1
|
Cairns SP, Lindinger MI. Lactic acidosis: implications for human exercise performance. Eur J Appl Physiol 2025:10.1007/s00421-025-05750-0. [PMID: 40088272 DOI: 10.1007/s00421-025-05750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/22/2025] [Indexed: 03/17/2025]
Abstract
During high-intensity exercise a lactic-acidosis occurs with raised myoplasmic and plasma concentrations of lactate- and protons ([lactate-], [H+] or pH). We critically evaluate whether this causes/contributes to fatigue during human exercise. Increases of [lactate-] per se (to 25 mM in plasma, 50 mM intracellularly) exert little detrimental effect on muscle performance while ingestion/infusion of lactate- can be ergogenic. An exercise-induced intracellular acidosis at the whole-muscle level (pHi falls from 7.1-7.0 to 6.9-6.3), incorporates small changes in slow-twitch fibres (pHi ~ 6.9) and large changes in fast-twitch fibres (pHi ~ 6.2). The relationship between peak force/power and acidosis during fatiguing contractions varies across exercise regimes implying that acidosis is not the sole cause of fatigue. Concomitant changes of other putative fatigue factors include phosphate metabolites, glycogen, ions and reactive oxygen species. Acidosis to pHi 6.7-6.6 at physiological temperatures (during recovery from exercise or induced in non-fatigued muscle), has minimal effect on force/power. Acidosis to pHi ~ 6.5-6.2 per se reduces maximum force (~12%), slows shortening velocity (~5%), and lowers peak power (~22%) in non-fatigued muscles/individuals. A pre-exercise induced-acidosis with ammonium chloride impairs exercise performance in humans and accelerates the decline of force/power (15-40% initial) in animal muscles stimulated repeatedly in situ. Raised [H+]i and diprotonated inorganic phosphate ([H2PO4-]i) act on myofilament proteins to reduce maximum cross-bridge activity, Ca2+-sensitivity, and myosin ATPase activity. Acidosis/[lactate-]o attenuates detrimental effects of large K+-disturbances on action potentials and force in non-fatigued muscle. We propose that depressive effects of acidosis and [H2PO4-]i on myofilament function dominate over the protective effects of acidosis/lactate- on action potentials during fatigue. Raised extracellular [H+]/[lactate-] do not usually cause central fatigue but do contribute to elevated perceived exertion and fatigue sensations by activating group III/IV muscle afferents. Modulation of H+/lactate- regulation (via extracellular H+-buffers, monocarboxylate transporters, carbonic anhydrase, carnosine) supports a role for intracellular acidosis in fatigue. In conclusion, current evidence advocates that severe acidosis in fast-twitch fibres can contribute to force/power fatigue during intense human exercise.
Collapse
Affiliation(s)
- Simeon P Cairns
- Sport and Recreation Research Institute New Zealand, School of Sport and Recreation, Faculty of Health and Environmental Sciences, Auckland University of Technology, Private Bag 92006, Auckland, 1020, New Zealand.
- Health and Rehabilitation Research Institute, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, 1020, New Zealand.
| | - Michael I Lindinger
- Research and Development, The Nutraceutical Alliance Inc, Guelph, ON, L8N 3Z5, Canada
| |
Collapse
|
2
|
Chiu IM, Sokol CL. Neuroimmune recognition of allergens. Curr Opin Immunol 2024; 90:102458. [PMID: 39213825 PMCID: PMC11423315 DOI: 10.1016/j.coi.2024.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Interactions between the nervous system and the immune system play crucial roles in initiating and directing the type 2 immune response. Sensory neurons can initiate innate and adaptive type 2 immunity through their ability to detect allergens and promote dendritic cell and mast cell responses. Neurons also indirectly promote type 2 inflammation through suppression of type 1 immune responses. Type 2 cytokines promote neuronal function by directly activating or sensitizing neurons. This positive neuroimmune feedback loop may not only enhance allergic inflammation but also promote the system-wide responses of aversion, anaphylaxis, and allergen polysensitization that are characteristic of allergic immunity.
Collapse
Affiliation(s)
- Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Caroline L Sokol
- Center for Immunology & Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
3
|
Wu Z, Pope SD, Ahmed NS, Leung DL, Hajjar S, Yue Q, Anand DM, Kopp EB, Okin D, Ma W, Kagan JC, Hargreaves DC, Medzhitov R, Zhou X. Control of Inflammatory Response by Tissue Microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.592432. [PMID: 38798655 PMCID: PMC11118380 DOI: 10.1101/2024.05.10.592432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Inflammation is an essential defense response but operates at the cost of normal functions. Whether and how the negative impact of inflammation is monitored remains largely unknown. Acidification of the tissue microenvironment is associated with inflammation. Here we investigated whether macrophages sense tissue acidification to adjust inflammatory responses. We found that acidic pH restructured the inflammatory response of macrophages in a gene-specific manner. We identified mammalian BRD4 as a novel intracellular pH sensor. Acidic pH disrupts the transcription condensates containing BRD4 and MED1, via histidine-enriched intrinsically disordered regions. Crucially, decrease in macrophage intracellular pH is necessary and sufficient to regulate transcriptional condensates in vitro and in vivo, acting as negative feedback to regulate the inflammatory response. Collectively, these findings uncovered a pH-dependent switch in transcriptional condensates that enables environmental sensing to directly control inflammation, with a broader implication for calibrating the magnitude and quality of inflammation by the inflammatory cost.
Collapse
Affiliation(s)
- Zhongyang Wu
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Scott D. Pope
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Nasiha S. Ahmed
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Diana L. Leung
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Stephanie Hajjar
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Qiuyu Yue
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Diya M. Anand
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Elizabeth B. Kopp
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Daniel Okin
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, 02115
| | - Weiyi Ma
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jonathan C. Kagan
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Diana C. Hargreaves
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine
| | - Xu Zhou
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
4
|
Alsaadi H, Peller J, Ghasemlou N, Kawaja MD. Immunohistochemical phenotype of sensory neurons associated with sympathetic plexuses in the trigeminal ganglia of adult nerve growth factor transgenic mice. J Comp Neurol 2024; 532:e25563. [PMID: 37986234 DOI: 10.1002/cne.25563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Following peripheral nerve injury, postganglionic sympathetic axons sprout into the affected sensory ganglia and form perineuronal sympathetic plexuses with somata of sensory neurons. This sympathosensory coupling contributes to the onset and persistence of injury-induced chronic pain. We have documented the presence of similar sympathetic plexuses in the trigeminal ganglia of adult mice that ectopically overexpress nerve growth factor (NGF), in the absence of nerve injury. In this study, we sought to further define the phenotype(s) of these trigeminal sensory neurons having sympathetic plexuses in our transgenic mice. Using quantitative immunofluorescence staining analyses, we show that the invading sympathetic axons specifically target sensory somata immunopositive for several biomarkers: NGF high-affinity receptor tyrosine kinase A (trkA), calcitonin gene-related peptide (CGRP), neurofilament heavy chain (NFH), and P2X purinoceptor 3 (P2X3). Based on these phenotypic characteristics, the majority of the sensory somata surrounded by sympathetic plexuses are likely to be NGF-responsive nociceptors (i.e., trkA expressing) that are peptidergic (i.e., CGRP expressing), myelinated (i.e., NFH expressing), and ATP sensitive (i.e., P2X3 expressing). Our data also show that very few sympathetic plexuses surround sensory somata expressing other nociceptive (pain) biomarkers, including substance P and acid-sensing ion channel 3. No sympathetic plexuses are associated with sensory somata that display isolectin B4 binding. Though the cellular mechanisms that trigger the formation of sympathetic plexus (with and without nerve injury) remain unknown, our new observations yield an unexpected specificity with which invading sympathetic axons appear to target a precise subtype of nociceptors. This selectivity likely contributes to pain development and maintenance associated with sympathosensory coupling.
Collapse
Affiliation(s)
- Hanin Alsaadi
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Jacob Peller
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Michael D Kawaja
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
5
|
Anselmi L, Ducrocq GP, Ruiz-Velasco V, Stocker SD, Higgins SP, Kaufman MP. Functional knockout of the TRPV1 channel has no effect on the exercise pressor reflex in rats. J Physiol 2023; 601:5241-5256. [PMID: 37878364 PMCID: PMC12042824 DOI: 10.1113/jp285267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023] Open
Abstract
The role played by the transient receptor potential vanilloid 1 (TRPV1) channel on the thin fibre afferents evoking the exercise pressor reflex is controversial. To shed light on this controversy, we compared the exercise pressor reflex between newly developed TRPV1+/+ , TRPV1+/- and TRPV1-/- rats. Carotid arterial injection of capsaicin (0.5 μg), evoked significant pressor responses in TRPV1+/+ and TRPV1+/- rats, but not in TRPV1-/- rats. In acutely isolated dorsal root ganglion neurons innervating the gastrocnemius muscles, capsaicin evoked inward currents in neurons isolated from TRPV1+/+ and TRPV1+/- rats but not in neurons isolated from TRPV1-/- rats. The reflex was evoked by stimulating the tibial nerve in decerebrated rats whose femoral artery was either freely perfused or occluded. We found no difference between the reflex in the three groups of rats regardless of the patency of the femoral artery. For example, the peak pressor responses to contraction in TRPV1+/+ , TRPV1+/- and TRPV1-/- rats with patent femoral arteries averaged 17.1 ± 7.2, 18.9 ± 12.4 and 18.4 ± 8.6 mmHg, respectively. Stimulation of the tibial nerve after paralysis with pancuronium had no effect on arterial pressure, findings which indicated that the pressor responses to contraction were not caused by electrical stimulation of afferent tibial nerve axons. We also found that expression levels of acid-sensing ion channel 1 and endoperoxide 4 receptor in the L4 and 5 dorsal root ganglia were not upregulated in the TRPV1-/- rats. We conclude that TRPV1 is not needed to evoke the exercise pressor reflex in rats whose contracting muscles have either a patent or an occluded arterial blood supply. KEY POINTS: A reflex arising in contracting skeletal muscle contributes to the increases in arterial blood pressure, cardiac output and breathing evoked by exercise. The sensory arm of the reflex comprises both mechanoreceptors and metaboreceptors, of which the latter signals that blood flow to exercising muscle is not meeting its metabolic demand. The nature of the channel on the metaboreceptor sensing a mismatch between supply and demand is controversial; some believe that it is the transient receptor potential vanilloid 1 (TRPV1) channel. Using genetically engineered rats in which the TRPV1 channel is rendered non-functional, we have shown that it is not needed to evoke the metaboreflex.
Collapse
Affiliation(s)
- Laura Anselmi
- Heart and Vascular Institute Penn State College of Medicine, Hershey, PA, USA
| | - Guillaume P Ducrocq
- Heart and Vascular Institute Penn State College of Medicine, Hershey, PA, USA
- Mitochondria, Oxidative Stress and Muscular Protection Laboratory (UR 3072), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Victor Ruiz-Velasco
- Heart and Vascular Institute Penn State College of Medicine, Hershey, PA, USA
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Sean D Stocker
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shannon P Higgins
- Heart and Vascular Institute Penn State College of Medicine, Hershey, PA, USA
| | - Marc P Kaufman
- Heart and Vascular Institute Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
6
|
Khataei T, Benson CJ. ASIC3 plays a protective role in delayed-onset muscle soreness (DOMS) through muscle acid sensation during exercise. FRONTIERS IN PAIN RESEARCH 2023; 4:1215197. [PMID: 37795390 PMCID: PMC10546048 DOI: 10.3389/fpain.2023.1215197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/10/2023] [Indexed: 10/06/2023] Open
Abstract
Immediate exercise-induced pain (IEIP) and DOMS are two types of exercise-induced muscle pain and can act as barriers to exercise. The burning sensation of IEIP occurs during and immediately after intensive exercise, whereas the soreness of DOMS occurs later. Acid-sensing ion channels (ASICs) within muscle afferents are activated by H+ and other chemicals and have been shown to play a role in various chronic muscle pain conditions. Here, we further defined the role of ASICs in IEIP, and also tested if ASIC3 is required for DOMS. After undergoing exhaustive treadmill exercise, exercise-induced muscle pain was assessed in wild-type (WT) and ASIC3-/- mice at baseline via muscle withdrawal threshold (MWT), immediately, and 24 h after exercise. Locomotor movement, grip strength, and repeat exercise performance were tested at baseline and 24 h after exercise to evaluate DOMS. We found that ASIC3-/- had similar baseline muscle pain, locomotor activity, grip strength, and exercise performance as WT mice. WT showed diminished MWT immediately after exercise indicating they developed IEIP, but ASIC3-/- mice did not. At 24 h after baseline exercise, both ASIC3-/- and WT had similarly lower MWT and grip strength, however, ASIC3-/- displayed significantly lower locomotor activity and repeat exercise performance at 24 h time points compared to WT. In addition, ASIC3-/- mice had higher muscle injury as measured by serum lactate dehydrogenase and creatine kinase levels at 24 h after exercise. These results show that ASIC3 is required for IEIP, but not DOMS, and in fact might play a protective role to prevent muscle injury associated with strenuous exercise.
Collapse
Affiliation(s)
- Tahsin Khataei
- Department of Internal Medicine, Roy J and Lucile A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa City VA Healthcare System, Iowa City, IA, United States
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, United States
| | - Christopher J. Benson
- Department of Internal Medicine, Roy J and Lucile A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa City VA Healthcare System, Iowa City, IA, United States
| |
Collapse
|
7
|
Filipović N, Marinović Guić M, Košta V, Vukojević K. Cardiac innervations in diabetes mellitus-Anatomical evidence of neuropathy. Anat Rec (Hoboken) 2023; 306:2345-2365. [PMID: 36251628 DOI: 10.1002/ar.25090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/09/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022]
Abstract
The extensive innervations of the heart include a complex network of sympathetic, parasympathetic, and sensory nerves connected in loops that serve to regulate cardiac output. Metabolic dysfunction in diabetes affects many different organ systems, including the cardiovascular system; it causes cardiac arrhythmias, silent myocardial ischemia, and sudden cardiac death, among others. These conditions are associated with damage to the nerves that innervate the heart, cardiac autonomic neuropathy (CAN), which is caused by various pathophysiological mechanisms. In this review, the main facts about the anatomy of cardiac innervations and the current knowledge of CAN, its pathophysiological mechanisms, and its diagnostic approach are discussed. In addition, anatomical evidence for CAN from human and animal studies has been summarized.
Collapse
Affiliation(s)
- Natalija Filipović
- Department of Anatomy, Histology and Embryology, Laboratory for Experimental Neurocardiology, University of Split School of Medicine, Split, Croatia
| | - Maja Marinović Guić
- Department of Diagnostic and Interventional Radiology, University Hospital of Split, Split, Croatia
- University Department of Health Studies, University of Split, Split, Croatia
| | - Vana Košta
- Department of Neurology, University Hospital of Split, Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, Laboratory for Experimental Neurocardiology, University of Split School of Medicine, Split, Croatia
| |
Collapse
|
8
|
Cervini GA, Rice M, Jasperse JL. Potential Local Mechanisms for Exercise-Induced Hypoalgesia in Response to Blood Flow Restriction Training. Cureus 2023; 15:e43219. [PMID: 37692724 PMCID: PMC10490383 DOI: 10.7759/cureus.43219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Overall, there is a great need within sports medicine to ensure that athletes can return from injury in an efficient, yet thorough manner. It is crucial to not avoid necessary difficulties in this process but also to ensure time-efficient rehabilitation. One of the more promising techniques to achieve timely recovery is blood flow restriction (BFR) training. BFR training is a growing and novel development that could be a vital tool to lighten the burden of recovery from injury in athletes. BFR utilizes a pneumatic tourniquet to limit blood flow in specific areas of the body. The use of BFR has been shown to potentially enhance the analgesic effects of exercise-induced hypoalgesia (EIH). By limiting pain, athletes will be less burdened by mobility and loading exercises required for them to effectively return to play. In a field where time away from sports can have massive implications, the need for tools to assist in the acceleration of the rehabilitation process is vital. Much of the work that has already been done in the field has been able to exploit the benefits of EIH and further enhance the body's capabilities through BFR. Studies have compared EIH at low- and high-intensity settings utilizing BFR with both resistance and aerobic exercise. The results of these studies show comparable beta-endorphin levels with high-intensity exercise without BFR and low-intensity exercise with BFR. Low-intensity training with BFR had greater local pain relief, perhaps indicating the promising effects of BFR in enhancing EIH. By reviewing the current literature on this topic, we hope that further progress can be made to better understand the mechanism behind BFR and its ability to enhance EIH. Currently, local metabolites are a major focus for the potential mechanism behind these effects. Mas-related G-protein-coupled receptors (Mrgprs) contribute to local pain pathways via mast cell degranulation. Similarly, chemokine receptor 2/chemokine ligand 2 (CCR2/CCL2) triggers mast cell degranulation and inflammation-induced pain. Finally, pain-reducing effects have been linked to anti-inflammatory IL-10 signaling and anaerobic metabolites via transient receptor potential vanilloid 1 (TRPV1). Through a better understanding of these metabolites and their mechanisms, it is possible to further exploit the use of BFR to not only serve athletes recovering from injury but also apply this information to better serve all patients.
Collapse
Affiliation(s)
- Giovanni A Cervini
- Biomedical Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Matthew Rice
- Biomedical Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Jeffrey L Jasperse
- Biomedical Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, USA
| |
Collapse
|
9
|
Hawashin A, Brakmann IC, Tian Y, Gründer S, Ortega-Ramírez AM. Modulation of Acid-Sensing Ion Channels by Tannic Acid and Green Tea via a Membrane-Mediated Mechanism. ACS Chem Neurosci 2023. [PMID: 37379568 DOI: 10.1021/acschemneuro.3c00032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated ion channels that contribute to pain perception and neurotransmission. Being involved in sensing inflammation and ischemia, ASIC1a and ASIC3 are promising drug targets. Polyphenol tannic acid (TA) as well as green tea can interact with a variety of ion channels, but their effect on ASICs remains unknown. In addition, it is unknown whether they interact with ion channels via a common mechanism. Here, we show that TA is a potent modulator of ASICs. TA inhibited the transient current of rat ASIC3 expressed in HEK cells with an apparent IC50 of 2.2 ± 0.6 μM; it potentiated the sustained current and induced a slowly declining decay current. In addition, it produced an acidic shift of the pH-dependent activation of ASIC3 and inhibited the window current at pH 7.0. Moreover, TA inhibited the transient current of ASIC1a, ASIC1b, and ASIC2a. Pentagalloylglucose that is chemically identical to the central part of TA and a green tea extract both had effects on ASIC3 comparable to TA. TA and green tea inhibited inward currents generated by gramicidin channels, indicating interaction with the membrane. These results show that TA, pentagalloylglucose, and green tea modulate ASICs and identify alteration of the membrane as the potential common mechanism of this modulation. These properties will limit clinical application of these molecules.
Collapse
Affiliation(s)
- Ammar Hawashin
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, Aachen D-52074, Germany
| | - Ilka C Brakmann
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, Aachen D-52074, Germany
| | - Yuemin Tian
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, Aachen D-52074, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, Aachen D-52074, Germany
| | | |
Collapse
|
10
|
López-Ramírez O, González-Garrido A. The role of acid sensing ion channels in the cardiovascular function. Front Physiol 2023; 14:1194948. [PMID: 37389121 PMCID: PMC10300344 DOI: 10.3389/fphys.2023.1194948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023] Open
Abstract
Acid Sensing Ion Channels (ASIC) are proton sensors involved in several physiological and pathophysiological functions including synaptic plasticity, sensory systems and nociception. ASIC channels have been ubiquitously localized in neurons and play a role in their excitability. Information about ASIC channels in cardiomyocyte function is limited. Evidence indicates that ASIC subunits are expressed in both, plasma membrane and intracellular compartments of mammalian cardiomyocytes, suggesting unrevealing functions in the cardiomyocyte physiology. ASIC channels are expressed in neurons of the peripheral nervous system including the nodose and dorsal root ganglia (DRG), both innervating the heart, where they play a dual role as mechanosensors and chemosensors. In baroreceptor neurons from nodose ganglia, mechanosensation is directly associated with ASIC2a channels for detection of changes in arterial pressure. ASIC channels expressed in DRG neurons have several roles in the cardiovascular function. First, ASIC2a/3 channel has been proposed as the molecular sensor of cardiac ischemic pain for its pH range activation, kinetics and the sustained current. Second, ASIC1a seems to have a critical role in ischemia-induced injury. And third, ASIC1a, 2 and 3 are part of the metabolic component of the exercise pressure reflex (EPR). This review consists of a summary of several reports about the role of ASIC channels in the cardiovascular system and its innervation.
Collapse
Affiliation(s)
- Omar López-Ramírez
- Instituto de Oftalmología Fundación de Asistencia Privada Conde de Valenciana, I.A.P., Mexico City, Mexico
| | - Antonia González-Garrido
- Laboratorio de Enfermedades Mendelianas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
11
|
Hung CH, Chin Y, Fong YO, Lee CH, Han DS, Lin JH, Sun WH, Chen CC. Acidosis-related pain and its receptors as targets for chronic pain. Pharmacol Ther 2023; 247:108444. [PMID: 37210007 DOI: 10.1016/j.pharmthera.2023.108444] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Sensing acidosis is an important somatosensory function in responses to ischemia, inflammation, and metabolic alteration. Accumulating evidence has shown that acidosis is an effective factor for pain induction and that many intractable chronic pain diseases are associated with acidosis signaling. Various receptors have been known to detect extracellular acidosis and all express in the somatosensory neurons, such as acid sensing ion channels (ASIC), transient receptor potential (TRP) channels and proton-sensing G-protein coupled receptors. In addition to sense noxious acidic stimulation, these proton-sensing receptors also play a vital role in pain processing. For example, ASICs and TRPs are involved in not only nociceptive activation but also anti-nociceptive effects as well as some other non-nociceptive pathways. Herein, we review recent progress in probing the roles of proton-sensing receptors in preclinical pain research and their clinical relevance. We also propose a new concept of sngception to address the specific somatosensory function of acid sensation. This review aims to connect these acid-sensing receptors with basic pain research and clinical pain diseases, thus helping with better understanding the acid-related pain pathogenesis and their potential therapeutic roles via the mechanism of acid-mediated antinociception.
Collapse
Affiliation(s)
- Chih-Hsien Hung
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin Chin
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-On Fong
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Der-Shen Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Jiann-Her Lin
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei-Hsin Sun
- Department of Life Science & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan; Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
12
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
13
|
Messina DN, Peralta ED, Seltzer AM, Patterson SI, Acosta CG. Age-dependent and modality-specific changes in the phenotypic markers Nav1.8, ASIC3, P2X3 and TRPM8 in male rat primary sensory neurons during healthy aging. Biogerontology 2023; 24:111-136. [PMID: 36478541 DOI: 10.1007/s10522-022-10000-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022]
Abstract
The effects during healthy aging of the tetrodotoxin-resistant voltage-gated sodium channel 1.8 (Nav1.8), the acid-sensing ion channel-3 (ASIC3), the purinergic-receptor 2X3 (P2X3) and transient receptor potential of melastatin-8 (TRPM8) on responses to non-noxious stimuli are poorly understood. These effects will influence the transferability to geriatric subjects of findings obtained using young animals. To evaluate the involvement of these functional markers in mechanical and cold sensitivity to non-noxious stimuli and their underlying mechanisms, we used a combination of immunohistochemistry and quantitation of immunostaining in sub-populations of neurons of the dorsal root ganglia (DRG), behavioral tests, pharmacological interventions and Western-blot in healthy male Wistar rats from 3 to 24 months of age. We found significantly decreased sensitivity to mechanical and cold stimuli in geriatric rats. These behavioural alterations occurred simultaneously with differing changes in the expression of Nav1.8, ASIC3, P2X3 and TRPM8 in the DRG at different ages. Using pharmacological blockade in vivo we demonstrated the involvement of ASIC3 and P2X3 in normal mechanosensation and of Nav1.8 and ASIC3 in cold sensitivity. Geriatric rats also exhibited reductions in the number of A-like large neurons and in the proportion of peptidergic to non-peptidergic neurons. The changes in normal sensory physiology in geriatric rats we report here strongly support the inclusion of aged rodents as an important group in the design of pre-clinical studies evaluating pain treatments.
Collapse
Affiliation(s)
- Diego N Messina
- Laboratorio de Estudios Neurobiológicos (LABENE), Facultad de Ciencias Médicas, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
| | - Emanuel D Peralta
- Laboratorio de Estudios Neurobiológicos (LABENE), Facultad de Ciencias Médicas, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
| | - Alicia M Seltzer
- Laboratorio de Estudios Neurobiológicos (LABENE), Facultad de Ciencias Médicas, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
| | - Sean I Patterson
- Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Histología y Embriología - CONICET, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
| | - Cristian G Acosta
- Laboratorio de Estudios Neurobiológicos (LABENE), Facultad de Ciencias Médicas, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Universidad Nacional de Cuyo, 5500, Mendoza, Argentina. .,Histology Laboratory 107, IHEM-Faculty of Medical Sciences, National University of Cuyo, Av. del Libertador 80, 5500, Mendoza, Argentina.
| |
Collapse
|
14
|
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
|
15
|
Pidoux L, Delanoe K, Barbier J, Marchand F, Lingueglia E, Deval E. Single Subcutaneous Injection of Lysophosphatidyl-Choline Evokes ASIC3-Dependent Increases of Spinal Dorsal Horn Neuron Activity. Front Mol Neurosci 2022; 15:880651. [PMID: 35774865 PMCID: PMC9239072 DOI: 10.3389/fnmol.2022.880651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022] Open
Abstract
Lysophosphatidyl-choline (LPC), a member of the phospholipid family, is an emerging player in pain. It is known to modulate different pain-related ion channels, including Acid-Sensing Ion Channel 3 (ASIC3), a cationic channel mainly expressed in peripheral sensory neurons. LPC potentiates ASIC3 current evoked by mild acidifications, but can also activate the channel at physiological pH. Very recently, LPC has been associated to chronic pain in patients suffering from fibromyalgia or osteoarthritis. Accordingly, repetitive injections of LPC within mouse muscle or joint generate both persistent pain-like and anxiety-like behaviors in an ASIC3-dependent manner. LPC has also been reported to generate acute pain behaviors when injected intraplantarly in rodents. Here, we explore the mechanism of action of a single cutaneous injection of LPC by studying its effects on spinal dorsal horn neurons. We combine pharmacological, molecular and functional approaches including in vitro patch clamp recordings and in vivo recordings of spinal neuronal activity. We show that a single cutaneous injection of LPC exclusively affects the nociceptive pathway, inducing an ASIC3-dependent sensitization of nociceptive fibers that leads to hyperexcitabilities of both high threshold (HT) and wide dynamic range (WDR) spinal neurons. ASIC3 is involved in LPC-induced increase of WDR neuron’s windup as well as in WDR and HT neuron’s mechanical hypersensitivity, and it participates, together with TRPV1, to HT neuron’s thermal hypersensitivity. The nociceptive input induced by a single LPC cutaneous rather induces short-term sensitization, contrary to previously described injections in muscle and joint. If the effects of peripheral LPC on nociceptive pathways appear to mainly depend on peripheral ASIC3 channels, their consequences on pain may also depend on the tissue injected. Our findings contribute to a better understanding of the nociceptive signaling pathway activated by peripheral LPC via ASIC3 channels, which is an important step regarding the ASIC3-dependent roles of this phospholipid in acute and chronic pain conditions.
Collapse
Affiliation(s)
- Ludivine Pidoux
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
| | - Kevin Delanoe
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
| | - Julie Barbier
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Fabien Marchand
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Eric Lingueglia
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
| | - Emmanuel Deval
- Université Côte d’Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France
- *Correspondence: Emmanuel Deval,
| |
Collapse
|
16
|
Albrecht PJ, Houk G, Ruggiero E, Dockum M, Czerwinski M, Betts J, Wymer JP, Argoff CE, Rice FL. Keratinocyte Biomarkers Distinguish Painful Diabetic Peripheral Neuropathy Patients and Correlate With Topical Lidocaine Responsiveness. FRONTIERS IN PAIN RESEARCH 2021; 2:790524. [PMID: 35295428 PMCID: PMC8915676 DOI: 10.3389/fpain.2021.790524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 01/11/2023] Open
Abstract
This study investigated quantifiable measures of cutaneous innervation and algesic keratinocyte biomarkers to determine correlations with clinical measures of patient pain perception, with the intent to better discriminate between diabetic patients with painful diabetic peripheral neuropathy (PDPN) compared to patients with low-pain diabetic peripheral neuropathy (lpDPN) or healthy control subjects. A secondary objective was to determine if topical treatment with a 5% lidocaine patch resulted in correlative changes among the quantifiable biomarkers and clinical measures of pain perception, indicative of potential PDPN pain relief. This open-label proof-of-principle clinical research study consisted of a pre-treatment skin biopsy, a 4-week topical 5% lidocaine patch treatment regimen for all patients and controls, and a post-treatment skin biopsy. Clinical measures of pain and functional interference were used to monitor patient symptoms and response for correlation with quantitative skin biopsy biomarkers of innervation (PGP9.5 and CGRP), and epidermal keratinocyte biomarkers (Nav1.6, Nav1.7, CGRP). Importantly, comparable significant losses of epidermal neural innervation (intraepidermal nerve fibers; IENF) and dermal innervation were observed among PDPN and lpDPN patients compared with control subjects, indicating that innervation loss alone may not be the driver of pain in diabetic neuropathy. In pre-treatment biopsies, keratinocyte Nav1.6, Nav1.7, and CGRP immunolabeling were all significantly increased among PDPN patients compared with control subjects. Importantly, no keratinocyte biomarkers were significantly increased among the lpDPN group compared with control. In post-treatment biopsies, the keratinocyte Nav1.6, Nav1.7, and CGRP immunolabeling intensities were no longer different between control, lpDPN, or PDPN cohorts, indicating that lidocaine treatment modified the PDPN-related keratinocyte increases. Analysis of the PDPN responder population demonstrated that increased pretreatment keratinocyte biomarker immunolabeling for Nav1.6, Nav1.7, and CGRP correlated with positive outcomes to topical lidocaine treatment. Epidermal keratinocytes modulate the signaling of IENF, and several analgesic and algesic signaling systems have been identified. These results further implicate epidermal signaling mechanisms as modulators of neuropathic pain conditions, highlight a novel potential mode of action for topical treatments, and demonstrate the utility of comprehensive skin biopsy evaluation to identify novel biomarkers in clinical pain studies.
Collapse
Affiliation(s)
- Phillip J. Albrecht
- Neuroscience and Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, United States
- Division of Health Sciences, University at Albany, Albany, NY, United States
- *Correspondence: Phillip J. Albrecht
| | - George Houk
- Neuroscience and Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, United States
| | - Elizabeth Ruggiero
- Neuroscience and Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, United States
| | - Marilyn Dockum
- Neuroscience and Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, United States
| | | | - Joseph Betts
- Neuroscience and Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, United States
| | - James P. Wymer
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Charles E. Argoff
- Department of Neurology, Albany Medical Center, Albany, NY, United States
| | - Frank L. Rice
- Neuroscience and Pain Research Group, Integrated Tissue Dynamics, LLC, Rensselaer, NY, United States
- Division of Health Sciences, University at Albany, Albany, NY, United States
| |
Collapse
|
17
|
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
|
18
|
Páez O, Segura-Chama P, Almanza A, Pellicer F, Mercado F. Properties and Differential Expression of H + Receptors in Dorsal Root Ganglia: Is a Labeled-Line Coding for Acid Nociception Possible? Front Physiol 2021; 12:733267. [PMID: 34764880 PMCID: PMC8576393 DOI: 10.3389/fphys.2021.733267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
Pain by chemical irritants is one of the less well-described aspects of nociception. The acidic substance is the paradigm of the chemical noxious compound. An acidic insult on cutaneous, subcutaneous and muscle tissue results in pain sensation. Acid (or H+) has at least two main receptor channels in dorsal root ganglia (DRG) nociceptors: the heat receptor transient receptor potential vanilloid 1 (TRPV1) and the acid-sensing ionic channels (ASICs). TRPV1 is a low-sensitivity H+ receptor, whereas ASIC channels display a higher H+ sensitivity of at least one order of magnitude. In this review, we first describe the functional and structural characteristics of these and other H+-receptor candidates and the biophysics of their responses to low pH. Additionally, we compile reports of the expression of these H+-receptors (and other possible complementary proteins) within the DRG and compare these data with mRNA expression profiles from single-cell sequencing datasets for ASIC3, ASIC1, transient receptor potential Ankiryn subtype 1 (TRPA1) and TRPV1. We show that few nociceptor subpopulations (discriminated by unbiased classifications) combine acid-sensitive channels. This comparative review is presented in light of the accumulating evidence for labeled-line coding for most noxious sensory stimuli.
Collapse
Affiliation(s)
- Omar Páez
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Nuerociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Pedro Segura-Chama
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Nuerociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
- Cátedras CONACyT, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Angélica Almanza
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Nuerociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Francisco Pellicer
- Laboratorio de Neurofisiología Integrativa, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Francisco Mercado
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Nuerociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| |
Collapse
|
19
|
Morgan M, Thai J, Trinh P, Habib M, Effendi KN, Ivanusic JJ. ASIC3 inhibition modulates inflammation-induced changes in the activity and sensitivity of Aδ and C fiber sensory neurons that innervate bone. Mol Pain 2021; 16:1744806920975950. [PMID: 33280501 PMCID: PMC7724402 DOI: 10.1177/1744806920975950] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Acid Sensing Ion Channel 3 (ASIC3) is a non-selective cation channel that is
activated by acidification, and is known to have a role in regulating
inflammatory pain. It has pro-algesic roles in a range of conditions that
present with bone pain, but the mechanism for this has not yet been
demonstrated. We aimed to determine if ASIC3 is expressed in Aδ and/or C fiber
bone afferent neurons, and to explore its role in the activation and
sensitization of bone afferent neurons after acute inflammation. A combination
of retrograde tracing and immunohistochemistry was used to determine expression
of ASIC3 in the soma of bone afferent neurons. A novel, in
vivo, electrophysiological bone-nerve preparation was used to make
recordings of the activity and sensitivity of bone afferent neurons in the
presence of carrageenan-induced inflammation, with and without the selective
ASIC3 inhibitor APET×2. A substantial proportion of bone afferent neurons
express ASIC3, including unmyelinated (neurofilament poor) and small diameter
myelinated (neurofilament rich) neurons that are likely to be C and Aδ nerve
fibers respectively. Electrophysiological recordings revealed that application
of APET×2 to the marrow cavity inhibited carrageenan-induced spontaneous
activity of C and Aδ fiber bone afferent neurons. APET×2 also inhibited
carrageenan-induced sensitization of Aδ and C fiber bone afferent neurons to
mechanical stimulation, but had no effect on the sensitivity of bone afferent
neurons in the absence of inflammation. This evidence supports a role for ASIC3
in the pathogenesis of pain associated with inflammation of the bone.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Phu Trinh
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Mohamed Habib
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Kelly N Effendi
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Fibromyalgia: Pathogenesis, Mechanisms, Diagnosis and Treatment Options Update. Int J Mol Sci 2021; 22:ijms22083891. [PMID: 33918736 PMCID: PMC8068842 DOI: 10.3390/ijms22083891] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Fibromyalgia is a syndrome characterized by chronic and widespread musculoskeletal pain, often accompanied by other symptoms, such as fatigue, intestinal disorders and alterations in sleep and mood. It is estimated that two to eight percent of the world population is affected by fibromyalgia. From a medical point of view, this pathology still presents inexplicable aspects. It is known that fibromyalgia is caused by a central sensitization phenomenon characterized by the dysfunction of neuro-circuits, which involves the perception, transmission and processing of afferent nociceptive stimuli, with the prevalent manifestation of pain at the level of the locomotor system. In recent years, the pathogenesis of fibromyalgia has also been linked to other factors, such as inflammatory, immune, endocrine, genetic and psychosocial factors. A rheumatologist typically makes a diagnosis of fibromyalgia when the patient describes a history of pain spreading in all quadrants of the body for at least three months and when pain is caused by digital pressure in at least 11 out of 18 allogenic points, called tender points. Fibromyalgia does not involve organic damage, and several diagnostic approaches have been developed in recent years, including the analysis of genetic, epigenetic and serological biomarkers. Symptoms often begin after physical or emotional trauma, but in many cases, there appears to be no obvious trigger. Women are more prone to developing the disease than men. Unfortunately, the conventional medical therapies that target this pathology produce limited benefits. They remain largely pharmacological in nature and tend to treat the symptomatic aspects of various disorders reported by the patient. The statistics, however, highlight the fact that 90% of people with fibromyalgia also turn to complementary medicine to manage their symptoms.
Collapse
|
21
|
Ishizawa R, Kim HK, Hotta N, Iwamoto GA, Mitchell JH, Smith SA, Vongpatanasin W, Mizuno M. TRPV1 (Transient Receptor Potential Vanilloid 1) Sensitization of Skeletal Muscle Afferents in Type 2 Diabetic Rats With Hyperglycemia. Hypertension 2021; 77:1360-1371. [PMID: 33641357 DOI: 10.1161/hypertensionaha.120.15672] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Rie Ishizawa
- From the Departments of Applied Clinical Research (R.I., S.A.S., M.M.), University of Texas Southwestern Medical Center, Dallas
| | - Han-Kyul Kim
- Internal Medicine (H.-K.K., J.H.M., S.A.S., W.V., M.M.), University of Texas Southwestern Medical Center, Dallas
| | - Norio Hotta
- College of Life and Health Sciences, Chubu University, Kasugai, Japan (N.H.)
| | - Gary A Iwamoto
- Cell Biology (G.A.I.), University of Texas Southwestern Medical Center, Dallas
| | - Jere H Mitchell
- Internal Medicine (H.-K.K., J.H.M., S.A.S., W.V., M.M.), University of Texas Southwestern Medical Center, Dallas
| | - Scott A Smith
- From the Departments of Applied Clinical Research (R.I., S.A.S., M.M.), University of Texas Southwestern Medical Center, Dallas.,Internal Medicine (H.-K.K., J.H.M., S.A.S., W.V., M.M.), University of Texas Southwestern Medical Center, Dallas
| | - Wanpen Vongpatanasin
- Internal Medicine (H.-K.K., J.H.M., S.A.S., W.V., M.M.), University of Texas Southwestern Medical Center, Dallas
| | - Masaki Mizuno
- From the Departments of Applied Clinical Research (R.I., S.A.S., M.M.), University of Texas Southwestern Medical Center, Dallas.,Internal Medicine (H.-K.K., J.H.M., S.A.S., W.V., M.M.), University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
22
|
Hung CH, Lee CH, Tsai MH, Chen CH, Lin HF, Hsu CY, Lai CL, Chen CC. Activation of acid-sensing ion channel 3 by lysophosphatidylcholine 16:0 mediates psychological stress-induced fibromyalgia-like pain. Ann Rheum Dis 2020; 79:1644-1656. [PMID: 32907805 PMCID: PMC7677496 DOI: 10.1136/annrheumdis-2020-218329] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Fibromyalgia is commonly considered a stress-related chronic pain disorder, and daily stressors are known triggers. However, the relation between stress and pain development remains poorly defined by clinical approaches. Also, the aetiology remains largely unknown. METHODS We used a newly developed mouse model and lipidomic approaches to probe the causation and explore the biological plausibility for how perceived stress translates into chronic non-inflammatory pain. Clinical and lipidomic investigations of fibromyalgia were conducted for human validation. RESULTS Using non-painful sound stimuli as psychological stressors, we demonstrated that mice developed long-lasting non-inflammatory hyperalgesia after repeated and intermittent sound stress exposure. Elevated serum malondialdehyde level in stressed mice indicated excessive oxidative stress and lipid oxidative damage. Lipidomics revealed upregulation of lysophosphatidylcholine 16:0 (LPC16:0), a product of lipid oxidisation, in stressed mice. Intramuscular LPC16:0 injection triggered nociceptive responses and a hyperalgesic priming-like effect that caused long-lasting hypersensitivity. Pharmacological or genetic inhibition of acid-sensing ion channel 3 impeded the development of LPC16:0-induced chronic hyperalgesia. Darapladib and antioxidants could effectively alleviate the stress-induced hyperalgesia by inhibiting LPC16:0 synthesis. Clinical investigations showed that excessive oxidative stress and LPC16:0 expression also exist in patients with fibromyalgia. Moreover, LPC16:0 expression was correlated with pain symptoms in patients with high oxidative stress and disease severity. CONCLUSIONS Our study provides experimental evidence for the causal effect of psychological stressors on chronic pain development. The findings identify a possible pathophysiological mechanism of stress-induced chronic non-inflammatory pain at molecular, behavioural and clinical levels that might indicate a new therapeutic approach for fibromyalgia.
Collapse
Affiliation(s)
- Chih-Hsien Hung
- Department of Neurology, Kaohsiung Medical University Hospital; Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- PhD program in Translational Medicine, Kaohsiung Medical University and Academia Sinica, Kaohsiung / Taipei, Taiwan
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ming-Hsien Tsai
- Department of Child Care, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, Texas, USA
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsiu-Fen Lin
- Department of Neurology, Kaohsiung Medical University Hospital; Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Yao Hsu
- Department of Neurology, Kaohsiung Medical University Hospital; Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chiou-Lian Lai
- Department of Neurology, Kaohsiung Medical University Hospital; Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD program in Translational Medicine, Kaohsiung Medical University and Academia Sinica, Kaohsiung / Taipei, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- PhD program in Translational Medicine, Kaohsiung Medical University and Academia Sinica, Kaohsiung / Taipei, Taiwan
- Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
23
|
Peterson RA, König C, Zimmermann K, Barry CM, Wiklendt L, Brookes SJH. Effects of Lactate on One Class of Group III (CT3) Muscle Afferents. Front Cell Neurosci 2020; 14:215. [PMID: 32848615 PMCID: PMC7424053 DOI: 10.3389/fncel.2020.00215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
A class of Group III muscle afferent neurons has branching sensory terminals in the connective tissue between layers of mouse abdominal muscles (“CT3 muscle afferents”). These sensory endings are both mechanosensitive and metabosensitive. In the present study, responses of CT3 afferents to lactate ions and changes in temperature were recorded. Raising muscle temperature from 32.7°C to 37°C had no consistent effects on CT3 afferent basal firing rate or responses to either von Frey hair stimulation or to an applied load. Superfusion with lactate ions (15 mM, pH 7.4) was associated with an increase in firing from 6 ± 0.7 Hz to 11.7 ± 6.7 Hz (14 units, n = 13, P < 0.05, P = 0.0484) but with considerable variability in the nature and latency of response. Reducing the concentration of extracellular divalent cations, which mimicked the chelating effects of lactate, did not increase firing. Raised concentrations of divalent cations (to compensate for chelation) did not block excitatory effects of lactate on CT3 afferents, suggesting that effects via ASIC3 were not involved. Messenger RNA for the G-protein coupled receptor, hydroxyl carboxylic acid receptor 1 (HCAR1) was detected in dorsal root ganglia and HCAR1-like immunoreactivity was present in spinal afferent nerve cell bodies retrogradely labeled from mouse abdominal muscles. HCAR1-like immunoreactivity was also present in axons in mouse abdominal muscles. This raises the possibility that some effects of lactate on group III muscle afferents may be mediated by HCAR1.
Collapse
Affiliation(s)
- Rochelle A Peterson
- Neurogastroentrology Laboratory, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Christine König
- Klinik für Anästhesiologie am Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Katharina Zimmermann
- Klinik für Anästhesiologie am Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christine M Barry
- Musculoskeletal Neurobiology Laboratory, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Lukasz Wiklendt
- Neurogastroentrology Laboratory, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Simon J H Brookes
- Neurogastroentrology Laboratory, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
24
|
Grotle AK, Macefield VG, Farquhar WB, O'Leary DS, Stone AJ. Recent advances in exercise pressor reflex function in health and disease. Auton Neurosci 2020; 228:102698. [PMID: 32861944 DOI: 10.1016/j.autneu.2020.102698] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 01/11/2023]
Abstract
Autonomic alterations at the onset of exercise are critical to redistribute cardiac output towards the contracting muscles while preventing a fall in arterial pressure due to excessive vasodilation within the contracting muscles. Neural mechanisms responsible for these adjustments include central command, the exercise pressor reflex, and arterial and cardiopulmonary baroreflexes. The exercise pressor reflex evokes reflex increases in sympathetic activity to the heart and systemic vessels and decreases in parasympathetic activity to the heart, which increases blood pressure (BP), heart rate, and total peripheral resistance through vasoconstriction of systemic vessels. In this review, we discuss recent advancements in our understanding of exercise pressor reflex function in health and disease. Specifically, we discuss emerging evidence suggesting that sympathetic vasoconstrictor drive to the contracting and non-contracting skeletal muscle is differentially controlled by central command and the metaboreflex in healthy conditions. Further, we discuss evidence from animal and human studies showing that cardiovascular diseases, including hypertension, diabetes, and heart failure, lead to an altered exercise pressor reflex function. We also provide an update on the mechanisms thought to underlie this altered exercise pressor reflex function in each of these diseases. Although these mechanisms are complex, multifactorial, and dependent on the etiology of the disease, there is a clear consensus that several mechanisms are involved. Ultimately, approaches targeting these mechanisms are clinically significant as they provide alternative therapeutic strategies to prevent adverse cardiovascular events while also reducing symptoms of exercise intolerance.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States of America
| | | | - William B Farquhar
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE, United States of America
| | - Donal S O'Leary
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Audrey J Stone
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States of America.
| |
Collapse
|
25
|
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
|
26
|
Khataei T, Harding AMS, Janahmadi M, El-Geneidy M, Agha-Alinejad H, Rajabi H, Snyder PM, Sluka KA, Benson CJ. ASICs are required for immediate exercise-induced muscle pain and are downregulated in sensory neurons by exercise training. J Appl Physiol (1985) 2020; 129:17-26. [PMID: 32463731 DOI: 10.1152/japplphysiol.00033.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exercise training is an effective therapy for many pain-related conditions, and trained athletes have lower pain perception compared with unconditioned people. Some painful conditions, including strenuous exercise, are associated with elevated levels of protons, metabolites, and inflammatory factors, which may activate receptors and/or ion channels, including acid-sensing ion channels (ASICs), on nociceptive sensory neurons. We hypothesized that ASICs are required for immediate exercise-induced muscle pain (IEIP) and that exercise training diminishes IEIP by modulating ASICs within muscle afferents. We found high-intensity interval training (HIIT) reduced IEIP in C57BL/6 mice and diminished ASIC mRNA levels in lumber dorsal root ganglia, and this downregulation of ASICs correlated with improved exercise capacity. Additionally, we found that ASIC3 -/- mice did not develop IEIP; however, the exercise capacity of ASIC3 -/- was similar to wild-type mice. These results suggest that ASICs are required for IEIP and that diminishment of IEIP after exercise training correlates with downregulation of ASICs in sensory neurons.NEW & NOTEWORTHY Exercise performance can be limited by the sensations of muscle fatigue and pain transmitted by muscle afferents. It has been proposed that exercise training abrogates these negative feedback signals. We found that acid-sensing ion channels (ASICs) are required for immediate exercise-induced muscle pain (IEIP). Moreover, exercise training prevented IEIP and was correlated with downregulation of ASICs in sensory neurons.
Collapse
Affiliation(s)
- Tahsin Khataei
- Department of Exercise Physiology, Tarbiat Modares University, Tehran, Iran.,Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | - Anne Marie S Harding
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | - Mahyar Janahmadi
- Department of Physiology and Neuroscience Research Center, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maram El-Geneidy
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | | | - Hamid Rajabi
- Department of Exercise Physiology, Kharazmi University, Tehran, Iran
| | - Peter M Snyder
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | - Kathleen A Sluka
- Department of Physical Therapy and Rehabilitation Science, The University of Iowa, Iowa City, Iowa.,Neuroscience Institute, The University of Iowa, Iowa City, Iowa
| | - Christopher J Benson
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa.,Department of Pharmacology, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
27
|
Kim JS, Ducrocq GP, Kaufman MP. Functional knockout of ASIC3 attenuates the exercise pressor reflex in decerebrated rats with ligated femoral arteries. Am J Physiol Heart Circ Physiol 2020; 318:H1316-H1324. [PMID: 32302492 PMCID: PMC7346538 DOI: 10.1152/ajpheart.00137.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/23/2020] [Accepted: 04/12/2020] [Indexed: 11/22/2022]
Abstract
The exercise pressor reflex arises from contracting muscle and is manifested by increases in arterial pressure, heart rate, and cardiac contractility. In patients with peripheral artery disease, the exercise pressor reflex is exaggerated. This effect is believed to be caused by a metabolite whose concentration is increased when the working muscles are inadequately perfused. Previous work in rats with simulated peripheral artery disease has shown that pharmacological blockade of acid-sensing ion channel 3 (ASIC3), which is found on group III and IV afferents, prevented the exaggeration of the exercise pressor reflex. Blockade of ASIC3, however, may have off-target effects that preclude a conclusion that ASIC3 plays a role in evoking the reflex in rats with simulated peripheral artery disease. In the present experiments performed in decerebrated rats with simulated peripheral artery disease, we compared the exercise pressor reflex in rats with a functional knockout of the ASIC3 (KO) with the reflex in their wild-type counterparts (WT). We found that the exercise pressor reflex in ASIC3 KO rats was significantly lower than the exercise pressor reflex in their WT counterparts (P < 0.05). ASIC 3 KO rats demonstrated lower pressor responses to intra-arterial injection of diprotonated phosphate (86 mM; pH 6.0), lactic acid (12 mM; pH 2.85), and capsaicin (0.2 μg; pH 7.2) (P < 0.05). In contrast, both ligated WT and ASIC3 KO rats displayed similar pressor responses to tendon stretch (P > 0.05). We conclude that ASIC3 play an important role in evoking the exaggerated exercise pressor reflex in rats with peripheral artery disease.NEW & NOTEWORTHY We used a genetic approach to test the hypothesis that the magnitude of the exercise pressor reflex evoked in ligated ASIC3 KO rats was significantly lower than the magnitude of the exercise pressor reflex evoked in their ligated wild-type (WT) counterparts. The pressor response to contraction in ligated ASIC3 KO rats was significantly smaller than was the pressor response to contraction in ligated WT rats.
Collapse
Affiliation(s)
- Joyce S Kim
- Heart and Vascular Institute and Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania
| | - Guillaume P Ducrocq
- Heart and Vascular Institute and Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania
| | - Marc P Kaufman
- Heart and Vascular Institute and Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
28
|
Garami A, Shimansky YP, Rumbus Z, Vizin RCL, Farkas N, Hegyi J, Szakacs Z, Solymar M, Csenkey A, Chiche DA, Kapil R, Kyle DJ, Van Horn WD, Hegyi P, Romanovsky AA. Hyperthermia induced by transient receptor potential vanilloid-1 (TRPV1) antagonists in human clinical trials: Insights from mathematical modeling and meta-analysis. Pharmacol Ther 2020; 208:107474. [PMID: 31926897 DOI: 10.1016/j.pharmthera.2020.107474] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
Antagonists of the transient receptor potential vanilloid-1 (TRPV1) channel alter body temperature (Tb) in laboratory animals and humans: most cause hyperthermia; some produce hypothermia; and yet others have no effect. TRPV1 can be activated by capsaicin (CAP), protons (low pH), and heat. First-generation (polymodal) TRPV1 antagonists potently block all three TRPV1 activation modes. Second-generation (mode-selective) TRPV1 antagonists potently block channel activation by CAP, but exert different effects (e.g., potentiation, no effect, or low-potency inhibition) in the proton mode, heat mode, or both. Based on our earlier studies in rats, only one mode of TRPV1 activation - by protons - is involved in thermoregulatory responses to TRPV1 antagonists. In rats, compounds that potently block, potentiate, or have no effect on proton activation cause hyperthermia, hypothermia, or no effect on Tb, respectively. A Tb response occurs when a TRPV1 antagonist blocks (in case of hyperthermia) or potentiates (hypothermia) the tonic TRPV1 activation by protons somewhere in the trunk, perhaps in muscles, and - via the acido-antithermogenic and acido-antivasoconstrictor reflexes - modulates thermogenesis and skin vasoconstriction. In this work, we used a mathematical model to analyze Tb data from human clinical trials of TRPV1 antagonists. The analysis suggests that, in humans, the hyperthermic effect depends on the antagonist's potency to block TRPV1 activation not only by protons, but also by heat, while the CAP activation mode is uninvolved. Whereas in rats TRPV1 drives thermoeffectors by mediating pH signals from the trunk, but not Tb signals, our analysis suggests that TRPV1 mediates both pH and thermal signals driving thermoregulation in humans. Hence, in humans (but not in rats), TRPV1 is likely to serve as a thermosensor of the thermoregulation system. We also conducted a meta-analysis of Tb data from human trials and found that polymodal TRPV1 antagonists (ABT-102, AZD1386, and V116517) increase Tb, whereas the mode-selective blocker NEO6860 does not. Several strategies of harnessing the thermoregulatory effects of TRPV1 antagonists in humans are discussed.
Collapse
Affiliation(s)
- Andras Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary.
| | - Yury P Shimansky
- Department of Neurobiology, Barrow Neurological Institute, Dignity Health, Phoenix, AZ, USA
| | - Zoltan Rumbus
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Robson C L Vizin
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA
| | - Nelli Farkas
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Judit Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Zsolt Szakacs
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Margit Solymar
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Alexandra Csenkey
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | | | | | | | - Wade D Van Horn
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Peter Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary; Department of Translational Medicine, First Department of Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Andrej A Romanovsky
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA; School of Molecular Sciences, Arizona State University, Tempe, AZ, USA; Zharko Pharma Inc., Olympia, WA, USA.
| |
Collapse
|
29
|
Zera T, Moraes DJA, da Silva MP, Fisher JP, Paton JFR. The Logic of Carotid Body Connectivity to the Brain. Physiology (Bethesda) 2020; 34:264-282. [PMID: 31165684 DOI: 10.1152/physiol.00057.2018] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The carotid body has emerged as a therapeutic target for cardio-respiratory-metabolic diseases. With the expansive functions of the chemoreflex, we sought mechanisms to explain differential control of individual responses. We purport a remarkable correlation between phenotype of a chemosensory unit (glomus cell-sensory afferent) with a distinct component of the reflex response. This logic could permit differential modulation of distinct chemoreflex responses, a strategy ideal for therapeutic exploitation.
Collapse
Affiliation(s)
- Tymoteusz Zera
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw , Warsaw , Poland
| | - Davi J A Moraes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo , São Paulo , Brazil
| | - Melina P da Silva
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo , São Paulo , Brazil
| | - James P Fisher
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland , Auckland , New Zealand
| | - Julian F R Paton
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland , Auckland , New Zealand
| |
Collapse
|
30
|
A dual role for peripheral GDNF signaling in nociception and cardiovascular reflexes in the mouse. Proc Natl Acad Sci U S A 2019; 117:698-707. [PMID: 31848242 DOI: 10.1073/pnas.1910905116] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Group III/IV muscle afferents transduce nociceptive signals and modulate exercise pressor reflexes (EPRs). However, the mechanisms governing afferent responsiveness to dually modulate these processes are not well characterized. We and others have shown that ischemic injury can induce both nociception-related behaviors and exacerbated EPRs in the same mice. This correlated with primary muscle afferent sensitization and increased expression of glial cell line-derived neurotrophic factor (GDNF) in injured muscle and increased expression of GDNF family receptor α1 (GFRα1) in dorsal root ganglia (DRG). Here, we report that increased GDNF/GFRα1 signaling to sensory neurons from ischemia/reperfusion-affected muscle directly modulated nociceptive-like behaviors and increased exercise-mediated reflexes and group III/IV muscle afferent sensitization. This appeared to have taken effect through increased cyclic adenosine monophosphate (cAMP) response element binding (CREB)/CREB binding protein-mediated expression of the purinergic receptor P2X5 in the DRGs. Muscle GDNF signaling to neurons may, therefore, play an important dual role in nociception and sympathetic reflexes and could provide a therapeutic target for treating complications from ischemic injuries.
Collapse
|
31
|
Zhang Y, Huang L, Kozlov SA, Rubini P, Tang Y, Illes P. Acupuncture alleviates acid- and purine-induced pain in rodents. Br J Pharmacol 2019; 177:77-92. [PMID: 31444978 DOI: 10.1111/bph.14847] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Affiliation(s)
- Ying Zhang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Lumei Huang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Sergey A Kozlov
- Shemyakin-Ovchinikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Patrizia Rubini
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Peter Illes
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China.,Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
32
|
Hsu WH, Lee CH, Chao YM, Kuo CH, Ku WC, Chen CC, Lin YL. ASIC3-dependent metabolomics profiling of serum and urine in a mouse model of fibromyalgia. Sci Rep 2019; 9:12123. [PMID: 31431652 PMCID: PMC6702159 DOI: 10.1038/s41598-019-48315-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022] Open
Abstract
Fibromyalgia (FM) is characterized by chronic widespread pain. The pathogenesis of FM remains unclear. No specific biomarkers are available. Animal models of FM may provide an opportunity to explore potential biomarkers in a relative homogenous disease condition. Here, we probed the metabolomics profiles of serum and urine in a mouse model of FM induced by intermittent cold stress (ICS). We focused on the role of acid-sensing ion channel 3 (ASIC3) in the metabolomics profiling because ICS treatment induced chronic widespread muscle pain lasting for 1 month in wild-type (Asic3+/+) but not Asic3-knockout (Asic3−/−) mice. Serum and urine samples were collected from both genotypes at different ICS stages, including before ICS (basal level) and post-ICS at days 10 (middle phase, P10) and 40 (recovery phase, P40). Control naïve mice and ICS-induced FM mice differed in 1H-NMR- and LC-MS-based metabolomics profiling. On pathway analysis, the leading regulated pathways in Asic3+/+ mice were taurine and hypotaurine, cysteine and methionine, glycerophospholipid, and ascorbate and aldarate metabolisms, and the major pathways in Asic3−/− mice involved amino acid-related metabolism. Finally, we developed an algorithm for the impactful metabolites in the FM model including cis-aconitate, kynurenate, taurine, pyroglutamic acid, pyrrolidonecarboxylic acid, and 4-methoxyphenylacetic acid in urine as well as carnitine, deoxycholic acid, lysoPC(16:0), lysoPC(20:3), oleoyl-L-carnitine, and trimethylamine N-oxide in serum. Asic3−/− mice were impaired in only muscle allodynia development but not other pain symptoms in the ICS model, so the ASIC3-dependent metabolomics changes could be useful for developing diagnostic biomarkers specific to chronic widespread muscle pain, the core symptom of FM. Further pharmacological validations are needed to validate these metabolomics changes as potential biomarkers for FM diagnosis and/or treatment responses.
Collapse
Affiliation(s)
- Wei-Hsiang Hsu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Yen-Ming Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan
| | - Ching-Hua Kuo
- Department of Pharmacy, National Taiwan University, Taipei, 100, Taiwan
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, 24205, Taiwan
| | - Chih-Cheng Chen
- 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.
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
33
|
Matsubara T, Hayashi K, Wakatsuki K, Abe M, Ozaki N, Yamanaka A, Mizumura K, Taguchi T. Thin-fibre receptors expressing acid-sensing ion channel 3 contribute to muscular mechanical hypersensitivity after exercise. Eur J Pain 2019; 23:1801-1813. [PMID: 31314951 DOI: 10.1002/ejp.1454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/25/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Delayed onset muscle soreness (DOMS) is characterized by mechanical hyperalgesia after lengthening contractions (LC). It is relatively common and causes disturbance for many people who require continuous exercise, yet its molecular and peripheral neural mechanisms are poorly understood. METHODS We examined whether muscular myelinated Aδ-fibres, in addition to unmyelinated C-fibres, are involved in LC-induced mechanical hypersensitivity, and whether acid-sensing ion channel (ASIC)-3 expressed in thin-fibre afferents contributes to this type of pain using a rat model of DOMS. The peripheral contribution of ASIC3 was investigated using single-fibre electrophysiological recordings in extensor digitorum longus muscle-peroneal nerve preparations in vitro. RESULTS Behavioural tests demonstrated a significant decrease of the muscular mechanical withdrawal threshold following LC to ankle extensor muscles, and it was improved by intramuscular injection of APETx2 (2.2 μM), a selective blocker of ASIC3. The lower concentration of APETx2 (0.22 µM) and its vehicle had no effect on the threshold. Intramuscular injection of APETx2 (2.2 μM) in naïve rats without LC did not affect the withdrawal threshold. In the ankle extensor muscles that underwent LC one day before the electrophysiological recordings, the mechanical response of Aδ- and C-fibres was significantly facilitated (i.e. decreased response threshold and increased magnitude of the response). The facilitated mechanical response of the Aδ- and C-fibres was significantly suppressed by selective blockade of ASIC3 with APETx2, but not by its vehicle. CONCLUSIONS These results clearly indicate that ASIC3 contributes to the augmented mechanical response of muscle thin-fibre receptors in delayed onset muscular mechanical hypersensitivity after LC. SIGNIFICANCE Here, we show that not only C- but also Aδ-fibre nociceptors in the muscle are involved in mechanical hypersensitivity after lengthening contractions, and that acid-sensing ion channel (ASIC)-3 expressed in the thin-fibre nociceptors is responsible for the mechanical hypersensitivity. ASIC3 might be a novel pharmacological target for pain after exercise.
Collapse
Affiliation(s)
- Takanori Matsubara
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Functional Anatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Koei Hayashi
- Department of Functional Anatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Koji Wakatsuki
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Masahiro Abe
- Medical Information Department, Vitacain Pharmaceutical Co. Ltd, Osaka, Japan
| | - Noriyuki Ozaki
- Department of Functional Anatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazue Mizumura
- Department of Physical Therapy, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Toru Taguchi
- Department of Physical Therapy, Faculty of Rehabilitation, Niigata University of Health and Welfare, Niigata, Japan
| |
Collapse
|
34
|
Kim JS, Harms JE, Ruiz-Velasco V, Kaufman MP. Effect of knockout of the ASIC3 on cardiovascular reflexes arising from hindlimb muscle in decerebrated rats. Am J Physiol Regul Integr Comp Physiol 2019; 317:R641-R648. [PMID: 31347922 DOI: 10.1152/ajpregu.00148.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The exercise pressor reflex is initiated by the contraction-induced activation of group III and IV muscle afferents. The reflex is manifested by increases in arterial blood pressure and cardiac output, which, in turn, are generated by increases in the sympathetic outflow to the heart and vasculature and decreases in the vagal outflow to the heart. In previous experiments, we used a pharmacological approach to assess the role played by the acid-sensing ion channel 3 (ASIC3) on group III and IV afferents in evoking the exercise pressor reflex. In the present experiments, we used an alternative approach, namely functional knockout (KO) of the ASIC3 gene, to confirm and extend our previous finding that pharmacological blockade of the ASIC3 had only a small impact on the expression of the exercise pressor reflex when the arterial supply to the contracting hindlimb muscles of rats was patent. Using this alternative approach, we compared the magnitude of the exercise pressor reflex evoked in ASIC3 KO rats with that evoked in their wild-type (WT) counterparts. We found both WT and ASIC3 KO rats displayed similar pressor responses to static contraction (WT, n = 10, +12 ± 2 mmHg; KO, n = 9, +11 ± 2 mmHg) and calcaneal tendon stretch (WT, n = 9, +13 ± 2 mmHg; KO, n = 7, +11 ± 2 mmHg). Likewise, both WT and ASIC3 KO displayed similar pressor responses to intra-arterial injection of 12 mM lactic acid (WT, n = 9, +14 ± 3 mmHg; KO, n = 8, +18 ± 5 mmHg), 24 mM lactic acid (WT, n = 9,+24 ± 2 mmHg; KO, n = 8, +20 ± 5 mmHg), capsaicin (WT, n = 9,+27 ± 5 mmHg; KO, n = 10, +29 ± 5 mmHg), and diprotonated phosphate ([Formula: see text]; WT, n = 6,+22 ± 3 mmHg; KO, n = 6, +32 ± 6 mmHg). We conclude that redundant receptors are responsible for evoking the pressor reflexes arising from group III and IV afferents.
Collapse
Affiliation(s)
- Joyce S Kim
- Heart and Vascular Institute and the Departments of Medicine and Anesthesiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Jonathan E Harms
- Heart and Vascular Institute and the Departments of Medicine and Anesthesiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Heart and Vascular Institute and the Departments of Medicine and Anesthesiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Marc P Kaufman
- Heart and Vascular Institute and the Departments of Medicine and Anesthesiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
35
|
Roles of ASICs in Nociception and Proprioception. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1099:37-47. [PMID: 30306513 DOI: 10.1007/978-981-13-1756-9_4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acid-sensing ion channels (ASICs) are a group of proton-gated ion channels belonging to the degenerin/epithelial sodium channel (DED/ENaC) family. There are at least six ASIC subtypes - ASIC1a, ASIC1b, ASIC2a, ASIC2b, ASIC3, and ASIC4 - all expressed in somatosensory neurons. ASIC3 is the most abundant in dorsal root ganglia (DRG) and the most sensitive to extracellular acidification. ASICs were found as the major player involved in acid-induced pain in humans. Accumulating evidence has further shown ASIC3 as the molecular determinant involved in pain-associated tissue acidosis in rodent models. Besides having a role in nociception, members of the DEG/ENaC family have been demonstrated as essential mechanotransducers in the nematode Caenorhabditis elegans and fly Drosophila melanogaster. ASICs are mammalian homologues of DEG/ENaC and therefore may play a role in mechanotransduction. However, the role of ASICs in neurosensory mechanotransduction is disputed. Here we review recent studies to probe the roles of ASICs in acid nociception and neurosensory mechanotransduction. In reviewing genetic models and delicate electrophysiology approaches, we show ASIC3 as a dual-function protein for both acid-sensing and mechano-sensing in somatosensory nerves and therefore involved in regulating both nociception and proprioception.
Collapse
|
36
|
The evolution and multi-molecular properties of NF1 cutaneous neurofibromas originating from C-fiber sensory endings and terminal Schwann cells at normal sites of sensory terminations in the skin. PLoS One 2019; 14:e0216527. [PMID: 31107888 PMCID: PMC6527217 DOI: 10.1371/journal.pone.0216527] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 04/24/2019] [Indexed: 12/30/2022] Open
Abstract
In addition to large plexiform neurofibromas (pNF), NF1 patients are frequently disfigured by cutaneous neurofibromas (cNF) and are often afflicted with chronic pain and itch even from seemingly normal skin areas. Both pNFs and cNF consist primarily of benign hyperproliferating nonmyelinating Schwann cells (nSC). While pNF clearly arise within deep nerves and plexuses, the role of cutaneous innervation in the origin of cNF and in chronic itch and pain is unknown. First, we conducted a comprehensive, multi-molecular, immunofluorescence (IF) analyses on 3mm punch biopsies from three separate locations in normal appearing, cNF-free skin in 19 NF1 patients and skin of 16 normal subjects. At least one biopsy in 17 NF1 patients had previously undescribed micro-lesions consisting of a small, dense cluster of nonpeptidergic C-fiber endings and the affiliated nSC consistently adjoining adnexal structures—dermal papillae, hair follicles, sweat glands, sweat ducts, and arterioles—where C-fiber endings normally terminate. Similar micro-lesions were detected in hind paw skin of mice with conditionally-induced SC Nf1-/- mutations. Hypothesizing that these microlesions were pre-cNF origins of cNF, we subsequently analyzed numerous overt, small cNF (s-cNF, 3–6 mm) and discovered that each had an adnexal structure at the epicenter of vastly increased nonpeptidergic C-fiber terminals, accompanied by excessive nSC. The IF and functional genomics assays indicated that neurturin (NTRN) and artemin (ARTN) signaling through cRET kinase and GFRα2 and GFRα3 co-receptors on the aberrant C-fiber endings and nSC may mutually promote the onset of pre-cNF and their evolution to s-cNF. Moreover, TrpA1 and TrpV1 receptors may, respectively, mediate symptoms of chronic itch and pain. These newly discovered molecular characteristics might be targeted to suppress the development of cNF and to treat chronic itch and pain symptoms in NF1 patients.
Collapse
|
37
|
Zaremba M, Ruiz-Velasco V. Opioid-Mediated Modulation of Acid-Sensing Ion Channel Currents in Adult Rat Sensory Neurons. Mol Pharmacol 2019; 95:519-527. [PMID: 30808671 PMCID: PMC6442318 DOI: 10.1124/mol.118.114918] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/20/2019] [Indexed: 01/10/2023] Open
Abstract
Muscle ischemia, associated with peripheral artery disease (PAD), leads to the release of proinflammatory mediators that decrease extracellular pH and trigger the activation of proton-activated acid-sensing ion channels (ASIC). Claudication pain, linked with low blood flow, can be partially relieved by endogenous opioid peptide release. However, we previously reported that sustained ASIC currents in dorsal root ganglion (DRG) neurons were enhanced by naturally occurring endomorphin-1 and -2 opioid peptides, indicating a role of opioid involvement in hyperalgesia. The present study examined whether clinically employed synthetic (fentanyl, remifentanil) and the semisynthetic opioid (oxycodone) would also potentiate sustained ASIC currents, which arise from ASIC3 channel isoforms. Here, we show that exposure of each opioid to DRG neurons resulted in potentiation of the sustained ASIC currents. On the other hand, the potentiation was not observed in DRG neurons from ASIC3 knockout rats. Further, the enhancement of the ASIC currents was resistant to pertussis toxin treatment, suggesting that Gα i/Gα o G-proteins are not involved. Additionally, the potentiation of sustained ASIC currents was greater in DRG neurons isolated from rats with ligated femoral arteries (a model of PAD). The effect of all three opioids on the transient ASIC peak current was mixed (increase, decrease, no effect). The inhibitory action appears to be mediated by the presence of ASIC1 isoform, while the potentiating effect is primarily due to ASIC3 isoform expression. These findings reveal that, under certain conditions, these three opioids can increase ASIC channel activity, possibly giving rise to opioid-induced hyperalgesia.
Collapse
Affiliation(s)
- Malgorzata Zaremba
- Ruiz-Velasco Laboratory, Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Ruiz-Velasco Laboratory, Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
38
|
Abstract
Clinical conditions resulting in musculoskeletal pain show important sex differences in both prevalence and degree of functional disability. The underlying mechanisms for these distinctions in pain manifestation are not fully known. However, recent preclinical studies have shown at the primary afferent level that males and females present fundamental differences in their peripheral response properties and injury-related gene expression patterns that may underlie observed afferent sensitization. At the spinal cord level, studies in various models of pain suggest important roles for the immune system, glutamate signaling and hormones in modulating sex differences. While preclinical studies have been able to characterize some of the basic underlying molecular mechanisms of sex differences in muscle pain, human studies have relied mainly on functional brain imaging studies to explain differences. Further complicating our understanding of how sex influences muscle pain is the notion that the type of injury sustained, or clinical condition may differentially activate distinct mechanisms of muscle pain development in males versus females. More research is necessary to better understand how the sexes differ in their perception of muscle pain. This review highlights recent advances in both human and animal studies of sex differences in muscle pain.
Collapse
|
39
|
Interleukin 1β inhibition contributes to the antinociceptive effects of voluntary exercise on ischemia/reperfusion-induced hypersensitivity. Pain 2019; 159:380-392. [PMID: 29112534 DOI: 10.1097/j.pain.0000000000001094] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Issues of peripheral circulation have been increasingly suggested as an underlying cause of musculoskeletal pain in many conditions, including sickle cell anemia and peripheral vascular disease. We have previously shown in our model of transient ischemia and reperfusion (I/R) injury of the forelimb that individual group III and IV muscle afferents display altered chemosensitivity and mechanical thresholds 1 day after injury. Functional alterations corresponded to increased evoked and spontaneous pain-related behaviors and decreased muscle strength and voluntary activity-all actions that echo clinical symptoms of ischemic myalgia. These behavioral and physiological changes appeared to originate in part from the action of increased interleukin 1β (IL1β) in the injured muscles at its upregulated IL1 receptor 1 within the dorsal root ganglion. Here, we describe that two days of voluntary wheel running prior to I/R blocks both injury-induced IL1β enhancement and the subsequent development of ischemic myalgia-like behaviors. Furthermore, the protective effects of 2 days prior exercise on the I/R-evoked increases in pain-related behaviors were also paralleled with systemic injection of the IL1 receptor antagonist during I/R. Interleukin 1 receptor antagonist treatment additionally prevented the I/R-induced changes in mechanical and chemical sensitivity of individual primary muscle afferents. Altogether, these data strengthen the evidence that transient I/R injury sensitizes group III and IV muscle afferents via increased IL1β in the muscles to stimulate ischemic myalgia development. Targeting IL1β may, therefore, be an effective treatment strategy for this insidious type of muscle pain.
Collapse
|
40
|
Fabbretti E. P2X3 receptors are transducers of sensory signals. Brain Res Bull 2019; 151:119-124. [PMID: 30660716 DOI: 10.1016/j.brainresbull.2018.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/27/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022]
Abstract
Peripheral stimuli are transduced by specific receptors expressed by sensory neurons and are further processed in the dorsal horn of spinal cord before to be transmitted to the brain. While relative few receptor subtypes mediate the initial depolarisation of sensory neurons, an impressive number of molecules and ion channels integrate these inputs into coded signals. Soluble mediators and ambient conditions further shape these processes, potentially triggering peripheral and central sensitisation, or sensory downregulation. Extracellular ATP is a major signaling molecule that acts via purinergic receptors and is a powerful modulator of cell communication as well as a neurotransmitter at peripheral/central synapses. In particular, ATP-mediated signals are transduced by P2X3 receptors expressed mainly by peripheral sensory neurons. Recent evidence suggests that P2X3 receptor function not only induces neuron depolarisation and firing with consequent neurotransmitter release, but it also triggers intracellular molecular changes that amplify purinergic signaling with important consequences.
Collapse
Affiliation(s)
- Elsa Fabbretti
- Department of Life Science, University of Trieste, via Giorgieri 5, 34127, Trieste, Italy.
| |
Collapse
|
41
|
Wen X, Qian C, Zhang Y, Wu R, Lu L, Zhu C, Cheng X, Cui R, You H, Mei F, Gao J, Li F, Bu L, Qu S. Key pathway and gene alterations in the gastric mucosa associated with obesity and obesity-related diabetes. J Cell Biochem 2018; 120:6763-6771. [PMID: 30450618 DOI: 10.1002/jcb.27976] [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: 08/27/2018] [Accepted: 10/02/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE The stomach plays an important role in obesity and obesity-related diabetes; yet, little is known about key pathways in the gastric mucosa associated with obesity and diabetes. METHODS We performed gene microarray and real time-polymerase chain reaction (RT-PCR) on gut mucosa samples from control subjects (CON), patients with simple obesity (OB), and patients with obesity and comorbid diabetes (OD) (n = 3 per group). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were used to predict the functional significance of differentially expressed genes. RESULTS In total, 262 genes were upregulated and 265 genes were downregulated in the OB group whereas 1756 genes were upregulated and 1053 genes were downregulated in the OD group compared with the CON group. Of these, 23 were co-regulated in both comparisons. Seven differentially expressed genes were validated by RT-PCR (NRIP3, L1CAM, TPO, P2RY1, OR8A1, ADAMTS19, and ASIC3). A functional analysis revealed that genes differentially expressed between the OB or OD and CON groups played crucial roles in metabolic, T cell, and G-protein coupled receptor biological processes, and primarily participated in the PI3K-Akt and AGE-RAGE signaling pathways. CONCLUSIONS Obesity and obesity-related diabetes are associated with important gene expression and pathway alterations in the stomach.
Collapse
Affiliation(s)
- Xin Wen
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Chunhua Qian
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Yi Zhang
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, National key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ruijin Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liesheng Lu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cuiling Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Xiaoyun Cheng
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Rai Cui
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Hui You
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Fangyun Mei
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Jingyang Gao
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Feng Li
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Le Bu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,National Metabolic Management Center, Shanghai Tenth People's Hospital, Shanghai, China.,Thyroid Research Center of Shanghai, Shanghai Tenth People's Hospital, China
| |
Collapse
|
42
|
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
|
43
|
Fierro L. Serotonin-gated inward currents are three times more frequent in rat hairy skin sensory afferents than in those innervating the skeletal muscle. Mol Pain 2018; 13:1744806917729055. [PMID: 28868961 PMCID: PMC5588798 DOI: 10.1177/1744806917729055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Tight whole-cell patch clamp was performed in 191 DiI (1,1'-dioctadecyl-3,3,3'3'-tetramethylindocarbocyanine perchlorate) retrogradely labeled rat sensory afferents from skin shoulders ( n = 93) and biceps femoris muscles ( n = 98). 5-HT-gated inward currents were evoked with 50-µM serotonin (5-HT; 5-hydroxytryptamine), and their frequency and current densities were compared between skin and skeletal muscle sensory afferents. To evaluate if 5-HT-gated inward currents coexist with other ligand-gated currents, the skin and skeletal muscle sensory afferents were also sequentially exposed to external solution at pH 6.8, ATP (50 µM), and capsaicin (1 µM). 5-HT evoked inward currents in 72% (67 of 93) of hairy skin sensory afferents and in only 24% (24 of 98) of skeletal muscle sensory afferents, and this difference was statistically significant ( p < 0.0000, chi-square test). The current densities obtained in hairy skin and skeletal muscle sensory afferents were not significantly different. They were -45.8 ± 7.7 and -32.4 ± 10.5 pA/pF, respectively (mean ± SEM, p < 0.30734). These results indicate that 5-HT-gated inward currents are three times more frequently evoked in small- to medium-sized sensory afferents (25-40 µm) innervating the hairy skin than on those innervating the skeletal muscle. When cells were gathered in two clusters, the difference was four times larger in the small-sized cluster (25-32 µm) and two times larger in the medium-sized cluster (33-40 µm). The results can be explained if the superficial somatic (cutaneous) nociceptive system is more exposed than the deep somatic nociceptive system (musculoskeletal) to physical and chemical stimuli inducing 5-HT-mediated inflammatory pain.
Collapse
Affiliation(s)
- Leonardo Fierro
- Leonardo Fierro, Universidad del Valle, Calle 4 B No. 36–00, Edificio 116, Oficina 5001, Cali, Valle del Cauca 760042, Colombia.
| |
Collapse
|
44
|
Garami A, Pakai E, McDonald HA, Reilly RM, Gomtsyan A, Corrigan JJ, Pinter E, Zhu DXD, Lehto SG, Gavva NR, Kym PR, Romanovsky AA. TRPV1 antagonists that cause hypothermia, instead of hyperthermia, in rodents: Compounds' pharmacological profiles, in vivo targets, thermoeffectors recruited and implications for drug development. Acta Physiol (Oxf) 2018; 223:e13038. [PMID: 29352512 PMCID: PMC6032921 DOI: 10.1111/apha.13038] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/11/2018] [Accepted: 01/13/2018] [Indexed: 01/03/2023]
Abstract
AIM Thermoregulatory side effects hinder the development of transient receptor potential vanilloid-1 (TRPV1) antagonists as new painkillers. While many antagonists cause hyperthermia, a well-studied effect, some cause hypothermia. The mechanisms of this hypothermia are unknown and were studied herein. METHODS Two hypothermia-inducing TRPV1 antagonists, the newly synthesized A-1165901 and the known AMG7905, were used in physiological experiments in rats and mice. Their pharmacological profiles against rat TRPV1 were studied in vitro. RESULTS Administered peripherally, A-1165901 caused hypothermia in rats by either triggering tail-skin vasodilation (at thermoneutrality) or inhibiting thermogenesis (in the cold). A-1165901-induced hypothermia did not occur in rats with desensitized (by an intraperitoneal dose of the TRPV1 agonist resiniferatoxin) sensory abdominal nerves. The hypothermic responses to A-1165901 and AMG7905 (administered intragastrically or intraperitoneally) were absent in Trpv1-/- mice, even though both compounds evoked pronounced hypothermia in Trpv1+/+ mice. In vitro, both A-1165901 and AMG7905 potently potentiated TRPV1 activation by protons, while potently blocking channel activation by capsaicin. CONCLUSION TRPV1 antagonists cause hypothermia by an on-target action: on TRPV1 channels on abdominal sensory nerves. These channels are tonically activated by protons and drive the reflectory inhibition of thermogenesis and tail-skin vasoconstriction. Those TRPV1 antagonists that cause hypothermia further inhibit these cold defences, thus decreasing body temperature. SIGNIFICANCE TRPV1 antagonists (of capsaicin activation) are highly unusual in that they can cause both hyper- and hypothermia by modulating the same mechanism. For drug development, this means that both side effects can be dealt with simultaneously, by minimizing these compounds' interference with TRPV1 activation by protons.
Collapse
Affiliation(s)
- A. Garami
- Systemic Inflammation Laboratory (FeverLab); Trauma Research; St. Joseph's Hospital and Medical Center; Phoenix AZ USA
- Institute for Translational Medicine; Medical School; University of Pecs; Pecs Hungary
| | - E. Pakai
- Systemic Inflammation Laboratory (FeverLab); Trauma Research; St. Joseph's Hospital and Medical Center; Phoenix AZ USA
- Institute for Translational Medicine; Medical School; University of Pecs; Pecs Hungary
| | - H. A. McDonald
- Neuroscience Research; Global Pharmaceutical Research and Development; AbbVie; North Chicago IL USA
| | - R. M. Reilly
- Neuroscience Research; Global Pharmaceutical Research and Development; AbbVie; North Chicago IL USA
| | - A. Gomtsyan
- Neuroscience Research; Global Pharmaceutical Research and Development; AbbVie; North Chicago IL USA
| | - J. J. Corrigan
- Systemic Inflammation Laboratory (FeverLab); Trauma Research; St. Joseph's Hospital and Medical Center; Phoenix AZ USA
| | - E. Pinter
- Department of Pharmacology and Pharmacotherapy; Medical School and Janos Szentagothai Research Centre; University of Pecs; Pecs Hungary
| | - D. X. D. Zhu
- Department of Neuroscience; Amgen; Thousand Oaks CA USA
| | - S. G. Lehto
- Department of Neuroscience; Amgen; Thousand Oaks CA USA
| | - N. R. Gavva
- Department of Neuroscience; Amgen; Thousand Oaks CA USA
| | - P. R. Kym
- Neuroscience Research; Global Pharmaceutical Research and Development; AbbVie; North Chicago IL USA
| | - A. A. Romanovsky
- Systemic Inflammation Laboratory (FeverLab); Trauma Research; St. Joseph's Hospital and Medical Center; Phoenix AZ USA
| |
Collapse
|
45
|
Ni L, Fang P, Hu ZL, Zhou HY, Chen JG, Wang F, Jin Y. Identification and Function of Acid-sensing Ion Channels in RAW 264.7 Macrophage Cells. Curr Med Sci 2018; 38:436-442. [PMID: 30074209 DOI: 10.1007/s11596-018-1897-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/07/2017] [Indexed: 12/20/2022]
Abstract
Activation of acid-sensing ion channels (ASICs) plays an important role in neuroinflammation. Macrophage recruitment to the sites of inflammation is an essential step in host defense. ASIC1 and ASIC3 have been reported to mediate the endocytosis and maturation of bone marrow derived macrophages. However, the expression and inflammation-related functions of ASICs in RAW 264.7 cells, another common macrophage, are still elusive. In the present study, we first demonstrated the presence of ASIC1, ASIC2a and ASIC3 in RAW 264.7 macrophage cell line by using reverse transcriptase polymerase chain reaction (RT-PCR), Western blotting and immunofluorescence experiments. The non-specific ASICs inhibitor amiloride and specific homomeric ASICla blocker PcTxl reduced the production of iNOS and COX-2 by LPS-induced activating RAW 264.7 cells. Furthermore, not only amiloride but also PcTxl inhibited the migration and LPS-induced apoptosis of RAW 264.7 cells. Taken together, our findings suggest that ASICs promote the inflammatory response and apoptosis of RAW 264.7 cells, and ASICs may serve as a potential novel target for immunological disease therapy.
Collapse
Affiliation(s)
- Lan Ni
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peng Fang
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuang-Li Hu
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hai-Yun Zhou
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Guo Chen
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang Wang
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - You Jin
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
46
|
Pin-Barre C, Pellegrino C, Laurin F, Laurin J. Cerebral Ischemia Changed the Effect of Metabosensitive Muscle Afferents on Somatic Reflex Without Affecting Thalamic Activity. Front Physiol 2018; 9:638. [PMID: 29896119 PMCID: PMC5986926 DOI: 10.3389/fphys.2018.00638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/11/2018] [Indexed: 12/14/2022] Open
Abstract
The purpose of the present study was to examine the contribution of group III and IV metabosensitive afferents at spinal and supraspinal levels in rats subjected to middle cerebral artery occlusion (MCAO) with reperfusion during the acute phase. Animals were randomized in Control (n = 23), SHAM (n = 18), MCAO-D1 (n = 10), and MCAO-D7 (n = 20) groups. Rats performed the Electrical Von Frey and the Adhesive removal tests before the surgery and at day 1 (D1), D3, and D7 after MCAO. Animals were subjected to electrophysiological recordings including the responses of group III/IV metabosensitive afferents to combinations of chemical activators and the triceps brachii somatic reflex activity at D1 or D7. The response of ventral posterolateral (VPL) thalamic nuclei was also recorded after group III/IV afferent activation. Histological measurements were performed to assess the infarct size and to confirm the location of the recording electrodes into the VPL. Behavioral results indicated that MCAO induced disorders of both mechanical sensibility and motor coordination of paretic forepaw during 7 days. Moreover, injured animals exhibited an absence of somatic reflex inhibition from the group III/IV afferents at D1, without affecting the response of both these afferents and the VPL. Finally, the regulation of the central motor drive by group III/IV afferents was modified at spinal level during the acute phase of cerebral ischemia and it might contribute to the observed behavioral disturbances.
Collapse
|
47
|
Cheng YR, Jiang BY, Chen CC. Acid-sensing ion channels: dual function proteins for chemo-sensing and mechano-sensing. J Biomed Sci 2018; 25:46. [PMID: 29793480 PMCID: PMC5966886 DOI: 10.1186/s12929-018-0448-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
Background Acid-sensing ion channels (ASICs) are a group of amiloride-sensitive ligand-gated ion channels belonging to the family of degenerin/epithelial sodium channels. ASICs are predominantly expressed in both the peripheral and central nervous system and have been characterized as potent proton sensors to detect extracellular acidification in the periphery and brain. Main body Here we review the recent studies focusing on the physiological roles of ASICs in the nervous system. As the major acid-sensing membrane proteins in the nervous system, ASICs detect tissue acidosis occurring at tissue injury, inflammation, ischemia, stroke, and tumors as well as fatiguing muscle to activate pain-sensing nerves in the periphery and transmit pain signals to the brain. Arachidonic acid and lysophosphocholine have been identified as endogenous non-proton ligands activating ASICs in a neutral pH environment. On the other hand, ASICs are found involved in the tether model mechanotransduction, in which the extracellular matrix and cytoplasmic cytoskeletons act like a gating-spring to tether the mechanically activated ion channels and thus transmit the stimulus force to the channels. Accordingly, accumulating evidence has shown ASICs play important roles in mechanotransduction of proprioceptors, mechanoreceptors and nociceptors to monitor the homoeostatic status of muscle contraction, blood volume, and blood pressure as well as pain stimuli. Conclusion Together, ASICs are dual-function proteins for both chemosensation and mechanosensation involved in monitoring physiological homoeostasis and pathological signals.
Collapse
Affiliation(s)
- Yuan-Ren Cheng
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, 128, Academia Rd. Sec. 2, Taipei, 115, Taiwan
| | - Bo-Yang Jiang
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, 128, Academia Rd. Sec. 2, Taipei, 115, Taiwan
| | - Chih-Cheng Chen
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, 128, Academia Rd. Sec. 2, Taipei, 115, Taiwan. .,Taiwan Mouse Clinic - National Comprehensive Mouse Phenotyping and Drug Testing Center, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
48
|
Stephan G, Huang L, Tang Y, Vilotti S, Fabbretti E, Yu Y, Nörenberg W, Franke H, Gölöncsér F, Sperlágh B, Dopychai A, Hausmann R, Schmalzing G, Rubini P, Illes P. The ASIC3/P2X3 cognate receptor is a pain-relevant and ligand-gated cationic channel. Nat Commun 2018; 9:1354. [PMID: 29636447 PMCID: PMC5893604 DOI: 10.1038/s41467-018-03728-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 03/09/2018] [Indexed: 12/21/2022] Open
Abstract
Two subclasses of acid-sensing ion channels (ASIC3) and of ATP-sensitive P2X receptors (P2X3Rs) show a partially overlapping expression in sensory neurons. Here we report that both recombinant and native receptors interact with each other in multiple ways. Current measurements with the patch-clamp technique prove that ASIC3 stimulation strongly inhibits the P2X3R current partly by a Ca2+-dependent mechanism. The proton-binding site is critical for this effect and the two receptor channels appear to switch their ionic permeabilities during activation. Co-immunoprecipation proves the close association of the two protein structures. BN-PAGE and SDS-PAGE analysis is also best reconciled with the view that ASIC3 and P2X3Rs form a multiprotein structure. Finally, in vivo measurements in rats reveal the summation of pH and purinergically induced pain. In conclusion, the receptor subunits do not appear to form a heteromeric channel, but tightly associate with each other to form a protein complex, mediating unidirectional inhibition. Two subclasses of ligand-gated ion channels (ASIC3 and P2X3) are both present at sensory neurons and might be therefore subject to receptor crosstalk. Here authors use electrophysiology, biochemistry and co-immunoprecipitation to show that the two ion channels interact and affect P2X3 currents.
Collapse
Affiliation(s)
- Gabriele Stephan
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Lumei Huang
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany.,Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Sandra Vilotti
- Neurobiology Sector, International School for Advanced Studies, Trieste, 34136, Italy
| | - Elsa Fabbretti
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Ye Yu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai, 100025, China
| | - Wolfgang Nörenberg
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Heike Franke
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Flóra Gölöncsér
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1043, Hungary.,János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Budapest, 1043, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1043, Hungary
| | - Anke Dopychai
- Molecular Pharmacology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, 52072, Germany
| | - Ralf Hausmann
- Molecular Pharmacology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, 52072, Germany
| | - Günther Schmalzing
- Molecular Pharmacology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, 52072, Germany
| | - Patrizia Rubini
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany
| | - Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, University of Leipzig, Leipzig, 04107, Germany.
| |
Collapse
|
49
|
Montalbetti N, Rooney JG, Marciszyn AL, Carattino MD. ASIC3 fine-tunes bladder sensory signaling. Am J Physiol Renal Physiol 2018; 315:F870-F879. [PMID: 29561183 DOI: 10.1152/ajprenal.00630.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are trimeric proton-activated, cation-selective neuronal channels that are considered to play important roles in mechanosensation and nociception. Here we investigated the role of ASIC3, a subunit primarily expressed in sensory neurons, in bladder sensory signaling and function. We found that extracellular acidification evokes a transient increase in current, consistent with the kinetics of activation and desensitization of ASICs, in ~25% of the bladder sensory neurons harvested from both wild-type (WT) and ASIC3 knockout (KO) mice. The absence of ASIC3 increased the magnitude of the peak evoked by extracellular acidification and reduced the rate of decay of the ASIC-like currents. These findings suggest that ASICs are assembled as heteromers and that the absence of ASIC3 alters the composition of these channels in bladder sensory neurons. Consistent with the notion that ASIC3 serves as a proton sensor, 59% of the bladder sensory neurons harvested from WT, but none from ASIC3 KO mice, fired action potentials in response to extracellular acidification. Studies of bladder function revealed that ASIC3 deletion reduces voiding volume and the pressure required to trigger micturition. In summary, our findings indicate that ASIC3 plays a role in the control of bladder function by modulating the response of afferents to filling.
Collapse
Affiliation(s)
- Nicolas Montalbetti
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - James G Rooney
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Allison L Marciszyn
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania.,Department of Cell Biology, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
50
|
Abstract
Cell bodies of trigeminal mesencephalic nucleus (Vmes) neurons are located within the central nervous system, and therefore, peripheral as well as central acidosis can modulate the excitability of Vmes neurons. Here, we report the effect of acidic pH on voltage-gated Na channels in acutely isolated rat Vmes neurons using a conventional whole-cell patch clamp technique. Acidic pH (pH 6.0) slightly but significantly shifted both the activation and steady-state fast inactivation relationships toward depolarized potentials. However, acidic pH (pH 6.0) had a minor effect on the inactivation kinetics of voltage-gated Na channels. Less sensitivity of voltage-gated Na channels to acidic pH may allow Vmes neurons to transduce the precise proprioceptive information even under acidic pH conditions.
Collapse
|