1
|
Bandarupalli R, Roth R, Klipp RC, Bankston JR, Li J. Molecular Insights into Single-Chain Lipid Modulation of Acid-Sensing Ion Channel 3. J Phys Chem B 2024; 128:12685-12697. [PMID: 39666997 DOI: 10.1021/acs.jpcb.4c04289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Polyunsaturated fatty acids (PUFAs) and their analogs play a significant role in modulating the activity of diverse ion channels, and recent studies show that these lipids potentiate acid-sensing ion channels (ASICs), leading to increased activity. The potentiation of the channel stems from multiple gating changes, but the exact mechanism of these effects remains uncertain. We posit a mechanistic explanation for one of these changes in channel function, the increase in the maximal current, by applying a combination of electrophysiology and all-atom molecular dynamics simulations on open-state hASIC3. Microsecond-scale simulations were performed on open-state hASIC3 in the absence and presence of a PUFA, docosahexaenoic acid (DHA), and a PUFA analogue, N-arachidonyl glycine (AG). Intriguingly, our simulations in the absence of PUFA or PUFA analogs reveal that a tail from the membrane phospholipid POPC inserts itself into the pore of the channel through lateral fenestrations on the sides of the transmembrane segments, obstructing ion permeation through the channel. The binding of either DHA or AG prevented POPC from accessing the pore in our simulations, which relied on the block of ionic conduction by phospholipids. Finally, we use single-channel recording to show that DHA increases the amplitude of the single-channel currents in ASIC3, which is consistent with our hypothesis that PUFAs relieve the pore block of the channel induced by POPCs. Together, these findings offer a potential mechanistic explanation of how PUFAs modulate the ASIC maximal current, revealing a novel mechanism of action for PUFA-induced modulation of ion channels.
Collapse
Affiliation(s)
- Ramya Bandarupalli
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi 38677, United States
| | - Rebecca Roth
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Robert C Klipp
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Jing Li
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi 38677, United States
| |
Collapse
|
2
|
Bandarupalli R, Roth R, Klipp RC, Bankston JR, Li J. Molecular Insights into Single Chain Lipid Modulation of Acid-Sensing Ion Channel 3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610156. [PMID: 39257759 PMCID: PMC11383688 DOI: 10.1101/2024.08.29.610156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Polyunsaturated fatty acids (PUFAs) and their analogs play a significant role in modulating the activity of diverse ion channels, and recent studies show that these lipids potentiate acid-sensing ion channels (ASICs), leading to increased activity. The potentiation of the channel stems from multiple gating changes, but the exact mechanism of these effects remains uncertain. We posit a mechanistic explanation for one of these changes in channel function, the increase in the maximal current, by applying a combination of electrophysiology and all-atom molecular dynamics simulations on the open-state hASIC3. Microsecond-scale simulations were performed on open-state hASIC3 in the absence and presence of a PUFA, docosahexaenoic acid (DHA), and a PUFA analog, N-arachidonyl glycine (AG). Intriguingly, our simulations in the absence of PUFA or PUFA analogs reveal that a tail from the membrane phospholipid POPC inserts itself into the pore of the channel through lateral fenestrations on the sides of the transmembrane segments, obstructing ion permeation through the channel. The binding of either DHA or AG prevented POPC from accessing the pore in our simulations, relieving the block of ionic conduction by phospholipids. Finally, we use the single-channel recording to show that DHA increases the amplitude of the single-channel currents in ASIC3, which is consistent with our hypothesis that PUFAs relieve the pore block of the channel induced by POPCs. Together, these findings offer a potential mechanistic explanation of how PUFAs modulate ASIC maximal current, revealing a novel mechanism of action for PUFA-induced modulation of ion channels.
Collapse
Affiliation(s)
- Ramya Bandarupalli
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS
| | - Rebecca Roth
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Robert C Klipp
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jing Li
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS
| |
Collapse
|
3
|
Bang J, Kim G, Young Park S, Jung HR, Kim SH, Kim JM. GCSB-5 regulates inflammatory arthritis and pain by modulating the mitogen-activated protein kinase signaling pathway in a murine model of rheumatoid arthritis. Arch Rheumatol 2023; 38:566-578. [PMID: 38125068 PMCID: PMC10728744 DOI: 10.46497/archrheumatol.2023.9643] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/15/2022] [Indexed: 12/23/2023] Open
Abstract
Objectives This study aimed to determine whether GCSB-5 has anti-inflammatory and antinociceptive effects in mice with collagen-induced arthritis (CIA), an animal model of rheumatoid arthritis (RA), and investigate the influence of GCSB-5 on the mitogen-activated protein kinase (MAPK) pathway. Materials and methods The experimental animal study was designed to include five groups: CIA mice treated with GCSB-5 (300 mg/kg), GCSB-5 (600 mg/kg), celecoxib (60 mg/kg), or saline for four weeks, and nontreated control mice. The clinical severity of arthritis was scored. Nociceptive thresholds were measured by using a von Frey dynamic plantar analgesimeter. The MAPK pathway was evaluated in mouse synovium. The expression of channels associated with pain signaling was assessed by western blot and immunohistochemical staining. Results GCSB-5 treatment diminished the severity of clinical arthritis and increased the nociceptive threshold in mice with CIA. Celecoxib, a positive control drug, also showed comparable changes. Clinical arthritis scores were inversely related to mechanical thresholds. GCSB-5 administration decreased the levels of anti-type II collagen antibody and inflammatory cytokines in the sera of mice with CIA. Furthermore, ERK, p38 MAPK, and JNK phosphorylation were downregulated and TRPV1 and ASIC3 expression were decreased in the synovium of GCSB-5-treated mice compared to salinetreated mice. Interleukin-6-induced TRPV1 and ASIC3 upregulation were also inhibited by GCSB-5 in human RA fibroblast-like synoviocytes in vitro. Conclusion GCSB-5 decreased inflammatory arthritis and pain in a murine model of RA. The results present evidence that GCSB-5 may be beneficial for relieving pain as well as decreasing inflammation in autoimmune arthritis, such as RA.
Collapse
Affiliation(s)
- Jihye Bang
- Department of Chronic Disease Convergence Research, Division of Allergy and Respiratory Disease Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Gyeonghwa Kim
- Division of Rheumatology, Department of Internal Medicine, School of Medicine & Institute for Medical Science, Keimyung University, Daegu, South Korea
| | - Soo Young Park
- Department of Internal Medicine,School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Hye Ra Jung
- Department of Pathology, Keimyung University School of Medicine, Daegu, South Korea
| | - Sang-Hyon Kim
- Division of Rheumatology, Department of Internal Medicine, School of Medicine & Institute for Medical Science, Keimyung University, Daegu, South Korea
| | - Ji-Min Kim
- Division of Rheumatology, Department of Internal Medicine, School of Medicine & Institute for Medical Science, Keimyung University, Daegu, South Korea
| |
Collapse
|
4
|
Obeidat AM, Wood MJ, Adamczyk NS, Ishihara S, Li J, Wang L, Ren D, Bennett DA, Miller RJ, Malfait AM, Miller RE. Piezo2 expressing nociceptors mediate mechanical sensitization in experimental osteoarthritis. Nat Commun 2023; 14:2479. [PMID: 37120427 PMCID: PMC10148822 DOI: 10.1038/s41467-023-38241-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 04/17/2023] [Indexed: 05/01/2023] Open
Abstract
Non-opioid targets are needed for addressing osteoarthritis pain, which is mechanical in nature and associated with daily activities such as walking and climbing stairs. Piezo2 has been implicated in the development of mechanical pain, but the mechanisms by which this occurs remain poorly understood, including the role of nociceptors. Here we show that nociceptor-specific Piezo2 conditional knock-out mice were protected from mechanical sensitization associated with inflammatory joint pain in female mice, joint pain associated with osteoarthritis in male mice, as well as both knee swelling and joint pain associated with repeated intra-articular injection of nerve growth factor in male mice. Single cell RNA sequencing of mouse lumbar dorsal root ganglia and in situ hybridization of mouse and human lumbar dorsal root ganglia revealed that a subset of nociceptors co-express Piezo2 and Ntrk1 (the gene that encodes the nerve growth factor receptor TrkA). These results suggest that nerve growth factor-mediated sensitization of joint nociceptors, which is critical for osteoarthritic pain, is also dependent on Piezo2, and targeting Piezo2 may represent a therapeutic option for osteoarthritis pain control.
Collapse
Affiliation(s)
- Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Matthew J Wood
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Natalie S Adamczyk
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Jun Li
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, Chicago, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center and Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Chicago, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA.
| |
Collapse
|
5
|
Malange KF, Navia-Pelaez JM, Dias EV, Lemes JBP, Choi SH, Dos Santos GG, Yaksh TL, Corr M. Macrophages and glial cells: Innate immune drivers of inflammatory arthritic pain perception from peripheral joints to the central nervous system. FRONTIERS IN PAIN RESEARCH 2022; 3:1018800. [PMID: 36387416 PMCID: PMC9644179 DOI: 10.3389/fpain.2022.1018800] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Millions of people suffer from arthritis worldwide, consistently struggling with daily activities due to debilitating pain evoked by this disease. Perhaps the most intensively investigated type of inflammatory arthritis is rheumatoid arthritis (RA), where, despite considerable advances in research and clinical management, gaps regarding the neuroimmune interactions that guide inflammation and chronic pain in this disease remain to be clarified. The pain and inflammation associated with arthritis are not isolated to the joints, and inflammatory mechanisms induced by different immune and glial cells in other tissues may affect the development of chronic pain that results from the disease. This review aims to provide an overview of the state-of-the-art research on the roles that innate immune, and glial cells play in the onset and maintenance of arthritis-associated pain, reviewing nociceptive pathways from the joint through the dorsal root ganglion, spinal circuits, and different structures in the brain. We will focus on the cellular mechanisms related to neuroinflammation and pain, and treatments targeting these mechanisms from the periphery and the CNS. A comprehensive understanding of the role these cells play in peripheral inflammation and initiation of pain and the central pathways in the spinal cord and brain will facilitate identifying new targets and pathways to aide in developing therapeutic strategies to treat joint pain associated with RA.
Collapse
Affiliation(s)
- Kaue Franco Malange
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | | | - Elayne Vieira Dias
- Department of Neurology, University of California, San Francisco, CA, United States
| | | | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, CA, United States
| | | | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|
6
|
Verkest C, Salinas M, Diochot S, Deval E, Lingueglia E, Baron A. Mechanisms of Action of the Peptide Toxins Targeting Human and Rodent Acid-Sensing Ion Channels and Relevance to Their In Vivo Analgesic Effects. Toxins (Basel) 2022; 14:toxins14100709. [PMID: 36287977 PMCID: PMC9612379 DOI: 10.3390/toxins14100709] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/16/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are voltage-independent H+-gated cation channels largely expressed in the nervous system of rodents and humans. At least six isoforms (ASIC1a, 1b, 2a, 2b, 3 and 4) associate into homotrimers or heterotrimers to form functional channels with highly pH-dependent gating properties. This review provides an update on the pharmacological profiles of animal peptide toxins targeting ASICs, including PcTx1 from tarantula and related spider toxins, APETx2 and APETx-like peptides from sea anemone, and mambalgin from snake, as well as the dimeric protein snake toxin MitTx that have all been instrumental to understanding the structure and the pH-dependent gating of rodent and human cloned ASICs and to study the physiological and pathological roles of native ASICs in vitro and in vivo. ASICs are expressed all along the pain pathways and the pharmacological data clearly support a role for these channels in pain. ASIC-targeting peptide toxins interfere with ASIC gating by complex and pH-dependent mechanisms sometimes leading to opposite effects. However, these dual pH-dependent effects of ASIC-inhibiting toxins (PcTx1, mambalgin and APETx2) are fully compatible with, and even support, their analgesic effects in vivo, both in the central and the peripheral nervous system, as well as potential effects in humans.
Collapse
Affiliation(s)
- Clément Verkest
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Miguel Salinas
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Sylvie Diochot
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Emmanuel Deval
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Eric Lingueglia
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
| | - Anne Baron
- CNRS (Centre National de la Recherche Scientifique), IPMC (Institut de Pharmacologie Moléculaire et Cellulaire), LabEx ICST (Laboratory of Excellence in Ion Channel Science and Therapeutics), FHU InovPain (Fédération Hospitalo-Universitaire “Innovative Solutions in Refractory Chronic Pain”), Université Côte d’Azur, 660 Route des Lucioles, Sophia-Antipolis, 06560 Nice, France
- Correspondence:
| |
Collapse
|
7
|
Lysophosphatidylcholine 16:0 mediates chronic joint pain associated to rheumatic diseases through acid-sensing ion channel 3. Pain 2022; 163:1999-2013. [PMID: 35086123 PMCID: PMC9479040 DOI: 10.1097/j.pain.0000000000002596] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/08/2021] [Indexed: 02/04/2023]
Abstract
ABSTRACT Rheumatic diseases are often associated to debilitating chronic pain, which remains difficult to treat and requires new therapeutic strategies. We had previously identified lysophosphatidylcholine (LPC) in the synovial fluids from few patients and shown its effect as a positive modulator of acid-sensing ion channel 3 (ASIC3) able to induce acute cutaneous pain in rodents. However, the possible involvement of LPC in chronic joint pain remained completely unknown. Here, we show, from 2 independent cohorts of patients with painful rheumatic diseases, that the synovial fluid levels of LPC are significantly elevated, especially the LPC16:0 species, compared with postmortem control subjects. Moreover, LPC16:0 levels correlated with pain outcomes in a cohort of osteoarthritis patients. However, LPC16:0 do not appear to be the hallmark of a particular joint disease because similar levels are found in the synovial fluids of a second cohort of patients with various rheumatic diseases. The mechanism of action was next explored by developing a pathology-derived rodent model. Intra-articular injections of LPC16:0 is a triggering factor of chronic joint pain in both male and female mice, ultimately leading to persistent pain and anxiety-like behaviors. All these effects are dependent on ASIC3 channels, which drive sufficient peripheral inputs to generate spinal sensitization processes. This study brings evidences from mouse and human supporting a role for LPC16:0 via ASIC3 channels in chronic pain arising from joints, with potential implications for pain management in osteoarthritis and possibly across other rheumatic diseases.
Collapse
|
8
|
Calcium-Permeable Channels Cooperation for Rheumatoid Arthritis: Therapeutic Opportunities. Biomolecules 2022; 12:biom12101383. [PMID: 36291594 PMCID: PMC9599458 DOI: 10.3390/biom12101383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis is a common autoimmune disease that results from the deposition of antibodies–autoantigens in the joints, leading to long-lasting inflammation. The main features of RA include cartilage damage, synovial invasion and flare-ups of intra-articular inflammation, and these pathological processes significantly reduce patients’ quality of life. To date, there is still no drug target that can act in rheumatoid arthritis. Therefore, the search for novel drug targets has become urgent. Due to their unique physicochemical properties, calcium ions play an important role in all cellular activities and the body has evolved a rigorous calcium signaling system. Calcium-permeable channels, as the main operators of calcium signaling, are widely distributed in cell membranes, endoplasmic reticulum membranes and mitochondrial membranes, and mediate the efflux and entry of Ca2+. Over the last century, more and more calcium-permeable channels have been identified in human cells, and the role of this large family of calcium-permeable channels in rheumatoid arthritis has gradually become clear. In this review, we briefly introduce the major calcium-permeable channels involved in the pathogenesis of RA (e.g., acid-sensitive ion channel (ASIC), transient receptor potential (TRP) channel and P2X receptor) and explain the specific roles and mechanisms of these calcium-permeable channels in the pathogenesis of RA, providing more comprehensive ideas and targets for the treatment of RA.
Collapse
|
9
|
Lombardi AF, Ma Y, Jang H, Jerban S, Tang Q, Searleman AC, Meyer RS, Du J, Chang EY. AcidoCEST-UTE MRI Reveals an Acidic Microenvironment in Knee Osteoarthritis. Int J Mol Sci 2022; 23:4466. [PMID: 35457284 PMCID: PMC9027981 DOI: 10.3390/ijms23084466] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 02/01/2023] Open
Abstract
A relationship between an acidic pH in the joints, osteoarthritis (OA), and pain has been previously demonstrated. Acidosis Chemical Exchange Saturation Transfer (acidoCEST) indirectly measures the extracellular pH through the assessment of the exchange of protons between amide groups on iodinated contrast agents and bulk water. It is possible to estimate the extracellular pH in the osteoarthritic joint using acidoCEST MRI. However, conventional MR sequences cannot image deep layers of cartilage, meniscus, ligaments, and other musculoskeletal tissues that present with short echo time and fast signal decay. Ultrashort echo time (UTE) MRI, on the other hand, has been used successfully to image those joint tissues. Here, our goal is to compare the pH measured in the knee joints of volunteers without OA and patients with severe OA using acidoCEST-UTE MRI. Patients without knee OA and patients with severe OA were examined using acidoCEST-UTE MRI and the mean pH of cartilage, meniscus, and fluid was calculated. Additionally, the relationship between the pH measurements and the Knee Injury and Osteoarthritis Outcome Score (KOOS) was investigated. AcidoCEST-UTE MRI can detect significant differences in the pH of knee cartilage, meniscus, and fluid between joints without and with OA, with OA showing lower pH values. In addition, symptoms and knee-joint function become worse at lower pH measurements.
Collapse
Affiliation(s)
- Alecio F. Lombardi
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (E.Y.C.)
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| | - Yajun Ma
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| | - Hyungseok Jang
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| | - Saeed Jerban
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| | - Qingbo Tang
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (E.Y.C.)
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| | - Adam C. Searleman
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| | - Robert Scott Meyer
- Orthopaedic Surgery Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA;
| | - Jiang Du
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| | - Eric Y. Chang
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (E.Y.C.)
- Department of Radiology, University of California San Diego, San Diego, CA 92161, USA; (Y.M.); (H.J.); (S.J.); (A.C.S.); (J.D.)
| |
Collapse
|
10
|
Dulai JS, Smith ESJ, Rahman T. Acid-sensing ion channel 3: An analgesic target. Channels (Austin) 2021; 15:94-127. [PMID: 33258401 PMCID: PMC7801124 DOI: 10.1080/19336950.2020.1852831] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Acid-sensing ion channel 3 (ASIC3) belongs to the epithelial sodium channel/degenerin (ENaC/DEG) superfamily. There are 7 different ASIC subunits encoded by 5 different genes. Most ASIC subunits form trimeric ion channels that upon activation by extracellular protons mediate a transient inward current inducing cellular excitability. ASIC subunits exhibit differential tissue expression and biophysical properties, and the ability of subunits to form homo- and heteromeric trimers further increases the complexity of currents measured and their pharmacological properties. ASIC3 is of particular interest, not only because it exhibits high expression in sensory neurones, but also because upon activation it does not fully inactivate: a transient current is followed by a sustained current that persists during a period of extracellular acidity, i.e. ASIC3 can encode prolonged acidosis as a nociceptive signal. Furthermore, certain mediators sensitize ASIC3 enabling smaller proton concentrations to activate it and other mediators can directly activate the channel at neutral pH. Moreover, there is a plethora of evidence using transgenic mouse models and pharmacology, which supports ASIC3 as being a potential target for development of analgesics. This review will focus on current understanding of ASIC3 function to provide an overview of how ASIC3 contributes to physiology and pathophysiology, examining the mechanisms by which it can be modulated, and highlighting gaps in current understanding and future research directions.
Collapse
Affiliation(s)
| | | | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
11
|
Ishihara S, Obeidat AM, Wokosin DL, Ren D, Miller RJ, Malfait AM, Miller RE. The role of intra-articular neuronal CCR2 receptors in knee joint pain associated with experimental osteoarthritis in mice. Arthritis Res Ther 2021; 23:103. [PMID: 33827672 PMCID: PMC8025346 DOI: 10.1186/s13075-021-02486-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background C–C chemokine receptor 2 (CCR2) signaling plays a key role in pain associated with experimental murine osteoarthritis (OA) after destabilization of the medial meniscus (DMM). Here, we aimed to assess if CCR2 expressed by intra-articular sensory neurons contributes to knee hyperalgesia in the early stages of the model. Methods DMM surgery was performed in the right knee of 10-week-old male wild-type (WT), Ccr2 null, or Ccr2RFP C57BL/6 mice. Knee hyperalgesia was measured using a Pressure Application Measurement device. CCR2 receptor antagonist (CCR2RA) was injected systemically (i.p.) or intra-articularly (i.a.) at different times after DMM to test its ability to reverse knee hyperalgesia. In vivo Ca2+ imaging of the dorsal root ganglion (DRG) was performed to assess sensory neuron responses to CCL2 injected into the knee joint cavity. CCL2 protein in the knee was measured by ELISA. Ccr2RFP mice and immunohistochemical staining for the pan-neuronal marker, protein gene product 9.5 (PGP9.5), or the sensory neuron marker, calcitonin gene-related peptide (CGRP), were used to visualize the location of CCR2 on intra-articular afferents. Results WT, but not Ccr2 null, mice displayed knee hyperalgesia 2–16 weeks after DMM. CCR2RA administered i.p. alleviated established hyperalgesia in WT mice 4 and 8 weeks after surgery. Intra-articular injection of CCL2 excited sensory neurons in the L4-DRG, as determined by in vivo calcium imaging; responses to CCL2 increased in mice 20 weeks after DMM. CCL2, but not vehicle, injected i.a. rapidly caused transient knee hyperalgesia in naïve WT, but not Ccr2 null, mice. Intra-articular CCR2RA injection also alleviated established hyperalgesia in WT mice 4 and 7 weeks after surgery. CCL2 protein was elevated in the knees of both WT and Ccr2 null mice 4 weeks after surgery. Co-expression of CCR2 and PGP9.5 as well as CCR2 and CGRP was observed in the lateral synovium of naïve mice; co-expression was also observed in the medial compartment of knees 8 weeks after DMM. Conclusions The findings suggest that CCL2-CCR2 signaling locally in the joint contributes to knee hyperalgesia in experimental OA, and it is in part mediated through direct stimulation of CCR2 expressed by intra-articular sensory afferents. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02486-y.
Collapse
Affiliation(s)
- Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - David L Wokosin
- Department of Physiology, Northwestern University, Chicago, IL, 60611, USA
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA.
| |
Collapse
|
12
|
Qian X, Zhang Y, Tao J, Niu R, Song S, Wang C, Peng X, Chen F. Acidosis induces synovial fibroblasts to release vascular endothelial growth factor via acid-sensitive ion channel 1a. J Transl Med 2021; 101:280-291. [PMID: 32826932 DOI: 10.1038/s41374-020-0423-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/05/2020] [Accepted: 03/20/2020] [Indexed: 12/28/2022] Open
Abstract
Acid-sensitive ion channel 1a (ASIC1a) is a member of the extracellular H+ activated cation channel family. Studies have shown that tissue acidification contributes to the formation of microvessels in rheumatoid arthritis (RA) synovial tissue, but its underlying mechanisms remain unclear. The purpose of this study was to investigate the role of tissue acidification in microvascular formation of arthritic synovial tissue and the effect of ASIC1a on vascular endothelial growth factor (VEGF) release from arthritic synovial tissue. Our results indicate that ASIC1a expression, VEGF expression, and microvessel density (MVD) are elevated in RA synovial tissue and adjuvant arthritis (AA) rat synovial tissue. When AA rats were treated with ASIC1a-specific blocker psalmotoxin-1 (PcTx-1), the expression of ASIC1a, VEGF expression, and MVD were all reduced. Acidification of RA synovial fibroblasts (RASF) can promote the release of VEGF. PcTx-1 and ASIC1a-short hairpin RNA can inhibit acid-induced release of VEGF. In addition, the ASIC1a overexpression vector can promote acid-induced VEGF release. This indicates that extracellular acidification induces the release of VEGF by RASF via ASIC1a. These findings suggest that blocking ASIC1a mediates the release of VEGF from synoviocytes may provide a potential therapeutic strategy for RA therapy.
Collapse
Affiliation(s)
- Xuewen Qian
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Yihao Zhang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Jingjing Tao
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Ruowen Niu
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Sujing Song
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Cong Wang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Xiaoqing Peng
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Feihu Chen
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
13
|
Li H, Li H, Cheng J, Liu X, Zhang Z, Wu C. Acid-sensing Ion Channel 3 Overexpression in Incisions Regulated by Nerve Growth Factor Participates in Postoperative Nociception in Rats. Anesthesiology 2020; 133:1244-1259. [PMID: 32997750 DOI: 10.1097/aln.0000000000003576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Acid-sensing ion channel 3 (ASIC3) upregulation has been reported in dorsal root ganglion neurons after incision and contributes to postoperative nociception. This study hypothesized that upregulation of ASIC3 in incised tissues is induced by nerve growth factor through the phosphoinositide 3-kinase/protein kinase B signaling pathway. METHODS A plantar incision model was established in adult male and female Sprague-Dawley rats. ASIC3 was inhibited by APETx2 treatment, small interfering RNA treatment, or ASIC3 knockout. Sciatic nerve ligation was performed to analyze ASIC3 transport. A nerve growth factor antibody and a phosphoinositide 3-kinase inhibitor were used to investigate the mechanism by which nerve growth factor regulates ASIC3 expression. RESULTS Acid-sensing ion channel 3 inhibition decreased incisional guarding and mechanical nociception. ASIC3 protein levels were increased in skin and muscle 4 h after incision (mean ± SD: 5.4 ± 3.2-fold in skin, n = 6, P = 0.001; 4.3 ± 2.2-fold in muscle, n = 6, P = 0.001). Sciatic nerve ligation revealed bidirectional ASIC3 transport. Nerve growth factor antibody treatment inhibited the expression of ASIC3 (mean ± SD: antibody 2.3 ± 0.8-fold vs. vehicle 4.9 ± 2.4-fold, n = 6, P = 0.036) and phosphorylated protein kinase B (mean ± SD: antibody 0.8 ± 0.3-fold vs. vehicle 1.8 ± 0.8-fold, n = 6, P = 0.010) in incised tissues. Intraplantar injection of nerve growth factor increased the expression of ASIC3 and phosphorylated protein kinase B. ASIC3 expression and incisional pain-related behaviors were inhibited by pretreatment with the phosphoinositide 3-kinase inhibitor LY294002. CONCLUSIONS Acid-sensing ion channel 3 overexpression in incisions contributes to postoperative guarding and mechanical nociception. Bidirectional transport of ASIC3 between incised tissues and dorsal root ganglion neurons occurs through the sciatic nerve. Nerve growth factor regulates ASIC3 expression after plantar incision through the phosphoinositide 3-kinase/protein kinase B signaling pathway.
Collapse
|
14
|
Pain Mechanism in Rheumatoid Arthritis: From Cytokines to Central Sensitization. Mediators Inflamm 2020; 2020:2076328. [PMID: 33005097 PMCID: PMC7503123 DOI: 10.1155/2020/2076328] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/13/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Pain is the most common symptom in patients with rheumatoid arthritis (RA). Although in recent years, through the implementation of targeted treatment and the introduction of disease-modifying antirheumatic drugs (DMARDs), the treatment of RA patients has made a significant progress, a large proportion of patients still feel pain. Finding appropriate treatment to alleviate the pain is very important for RA patients. Current research showed that, in addition to inflammation, RA pain involves peripheral sensitization and abnormalities in the central nervous system (CNS) pain regulatory mechanisms. This review summarized the literature on pain mechanisms of RA published in recent years. A better understanding of pain mechanisms will help to develop new analgesic targets and deploy new and existing therapies.
Collapse
|
15
|
Chakrabarti S, Ai M, Henson FM, Smith ESJ. Peripheral mechanisms of arthritic pain: A proposal to leverage large animals for in vitro studies. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100051. [PMID: 32817908 PMCID: PMC7426561 DOI: 10.1016/j.ynpai.2020.100051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 04/14/2023]
Abstract
Pain arising from musculoskeletal disorders such as arthritis is one of the leading causes of disability. Whereas the past 20-years has seen an increase in targeted therapies for rheumatoid arthritis (RA), other arthritis conditions, especially osteoarthritis, remain poorly treated. Although modulation of central pain pathways occurs in chronic arthritis, multiple lines of evidence indicate that peripherally driven pain is important in arthritic pain. To understand the peripheral mechanisms of arthritic pain, various in vitro and in vivo models have been developed, largely in rodents. Although rodent models provide numerous advantages for studying arthritis pathogenesis and treatment, the anatomy and biomechanics of rodent joints differ considerably to those of humans. By contrast, the anatomy and biomechanics of joints in larger animals, such as dogs, show greater similarity to human joints and thus studying them can provide novel insight for arthritis research. The purpose of this article is firstly to review models of arthritis and behavioral outcomes commonly used in large animals. Secondly, we review the existing in vitro models and assays used to study arthritic pain, primarily in rodents, and discuss the potential for adopting these strategies, as well as likely limitations, in large animals. We believe that exploring peripheral mechanisms of arthritic pain in vitro in large animals has the potential to reduce the veterinary burden of arthritis in commonly afflicted species like dogs, as well as to improve translatability of pain research into the clinic.
Collapse
Affiliation(s)
- Sampurna Chakrabarti
- Department of Neuroscience, Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- Department of Pharmacology, University of Cambridge, UK
| | - Minji Ai
- Department of Veterinary Medicine, University of Cambridge, UK
| | | | | |
Collapse
|
16
|
Osmakov DI, Khasanov TA, Andreev YA, Lyukmanova EN, Kozlov SA. Animal, Herb, and Microbial Toxins for Structural and Pharmacological Study of Acid-Sensing Ion Channels. Front Pharmacol 2020; 11:991. [PMID: 32733241 PMCID: PMC7360831 DOI: 10.3389/fphar.2020.00991] [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: 04/24/2020] [Accepted: 06/19/2020] [Indexed: 12/22/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are of the most sensitive molecular sensors of extracellular pH change in mammals. Six isoforms of these channels are widely represented in membranes of neuronal and non-neuronal cells, where these molecules are involved in different important regulatory functions, such as synaptic plasticity, learning, memory, and nociception, as well as in various pathological states. Structural and functional studies of both wild-type and mutant ASICs are essential for human care and medicine for the efficient treatment of socially significant diseases and ensure a comfortable standard of life. Ligands of ASICs serve as indispensable tools for these studies. Such bioactive compounds can be synthesized artificially. However, to date, the search for such molecules has been most effective amongst natural sources, such as animal venoms or plants and microbial extracts. In this review, we provide a detailed and comprehensive structural and functional description of natural compounds acting on ASICs, as well as the latest information on structural aspects of their interaction with the channels. Many of the examples provided in the review demonstrate the undoubted fundamental and practical successes of using natural toxins. Without toxins, it would not be possible to obtain data on the mechanisms of ASICs' functioning, provide detailed study of their pharmacological properties, or assess the contribution of the channels to development of different pathologies. The selectivity to different isoforms and variety in the channel modulation mode allow for the appraisal of prospective candidates for the development of new drugs.
Collapse
Affiliation(s)
- Dmitry I Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Timur A Khasanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Yaroslav A Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ekaterina N Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Sergey A Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| |
Collapse
|
17
|
The Role of Acid-sensing Ion Channel 3 in the Modulation of Tooth Mechanical Hyperalgesia Induced by Orthodontic Tooth Movement. Neuroscience 2020; 442:274-285. [PMID: 32592826 DOI: 10.1016/j.neuroscience.2020.06.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/05/2023]
Abstract
This study aimed to explore the role of acid-sensing ion channel 3 (ASIC3) in the modulation of tooth mechanical hyperalgesia induced by orthodontic tooth movement. In male Sprague-Dawley rats, closed coil springs were ligated between mandibular incisors and molars to mimic orthodontic tooth movement. Bite force was assessed to evaluate tooth mechanical hyperalgesia. The alveolar bone, trigeminal ganglia, and trigeminal nucleus caudalis underwent immunohistochemical staining and immunoblotting for ASIC3. The inferior alveolar nerves were transected to explore the interaction between the periodontal sensory endings and trigeminal ganglia. The role of ASIC3 in trigeminal ganglia was further explored with lentivirus-mediated ASIC3 ribonucleic acid interference. Results showed that ASIC3 was expressed in the periodontal Ruffini endings and expression of ASIC3 protein was elevated in periodontal tissues, trigeminal ganglia, and trigeminal nucleus caudalis, following orthodontic tooth movement. ASIC3 agonists and antagonists significantly aggravated and mitigated tooth mechanical hyperalgesia, respectively. ASIC3 expression decreased after inferior alveolar nerve transection in periodontal tissues. Both in vitro and vivo, the lentivirus vector carrying ASIC3 shRNA inhibited ASIC3 expression and relieved tooth mechanical hyperalgesia. To conclude, ASIC3 is important in the modulation of tooth mechanical hyperalgesia induced by orthodontic tooth movement. Further, the role of ASIC3 in the modulation of pain in periodontal tissues is regulated by trigeminal ganglia. An adjuvant analgesic therapy targeting ASIC3 could alleviate orthodontic movement-associated mechanical hyperalgesia in rats.
Collapse
|
18
|
Oostinga D, Steverink JG, van Wijck AJM, Verlaan JJ. An understanding of bone pain: A narrative review. Bone 2020; 134:115272. [PMID: 32062002 DOI: 10.1016/j.bone.2020.115272] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022]
Abstract
Skeletal pathologies are often accompanied by bone pain, which has negative effects on the quality of life and functional status of patients. Bone pain can be caused by a wide variety of injuries and diseases including (poorly healed) fractures, bone cancer, osteoarthritis and also iatrogenic by skeletal interventions. Orthopedic interventions are considered to be the most painful surgical procedures overall. Two major groups of medication currently used to attenuate bone pain are NSAIDs and opioids. However, these systemic drugs frequently introduce adverse events, emphasizing the need for alternative therapies that are directed at the pathophysiological mechanisms underlying bone pain. The periosteum, cortical bone and bone marrow are mainly innervated by sensory A-delta fibers and C-fibers. These fibers are mostly present in the periosteum rendering this structure most sensitive to nociceptive stimuli. A-delta fibers and C-fibers can be activated upon mechanical distortion, acidic environment and increased intramedullary pressure. After activation, these fibers can be sensitized by inflammatory mediators, phosphorylation of acid-sensing ion channels and cytokine receptors, or by upregulation of transcription factors. This can result in a change of pain perception such that normally non-noxious stimuli are now perceived as noxious. Pathological conditions in the bone can produce neurotrophic factors that bind to receptors on A-delta fibers and C-fibers. These fibers then start to sprout and increase the innervation density of the bone, making it more sensitive to nociceptive stimuli. In addition, repetitive painful stimuli cause neurochemical and electrophysiological alterations in afferent sensory neurons in the spinal cord, which leads to central sensitization, and can contribute to chronic bone pain. Understanding the pathophysiological mechanisms underlying bone pain in different skeletal injuries and diseases is important for the development of alternative, targeted pain treatments. These pain mechanism-based alternatives have the potential to improve the quality of life of patients suffering from bone pain without introducing undesirable systemic effects.
Collapse
Affiliation(s)
- Douwe Oostinga
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Jasper G Steverink
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Albert J M van Wijck
- Department of Anesthesiology, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Jorrit-Jan Verlaan
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| |
Collapse
|
19
|
Callejo G, Pattison LA, Greenhalgh JC, Chakrabarti S, Andreopoulou E, Hockley JRF, Smith ESJ, Rahman T. In silico screening of GMQ-like compounds reveals guanabenz and sephin1 as new allosteric modulators of acid-sensing ion channel 3. Biochem Pharmacol 2020; 174:113834. [PMID: 32027884 PMCID: PMC7068650 DOI: 10.1016/j.bcp.2020.113834] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/31/2020] [Indexed: 11/25/2022]
Abstract
Acid-sensing ion channels (ASICs) are voltage-independent cation channels that detect decreases in extracellular pH. Dysregulation of ASICs underpins a number of pathologies. Of particular interest is ASIC3, which is recognised as a key sensor of acid-induced pain and is important in the establishment of pain arising from inflammatory conditions, such as rheumatoid arthritis. Thus, the identification of new ASIC3 modulators and the mechanistic understanding of how these compounds modulate ASIC3 could be important for the development of new strategies to counteract the detrimental effects of dysregulated ASIC3 activity in inflammation. Here, we report the identification of novel ASIC3 modulators based on the ASIC3 agonist, 2-guanidine-4-methylquinazoline (GMQ). Through a GMQ-guided in silico screening of Food and Drug administration (FDA)-approved drugs, 5 compounds were selected and tested for their modulation of rat ASIC3 (rASIC3) using whole-cell patch-clamp electrophysiology. Of the chosen drugs, guanabenz (GBZ), an α2-adrenoceptor agonist, produced similar effects to GMQ on rASIC3, activating the channel at physiological pH (pH 7.4) and potentiating its response to mild acidic (pH 7) stimuli. Sephin1, a GBZ derivative that lacks α2-adrenoceptor activity, has been proposed to act as a selective inhibitor of a regulatory subunit of the stress-induced protein phosphatase 1 (PPP1R15A) with promising therapeutic potential for the treatment of multiple sclerosis. However, we found that like GBZ, sephin1 activates rASIC3 at pH 7.4 and potentiates its response to acidic stimulation (pH 7), i.e. sephin1 is a novel modulator of rASIC3. Furthermore, docking experiments showed that, like GMQ, GBZ and sephin1 likely interact with the nonproton ligand sensor domain of rASIC3. Overall, these data demonstrate the utility of computational analysis for identifying novel ASIC3 modulators, which can be validated with electrophysiological analysis and may lead to the development of better compounds for targeting ASIC3 in the treatment of inflammatory conditions.
Collapse
Affiliation(s)
- Gerard Callejo
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Luke A Pattison
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Jack C Greenhalgh
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Sampurna Chakrabarti
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Evangelia Andreopoulou
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - James R F Hockley
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom.
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom.
| |
Collapse
|
20
|
Walters ET. Adaptive mechanisms driving maladaptive pain: how chronic ongoing activity in primary nociceptors can enhance evolutionary fitness after severe injury. Philos Trans R Soc Lond B Biol Sci 2019; 374:20190277. [PMID: 31544606 DOI: 10.1098/rstb.2019.0277] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chronic pain is considered maladaptive by clinicians because it provides no apparent protective or recuperative benefits. Similarly, evolutionary speculations have assumed that chronic pain represents maladaptive or evolutionarily neutral dysregulation of acute pain mechanisms. By contrast, the present hypothesis proposes that chronic pain can be driven by mechanisms that evolved to reduce increased vulnerability to attack from predators and aggressive conspecifics, which often target prey showing physical impairment after severe injury. Ongoing pain and anxiety persisting long after severe injury continue to enhance vigilance and behavioural caution, decreasing the heightened vulnerability to attack that results from motor impairment and disfigurement, thereby increasing survival and reproduction (fitness). This hypothesis is supported by evidence of animals surviving and reproducing after traumatic amputations, and by complex specializations that enable primary nociceptors to detect local and systemic signs of injury and inflammation, and to maintain low-frequency discharge that can promote ongoing pain indefinitely. Ongoing activity in nociceptors involves intricate electrophysiological and anatomical specializations, including inducible alterations in the expression of ion channels and receptors that produce persistent hyperexcitability and hypersensitivity to chemical signals of injury. Clinically maladaptive chronic pain may sometimes result from the recruitment of this powerful evolutionary adaptation to severe bodily injury. This article is part of the Theo Murphy meeting issue 'Evolution of mechanisms and behaviour important for pain'.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
21
|
|
22
|
Okada S, Saito H, Matsuura Y, Mikuzuki L, Sugawara S, Onose H, Asaka J, Ohara K, Lee J, Iinuma T, Katagiri A, Iwata K. Upregulation of calcitonin gene-related peptide, neuronal nitric oxide synthase, and phosphorylated extracellular signal-regulated kinase 1/2 in the trigeminal ganglion after bright light stimulation of the eye in rats. J Oral Sci 2019; 61:146-155. [PMID: 30918211 DOI: 10.2334/josnusd.18-0031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Bright light stimulation of the eye activates trigeminal subnucleus caudalis (Vc) neurons in rats. Sensory information is conveyed to the Vc via the trigeminal ganglion (TG). Thus, it is likely that TG neurons respond to photic stimulation and are involved in photic hypersensitivity. However, the mechanisms underlying this process are unclear. Therefore, the hypothesis in this study is bright light stimulation enhances the excitability of TG neurons involved in photic hypersensitivity. Expressions of calcitonin gene-related peptide (CGRP) and neuronal nitric oxide synthase (nNOS) were significantly higher in TG neurons from 5 min to 12 h after photic stimulation of the eye. Phosphorylation of extracellular signal-regulated kinase1/2 (pERK1/2) was enhanced in TG neurons within 5 min after photic stimulation, while pERK1/2 immunoreactivity in satellite glial cells (SGCs) persisted for more than 12 h after the stimulus. Activation of SGCs was observed from 5 min to 2 h. Expression of CGRP, nNOS, and pERK1/2 was observed in small and medium TG neurons, and activation of SGCs and pERK1/2-immunoreactive SGCs encircling large TG neurons was accelerated after stimulation. These results suggest that upregulation of CGRP, nNOS, and pERK1/2 within the TG is involved in photic hypersensitivity.
Collapse
Affiliation(s)
- Shinji Okada
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry.,Department of Physiology, Nihon University School of Dentistry
| | - Hiroto Saito
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry.,Department of Physiology, Nihon University School of Dentistry
| | - Yutaka Matsuura
- Department of Oral Physiology, Osaka University Graduate School of Dentistry
| | - Lou Mikuzuki
- Department of Physiology, Nihon University School of Dentistry.,Department of Psychosomatic Dentistry, Tokyo Medical and Dental University, Graduate School
| | - Shiori Sugawara
- Department of Physiology, Nihon University School of Dentistry.,Department of Psychosomatic Dentistry, Tokyo Medical and Dental University, Graduate School
| | - Hiroki Onose
- Department of Physiology, Nihon University School of Dentistry
| | - Junichi Asaka
- Department of Physiology, Nihon University School of Dentistry
| | - Kinuyo Ohara
- Department of Endodontics, Nihon University School of Dentistry
| | - Jun Lee
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry
| | - Toshimitsu Iinuma
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry
| | - Ayano Katagiri
- Department of Physiology, Nihon University School of Dentistry.,Department of Oral Physiology, Osaka University Graduate School of Dentistry
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry
| |
Collapse
|
23
|
Maatuf Y, Geron M, Priel A. The Role of Toxins in the Pursuit for Novel Analgesics. Toxins (Basel) 2019; 11:toxins11020131. [PMID: 30813430 PMCID: PMC6409898 DOI: 10.3390/toxins11020131] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic pain is a major medical issue which reduces the quality of life of millions and inflicts a significant burden on health authorities worldwide. Currently, management of chronic pain includes first-line pharmacological therapies that are inadequately effective, as in just a portion of patients pain relief is obtained. Furthermore, most analgesics in use produce severe or intolerable adverse effects that impose dose restrictions and reduce compliance. As the majority of analgesic agents act on the central nervous system (CNS), it is possible that blocking pain at its source by targeting nociceptors would prove more efficient with minimal CNS-related side effects. The development of such analgesics requires the identification of appropriate molecular targets and thorough understanding of their structural and functional features. To this end, plant and animal toxins can be employed as they affect ion channels with high potency and selectivity. Moreover, elucidation of the toxin-bound ion channel structure could generate pharmacophores for rational drug design while favorable safety and analgesic profiles could highlight toxins as leads or even as valuable therapeutic compounds themselves. Here, we discuss the use of plant and animal toxins in the characterization of peripherally expressed ion channels which are implicated in pain.
Collapse
Affiliation(s)
- Yossi Maatuf
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Matan Geron
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Avi Priel
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
24
|
Abstract
CGRP has long been suspected as a mediator of arthritis pain, although evidence that CGRP directly mediates human musculoskeletal pain remains circumstantial. This chapter describes in depth the evidence surrounding CGRP's association with pain in musculoskeletal disorders and also summarises evidence for CGRP being a direct cause of pain in other conditions. CGRP-immunoreactive nerves are present in musculoskeletal tissues, and CGRP expression is altered in musculoskeletal pain. CGRP modulates musculoskeletal pain through actions both in the periphery and central nervous system. Human observational studies, research on animal arthritis models and the few reported randomised controlled trials in humans of treatments that target CGRP provide the context of CGRP as a possible pain biomarker or mediator in conditions other than migraine.
Collapse
Affiliation(s)
- David A Walsh
- Pain Centre Versus Arthritis, NIHR Nottingham Biomedical Research Centre and Division of ROD, University of Nottingham, Nottingham, UK.
- Rheumatology, Sherwood Forest Hospitals NHS Foundation Trust, Nottinghamshire, UK.
| | - Daniel F McWilliams
- Pain Centre Versus Arthritis, NIHR Nottingham Biomedical Research Centre and Division of ROD, University of Nottingham, Nottingham, UK
| |
Collapse
|
25
|
Abstract
A large series of different ion channels have been identified and investigated as potential targets for new medicines for the treatment of a variety of human diseases, including pain. Among these channels, the voltage gated calcium channels (VGCC) are inhibited by drugs for the treatment of migraine, neuropathic pain or intractable pain. Transient receptor potential (TRP) channels are emerging as important pain transducers as they sense low pH media or oxidative stress and other mediators and are abundantly found at sites of inflammation or tissue injury. Low pH may also activate acid sensing ion channels (ASIC) and mechanical forces stimulate the PIEZO channels. While potent agonists of TRP channels due to their desensitizing action on pain transmission are used as topical applications, the potential of TRP antagonists as pain therapeutics remains an exciting field of investigation. The study of ASIC or PIEZO channels in pain signaling is in an early stage, whereas antagonism of the purinergic P2X3 channels has been reported to provide beneficial effects in chronic intractable cough. The present chapter covers these intriguing channels in great detail, highlighting their diverse mechanisms and broad potential for therapeutic utility.
Collapse
Affiliation(s)
- Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| |
Collapse
|
26
|
Reiners M, Margreiter MA, Oslender-Bujotzek A, Rossetti G, Gründer S, Schmidt A. The Conorfamide RPRFa Stabilizes the Open Conformation of Acid-Sensing Ion Channel 3 via the Nonproton Ligand-Sensing Domain. Mol Pharmacol 2018; 94:1114-1124. [PMID: 30012583 DOI: 10.1124/mol.118.112375] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/06/2018] [Indexed: 01/26/2023] Open
Abstract
Acid-sensing ion channel 3 (ASIC3) is a proton-gated Na+ channel with important roles in pain. ASIC3 quickly desensitizes in less than a second, limiting its capacity to sense sustained acidosis during pain. RFamide neuropeptides are modulators of ASIC3 that slow its desensitization and induce a variable sustained current. The molecular mechanism of slowed desensitization and the RFamide binding site on ASIC3 are unknown. RPRFamide, a RFamide from the venom of a cone snail, has a comparatively high affinity for ASIC3 and strongly slows its desensitization. Here we show that covalent binding of a UV-sensitive RPRFamide variant to ASIC3 prevents desensitization, suggesting that RPRFamide has to unbind from ASIC3 before it can desensitize. Moreover, we show by in silico docking to a homology model of ASIC3 that a cavity in the lower palm domain, which is also known as the nonproton ligand-sensing domain, is a potential binding site of RPRFamide. Finally, using extensive mutagenesis of residues lining the nonproton ligand-sensing domain, we confirm that this domain is essential for RPRFamide modulation of ASIC3. As comparative analysis of ASIC crystal structures in the open and in the desensitized conformation suggests that the lower palm domain contracts during desensitization, our results collectively suggest that RPRFamide, and probably also other RFamide neuropeptides, bind to the nonproton ligand-sensing domain to stabilize the open conformation of ASIC3.
Collapse
Affiliation(s)
- Melissa Reiners
- Institute of Physiology (M.R., A.O.-B., S.G., A.S.) and Department of Oncology, Hematology and Stem Cell Transplantation (G.R.), RWTH Aachen University, Aachen, Germany; and Computational Biomedicine - Institute for Advanced Simulation (IAS)/Institute of Neuroscience and Medicine (INM) and Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, Jülich, Germany (M.A.M., G.R.)
| | - Michael A Margreiter
- Institute of Physiology (M.R., A.O.-B., S.G., A.S.) and Department of Oncology, Hematology and Stem Cell Transplantation (G.R.), RWTH Aachen University, Aachen, Germany; and Computational Biomedicine - Institute for Advanced Simulation (IAS)/Institute of Neuroscience and Medicine (INM) and Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, Jülich, Germany (M.A.M., G.R.)
| | - Adrienne Oslender-Bujotzek
- Institute of Physiology (M.R., A.O.-B., S.G., A.S.) and Department of Oncology, Hematology and Stem Cell Transplantation (G.R.), RWTH Aachen University, Aachen, Germany; and Computational Biomedicine - Institute for Advanced Simulation (IAS)/Institute of Neuroscience and Medicine (INM) and Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, Jülich, Germany (M.A.M., G.R.)
| | - Giulia Rossetti
- Institute of Physiology (M.R., A.O.-B., S.G., A.S.) and Department of Oncology, Hematology and Stem Cell Transplantation (G.R.), RWTH Aachen University, Aachen, Germany; and Computational Biomedicine - Institute for Advanced Simulation (IAS)/Institute of Neuroscience and Medicine (INM) and Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, Jülich, Germany (M.A.M., G.R.)
| | - Stefan Gründer
- Institute of Physiology (M.R., A.O.-B., S.G., A.S.) and Department of Oncology, Hematology and Stem Cell Transplantation (G.R.), RWTH Aachen University, Aachen, Germany; and Computational Biomedicine - Institute for Advanced Simulation (IAS)/Institute of Neuroscience and Medicine (INM) and Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, Jülich, Germany (M.A.M., G.R.)
| | - Axel Schmidt
- Institute of Physiology (M.R., A.O.-B., S.G., A.S.) and Department of Oncology, Hematology and Stem Cell Transplantation (G.R.), RWTH Aachen University, Aachen, Germany; and Computational Biomedicine - Institute for Advanced Simulation (IAS)/Institute of Neuroscience and Medicine (INM) and Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, Jülich, Germany (M.A.M., G.R.)
| |
Collapse
|
27
|
Chakrabarti S, Pattison LA, Singhal K, Hockley JRF, Callejo G, Smith ESJ. Acute inflammation sensitizes knee-innervating sensory neurons and decreases mouse digging behavior in a TRPV1-dependent manner. Neuropharmacology 2018; 143:49-62. [PMID: 30240782 PMCID: PMC6277850 DOI: 10.1016/j.neuropharm.2018.09.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 01/20/2023]
Abstract
Ongoing, spontaneous pain is characteristic of inflammatory joint pain and reduces an individual's quality of life. To understand the neural basis of inflammatory joint pain, we made a unilateral knee injection of complete Freund's adjuvant (CFA) in mice, which reduced their natural digging behavior. We hypothesized that sensitization of knee-innervating dorsal root ganglion (DRG) neurons underlies this altered behavior. To test this hypothesis, we performed electrophysiological recordings on retrograde labeled knee-innervating primary DRG neuron cultures and measured their responses to a number of electrical and chemical stimuli. We found that 24-h after CFA-induced knee inflammation, knee neurons show a decreased action potential generation threshold, as well as increased GABA and capsaicin sensitivity, but have unaltered acid sensitivity. The inflammation-induced sensitization of knee neurons persisted for 24-h in culture, but was not observed after 48-h in culture. Through immunohistochemistry, we showed that the increased knee neuron capsaicin sensitivity correlated with enhanced expression of the capsaicin receptor, transient receptor potential vanilloid 1 (TRPV1) in knee-innervating neurons of the CFA-injected side. We also observed an increase in the co-expression of TRPV1 with tropomyosin receptor kinase A (TrkA), which is the receptor for nerve growth factor (NGF), suggesting that NGF partially induces the increased TRPV1 expression. Lastly, we found that systemic administration of the TRPV1 antagonist, A-425619, reversed the decrease in digging behavior induced by CFA injection, further confirming the role of TRPV1, expressed by knee neurons, in acute inflammatory joint pain. Knee inflammation decreases digging behavior in mice. Knee-innervating dorsal root ganglion neurons are hyperexcitable after inflammation. NGF-mediated increase in TRPV1 expression is observed in knee-innervating neurons. Systemic TRPV1 antagonist administration normalises digging behavior in mice.
Collapse
Affiliation(s)
| | - Luke A Pattison
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Kaajal Singhal
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Gerard Callejo
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
28
|
Yu GM, Liu D, Yuan N, Liu BH. Dual role of acid-sensing ion channels 3 in rheumatoid arthritis: destruction or protection? Immunopharmacol Immunotoxicol 2018; 40:273-277. [PMID: 30035658 DOI: 10.1080/08923973.2018.1485156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Acid-sensing ion channels (ASIC) are voltage-independent cationic channels that open in response to decrease in extracellular pH. Amongst different subtypes, ASIC3 has received much attention in joint inflammatory conditions including rheumatoid arthritis. There have been a number of studies showing that there is an increase in expression of ASIC3 on nerve afferents supplying joints in response to inflammatory stimulus. Accordingly, a number of selective as well as nonselective ASIC3 inhibitors have shown potential in attenuating pain and inflammation in animal models of rheumatoid arthritis. On the other hand, there have been studies showing that ASIC3 may exert protective effects in joint inflammation. ASIC-/- animals, without ASIC3 genes, exhibit more joint inflammation and destruction in comparison to ASIC+/+ animals. The present review discusses the dual nature of ASIC3 in joint inflammation with possible mechanisms.
Collapse
Affiliation(s)
- Gui-Mei Yu
- a Department of Rheumatology , The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun , PR China
| | - Di Liu
- a Department of Rheumatology , The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun , PR China
| | - Na Yuan
- a Department of Rheumatology , The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun , PR China
| | - Bao-Hua Liu
- b Department of Emergency , The First Hospital of Jilin University , Changchun , PR China
| |
Collapse
|
29
|
Yan XG, Li WG, Qi X, Zhu JJ, Huang C, Han SL, Jiang Q, Xu TL, Liu JH. Subtype-selective inhibition of acid-sensing ion channel 3 by a natural flavonoid. CNS Neurosci Ther 2018; 25:47-56. [PMID: 29781252 DOI: 10.1111/cns.12979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 01/10/2023] Open
Abstract
AIMS Acid-sensing ion channels (ASICs) are extracellular proton-gated cation channels that have been implicated in multiple physiological and pathological processes, and peripheral ASIC3 prominently participate into the pathogenesis of chronic pain, itch, and neuroinflammation, which necessitates the need for discovery and development of novel modulators in a subtype-specific manner. METHODS Whole-cell patch clamp recordings and behavioral assays were used to examine the effect of several natural compounds on the ASIC-mediated currents and acid-induced nocifensive behavior, respectively. RESULTS We identified a natural flavonoid compound, (-)-epigallocatechin gallate (EGCG, compound 11), that acts as a potent inhibitor for the ASIC3 channel in an isoform-specific way. The compound 11 inhibited ASIC3 currents with an apparent half maximal inhibitory concentration of 13.2 μmol/L when measured at pH 5.0. However, at the concentration up to 100 μmol/L, the compound 11 had no significant impacts on the homomeric ASIC1a, 1b, and 2a channels. In contrast to most of the known ASIC inhibitors that usually bear either basic or carboxylic groups, the compound 11 belongs to the polyphenolic family. In compound 11, both the chirality and the 3-hydroxyl group of its pyrogallol part, in addition to the integrity of the gallate part, are crucial for the inhibitory efficacy. Finally, EGCG was found significantly to decrease the acid-induced nocifensive behavior in mice. CONCLUSION Taken together, these results thus defined a novel backbone structure for small molecule drug design targeting ASIC3 channels to treat pain-related diseases.
Collapse
Affiliation(s)
- Xiao-Gang Yan
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Guang Li
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Qi
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Jie Zhu
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Huang
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shao-Ling Han
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Jiang
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian-Le Xu
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Hua Liu
- Departments of Chemistry, Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Wright AJ, Husson ZM, Hu D, Callejo G, Brindle KM, Smith ESJ. Increased hyperpolarized [1- 13 C] lactate production in a model of joint inflammation is not accompanied by tissue acidosis as assessed using hyperpolarized 13 C-labelled bicarbonate. NMR IN BIOMEDICINE 2018; 31:e3892. [PMID: 29380927 PMCID: PMC5887936 DOI: 10.1002/nbm.3892] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 05/11/2023]
Abstract
Arthritic conditions are a major source of chronic pain. Furthering our understanding of disease mechanisms creates the opportunity to develop more targeted therapeutics. In rheumatoid arthritis (RA), measurements of pH in human synovial fluid suggest that acidosis occurs, but that this is highly variable between individuals. Here we sought to determine if tissue acidosis occurs in a widely used rodent arthritis model: complete Freund's adjuvant (CFA)-induced inflammation. CFA robustly evoked paw and ankle swelling, concomitant with worsening clinical scores over time. We used magnetic resonance spectroscopic imaging of hyperpolarized [1-13 C]pyruvate metabolism to demonstrate that CFA induces an increase in the lactate-to-pyruvate ratio. This increase is indicative of enhanced glycolysis and an increased lactate concentration, as has been observed in the synovial fluid from RA patients, and which was correlated with acidosis. We also measured the 13 CO2 /H13 CO3- ratio, in animals injected with hyperpolarized H13 CO3- , to estimate extracellular tissue pH and showed that despite the apparent increase in glycolytic activity in CFA-induced inflammation there was no accompanying decrease in extracellular pH. The pH was 7.23 ± 0.06 in control paws and 7.32 ± 0.09 in inflamed paws. These results could explain why mice lacking acid-sensing ion channel subunits 1, 2 and 3 do not display any changes in mechanical or thermal hyperalgesia in CFA-induced inflammation.
Collapse
Affiliation(s)
- Alan J. Wright
- Cancer Research UK Cambridge InstituteUniversity of Cambridge, Li Ka Shing Centre, Robinson WayCambridgeUK
| | - Zoé M.A. Husson
- Department of PharmacologyUniversity of Cambridge, Tennis Court RoadCambridgeUK
| | - De‐En Hu
- Cancer Research UK Cambridge InstituteUniversity of Cambridge, Li Ka Shing Centre, Robinson WayCambridgeUK
| | - Gerard Callejo
- Department of PharmacologyUniversity of Cambridge, Tennis Court RoadCambridgeUK
| | - Kevin M. Brindle
- Cancer Research UK Cambridge InstituteUniversity of Cambridge, Li Ka Shing Centre, Robinson WayCambridgeUK
- Department of BiochemistryUniversity of Cambridge, Tennis Court RoadCambridgeUK
| | - Ewan St. John Smith
- Department of PharmacologyUniversity of Cambridge, Tennis Court RoadCambridgeUK
| |
Collapse
|
31
|
Abstract
Acid-sensing ion channels (ASICs) are a family of ion channels, consisting of four members; ASIC1 to 4. These channels are sensitive to changes in pH and are expressed throughout the central and peripheral nervous systems-including brain, spinal cord, and sensory ganglia. They have been implicated in a number of neurological conditions such as stroke and cerebral ischemia, traumatic brain injury, and epilepsy, and more recently in migraine. Their expression within areas of interest in the brain in migraine, such as the hypothalamus and PAG, their demonstrated involvement in preclinical models of meningeal afferent signaling, and their role in cortical spreading depression (the electrophysiological correlate of migraine aura), has enhanced research interest into these channels as potential therapeutic targets in migraine. Migraine is a disorder with a paucity of both acute and preventive therapies available, in which at best 50% of patients respond to available medications, and these medications often have intolerable side effects. There is therefore a great need for therapeutic development for this disabling condition. This review will summarize the understanding of the structure and CNS expression of ASICs, the mechanisms for their potential role in nociception, recent work in migraine, and areas for future research and drug development.
Collapse
Affiliation(s)
- Nazia Karsan
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Denmark Hill, London, SE5 9PJ, UK
| | - Eric B Gonzales
- TCU and UNTHSC School of Medicine (applicant for LCME accreditation), Department of Medical Education, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Road, BSB-14, Richardson, TX, 75080, USA.
| |
Collapse
|
32
|
Tackling Pain Associated with Rheumatoid Arthritis: Proton-Sensing Receptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1099:49-64. [PMID: 30306514 DOI: 10.1007/978-981-13-1756-9_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Rheumatoid arthritis (RA), characterized by chronic inflammation of synovial joints, is often associated with ongoing pain and increased pain sensitivity. Chronic pain that comes with RA turns independent, essentially becoming its own disease. It could partly explain that a significant number (50%) of RA patients fail to respond to current RA therapies that focus mainly on suppression of joint inflammation. The acute phase of pain seems to associate with joint inflammation in early RA. In established RA, the chronic phase of pain could be linked to inflammatory components of neuron-immune interactions and noninflammatory components. Accumulating evidence suggests that the initial inflammation and autoimmunity in RA (preclinical RA) begin outside of the joint and may originate at mucosal sites and alterations in the composition of microbiota located at mucosal sites could be essential for mucosal inflammation, triggering joint inflammation. Fibroblast-like synoviocytes in the inflamed joint respond to cytokines to release acidic components, lowering pH in synovial fluid. Extracellular proton binds to proton-sensing ion channels, and G-protein-coupled receptors in joint nociceptive fibers may contribute to sensory transduction and release of neurotransmitters, leading to pain and hyperalgesia. Activation of peripheral sensory neurons or nociceptors further modulates inflammation, resulting in neuroinflammation or neurogenic inflammation. Peripheral and central nerves work with non-neuronal cells (such as immune cells, glial cells) in concert to contribute to the chronic phase of RA-associated pain. This review will discuss actions of proton-sensing receptors on neurons or non-neuronal cells that modulate RA pathology and associated chronic pain, and it will be beneficial for the development of future therapeutic treatments.
Collapse
|
33
|
Miller RE, Kim YS, Tran PB, Ishihara S, Dong X, Miller RJ, Malfait AM. Visualization of Peripheral Neuron Sensitization in a Surgical Mouse Model of Osteoarthritis by In Vivo Calcium Imaging. Arthritis Rheumatol 2017; 70:88-97. [PMID: 28992367 DOI: 10.1002/art.40342] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To develop a method for analyzing sensory neuron responses to mechanical stimuli in vivo, and to evaluate whether these neuronal responses change after destabilization of the medial meniscus (DMM). METHODS DMM or sham surgery was performed in 10-week-old male C57BL/6 wild-type or Pirt-GCaMP3+/- mice. All experiments were performed 8 weeks after surgery. Knee and hind paw hyperalgesia were assessed in wild-type mice. The retrograde label DiI was injected into the ipsilateral knee to quantify the number of knee-innervating neurons in the L4 dorsal root ganglion (DRG) in wild-type mice. In vivo calcium imaging was performed on the ipsilateral L4 DRG of Pirt-GCaMP3+/- mice as mechanical stimuli (paw pinch, knee pinch, or knee twist) were applied to the ipsilateral hind limb. RESULTS Eight weeks after surgery, mice subjected to DMM had more hyperalgesia in the knee and hind paw compared to mice subjected to sham surgery. Intraarticular injection of DiI labeled similar numbers of neurons in the L4 DRG of mice subjected to sham surgery and mice subjected to DMM. Increased numbers of sensory neurons responded to all 3 mechanical stimuli in mice subjected to DMM, as assessed by in vivo calcium imaging. The majority of responses in mice subjected to sham surgery and mice subjected to DMM were in small to medium-sized neurons, consistent with the size of nociceptors. The magnitude of responses was similar between mice subjected to sham surgery and mice subjected to DMM. CONCLUSION Our findings indicate that increased numbers of small to medium-sized DRG neurons respond to mechanical stimuli 8 weeks after DMM surgery, suggesting that nociceptors have become sensitized by lowering the response threshold.
Collapse
Affiliation(s)
| | - Yu Shin Kim
- Johns Hopkins University School of Medicine, Baltimore, Maryland, and University of Texas, Medical Branch School of Medicine, Galveston
| | | | | | - Xinzhong Dong
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | |
Collapse
|
34
|
Hsieh WS, Kung CC, Huang SL, Lin SC, Sun WH. TDAG8, TRPV1, and ASIC3 involved in establishing hyperalgesic priming in experimental rheumatoid arthritis. Sci Rep 2017; 7:8870. [PMID: 28827659 PMCID: PMC5566336 DOI: 10.1038/s41598-017-09200-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/20/2017] [Indexed: 01/13/2023] Open
Abstract
Rheumatoid arthritis (RA), characterized by chronic inflammation of synovial joints, is often associated with ongoing pain and increased pain sensitivity. High hydrogen ion concentration (acidosis) found in synovial fluid in RA patients is associated with disease severity. Acidosis signaling acting on proton-sensing receptors may contribute to inflammation and pain. Previous studies focused on the early phase of arthritis (<5 weeks) and used different arthritis models, so elucidating the roles of different proton-sensing receptors in the chronic phase of arthritis is difficult. We intra-articularly injected complete Freund’s adjuvant into mice once a week for 4 weeks to establish chronic RA pain. Mice with knockout of acid-sensing ion channel 3 (ASIC3) or transient receptor potential/vanilloid receptor subtype 1 (TRPV1) showed attenuated chronic phase (>6 weeks) of RA pain. Mice with T-cell death-associated gene 8 (TDAG8) knockout showed attenuated acute and chronic phases of RA pain. TDAG8 likely participates in the initiation of RA pain, but all three genes, TDAG8, TRPV1, and ASIC3, are essential to establish hyperalgesic priming to regulate the chronic phase of RA pain.
Collapse
Affiliation(s)
- Wei-Shan Hsieh
- Department of Life Sciences, National Central University, Zhongli, Taoyuan city, Taiwan
| | - Chia-Chi Kung
- Department of Life Sciences, National Central University, Zhongli, Taoyuan city, Taiwan.,Department of Anesthesiology, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Shir-Ly Huang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Chang Lin
- Department of Immunology, Cathy General Hospital, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Sciences, National Central University, Zhongli, Taoyuan city, Taiwan.
| |
Collapse
|
35
|
Kim J, Yang S, Choi CY. The Evaluation of the Effect of Herbal Extract on Osteoarthritis: In Vitro and In Vivo Study. Prev Nutr Food Sci 2016; 21:310-316. [PMID: 28078252 PMCID: PMC5216881 DOI: 10.3746/pnf.2016.21.4.310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/28/2016] [Indexed: 12/28/2022] Open
Abstract
In this study, the anti-osteoarthritis effects of Cynanchum wilfordii, Phlomis umbrosa, and Angelica gigas extract (CPAE), observed and confirmed in previously clinical studies were further investigated by in vitro and in vivo studies. Anabolic biomarkers related to healthy cartilage maintenance, such as aggrecan, type II collagen α-1 (Col2a1), sex determining region Y-box-9 (Sox-9), and catabolic biomarkers related to osteoarthritis, such as cyclooxygenase-2 (Cox-2), matrix metalloproteinase-13 (Mmp13), and nuclear factor kappa-light-chain-enhancer of activated B cells (Nfκb), were evaluated by quantitative reverse transcriptase polymerase chain reaction and reporter gene assay. In vitro study results showed significant changes in both anabolic and catabolic biomarkers. For anabolic factors, significant changes in the level of aggrecan (P<0.05), Col2a1 (P<0.05), and Sox-9 (P<0.01) activation were shown after treatment of cartilage cells with CPAE (50 ng/mL) with similar efficacy compared to insulin growth factor, the positive control (100 ng/mL). For catabolic factors, significant changes in the inhibition activity of Cox-2 (P<0.05), Mmp13 (P<0.01), and Nfκb (P<0.05) were shown for CPAE (50 ng/mL) with similar efficacy compared to Celecoxib, the positive control (10 μM). In the in vivo carrageenan-induced paw edema model study results showed that CPAE-treated groups (100 mg/kg) and Celecoxib-treated groups (60 mg/kg) showed comparably significant efficacy of inhibition by 37.1% and 52.1%, respectively. Furthermore, CPAE (200 mg/kg) showed similar effect to Celecoxib (60 mg/kg) with an inhibition rate of 54.3%. This result confirms that CPAE effectively inhibited the inflammation-induced osteoarthritis symptoms.
Collapse
Affiliation(s)
- Jaeyong Kim
- Jeonnam Institute of Natural Resources Research, Jeonnam 59338, Korea
| | - Siyoung Yang
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Chul-Yung Choi
- Jeonnam Institute of Natural Resources Research, Jeonnam 59338, Korea
| |
Collapse
|
36
|
Abdelhamid RE, Sluka KA. ASICs Mediate Pain and Inflammation in Musculoskeletal Diseases. Physiology (Bethesda) 2016; 30:449-59. [PMID: 26525344 DOI: 10.1152/physiol.00030.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Chronic musculoskeletal pain is debilitating and affects ∼ 20% of adults. Tissue acidosis is present in painful musculoskeletal diseases like rheumatoid arthritis. ASICs are located on skeletal muscle and joint nociceptors as well as on nonneuronal cells in the muscles and joints, where they mediate nociception. This review discusses the properties of different types of ASICs, factors affecting their pH sensitivity, and their role in musculoskeletal hyperalgesia and inflammation.
Collapse
Affiliation(s)
- Ramy E Abdelhamid
- Department of Physical Therapy and Rehabilitation Science, Neuroscience Graduate Program, Pain Research Program, University of Iowa, Iowa City, Iowa
| | - Kathleen A Sluka
- Department of Physical Therapy and Rehabilitation Science, Neuroscience Graduate Program, Pain Research Program, University of Iowa, Iowa City, Iowa
| |
Collapse
|
37
|
Munkholm TK, Arendt-Nielsen L. The interaction between NGF-induced hyperalgesia and acid-provoked pain in the infrapatellar fat pad and tibialis anterior muscle of healthy volunteers. Eur J Pain 2016; 21:474-485. [PMID: 27634419 DOI: 10.1002/ejp.941] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND Tissue pH is lowered in inflamed tissues, and the increased proton concentration activates acid-sensing ion channels (ASICs), contributing to pain and hyperalgesia. ASICs can be upregulated by nerve growth factor (NGF). The aim of this study was to investigate two new human experimental pain models combining NGF- and acid-induced pain in a randomized, controlled, double-blind study. METHODS In experiment 1, volunteers (N = 16) received an injection of either NGF or isotonic saline in each infrapatellar fat pad (IFP). One day after 5 mL of phosphate-buffered acidic saline was infused into each IFP at a rate of 20 mL/h. In experiment 2, the tibialis anterior (TA) muscle of additional volunteers (N = 16) was examined, following the same procedure except that the volume and infusion rate of acid were different (10 mL, 30 mL/h). Continuous pain ratings were recorded during and after acid infusions. In addition, soreness scores on a Likert scale and pressure pain thresholds (PPTs) were assessed. RESULTS The PPT of the IFP was significantly decreased at the NGF injection site on day 1, but acid-provoked pain ratings and the change in PPT from pre- to postinfusion between the knees were similar. In the muscle pain model, local mechanical hyperalgesia developed 3 h after the NGF injection and a significant additional decrease in PPT was found after acid infusion compared to preinfusion. CONCLUSIONS NGF sensitization in the IFP was not facilitated by acid, whereas an acid-provoked enhancement of muscle hyperalgesia was found. NGF sensitization of adipose tissue responds differently to acid provocation compared to muscle tissue. SIGNIFICANCE Quantification of two novel pain models combining NGF and acid. Hyperalgesia developed after NGF injection in the infrapatellar fat pad, but it was not facilitated by acid provocation. Contrary, NGF-induced hyperalgesia in muscle tissue was enhanced by acid.
Collapse
Affiliation(s)
- T K Munkholm
- Center for Sensory-Motor Interaction (SMI), Aalborg University, Denmark
| | - L Arendt-Nielsen
- Center for Sensory-Motor Interaction (SMI), Aalborg University, Denmark
| |
Collapse
|
38
|
Bas DB, Su J, Wigerblad G, Svensson CI. Pain in rheumatoid arthritis: models and mechanisms. Pain Manag 2016; 6:265-84. [PMID: 27086843 DOI: 10.2217/pmt.16.4] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pain is one of the most challenging symptoms for patients with rheumatoid arthritis (RA). RA-related pain is frequently considered to be solely a consequence of inflammation in the joints; however, recent studies show that multiple mechanisms are involved. Indeed, RA pain may start even before the disease manifests, and frequently does not correlate with the degree of inflammation or pharmacological management. In this aspect, animal studies have the potential to provide new insights into the pathology that initiate and maintain pain in RA. The focus of this review is to describe the most commonly used animal models for studies of RA pathology, which have also been utilized in pain research, and to summarize findings providing potential clues to the mechanisms involved in the regulation of RA-induced pain.
Collapse
Affiliation(s)
- Duygu B Bas
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Jie Su
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Gustaf Wigerblad
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Camilla I Svensson
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
39
|
da Silva Serra I, Husson Z, Bartlett JD, Smith ESJ. Characterization of cutaneous and articular sensory neurons. Mol Pain 2016; 12:1744806916636387. [PMID: 27030722 PMCID: PMC4956179 DOI: 10.1177/1744806916636387] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 02/02/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND A wide range of stimuli can activate sensory neurons and neurons innervating specific tissues often have distinct properties. Here, we used retrograde tracing to identify sensory neurons innervating the hind paw skin (cutaneous) and ankle/knee joints (articular), and combined immunohistochemistry and electrophysiology analysis to determine the neurochemical phenotype of cutaneous and articular neurons, as well as their electrical and chemical excitability. RESULTS Immunohistochemistry analysis using RetroBeads as a retrograde tracer confirmed previous data that cutaneous and articular neurons are a mixture of myelinated and unmyelinated neurons, and the majority of both populations are peptidergic. In whole-cell patch-clamp recordings from cultured dorsal root ganglion neurons, voltage-gated inward currents and action potential parameters were largely similar between articular and cutaneous neurons, although cutaneous neuron action potentials had a longer half-peak duration (HPD). An assessment of chemical sensitivity showed that all neurons responded to a pH 5.0 solution, but that acid-sensing ion channel (ASIC) currents, determined by inhibition with the nonselective acid-sensing ion channel antagonist benzamil, were of a greater magnitude in cutaneous compared to articular neurons. Forty to fifty percent of cutaneous and articular neurons responded to capsaicin, cinnamaldehyde, and menthol, indicating similar expression levels of transient receptor potential vanilloid 1 (TRPV1), transient receptor potential ankyrin 1 (TRPA1), and transient receptor potential melastatin 8 (TRPM8), respectively. By contrast, significantly more articular neurons responded to ATP than cutaneous neurons. CONCLUSION This work makes a detailed characterization of cutaneous and articular sensory neurons and highlights the importance of making recordings from identified neuronal populations: sensory neurons innervating different tissues have subtly different properties, possibly reflecting different functions.
Collapse
Affiliation(s)
- Ines da Silva Serra
- Department of Pharmacology, University of Cambridge, Cambridge, UK School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| | - Zoé Husson
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
40
|
Zhou RP, Wu XS, Wang ZS, Xie YY, Ge JF, Chen FH. Novel Insights into Acid-Sensing Ion Channels: Implications for Degenerative Diseases. Aging Dis 2015; 7:491-501. [PMID: 27493834 DOI: 10.14336/ad.2015.1213] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/13/2015] [Indexed: 12/16/2022] Open
Abstract
Degenerative diseases often strike older adults and are characterized by progressive deterioration of cells, eventually leading to tissue and organ degeneration for which limited effective treatment options are currently available. Acid-sensing ion channels (ASICs), a family of extracellular H(+)-activated ligand-gated ion channels, play critical roles in physiological and pathological conditions. Aberrant activation of ASICs is reported to regulate cell apoptosis, differentiation and autophagy. Accumulating evidence has highlighted a dramatic increase and activation of ASICs in degenerative disorders, including multiple sclerosis, Parkinson's disease, Huntington's disease, intervertebral disc degeneration and arthritis. In this review, we have comprehensively discussed the critical roles of ASICs and their potential utility as therapeutic targets in degenerative diseases.
Collapse
Affiliation(s)
- Ren-Peng Zhou
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Xiao-Shan Wu
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Zhi-Sen Wang
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Ya-Ya Xie
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Jin-Fang Ge
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Fei-Hu Chen
- 1Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei 230032, China; 2The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| |
Collapse
|
41
|
Osmakov DI, Andreev YA, Kozlov SA. Acid-sensing ion channels and their modulators. BIOCHEMISTRY (MOSCOW) 2015; 79:1528-45. [PMID: 25749163 DOI: 10.1134/s0006297914130069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
According to a modern look acid-sensing ion channels (ASICs) are one of the most important receptors that perceive pH change in the body. ASICs represent proton-gated Na+-selective channels, which are expressed in neurons of the central and peripheral nervous system. These channels are attracting attention of researchers around the world, as they are involved in various physiological processes in the body. Drop of pH may occur in tissues in norm (e.g. the accumulation of lactic acid, the release of protons upon ATP hydrolysis) and pathology (inflammation, ischemic stroke, tissue damage and seizure). These processes are accompanied by unpleasant pain sensations, which may be short-lived or can lead to chronic inflammatory diseases. Modulators of ASIC channels activity are potential candidates for new effective analgesic and neuroprotection drugs. This review summarizes available information about structure, function, and physiological role of ASIC channels. In addition a description of all known ligands of these channels and their practical relevance is provided.
Collapse
Affiliation(s)
- D I Osmakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| | | | | |
Collapse
|
42
|
Cuesta A, Viña E, Cabo R, Vázquez G, Cobo R, García-Suárez O, García-Cosamalón J, Vega JA. Acid-sensing ion channel 2 (asic 2) and trkb interrelationships within the intervertebral disc. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:10305-10314. [PMID: 26617738 PMCID: PMC4637553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/28/2015] [Indexed: 06/05/2023]
Abstract
The cells of the intervertebral disc (IVD) have an unusual acidic and hyperosmotic microenvironment. They express acid-sensing ion channels (ASICs), gated by extracellular protons and mechanical forces, as well as neurotrophins and their signalling receptors. In the nervous tissues some neurotrophins regulate the expression of ASICs. The expression of ASIC2 and TrkB in human normal and degenerated IVD was assessed using quantitative-PCR, Western blot, and immunohistochemistry. Moreover, we investigated immunohistochemically the expression of ASIC2 in the IVD of TrkB-deficient mice. ASIC2 and TrkB mRNAs were found in normal human IVD and both increased significantly in degenerated IVD. ASIC2 and TrkB proteins were also found co-localized in a variable percentage of cells, being significantly higher in degenerated IVD than in controls. The murine IVD displayed ASIC2 immunoreactivity which was absent in the IVD of TrkB-deficient mice. Present results demonstrate the occurrence of ASIC2 and TrkB in the human IVD, and the increased expression of both in pathological IVD suggest their involvement in IVD degeneration. These data also suggest that TrkB-ligands might be involved in the regulation of ASIC2 expression, and therefore in mechanisms by which the IVD cells accommodate to low pH and hypertonicity.
Collapse
Affiliation(s)
- Antonio Cuesta
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
| | - Eliseo Viña
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
- Unidad de Cuidados Intensivos, Hospital de CabueñesGijón, Spain
| | - Roberto Cabo
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
| | - Gorka Vázquez
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
| | - Ramón Cobo
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
| | - Olivia García-Suárez
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
| | - José García-Cosamalón
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
- Servicio de Neurocirugía, Complejo Universitario Hospital de LeónLeón, Spain
- Fundación Leonesa ProneuroencienasLeón, Spain
| | - José A Vega
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de OviedoNorth Korea
- Fundación Leonesa ProneuroencienasLeón, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de ChileChile
| |
Collapse
|
43
|
Xu XX, Cao Y, Ding TT, Fu KY, Li Y, Xie QF. Role of TRPV1 and ASIC3 channels in experimental occlusal interference-induced hyperalgesia in rat masseter muscle. Eur J Pain 2015. [PMID: 26201614 DOI: 10.1002/ejp.758] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Masticatory muscle pain may occur following immediate occlusal alteration by dental treatment. The underlying mechanisms are poorly understood. Transient receptor potential vanilloid-1 (TRPV1) and acid-sensing ion channel-3 (ASIC3) mediate muscle hyperalgesia under various pathologic conditions. We have developed a rat model of experimental occlusal interference (EOI) that consistently induces mechanical hyperalgesia in jaw muscles. Whether TRPV1 and ASIC3 mediate this EOI-induced hyperalgesia is unknown. METHODS Rat model of EOI-induced masseter hyperalgesia was established. Real-time polymerase chain reaction, Western blot and retrograde labelling combined with immunofluorescence were performed to evaluate the modulation of TRPV1 and ASIC3 expression in trigeminal ganglia (TGs) and masseter afferents of rats after EOI. The effects of intramuscular administration of TRPV1 and ASIC3 antagonists on the EOI-induced hyperalgesia in masseter muscle were examined. RESULTS After EOI, gene expressions and protein levels of TRPV1 and ASIC3 in bilateral TGs were up-regulated. The percentage of ASIC3- (but not TRPV1-) positive neurons in masseter afferents increased after EOI. More small-sized and small to medium-sized masseter afferents expressed TRPV1 and ASIC3 separately following EOI. These changes peaked at day 7 and then returned to original status within 10 days after EOI. Intramuscular administration of the TRPV1 antagonist AMG-9810 partially reversed this mechanical hyperalgesia in masseter muscle. No improvement was exhibited after administration of the ASIC3 antagonist APETx2. Co-injection of AMG-9810 and APETx2 enhanced the effect of AMG-9810 administration alone. CONCLUSIONS Peripheral TRPV1 and ASIC3 contribute to the development of the EOI-induced mechanical hyperalgesia in masseter muscle.
Collapse
Affiliation(s)
- X X Xu
- Department of Prosthodontics, Peking University School & Hospital of Stomatology, Beijing, China.,Department of Biomedical Sciences, City University of Hong Kong, China
| | - Y Cao
- Department of Prosthodontics, Peking University School & Hospital of Stomatology, Beijing, China
| | - T T Ding
- Department of Prosthodontics, Peking University School & Hospital of Stomatology, Beijing, China
| | - K Y Fu
- Center for TMD and Orofacial Pain, Peking University School & Hospital of Stomatology, Beijing, China
| | - Y Li
- Department of Biomedical Sciences, City University of Hong Kong, China
| | - Q F Xie
- Department of Prosthodontics, Peking University School & Hospital of Stomatology, Beijing, China
| |
Collapse
|
44
|
Dussor G. ASICs as therapeutic targets for migraine. Neuropharmacology 2015; 94:64-71. [PMID: 25582295 PMCID: PMC4458434 DOI: 10.1016/j.neuropharm.2014.12.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/26/2014] [Accepted: 12/04/2014] [Indexed: 01/05/2023]
Abstract
Migraine is the most common neurological disorder and one of the most common chronic pain conditions. Despite its prevalence, the pathophysiology leading to migraine is poorly understood and the identification of new therapeutic targets has been slow. Several processes are currently thought to contribute to migraine including altered activity in the hypothalamus, cortical-spreading depression (CSD), and afferent sensory input from the cranial meninges. Decreased extracellular pH and subsequent activation of acid-sensing ion channels (ASICs) may contribute to each of these processes and may thus play a role in migraine pathophysiology. Although few studies have directly examined a role of ASICs in migraine, studies directly examining a connection have generated promising results including efficacy of ASIC blockers in both preclinical migraine models and in human migraine patients. The purpose of this review is to discuss the pathophysiology thought to contribute to migraine and findings that implicate decreased pH and/or ASICs in these events, as well as propose issues to be resolved in future studies of ASICs and migraine. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Greg Dussor
- The University of Texas at Dallas, School of Behavioral and Brain Sciences, GR-41, 800 West Campbell Road, Richardson, TX, 75080, USA.
| |
Collapse
|
45
|
Acidic microenvironment and bone pain in cancer-colonized bone. BONEKEY REPORTS 2015; 4:690. [PMID: 25987988 DOI: 10.1038/bonekey.2015.58] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/20/2015] [Indexed: 12/15/2022]
Abstract
Solid cancers and hematologic cancers frequently colonize bone and induce skeletal-related complications. Bone pain is one of the most common complications associated with cancer colonization in bone and a major cause of increased morbidity and diminished quality of life, leading to poor survival in cancer patients. Although the mechanisms responsible for cancer-associated bone pain (CABP) are poorly understood, it is likely that complex interactions among cancer cells, bone cells and peripheral nerve cells contribute to the pathophysiology of CABP. Clinical observations that specific inhibitors of osteoclasts reduce CABP indicate a critical role of osteoclasts. Osteoclasts are proton-secreting cells and acidify extracellular bone microenvironment. Cancer cell-colonized bone also releases proton/lactate to avoid intracellular acidification resulting from increased aerobic glycolysis known as the Warburg effect. Thus, extracellular microenvironment of cancer-colonized bone is acidic. Acidosis is algogenic for nociceptive sensory neurons. The bone is densely innervated by the sensory neurons that express acid-sensing nociceptors. Collectively, CABP is evoked by the activation of these nociceptors on the sensory neurons innervating bone by the acidic extracellular microenvironment created by bone-resorbing osteoclasts and bone-colonizing cancer cells. As current treatments do not satisfactorily control CABP and can elicit serious side effects, new therapeutic interventions are needed to manage CABP. Understanding of the cellular and molecular mechanism by which the acidic extracellular microenvironment is created in cancer-colonized bone and by which the expression and function of the acid-sensing nociceptors on the sensory neurons are regulated would facilitate to develop novel therapeutic approaches for the management of CABP.
Collapse
|
46
|
Increased expression of acid-sensing ion channel 3 within dorsal root ganglia in a rat model of bone cancer pain. Neuroreport 2015; 25:887-93. [PMID: 25006846 DOI: 10.1097/wnr.0000000000000182] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In an attempt to investigate the underlying mechanisms of cancer-induced bone pain, we investigated the presence of acid-sensing ion channel 3 (ASIC3) in dorsal root ganglia (DRG) neurons in an animal model of bone cancer pain. Forty-five female Sprague-Dawley rats were randomized into three groups: sham-operation group (sham), cancer-bearing animals killed after 7 days (C7), and cancer-bearing animals killed after 14 days (C14). After establishment of the bone cancer pain model, pain-related behavioral tests were performed to determine the paw withdrawal threshold of mechanical allodynia and thermal hyperalgesia, respectively. Reverse transcription-PCR, western blot, and immunofluorescence were used to determine mRNA and protein expression of ASIC3 in ipsilateral and contralateral lumbar 4-5 DRG neurons. Compared with the sham group, paw withdrawal threshold of mechanical allodynia and thermal hyperalgesia in the C14 group showed a significant decrease (P<0.01) from postoperation day 7 to the termination of the experiment. Compared with the sham group, the ipsilateral but not contralateral mRNA of ASIC3 was upregulated in the C14 group. Meanwhile, the ipsilateral protein expression of ASIC3 was increased in the C7 and C14 group compared with the sham group. Double-labeled immunofluorescence showed that ASIC3 and isolectin-B4 (IB4)-colocalized small DRG neurons in the C14 group were more than that in the sham group. Furthermore, we also found that there were more ASIC3 and neurofilament 200 (NF200)-colocalized DRG neurons in the C14 group than in the sham group. The upregulation of mRNA and protein levels of ASIC3 suggested its potential involvement in the development and maintenance of cancer-induced bone pain.
Collapse
|
47
|
Yoneda T, Hiasa M, Nagata Y, Okui T, White F. Contribution of acidic extracellular microenvironment of cancer-colonized bone to bone pain. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2677-84. [PMID: 25687976 DOI: 10.1016/j.bbamem.2015.02.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 02/01/2015] [Accepted: 02/05/2015] [Indexed: 12/31/2022]
Abstract
Solid and hematologic cancer colonized bone produces a number of pathologies. One of the most common complications is bone pain. Cancer-associated bone pain (CABP) is a major cause of increased morbidity and diminishes the quality of life and affects survival. Current treatments do not satisfactorily control CABP and can elicit adverse effects. Thus, new therapeutic interventions are needed to manage CABP. However, the mechanisms responsible for CABP are poorly understood. The observation that specific osteoclast inhibitors can reduce CABP in patients indicates a critical role of osteoclasts in the pathophysiology of CABP. Osteoclasts create an acidic extracellular microenvironment by secretion of protons via vacuolar proton pumps during bone resorption. In addition, bone-colonized cancer cells also release protons and lactate via plasma membrane pH regulators to avoid intracellular acidification resulting from increased aerobic glycolysis known as the Warburg effect. Since acidosis is algogenic for sensory neurons and bone is densely innervated by sensory neurons that express acid-sensing nociceptors, the acidic bone microenvironments can evoke CABP. Understanding of the mechanism by which the acidic extracellular microenvironment is created in cancer-colonized bone and the expression and function of the acid-sensing nociceptors are regulated should facilitate the development of novel approaches for management of CABP. Here, the contribution of the acidic microenvironment created in cancer-colonized bone to elicitation of CABP and potential therapeutic implications of blocking the development and recognition of acidic microenvironment will be described. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Toshiyuki Yoneda
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Masahiro Hiasa
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Yuki Nagata
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Tatsuo Okui
- Department of Medicine, Hematology/Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA.
| | - Fletcher White
- Department of Anesthesia, Paul and Carole Stark Neurosciences Research Institute, Indiana University, 320 West 15th Street, Indianapolis, IN 46202, USA.
| |
Collapse
|
48
|
Sluka KA, Gregory NS. The dichotomized role for acid sensing ion channels in musculoskeletal pain and inflammation. Neuropharmacology 2015; 94:58-63. [PMID: 25582293 DOI: 10.1016/j.neuropharm.2014.12.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 11/26/2014] [Accepted: 12/04/2014] [Indexed: 01/24/2023]
Abstract
Chronic muscle pain affects between 11 and 24% of the world's population with the majority of people experiencing musculoskeletal pain at some time in their life. Acid sensing ion channels (ASICs) are important sensors of modest decreases in extracellular pH that occur within the physiological range. These decreases in extracellular pH occur in response to inflammation, fatiguing exercise, and ischemia. Further, injection of acidic saline into muscle produces enhanced nociceptive behaviors in animals and pain in human subjects. Of the different types of ASICs, ASIC3 and ASIC1 have been implicated in transmission of nociceptive information from the musculoskeletal system. The current review will provide an overview of the evidence for ASIC3 and ASIC1 in musculoskeletal pain in both inflammatory and non-inflammatory models. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Kathleen A Sluka
- Department of Physical Therapy and Rehabilitation Science, Neuroscience Graduate Program, Pain Research Program, University of Iowa, Iowa City, IA 52242, USA.
| | - Nicholas S Gregory
- Department of Physical Therapy and Rehabilitation Science, Neuroscience Graduate Program, Pain Research Program, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
49
|
Lin SH, Sun WH, Chen CC. Genetic exploration of the role of acid-sensing ion channels. Neuropharmacology 2015; 94:99-118. [PMID: 25582292 DOI: 10.1016/j.neuropharm.2014.12.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/27/2022]
Abstract
Advanced gene targeting technology and related tools in mice have been incorporated into studies of acid-sensing ion channels (ASICs). A single ASIC subtype can be knocked out specifically and screened thoroughly for expression in the nervous system at the cellular level. Mapping studies have further shed light on the initiation and identification of related behavioral phenotypes. Here we review studies involving genetically engineered mouse models used to investigate the physiological function of individual ASIC subtypes: ASIC1 (and ASIC1a), ASIC2, ASIC3 and ASIC4. We discuss the detailed expression studies and significant phenotypes revealed with gene knockout for most known Asic subtypes. Each strategy designed to manipulate mouse genetics has advantages and disadvantages. We discuss the limitations of these Asic-knockout models and propose future directions to solve the genetic issues. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.
Collapse
Affiliation(s)
- Shing-Hong Lin
- Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.
| | - Wei-Hsin Sun
- Department of Life Sciences, National Central University, Jhongli 32054, Taiwan.
| | - Chih-Cheng Chen
- Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Academia Sinica, Taipei 115, Taiwan.
| |
Collapse
|
50
|
Sugimura N, Ikeuchi M, Izumi M, Kawano T, Aso K, Kato T, Ushida T, Yokoyama M, Tani T. Repeated intra-articular injections of acidic saline produce long-lasting joint pain and widespread hyperalgesia. Eur J Pain 2014; 19:629-38. [PMID: 25158678 DOI: 10.1002/ejp.584] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2014] [Indexed: 11/08/2022]
Abstract
BACKGROUND Synovial fluid in inflamed joint shows a drop in pH, which activates proton-gated ion channels in nociceptors. No studies have ever tried to develop and characterize acid-induced joint pain. METHODS Rats were injected intra-articularly with pH 4.0 acidic saline twice, 5 days apart. Pain-related behaviour tests including weight-bearing asymmetry, paw withdrawal threshold and knee compression threshold were conducted. To clarify the roles of proton-gated ion channels, rats were injected intra-articularly with selective antagonists for ASIC1a, ASIC3 and TRPV1 on day 5 (before the second injection) or on day 14. Underlying peripheral and central pain mechanisms were evaluated using joint histology, interleukin-1β concentrations in the synovium, single-fibre recording of the knee afferent and expression of phosphorylated cyclic adenosine monophosphate-responsive element-binding protein (p-CREB) in the spinal dorsal horn. RESULTS Repeated injections of acidic saline induced weight-bearing asymmetry, decrease in paw withdrawal threshold and knee compression threshold bilaterally, which lasted until day 28. Early administration of ASIC3 antagonist reduced the bilateral and long-lasting hyperalgesia. Neither articular degeneration nor synovial inflammation was observed. C-fibre of the knee afferent was activated by acidic saline, which was attenuated by pre-injection of ASIC3 antagonist. p-CREB expression was transiently up-regulated bilaterally on day 6, but not on day 14. CONCLUSION We developed and characterized a model of acid-induced long-lasting bilateral joint pain. Peripheral ASIC3 and spinal p-CREB played important roles for the development of hyperalgesia. This animal model gives insights into the mechanisms of joint pain, which is helpful in developing better pain treatments.
Collapse
Affiliation(s)
- N Sugimura
- Department of Orthopaedic Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|