRP channels, and TRPV1 in particular, should be explored as analgesic targets for patients experiencing pain after oral irradiation.
Collapse
Affiliation(s)
- Yen Lai
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Wolfgang Bäumer
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
- Institute of Pharmacology and Toxicology, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Constanza Meneses
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina
- Translational Research in Pain, North Carolina State University, Raleigh, North Carolina
| | - Donald M Roback
- Department of Radiation Oncology, Rex Cancer Center, Raleigh, North Carolina
| | - James B Robertson
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Santosh K Mishra
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, North Carolina
| | - B Duncan X Lascelles
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Translational Research in Pain, North Carolina State University, Raleigh, North Carolina
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, North Carolina
| | - Michael W Nolan
- Department of Clinical Sciences, North Carolina State University, Raleigh, North Carolina
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
9
|
Ramsden CE, Zamora D, Faurot KR, MacIntosh B, Horowitz M, Keyes GS, Yuan ZX, Miller V, Lynch C, Honvoh G, Park J, Levy R, Domenichiello AF, Johnston A, Majchrzak-Hong S, Hibbeln JR, Barrow DA, Loewke J, Davis JM, Mannes A, Palsson OS, Suchindran CM, Gaylord SA, Mann JD. Dietary alteration of n-3 and n-6 fatty acids for headache reduction in adults with migraine: randomized controlled trial. BMJ 2021; 374:n1448. [PMID: 34526307 PMCID: PMC8244542 DOI: 10.1136/bmj.n1448] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To determine whether dietary interventions that increase n-3 fatty acids with and without reduction in n-6 linoleic acid can alter circulating lipid mediators implicated in headache pathogenesis, and decrease headache in adults with migraine. DESIGN Three arm, parallel group, randomized, modified double blind, controlled trial. SETTING Ambulatory, academic medical center in the United States over 16 weeks. PARTICIPANTS 182 participants (88% women, mean age 38 years) with migraines on 5-20 days per month (67% met criteria for chronic migraine). INTERVENTIONS Three diets designed with eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and linoleic acid altered as controlled variables: H3 diet (n=61)-increase EPA+DHA to 1.5 g/day and maintain linoleic acid at around 7% of energy; H3-L6 diet (n=61)-increase n-3 EPA+DHA to 1.5 g/day and decrease linoleic acid to ≤1.8% of energy; control diet (n=60)-maintain EPA+DHA at <150 mg/day and linoleic acid at around 7% of energy. All participants received foods accounting for two thirds of daily food energy and continued usual care. MAIN OUTCOME MEASURES The primary endpoints (week 16) were the antinociceptive mediator 17-hydroxydocosahexaenoic acid (17-HDHA) in blood and the headache impact test (HIT-6), a six item questionnaire assessing headache impact on quality of life. Headache frequency was assessed daily with an electronic diary. RESULTS In intention-to-treat analyses (n=182), the H3-L6 and H3 diets increased circulating 17-HDHA (log ng/mL) compared with the control diet (baseline-adjusted mean difference 0.6, 95% confidence interval 0.2 to 0.9; 0.7, 0.4 to 1.1, respectively). The observed improvement in HIT-6 scores in the H3-L6 and H3 groups was not statistically significant (-1.6, -4.2 to 1.0, and -1.5, -4.2 to 1.2, respectively). Compared with the control diet, the H3-L6 and H3 diets decreased total headache hours per day (-1.7, -2.5 to -0.9, and -1.3, -2.1 to -0.5, respectively), moderate to severe headache hours per day (-0.8, -1.2 to -0.4, and -0.7, -1.1 to -0.3, respectively), and headache days per month (-4.0, -5.2 to -2.7, and -2.0, -3.3 to -0.7, respectively). The H3-L6 diet decreased headache days per month more than the H3 diet (-2.0, -3.2 to -0.8), suggesting additional benefit from lowering dietary linoleic acid. The H3-L6 and H3 diets altered n-3 and n-6 fatty acids and several of their nociceptive oxylipin derivatives in plasma, serum, erythrocytes or immune cells, but did not alter classic headache mediators calcitonin gene related peptide and prostaglandin E2. CONCLUSIONS The H3-L6 and H3 interventions altered bioactive mediators implicated in headache pathogenesis and decreased frequency and severity of headaches, but did not significantly improve quality of life. TRIAL REGISTRATION ClinicalTrials.gov NCT02012790.
Collapse
Affiliation(s)
- Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daisy Zamora
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
- Department of Psychiatry, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Keturah R Faurot
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Beth MacIntosh
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Metabolic and Nutrition Research Core, UNC Medical Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark Horowitz
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Zhi-Xin Yuan
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Vanessa Miller
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chanee Lynch
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gilson Honvoh
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinyoung Park
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Russell Levy
- Cytokine Analysis Core, UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anthony F Domenichiello
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Angela Johnston
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sharon Majchrzak-Hong
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - Joseph R Hibbeln
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - David A Barrow
- Cytokine Analysis Core, UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James Loewke
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - John M Davis
- Department of Psychiatry, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Mannes
- Department of Perioperative Medicine, NIH Clinical Center, Bethesda, MD, USA
| | - Olafur S Palsson
- Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chirayath M Suchindran
- Department of Biostatistics, Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Susan A Gaylord
- Department of Physical Medicine and Rehabilitation, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J Douglas Mann
- Department of Neurology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
10
|
Abstract
Chronic posttraumatic headache (PTH) is among the most common and disabling sequelae of traumatic brain injury (TBI). Current PTH treatments are often only partially effective and have problematic side effects. We previously showed in a small randomized trial of patients with chronic nontraumatic headaches that manipulation of dietary fatty acids decreased headache frequency, severity, and pain medication use. Pain reduction was associated with alterations in oxylipins derived from n-3 and n-6 fatty acids, suggesting that oxylipins could potentially mediate clinical pain reduction. The objective of this study was to investigate whether circulating oxylipins measured in the acute setting after TBI could serve as prognostic biomarkers for developing chronic PTH. Participants enrolled in the Traumatic Head Injury Neuroimaging Classification Protocol provided serum within 3 days of TBI and were followed up at 90 days postinjury with a neurobehavioral symptom inventory (NSI) and satisfaction with life survey. Liquid chromatography-tandem mass spectrometry methods profiled 39 oxylipins derived from n-3 docosahexaenoic acid (DHA), and n-6 arachidonic acid and linoleic acid. Statistical analyses assessed the association of oxylipins with headache severity (primary outcome, measured by headache question on NSI) as well as associations between oxylipins and total NSI or satisfaction with life survey scores. Among oxylipins, 4-hydroxy-DHA and 19,20-epoxy-docosapentaenoate (DHA derivatives) were inversely associated with headache severity, and 11-hydroxy-9-epoxy-octadecenoate (a linoleic acid derivative) was positively associated with headache severity. These findings support a potential for DHA-derived oxylipins as prognostic biomarkers for development of chronic PTH.
Collapse
|
11
|
Khasabova IA, Golovko MY, Golovko SA, Simone DA, Khasabov SG. Intrathecal administration of Resolvin D1 and E1 decreases hyperalgesia in mice with bone cancer pain: Involvement of endocannabinoid signaling. Prostaglandins Other Lipid Mediat 2020; 151:106479. [PMID: 32745525 PMCID: PMC7669692 DOI: 10.1016/j.prostaglandins.2020.106479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/07/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Pain produced by bone cancer is often severe and difficult to treat. Here we examined effects of Resolvin D1 (RvD1) or E1 (RvE1), antinociceptive products of ω-3 polyunsaturated fatty acids, on cancer-induced mechanical allodynia and heat hyperalgesia. Experiments were performed using a mouse model of bone cancer produced by implantation of osteolytic ficrosarcoma into and around the calcaneus bone. Mechanical allodynia and heat hyperalgesia in the tumor-bearing paw were assessed by measuring withdrawal responses to a von Frey monofilament and to radiant heat applied on the plantar hind paw. RvD1, RvE1, and cannabinoid receptor antagonists were injected intrathecally. Spinal content of endocannabinoids was evaluated using UPLC-MS/MS analysis. RvD1 and RvE1 had similar antinociceptive potencies. ED50s for RvD1 and RvE1 in reducing mechanical allodynia were 0.2 pg (0.53 fmol) and 0.6 pg (1.71 fmol), respectively, and were 0.3 pg (0.8 fmol) and 0.2 pg (0.57 fmol) for reducing heat hyperalgesia. Comparisons of dose-response relationships showed equal efficacy for reducing mechanical allodynia, however, efficacy for reducing heat hyperalgesia was greater for of RvD1. Using UPLC-MS/MS we determined that RvD1, but not RvE1, increased levels of the endocannabinoids Anandamide and 2-Arachidonoylglycerol in the spinal cord. Importantly, Resolvins did not alter acute nociception or motor function in naïve mice. Our data indicate, that RvD1 and RvE1 produce potent antiallodynia and antihyperalgesia in a model of bone cancer pain. RvD1 also triggers spinal upregulation of endocannabinoids that produce additional antinociception predominantly through CB2 receptors.
Collapse
Affiliation(s)
- Iryna A Khasabova
- Department of Diagnostic and Biological Sciences, University of Minnesota, School of Dentistry, Minneapolis, MN, USA
| | - Mikhail Y Golovko
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Svetlana A Golovko
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, University of Minnesota, School of Dentistry, Minneapolis, MN, USA
| | - Sergey G Khasabov
- Department of Diagnostic and Biological Sciences, University of Minnesota, School of Dentistry, Minneapolis, MN, USA.
| |
Collapse
|
12
|
Molecular Pathways Linking Oxylipins to Nociception in Rats. THE JOURNAL OF PAIN 2020; 22:275-299. [PMID: 33031942 DOI: 10.1016/j.jpain.2020.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/31/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Oxylipins are lipid peroxidation products that participate in nociceptive, inflammatory, and vascular responses to injury. Effects of oxylipins depend on tissue-specific differences in accumulation of precursor polyunsaturated fatty acids and the expression of specific enzymes to transform the precursors. The study of oxylipins in nociception has presented technical challenges leading to critical knowledge gaps in the way these molecules operate in nociception. We applied a systems-based approach to characterize oxylipin precursor fatty acids, and expression of genes coding for proteins involved in biosynthesis, transport, signaling and inactivation of pro- and antinociceptive oxylipins in pain circuit tissues. We further linked these pathways to nociception by demonstrating intraplantar carrageenan injection induced gene expression changes in oxylipin biosynthetic pathways. We determined functional-biochemical relevance of the proposed pathways in rat hind paw and dorsal spinal cord by measuring basal and stimulated levels of oxylipins throughout the time-course of carrageenan-induced inflammation. Finally, when oxylipins were administered by intradermal injection we observed modulation of nociceptive thermal hypersensitivity, providing a functional-behavioral link between oxylipins, their molecular biosynthetic pathways, and involvement in pain and nociception. Together, these findings advance our understanding of molecular lipidomic systems linking oxylipins and their precursors to nociceptive and inflammatory signaling pathways in rats. PERSPECTIVE: We applied a systems approach to characterize molecular pathways linking precursor lipids and oxylipins to nociceptive signaling. This systematic, quantitative evaluation of the molecular pathways linking oxylipins to nociception provides a framework for future basic and clinical research investigating the role of oxylipins in pain.
Collapse
|
13
|
Mecklenburg J, Zou Y, Wangzhou A, Garcia D, Lai Z, Tumanov AV, Dussor G, Price TJ, Akopian AN. Transcriptomic sex differences in sensory neuronal populations of mice. Sci Rep 2020; 10:15278. [PMID: 32943709 PMCID: PMC7499251 DOI: 10.1038/s41598-020-72285-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
Many chronic pain conditions show sex differences in their epidemiology. This could be attributed to sex-dependent differential expression of genes (DEGs) involved in nociceptive pathways, including sensory neurons. This study aimed to identify sex-dependent DEGs in estrous female versus male sensory neurons, which were prepared by using different approaches and ganglion types. RNA-seq on non-purified sensory neuronal preparations, such as whole dorsal root ganglion (DRG) and hindpaw tissues, revealed only a few sex-dependent DEGs. Sensory neuron purification increased numbers of sex-dependent DEGs. These DEG sets were substantially influenced by preparation approaches and ganglion types [DRG vs trigeminal ganglia (TG)]. Percoll-gradient enriched DRG and TG neuronal fractions produced distinct sex-dependent DEG groups. We next isolated a subset of sensory neurons by sorting DRG neurons back-labeled from paw and thigh muscle. These neurons have a unique sex-dependent DEG set, yet there is similarity in biological processes linked to these different groups of sex-dependent DEGs. Female-predominant DEGs in sensory neurons relate to inflammatory, synaptic transmission and extracellular matrix reorganization processes that could exacerbate neuro-inflammation severity, especially in TG. Male-selective DEGs were linked to oxidative phosphorylation and protein/molecule metabolism and production. Our findings catalog preparation-dependent sex differences in neuronal gene expressions in sensory ganglia.
Collapse
Affiliation(s)
- Jennifer Mecklenburg
- Department of Endodontics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Yi Zou
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Dawn Garcia
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Alexei V Tumanov
- Departments of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Armen N Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA.
- Department of Pharmacology, The School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
14
|
Iwata K, Sessle BJ. The Evolution of Neuroscience as a Research Field Relevant to Dentistry. J Dent Res 2020; 98:1407-1417. [PMID: 31746682 DOI: 10.1177/0022034519875724] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The field of neuroscience did not exist as such when the Journal of Dental Research was founded 100 y ago. It has emerged as an important scientific field relevant to dentistry in view of the many neurally based functions manifested in the orofacial area (e.g., pain, taste, chewing, swallowing, salivation). This article reviews many of the novel insights that have been gained through neuroscience research into the neural basis of these functions and their clinical relevance to the diagnosis and management of pain and sensorimotor disorders. These include the neural pathways and brain circuitry underlying each of these functions and the role of nonneural as well as neural processes and their "plasticity" in modulating these functions and allowing for adaptation to tissue injury and pain and for learning or rehabilitation of orofacial functions.
Collapse
Affiliation(s)
- K Iwata
- Department of Physiology, Nihon University, School of Dentistry, Tokyo, Japan
| | - B J Sessle
- Faculty of Dentistry and Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Filippini HF, Molska GR, Zanjir M, Arudchelvan Y, Gong SG, Campos MM, Avivi-Arber L, Sessle BJ. Toll-Like Receptor 4 in the Rat Caudal Medulla Mediates Tooth Pulp Inflammatory Pain. Front Neurosci 2020; 14:643. [PMID: 32655361 PMCID: PMC7324534 DOI: 10.3389/fnins.2020.00643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/25/2020] [Indexed: 12/20/2022] Open
Abstract
The aims of this study were to investigate if Toll-like receptor 4 (TLR4) is expressed in the medullary dorsal horn (MDH) and if medullary application of a TLR4 antagonist (lipopolysaccharides from Rhodobacter sphaeroides, LPS-RS) can attenuate changes in nociceptive sensorimotor responses or TLR4 expression that might be evoked by mustard oil (MO) application to the right maxillary first molar tooth pulp. Of 41 adult male Sprague-Dawley rats used in the study, 23 received intrathecal application of the TLR4 antagonist LPS-RS (25 μg/10 μl; LPS-RS group) or isotonic saline (10 μl; vehicle control group) 10 min before pulpal application of MO (95%; 0.2 μl). Bilateral electromyographic (EMG) activities of the anterior digastric and masseter muscles were recorded continuously before and until 15 min after the MO application to the pulp. In 6 of these 23 rats and an additional 18 rats, the caudal medulla containing the ipsilateral and contralateral MDH was removed after euthanasia for subsequent Western Blot analysis of TLR4 expression in LPS-RS (n = 8) and vehicle (n = 8) groups and a naïve group (n = 8). The % change from baseline in the MO-evoked EMG activities within the anterior digastric muscles were significantly smaller in the LPS-RS group than the control group (two-way ANOVA, post hoc Bonferroni, P < 0.0001). Western Blot analysis revealed similar levels of TLR4 expression in the caudal medulla of the naïve, vehicle and LPS-RS groups. These novel findings suggest that TLR4 signaling in the caudal medulla may mediate MO-induced acute dental inflammatory pain in rats.
Collapse
Affiliation(s)
- Helena F Filippini
- Programa de Pós-graduação em Odontologia, Escola de Ciência da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | | | - Maryam Zanjir
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | | | - Siew-Ging Gong
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Maria M Campos
- Programa de Pós-graduação em Odontologia, Escola de Ciência da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Limor Avivi-Arber
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.,Centre for the Study of Pain, University of Toronto, Toronto, ON, Canada
| | - Barry J Sessle
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.,Centre for the Study of Pain, University of Toronto, Toronto, ON, Canada.,Departament of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Doolen S, Keyes GS, Ramsden CE. Hydroxy-epoxide and keto-epoxide derivatives of linoleic acid activate trigeminal neurons. NEUROBIOLOGY OF PAIN 2020; 7:100046. [PMID: 32478201 PMCID: PMC7248286 DOI: 10.1016/j.ynpai.2020.100046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 02/08/2023]
Abstract
11-hydroxy- and 11-keto-epoxide-LA derivatives elicit Ca2+ transients in trigeminal neuron subpopulations. 11H-12,13E-LA, 11 K-12,13E-LA, and 11H-9,10E-LA produce Ca2+ responses in higher proportions of neurons than linoleic acid or 9-HODE. 11-hydroxy-epoxide- and 11-keto-epoxide derivatives of linoleic acid potentially contribute to nociception.
Endogenous lipid mediators are proposed to contribute to headache and facial pain by activating trigeminal neurons (TN). We recently identified 11-hydroxy-epoxide- and 11-keto-epoxide derivatives of linoleic acid (LA) that are present in human skin and plasma and potentially contribute to nociception. Here we expand upon initial findings by examining the effects of 11-hydroxy- and 11-keto-epoxide-LA derivatives on TN activation in comparison to LA, the LA derivative [9-hydroxy-octadecadienoic acid (9-HODE)] and prostaglandin E2 (PGE2). 11-hydroxy- and 11-keto-epoxide-LA derivatives elicited Ca2+ transients in TN subpopulations. The proportion of neurons responding to test compounds (5 μM, 5 min) ranged from 16.2 ± 3.8 cells (11 K-9,10E-LA) to 34.1 ± 2.4 cells (11H-12,13E-LA). LA and 9-HODE (5 μM, 5 min) elicited responses in 11.6 ± 3.1% and 9.7 ± 3.4% of neurons, respectively. 11H-12,13E-LA, 11K-12,13E-LA, and 11H-9,10E-LA produced Ca2+ responses in significantly higher proportions of neurons compared to either LA or 9-HODE (F (6, 36) = 5.12, P = 0.0007). 11H-12,13E-LA and 11H-9,10E-LA increased proportions of responsive neurons in a concentration-dependent fashion, similar to PGE2. Most sensitive neurons responded to additional algesic agents (32.9% to capsaicin, 40.1% to PGE2, 58.0% to AITC), however 20.6% did not respond to any other agent. In summary, 11-hydroxy-epoxide derivatives of LA increase trigeminal neuron excitability, suggesting a potential role in headache or facial pain.
Collapse
Key Words
- 11-HEL, 11-hydroxy-epoxide-linoleic acid
- 11H-12,13E-LA, 11-hydroxy-12,13-trans-epoxy-(9Z)-octadecenoate
- 11H-9,10E-LA, 11-hydroxy-9,10-trans-epoxy-(12Z)-octadecenoate
- 9-HODE, 9-hydroxy-octadecadienoic acid
- CGRP, calcitonin gene related peptide
- DiHOMEs, dihydroxy-octadecenoic acids
- EpOMEs, epoxy-octadecenoic
- HODEs, octadecadienoic acids
- HpODEs, hydroperoxy-octadecadienoic acids
- Hyperalgesia
- LA, linoleic acid
- Linoleic acid
- Oxylipin
- PGE2, prostaglandin E2
- Pain
- Peroxidation
- TN, trigeminal neuron
- aCSF, artificial cerebrospinal fluid
Collapse
Affiliation(s)
- Suzanne Doolen
- Department of Physiology, University of Kentucky, 800 Rose Street, Lexington, KY 40536-0298, United States
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD 21224, USA
| | - Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD 21224, USA.,Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20814, USA
| |
Collapse
|
17
|
Foster JR, Ueno S, Chen MX, Harvey J, Dowell SJ, Irving AJ, Brown AJ. N-Palmitoylglycine and other N-acylamides activate the lipid receptor G2A/GPR132. Pharmacol Res Perspect 2019; 7:e00542. [PMID: 31768260 PMCID: PMC6868653 DOI: 10.1002/prp2.542] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/01/2019] [Accepted: 10/15/2019] [Indexed: 01/22/2023] Open
Abstract
The G-protein-coupled receptor GPR132, also known as G2A, is activated by 9-hydroxyoctadecadienoic acid (9-HODE) and other oxidized fatty acids. Other suggested GPR132 agonists including lysophosphatidylcholine (LPC) have not been readily reproduced. Here, we identify N-acylamides in particular N-acylglycines, as lipid activators of GPR132 with comparable activity to 9-HODE. The order-of-potency is N-palmitoylglycine > 9-HODE ≈ N-linoleoylglycine > linoleamide > N-oleoylglycine ≈ N-stereoylglycine > N-arachidonoylglycine > N-docosehexanoylglycine. Physiological concentrations of N-acylglycines in tissue are sufficient to activate GPR132. N-linoleoylglycine and 9-HODE also activate rat and mouse GPR132, despite limited sequence conservation to human. We describe pharmacological tools for GPR132, identified through drug screening. SKF-95667 is a novel GPR132 agonist. SB-583831 and SB-583355 are peptidomimetic molecules containing core amino acids (glycine and phenylalanine, respectively), and structurally related to previously described ligands. A telmisartan analog, GSK1820795A, antagonizes the actions of N-acylamides at GPR132. The synthetic cannabinoid CP-55 940 also activates GPR132. Molecular docking to a homology model suggested a site for lipid binding, predicting the acyl side-chain to extend into the membrane bilayer between TM4 and TM5 of GPR132. Small-molecule ligands are envisaged to occupy a "classical" site encapsulated in the 7TM bundle. Structure-directed mutagenesis indicates a critical role for arginine at position 203 in transmembrane domain 5 to mediate GPR132 activation by N-acylamides. Our data suggest distinct modes of binding for small-molecule and lipid agonists to the GPR132 receptor. Antagonists, such as those described here, will be vital to understand the physiological role of this long-studied target.
Collapse
Affiliation(s)
- James R. Foster
- GlaxoSmithKline R&D Ltd, Medicines Research CentreStevenageUK
- School of MedicineNinewells Hospital and Medical SchoolDundee UniversityDundeeUK
| | - Shohta Ueno
- GlaxoSmithKline R&D Ltd, Medicines Research CentreStevenageUK
- Present address:
RegeneronUxbridgeUK
| | - Mao Xiang Chen
- GlaxoSmithKline R&D Ltd, Medicines Research CentreStevenageUK
| | - Jenni Harvey
- School of MedicineNinewells Hospital and Medical SchoolDundee UniversityDundeeUK
| | - Simon J. Dowell
- GlaxoSmithKline R&D Ltd, Medicines Research CentreStevenageUK
| | - Andrew J. Irving
- School of Biomolecular and Biomedical ScienceThe Conway InstituteUniversity College DublinDublinIreland
| | - Andrew J. Brown
- GlaxoSmithKline R&D Ltd, Medicines Research CentreStevenageUK
| |
Collapse
|
18
|
Price TJ, Gold MS. From Mechanism to Cure: Renewing the Goal to Eliminate the Disease of Pain. PAIN MEDICINE 2019; 19:1525-1549. [PMID: 29077871 DOI: 10.1093/pm/pnx108] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective Persistent pain causes untold misery worldwide and is a leading cause of disability. Despite its astonishing prevalence, pain is undertreated, at least in part because existing therapeutics are ineffective or cause intolerable side effects. In this review, we cover new findings about the neurobiology of pain and argue that all but the most transient forms of pain needed to avoid tissue damage should be approached as a disease where a cure can be the goal of all treatment plans, even if attaining this goal is not yet always possible. Design We reviewed the literature to highlight recent advances in the area of the neurobiology of pain. Results We discuss barriers that are currently hindering the achievement of this goal, as well as the development of new therapeutic strategies. We also discuss innovations in the field that are creating new opportunities to treat and even reverse persistent pain, some of which are in late-phase clinical trials. Conclusion We conclude that the confluence of new basic science discoveries and development of new technologies are creating a path toward pain therapeutics that should offer significant hope of a cure for patients and practitioners alike. Classification of Evidence. Our review points to new areas of inquiry for the pain field to advance the goal of developing new therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, Texas
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
Byers MR. Chewing causes rapid changes in immunoreactive nerve patterns in rat molar teeth: Implications for dental proprioception and pain. Arch Oral Biol 2019; 107:104511. [PMID: 31445382 DOI: 10.1016/j.archoralbio.2019.104511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE This study tests the hypothesis that normal use of teeth (chewing) causes changes in immunoreactive-(IR) patterns for endings of large Aβ and CGRP axons in rat molar cusps. DESIGN First, a new paradigm to test chewing in adult male rats was developed. Then IR patterns for large dental axons were analysed for a calcium-binding protein, parvalbumin (PV), heavy neurofilament protein-200 (NFP), and vesicle-release molecule synaptophysin (SYN) that all typify large dental axons and proprioceptors for comparison with endings of CGRP-IR neuropeptide axons. The behavior groups were: (1) daytime sleeping/fasting (Group:SF); (2) brief feeding after 8-11 h of daytime sleeping/fasting (Group:SF-C); (3) normal nocturnal feeding (Group:N); (4) nocturnal fasting (Group:NF); (5) brief feeding/chewing after nocturnal fasting (Group:NF-C). RESULTS Nerve endings with NFP-, PV-, or SYN-IR were lost or altered in pulp and dentin in all chewing groups. Other endings with CGRP-IR were near those with PV-, NFP- and SYN-IR at the pulp-dentin border and in dentin, and they also lost immunoreactivity in all chewing groups. The special beaded regions along the crown pulp/dentin borders lost neural labeling in all chewing groups. Nerves of molar roots and periodontal ligament were not changed. CONCLUSIONS Rapid neural reactions to chewing show extensive, reversible, non-nociceptive depletions of crown innervation. Those changes were rapid enough to occur during normal feeding followed by recovery during rest. The new dental paradigm related to chewing and fasting allows dissection of intradental proprioceptive-like mechanisms during normal tooth functions for comparison with nociceptive and mechanosensitive reactions after injury or inflammation.
Collapse
Affiliation(s)
- Margaret R Byers
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195-6540 USA.
| |
Collapse
|
20
|
Developmental Axon Degeneration Requires TRPV1-Dependent Ca 2+ Influx. eNeuro 2019; 6:eN-NWR-0019-19. [PMID: 30838324 PMCID: PMC6399429 DOI: 10.1523/eneuro.0019-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/16/2022] Open
Abstract
Development of the nervous system relies on a balance between axon and dendrite growth and subsequent pruning and degeneration. The developmental degeneration of dorsal root ganglion (DRG) sensory axons has been well studied in part because it can be readily modeled by removing the trophic support by nerve growth factor (NGF) in vitro. We have recently reported that axonal fragmentation induced by NGF withdrawal is dependent on Ca2+, and here, we address the mechanism of Ca2+ entry required for developmental axon degeneration of mouse embryonic DRG neurons. Our results show that the transient receptor potential vanilloid family member 1 (TRPV1) cation channel plays a critical role mediating Ca2+ influx in DRG axons withdrawn from NGF. We further demonstrate that TRPV1 activation is dependent on reactive oxygen species (ROS) generation that is driven through protein kinase C (PKC) and NADPH oxidase (NOX)-dependent pathways that become active upon NGF withdrawal. These findings demonstrate novel mechanistic links between NGF deprivation, PKC activation, ROS generation, and TRPV1-dependent Ca2+ influx in sensory axon degeneration.
Collapse
|
21
|
Jensen JR, Pitcher MH, Yuan ZX, Ramsden CE, Domenichiello AF. Concentrations of oxidized linoleic acid derived lipid mediators in the amygdala and periaqueductal grey are reduced in a mouse model of chronic inflammatory pain. Prostaglandins Leukot Essent Fatty Acids 2018; 135:128-136. [PMID: 30103924 PMCID: PMC6269101 DOI: 10.1016/j.plefa.2018.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 12/26/2022]
Abstract
Chronic pain is both a global public health concern and a serious source of personal suffering for which current treatments have limited efficacy. Recently, oxylipins derived from linoleic acid (LA), the most abundantly consumed polyunsaturated fatty acid in the modern diet, have been implicated as mediators of pain in the periphery and spinal cord. However, oxidized linoleic acid derived mediators (OXLAMs) remain understudied in the brain, particularly during pain states. In this study, we employed a mouse model of chronic inflammatory pain followed by a targeted lipidomic analysis of the animals' amygdala and periaqueductal grey (PAG) using LC-MS/MS to investigate the effect of chronic inflammatory pain on oxylipin concentrations in these two brain nuclei known to participate in pain sensation and perception. From punch biopsies of these brain nuclei, we detected twelve OXLAMs in both the PAG and amygdala and one arachidonic acid derived mediator, 15-HETE, in the amygdala only. In the amygdala, we observed an overall decrease in the concentration of the majority of OXLAMs detected, while in the PAG the concentrations of only the epoxide LA derived mediators, 9,10-EpOME and 12,13-EpOME, and one trihydroxy LA derived mediator, 9,10,11-TriHOME, were reduced. This data provides the first evidence that OXLAM concentrations in the brain are affected by chronic pain, suggesting that OXLAMs may be relevant to pain signaling and adaptation to chronic pain in pain circuits in the brain and that the current view of OXLAMs in nociception derived from studies in the periphery is incomplete.
Collapse
Affiliation(s)
- J R Jensen
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States
| | - M H Pitcher
- National Center for Complementary and Integrative Health, NIH, Bethesda, MD, United States
| | - Z X Yuan
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States
| | - C E Ramsden
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States; Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, United States
| | - A F Domenichiello
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States.
| |
Collapse
|
22
|
Madej MG, Ziegler CM. Dawning of a new era in TRP channel structural biology by cryo-electron microscopy. Pflugers Arch 2018; 470:213-225. [PMID: 29344776 DOI: 10.1007/s00424-018-2107-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022]
Abstract
Cryo-electron microscopy (cryo-EM) permits the determination of atomic protein structures by averaging large numbers of individual projection images recorded at cryogenic temperatures-a method termed single-particle analysis. The cryo-preservation traps proteins within a thin glass-like ice layer, making literally a freeze image of proteins in solution. Projections of randomly adopted orientations are merged to reconstruct a 3D density map. While atomic resolution for highly symmetric viruses was achieved already in 2009, the development of new sensitive and fast electron detectors has enabled cryo-EM for smaller and asymmetrical proteins including fragile membrane proteins. As one of the most important structural biology methods at present, cryo-EM was awarded in October 2017 with the Nobel Prize in Chemistry. The molecular understanding of Transient-Receptor-Potential (TRP) channels has been boosted tremendously by cryo-EM single-particle analysis. Several near-atomic and atomic structures gave important mechanistic insights, e.g., into ion permeation and selectivity, gating, as well as into the activation of this enigmatic and medically important membrane protein family by various chemical and physical stimuli. Lastly, these structures have set the starting point for the rational design of TRP channel-targeted therapeutics to counteract life-threatening channelopathies. Here, we attempt a brief introduction to the method, review the latest advances in cryo-EM structure determination of TRP channels, and discuss molecular insights into the channel function based on the wealth of TRP channel cryo-EM structures.
Collapse
Affiliation(s)
- M Gregor Madej
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstrasse 31, D-93053, Regensburg, Germany
| | - Christine M Ziegler
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstrasse 31, D-93053, Regensburg, Germany.
| |
Collapse
|
23
|
(−)-α-Bisabolol reduces orofacial nociceptive behavior in rodents. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:187-195. [DOI: 10.1007/s00210-016-1319-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/18/2016] [Indexed: 12/31/2022]
|
24
|
Affiliation(s)
- R. Dubner
- Department of Neural and Pain Sciences, University of Maryland Dental School, Baltimore, MD, USA
| |
Collapse
|