51
|
Ghosh D, Nieves-Cintrón M, Tajada S, Brust-Mascher I, Horne MC, Hell JW, Dixon RE, Santana LF, Navedo MF. Dynamic L-type Ca V1.2 channel trafficking facilitates Ca V1.2 clustering and cooperative gating. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1341-1355. [PMID: 29959960 PMCID: PMC6407617 DOI: 10.1016/j.bbamcr.2018.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/22/2018] [Accepted: 06/26/2018] [Indexed: 11/21/2022]
Abstract
L-type CaV1.2 channels are key regulators of gene expression, cell excitability and muscle contraction. CaV1.2 channels organize in clusters throughout the plasma membrane. This channel organization has been suggested to contribute to the concerted activation of adjacent CaV1.2 channels (e.g. cooperative gating). Here, we tested the hypothesis that dynamic intracellular and perimembrane trafficking of CaV1.2 channels is critical for formation and dissolution of functional channel clusters mediating cooperative gating. We found that CaV1.2 moves in vesicular structures of circular and tubular shape with diverse intracellular and submembrane trafficking patterns. Both microtubules and actin filaments are required for dynamic movement of CaV1.2 vesicles. These vesicles undergo constitutive homotypic fusion and fission events that sustain CaV1.2 clustering, channel activity and cooperative gating. Our study suggests that CaV1.2 clusters and activity can be modulated by diverse and unique intracellular and perimembrane vesicular dynamics to fine-tune Ca2+ signals.
Collapse
Affiliation(s)
- Debapriya Ghosh
- Department of Pharmacology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Madeline Nieves-Cintrón
- Department of Pharmacology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Sendoa Tajada
- Department of Physiology & Membrane Biology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Ingrid Brust-Mascher
- Advanced Imaging Facility, School of Veterinary Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Mary C Horne
- Department of Pharmacology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Johannes W Hell
- Department of Pharmacology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Rose E Dixon
- Department of Physiology & Membrane Biology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Luis F Santana
- Department of Physiology & Membrane Biology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA
| | - Manuel F Navedo
- Department of Pharmacology, School of Medicine, One Shields Avenue, University of California, Davis, CA 95616, USA.
| |
Collapse
|
52
|
Kádková A, Synytsya V, Krusek J, Zímová L, Vlachová V. Molecular basis of TRPA1 regulation in nociceptive neurons. A review. Physiol Res 2018; 66:425-439. [PMID: 28730837 DOI: 10.33549/physiolres.933553] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Transient receptor potential A1 (TRPA1) is an excitatory ion channel that functions as a cellular sensor, detecting a wide range of proalgesic agents such as environmental irritants and endogenous products of inflammation and oxidative stress. Topical application of TRPA1 agonists produces an acute nociceptive response through peripheral release of neuropeptides, purines and other transmitters from activated sensory nerve endings. This, in turn, further regulates TRPA1 activity downstream of G-protein and phospholipase C-coupled signaling cascades. Despite the important physiological relevance of such regulation leading to nociceptor sensitization and consequent pain hypersensitivity, the specific domains through which TRPA1 undergoes post-translational modifications that affect its activation properties are yet to be determined at a molecular level. This review aims at providing an account of our current knowledge on molecular basis of regulation by neuronal inflammatory signaling pathways that converge on the TRPA1 channel protein and through modification of its specific residues influence the extent to which this channel may contribute to pain.
Collapse
Affiliation(s)
- A Kádková
- Department of Cellular Neurophysiology, Institute of Physiology CAS, Prague, Czech Republic. or
| | | | | | | | | |
Collapse
|
53
|
Hall BE, Prochazkova M, Sapio MR, Minetos P, Kurochkina N, Binukumar BK, Amin ND, Terse A, Joseph J, Raithel SJ, Mannes AJ, Pant HC, Chung MK, Iadarola MJ, Kulkarni AB. Phosphorylation of the Transient Receptor Potential Ankyrin 1 by Cyclin-dependent Kinase 5 affects Chemo-nociception. Sci Rep 2018; 8:1177. [PMID: 29352128 PMCID: PMC5775258 DOI: 10.1038/s41598-018-19532-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/04/2018] [Indexed: 12/25/2022] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a key neuronal kinase that is upregulated during inflammation, and can subsequently modulate sensitivity to nociceptive stimuli. We conducted an in silico screen for Cdk5 phosphorylation sites within proteins whose expression was enriched in nociceptors and identified the chemo-responsive ion channel Transient Receptor Potential Ankyrin 1 (TRPA1) as a possible Cdk5 substrate. Immunoprecipitated full length TRPA1 was shown to be phosphorylated by Cdk5 and this interaction was blocked by TFP5, an inhibitor that prevents activation of Cdk5. In vitro peptide-based kinase assay revealed that four of six TRPA1 Cdk5 consensus sites acted as substrates for Cdk5, and modeling of the ankyrin repeats disclosed that phosphorylation would occur at characteristic pockets within the (T/S)PLH motifs. Calcium imaging of trigeminal ganglion neurons from genetically engineered mice overexpressing or lacking the Cdk5 activator p35 displayed increased or decreased responsiveness, respectively, to stimulation with the TRPA1 agonist allylisothiocyanate (AITC). AITC-induced chemo-nociceptive behavior was also heightened in vivo in mice overexpressing p35 while being reduced in p35 knockout mice. Our findings demonstrate that TRPA1 is a substrate of Cdk5 and that Cdk5 activity is also able to modulate TRPA1 agonist-induced calcium influx and chemo-nociceptive behavioral responses.
Collapse
Affiliation(s)
- Bradford E Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Michaela Prochazkova
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Paul Minetos
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.,Tulane University School of Medicine, New Orleans, LA, USA
| | | | - B K Binukumar
- Institute of Genomics and Integrative Biology, New Delhi, India
| | - Niranjana D Amin
- Neuronal Cytoskeletal Protein Regulation Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Anita Terse
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - John Joseph
- University of Maryland, School of Dentistry, Baltimore, MD, USA
| | - Stephen J Raithel
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Harish C Pant
- Neuronal Cytoskeletal Protein Regulation Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Man-Kyo Chung
- University of Maryland, School of Dentistry, Baltimore, MD, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
54
|
Wang S, Kobayashi K, Kogure Y, Yamanaka H, Yamamoto S, Yagi H, Noguchi K, Dai Y. Negative Regulation of TRPA1 by AMPK in Primary Sensory Neurons as a Potential Mechanism of Painful Diabetic Neuropathy. Diabetes 2018; 67:98-109. [PMID: 29025860 DOI: 10.2337/db17-0503] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 10/06/2017] [Indexed: 11/13/2022]
Abstract
AMPK is a widely expressed intracellular energy sensor that monitors and modulates energy expenditure. Transient receptor potential ankyrin 1 (TRPA1) channel is a widely recognized chemical and thermal sensor that plays vital roles in pain transduction. In this study, we discovered a functional link between AMPK and TRPA1 in dorsal root ganglion (DRG) neurons, in which AMPK activation rapidly resulted in downregulation of membrane-associated TRPA1 and its channel activity within minutes. Treatment with two AMPK activators, metformin or AICAR, inhibited TRPA1 activity in DRG neurons by decreasing the amount of membrane-associated TRPA1. Metformin induced a dose-dependent inhibition of TRPA1-mediated calcium influx. Conversely, in diabetic db/db mice, AMPK activity was impaired in DRG neurons, and this was associated with a concomitant increase in membrane-associated TRPA1 and mechanical allodynia. Notably, these molecular and behavioral changes were normalized following treatment with AMPK activators. Moreover, high-glucose exposure decreased activated AMPK levels and increased agonist-evoked TRPA1 currents in cultured DRG neurons, and these effects were prevented by treatment with AMPK activators. Our results identify AMPK as a previously unknown regulator of TRPA1 channels. AMPK modulation of TRPA1 could thus serve as an underlying mechanism and potential therapeutic molecular target in painful diabetic neuropathy.
Collapse
Affiliation(s)
- Shenglan Wang
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
- Traditional Medicine Research Center, Chinese Medicine Confucius Institute at Hyogo College of Medicine, Kobe, Hyogo, Japan
| | - Kimiko Kobayashi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yoko Kogure
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
| | - Hiroki Yamanaka
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Satoshi Yamamoto
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
| | - Hideshi Yagi
- Department of Anatomy and Cell Biology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Koichi Noguchi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yi Dai
- Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo, Japan
- Traditional Medicine Research Center, Chinese Medicine Confucius Institute at Hyogo College of Medicine, Kobe, Hyogo, Japan
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
55
|
Jiang M, Taghizadeh F, Steyger PS. Potential Mechanisms Underlying Inflammation-Enhanced Aminoglycoside-Induced Cochleotoxicity. Front Cell Neurosci 2017; 11:362. [PMID: 29209174 PMCID: PMC5702304 DOI: 10.3389/fncel.2017.00362] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/03/2017] [Indexed: 12/20/2022] Open
Abstract
Aminoglycoside antibiotics remain widely used for urgent clinical treatment of life-threatening infections, despite the well-recognized risk of permanent hearing loss, i.e., cochleotoxicity. Recent studies show that aminoglycoside-induced cochleotoxicity is exacerbated by bacteriogenic-induced inflammation. This implies that those with severe bacterial infections (that induce systemic inflammation), and are treated with bactericidal aminoglycosides are at greater risk of drug-induced hearing loss than previously recognized. Incorporating this novel comorbid factor into cochleotoxicity risk prediction models will better predict which individuals are more predisposed to drug-induced hearing loss. Here, we review the cellular and/or signaling mechanisms by which host-mediated inflammatory responses to infection could enhance the trafficking of systemically administered aminoglycosides into the cochlea to enhance the degree of cochleotoxicity over that in healthy preclinical models. Once verified, these mechanisms will be potential targets for novel pharmacotherapeutics that reduce the risk of drug-induced hearing loss (and acute kidney damage) without compromising the life-saving bactericidal efficacy of aminoglycosides.
Collapse
Affiliation(s)
- Meiyan Jiang
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Farshid Taghizadeh
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Peter S Steyger
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR, United States.,National Center for Rehabilitative Auditory Research, VA Portland Health Care System, Portland, OR, United States
| |
Collapse
|
56
|
Viana F. TRPA1 channels: molecular sentinels of cellular stress and tissue damage. J Physiol 2017; 594:4151-69. [PMID: 27079970 DOI: 10.1113/jp270935] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/31/2016] [Indexed: 01/08/2023] Open
Abstract
TRPA1 is a non-selective cation channel expressed in mammalian peripheral pain receptors, with a major role in chemonociception. TRPA1 has also been implicated in noxious cold and mechanical pain sensation. TRPA1 has an ancient origin and plays important functions in lower organisms, including thermotaxis, mechanotransduction and modulation of lifespan. Here we highlight the role of TRPA1 as a multipurpose sensor of harmful signals, including toxic bacterial products and UV light, and as a sensor of stress and tissue damage. Sensing roles span beyond the peripheral nervous system to include major barrier tissues: gut, skin and lung. Tissue injury, environmental irritants and microbial pathogens are danger signals that can threaten the health of organisms. These signals lead to the coordinated activation of the nociceptive and the innate immune system to provide a homeostatic response trying to re-establish physiological conditions including tissue repair. Activation of TRPA1 participates in protective neuroimmune interactions at multiple levels, sensing ROS and bacterial products and triggering the release of neuropeptides. However, an exaggerated response to danger signals is maladaptive and can lead to the development of chronic inflammatory conditions.
Collapse
Affiliation(s)
- Félix Viana
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Alicante, Spain
| |
Collapse
|
57
|
Xing J, Li J. TRPA1 Function in Skeletal Muscle Sensory Neurons Following Femoral Artery Occlusion. Cell Physiol Biochem 2017; 42:2307-2317. [PMID: 28848196 DOI: 10.1159/000480003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/22/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND/AIMS Transient receptor potential channel A1 (TRPA1) is engaged in amplified autonomic responses evoked by stimulation of muscle afferent nerves in rats with experimental peripheral arterial disease. The purposes of this study were to characterize current responses induced by activation of TRPA1 in dorsal root ganglion (DRG) neurons of control limbs and limbs with femoral artery occlusion. METHODS DRG neurons from rats were labeled by injecting the fluorescence tracer DiI into the hindlimb muscles and whole-cell patch clamp experiments were performed to determine TRPA1 currents. RESULTS Data show that AITC (a TRPA1 agonist) from the concentrations of 50 µM to 200 µM produces a dose-dependent increase of amplitudes of inward current responses. Notably, the peak current amplitude induced by AITC is significantly larger in DRG neurons of ligated limbs than that in control limbs. AITC-induced current responses are observed in small and medium size DRG neurons, and there is no difference in size distribution of DRG neurons between control limbs and ligated limbs. However, femoral occlusion increases the percentage of the AITC-sensitive DRG neurons as compared to control. AITC-induced currents in DRG neurons are significantly attenuated by exposure to 10 µM of HC-030031, a potent and selective inhibitor of TRPA1, in both control and femoral occlusion groups. In addition, capsaicin (a TRPV1 agonist) evokes a greater increase in the amplitude of AITC-currents in DRG neurons of ligated limbs than that in control limbs. CONCLUSIONS A greater current response with activation of TRPA1 is developed in muscle afferent nerves when hindlimb arterial blood supply is deficient under ischemic conditions; and TRPV1 is partly responsible for augmented TRPA1 responses induced by arterial occlusion.
Collapse
Affiliation(s)
- Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, China.,Pennsylvania State Heart & Vascular Institute, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| | - Jianhua Li
- Pennsylvania State Heart & Vascular Institute, The Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
58
|
Singh U, Bernstein JA, Lorentz H, Sadoway T, Nelson V, Patel P, Salapatek AM. A Pilot Study Investigating Clinical Responses and Biological Pathways of Azelastine/Fluticasone in Nonallergic Vasomotor Rhinitis before and after Cold Dry Air Provocation. Int Arch Allergy Immunol 2017; 173:153-164. [DOI: 10.1159/000478698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/13/2017] [Indexed: 12/12/2022] Open
|
59
|
Semaphorin 4C Plexin-B2 signaling in peripheral sensory neurons is pronociceptive in a model of inflammatory pain. Nat Commun 2017; 8:176. [PMID: 28765520 PMCID: PMC5539317 DOI: 10.1038/s41467-017-00341-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 06/23/2017] [Indexed: 01/08/2023] Open
Abstract
Semaphorins and their transmembrane receptors, Plexins, are key regulators of axon guidance and development of neuronal connectivity. B-type Plexins respond to Class IV semaphorins and mediate a variety of developmental functions. Here we report that the expression of Plexin-B2 and its high-affinity ligand, Sema4C, persists in peripheral sensory neurons in adult life and is markedly increased in states of persistent pain in mice. Genetic deletion of Sema4C as well as adult-onset loss of Plexin-B2 leads to impairment of the development and duration of inflammatory hypersensitivity. Remarkably, unlike the neurodevelopmental functions of Plexin-B2 that solely rely on Ras signaling, we obtained genetic and pharmacological evidence for a requirement of RhoA-ROCK-dependent mechanisms as well as TRPA1 sensitization in pronociceptive functions of Sema4C-Plexin-B2 signaling in adult life. These results suggest important roles for Plexin-B2 signaling in sensory function that may be of therapeutic relevance in pathological pain. Semaphorins and their receptors are involved in neurodevelopment, but their functions in the adult nervous system are not fully understood. This study finds that semaphorin 4C and its receptor Plexin B are expressed in sensory neurons and are pronociceptive in a mouse model of inflammatory pain.
Collapse
|
60
|
Balemans D, Boeckxstaens GE, Talavera K, Wouters MM. Transient receptor potential ion channel function in sensory transduction and cellular signaling cascades underlying visceral hypersensitivity. Am J Physiol Gastrointest Liver Physiol 2017; 312:G635-G648. [PMID: 28385695 DOI: 10.1152/ajpgi.00401.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/21/2017] [Accepted: 04/01/2017] [Indexed: 01/31/2023]
Abstract
Visceral hypersensitivity is an important mechanism underlying increased abdominal pain perception in functional gastrointestinal disorders including functional dyspepsia, irritable bowel syndrome, and inflammatory bowel disease in remission. Although the exact pathophysiological mechanisms are poorly understood, recent studies described upregulation and altered functions of nociceptors and their signaling pathways in aberrant visceral nociception, in particular the transient receptor potential (TRP) channel family. A variety of TRP channels are present in the gastrointestinal tract (TRPV1, TRPV3, TRPV4, TRPA1, TRPM2, TRPM5, and TRPM8), and modulation of their function by increased activation or sensitization (decreased activation threshold) or altered expression in visceral afferents have been reported in visceral hypersensitivity. TRP channels directly detect or transduce osmotic, mechanical, thermal, and chemosensory stimuli. In addition, pro-inflammatory mediators released in tissue damage or inflammation can activate receptors of the G protein-coupled receptor superfamily leading to TRP channel sensitization and activation, which amplify pain and neurogenic inflammation. In this review, we highlight the present knowledge on the functional roles of neuronal TRP channels in visceral hypersensitivity and discuss the signaling pathways that underlie TRP channel modulation. We propose that a better understanding of TRP channels and their modulators may facilitate the development of more selective and effective therapies to treat visceral hypersensitivity.
Collapse
Affiliation(s)
- Dafne Balemans
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Guy E Boeckxstaens
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Karel Talavera
- Laboratory of Ion Channel Research and TRP Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven Belgium
| | - Mira M Wouters
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| |
Collapse
|
61
|
Abstract
Calcium signals control a plethora of essential cellular functions ranging from secretion and contraction to gene expression and sensory signaling cascades. An essential part of intracellular calcium signals originates from the transmembrane flux of calcium ions, which is mainly mediated through different calcium-permeable cation channels with variable calcium selectivity. Opening of individual calcium permeable channels induces a local cytosolic calcium rise that can be highly restricted in time and space. Here, we provide a short overview of the current knowledge about calcium permeation and localized calcium signals in transient receptor potential (TRP) channels. We also present a brief survey of some fundamental theoretical aspects of the local calcium signals generated upon opening of single calcium-permeable channels, and compare theoretical predictions with published experimental data on TRP channel-mediated local calcium signals.
Collapse
|
62
|
A-Kinase Anchoring Protein 79/150 Scaffolds Transient Receptor Potential A 1 Phosphorylation and Sensitization by Metabotropic Glutamate Receptor Activation. Sci Rep 2017; 7:1842. [PMID: 28500286 PMCID: PMC5431798 DOI: 10.1038/s41598-017-01999-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/05/2017] [Indexed: 01/20/2023] Open
Abstract
Mechanical pain serves as a base clinical symptom for many of the world’s most debilitating syndromes. Ion channels expressed by peripheral sensory neurons largely contribute to mechanical hypersensitivity. Transient Receptor Potential A 1 (TRPA1) is a ligand-gated ion channel that contributes to inflammatory mechanical hypersensitivity, yet little is known as to the post-translational mechanism behind its somatosensitization. Here, we utilize biochemical, electrophysiological, and behavioral measures to demonstrate that metabotropic glutamate receptor-induced sensitization of TRPA1 nociceptors stimulates targeted modification of the receptor. Type 1 mGluR5 activation increases TRPA1 receptor agonist sensitivity in an AKA-dependent manner. As a scaffolding protein for Protein Kinases A and C (PKA and PKC, respectively), AKAP facilitates phosphorylation and sensitization of TRPA1 in ex vivo sensory neuronal preparations. Furthermore, hyperalgesic priming of mechanical hypersensitivity requires both TRPA1 and AKAP. Collectively, these results identify a novel AKAP-mediated biochemical mechanism that increases TRPA1 sensitivity in peripheral sensory neurons, and likely contributes to persistent mechanical hypersensitivity.
Collapse
|
63
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
64
|
Meents JE, Fischer MJM, McNaughton PA. Sensitization of TRPA1 by Protein Kinase A. PLoS One 2017; 12:e0170097. [PMID: 28076424 PMCID: PMC5226813 DOI: 10.1371/journal.pone.0170097] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/28/2016] [Indexed: 01/08/2023] Open
Abstract
The TRPA1 ion channel is expressed in nociceptive (pain-sensitive) somatosensory neurons and is activated by a wide variety of chemical irritants, such as acrolein in smoke or isothiocyanates in mustard. Here, we investigate the enhancement of TRPA1 function caused by inflammatory mediators, which is thought to be important in lung conditions such as asthma and COPD. Protein kinase A is an important kinase acting downstream of inflammatory mediators to cause sensitization of TRPA1. By using site-directed mutagenesis, patch-clamp electrophysiology and calcium imaging we identify four amino acid residues, S86, S317, S428, and S972, as the principal targets of PKA-mediated phosphorylation and sensitization of TRPA1.
Collapse
Affiliation(s)
- Jannis E. Meents
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Institute of Physiology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Michael J. M. Fischer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Institute of Physiology and Pathophysiology, University of Erlangen-Nuremberg, Erlangen, Germany
- Center for Physiology and Pharmacology, Medical University Wien, Wien, Austria
| | - Peter A. McNaughton
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| |
Collapse
|
65
|
Avenali L, Abate Fulas O, Sondermann J, Narayanan P, Gomez-Varela D, Schmidt M. Nocistatin sensitizes TRPA1 channels in peripheral sensory neurons. Channels (Austin) 2017; 11:11-19. [PMID: 27362459 DOI: 10.1080/19336950.2016.1207025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The ability of sensory neurons to detect potentially harmful stimuli relies on specialized molecular signal detectors such as transient receptor potential (TRP) A1 ion channels. TRPA1 is critically implicated in vertebrate nociception and different pain states. Furthermore, TRPA1 channels are subject to extensive modulation and regulation - processes which consequently affect nociceptive signaling. Here we show that the neuropeptide Nocistatin sensitizes TRPA1-dependent calcium influx upon application of the TRPA1 agonist mustard oil (MO) in cultured sensory neurons of dorsal root ganglia (DRG). Interestingly, TRPV1-mediated cellular calcium responses are unaffected by Nocistatin. Furthermore, Nocistatin-induced TRPA1-sensitization is likely independent of the Nocistatin binding partner 4-Nitrophenylphosphatase domain and non-neuronal SNAP25-like protein homolog 1 (NIPSNAP1) as assessed by siRNA-mediated knockdown in DRG cultures. In conclusion, we uncovered the sensitization of TRPA1 by Nocistatin, which may represent a novel mechanism how Nocistatin can modulate pain.
Collapse
Affiliation(s)
- Luca Avenali
- a Max Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group , Goettingen , Germany
| | - Oli Abate Fulas
- a Max Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group , Goettingen , Germany
| | - Julia Sondermann
- a Max Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group , Goettingen , Germany
| | - Pratibha Narayanan
- a Max Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group , Goettingen , Germany
| | - David Gomez-Varela
- a Max Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group , Goettingen , Germany
| | - Manuela Schmidt
- a Max Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group , Goettingen , Germany
| |
Collapse
|
66
|
Leishman E, Kunkler PE, Manchanda M, Sangani K, Stuart JM, Oxford GS, Hurley JH, Bradshaw HB. Environmental Toxin Acrolein Alters Levels of Endogenous Lipids, Including TRP Agonists: A Potential Mechanism for Headache Driven by TRPA1 Activation. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2017; 1:28-36. [PMID: 29430557 PMCID: PMC5802349 DOI: 10.1016/j.ynpai.2017.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 06/08/2023]
Abstract
Exposure to airborne toxins can trigger headaches, but the mechanisms are not well understood. Some environmental toxins, such as acrolein, activate transient receptor potential ankyrin 1 (TRPA1), a receptor involved in pain sensation that is highly expressed in the trigeminovascular system. It has been shown in rat models that repeated exposure to acrolein induces trigeminovascular sensitization to both TRPA1 and TRP vanilloid 1 (TRPV1) agonists, a phenomenon linked to headache. In this study, we test the hypothesis that the sensitization of trigeminovascular responses in rats after acrolein exposure via inhalation is associated with changes in levels of endogenous lipids, including TRPV1 agonists, in the trigeminal ganglia, trigeminal nucleus, and cerebellum. Lipidomics analysis of 80 lipids was performed on each tissue after acute acrolein, chronic acrolein, or room air control. Both acute and chronic acrolein exposure drove widespread alterations in lipid levels. After chronic acrolein exposure, levels of all 6 N-acyl ethanolamines in the screening library, including the endogenous cannabinoid and TRPV1 agonist, N-arachidonoyl ethanolamine, were elevated in trigeminal tissue and in the cerebellum. This increase in TRPV1 ligands by acrolein exposure may indicate further downstream signaling, in that we also show here that a combination of these TRPV1 endogenous agonists increases the potency of the individual ligands in TRPV1-HEK cells. In addition to these TRPV1 agonists, 3 TRPV3 antagonists, 4 TRPV4 agonists, and 25 orphan lipids were up and down regulated after acrolein exposure. These data support the hypothesis that lipid signaling may represent a mechanism by which repeated exposure to the TRPA1 agonist and environmental toxin, acrolein, drives trigeminovascular sensitization.
Collapse
Affiliation(s)
- Emma Leishman
- Department of Psychological and Brain Sciences, Indiana University, 1101 East 10 Street, Bloomington, IN 47405, USA
| | - Phillip E. Kunkler
- Stark Neurosciences Institute, Indiana University School of Medicine, 320 West 15 Street, Indianapolis, IN 46202, USA
| | - Meera Manchanda
- Department of Psychological and Brain Sciences, Indiana University, 1101 East 10 Street, Bloomington, IN 47405, USA
| | - Kishan Sangani
- Department of Psychological and Brain Sciences, Indiana University, 1101 East 10 Street, Bloomington, IN 47405, USA
| | - Jordyn M. Stuart
- Department of Psychological and Brain Sciences, Indiana University, 1101 East 10 Street, Bloomington, IN 47405, USA
| | - Gerry S. Oxford
- Stark Neurosciences Institute, Indiana University School of Medicine, 320 West 15 Street, Indianapolis, IN 46202, USA
| | - Joyce H. Hurley
- Stark Neurosciences Institute, Indiana University School of Medicine, 320 West 15 Street, Indianapolis, IN 46202, USA
| | - Heather B. Bradshaw
- Department of Psychological and Brain Sciences, Indiana University, 1101 East 10 Street, Bloomington, IN 47405, USA
| |
Collapse
|
67
|
Mickle AD, Shepherd AJ, Mohapatra DP. Nociceptive TRP Channels: Sensory Detectors and Transducers in Multiple Pain Pathologies. Pharmaceuticals (Basel) 2016; 9:ph9040072. [PMID: 27854251 PMCID: PMC5198047 DOI: 10.3390/ph9040072] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 02/07/2023] Open
Abstract
Specialized receptors belonging to the transient receptor potential (TRP) family of ligand-gated ion channels constitute the critical detectors and transducers of pain-causing stimuli. Nociceptive TRP channels are predominantly expressed by distinct subsets of sensory neurons of the peripheral nervous system. Several of these TRP channels are also expressed in neurons of the central nervous system, and in non-neuronal cells that communicate with sensory nerves. Nociceptive TRPs are activated by specific physico-chemical stimuli to provide the excitatory trigger in neurons. In addition, decades of research has identified a large number of immune and neuromodulators as mediators of nociceptive TRP channel activation during injury, inflammatory and other pathological conditions. These findings have led to aggressive targeting of TRP channels for the development of new-generation analgesics. This review summarizes the complex activation and/or modulation of nociceptive TRP channels under pathophysiological conditions, and how these changes underlie acute and chronic pain conditions. Furthermore, development of small-molecule antagonists for several TRP channels as analgesics, and the positive and negative outcomes of these drugs in clinical trials are discussed. Understanding the diverse functional and modulatory properties of nociceptive TRP channels is critical to function-based drug targeting for the development of evidence-based and efficacious new generation analgesics.
Collapse
Affiliation(s)
- Aaron D Mickle
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | - Andrew J Shepherd
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | - Durga P Mohapatra
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
68
|
Shang S, Zhu F, Liu B, Chai Z, Wu Q, Hu M, Wang Y, Huang R, Zhang X, Wu X, Sun L, Wang Y, Wang L, Xu H, Teng S, Liu B, Zheng L, Zhang C, Zhang F, Feng X, Zhu D, Wang C, Liu T, Zhu MX, Zhou Z. Intracellular TRPA1 mediates Ca2+ release from lysosomes in dorsal root ganglion neurons. J Cell Biol 2016; 215:369-381. [PMID: 27799370 PMCID: PMC5100290 DOI: 10.1083/jcb.201603081] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/16/2016] [Accepted: 10/04/2016] [Indexed: 11/22/2022] Open
Abstract
Transient receptor potential A1 (TRPA1) is a nonselective cation channel implicated in thermosensation and inflammatory pain. In this study, we show that TRPA1 (activated by allyl isothiocyanate, acrolein, and 4-hydroxynonenal) elevates the intracellular Ca2+ concentration ([Ca2+]i) in dorsal root ganglion (DRG) neurons in the presence and absence of extracellular Ca2+ Pharmacological and immunocytochemical analyses revealed the presence of TRPA1 channels both on the plasma membrane and in endolysosomes. Confocal line-scan imaging demonstrated Ca2+ signals elicited from individual endolysosomes ("lysosome Ca2+ sparks") by TRPA1 activation. In physiological solutions, the TRPA1-mediated endolysosomal Ca2+ release contributed to ∼40% of the overall [Ca2+]i rise and directly triggered vesicle exocytosis and calcitonin gene-related peptide release, which greatly enhanced the excitability of DRG neurons. Thus, in addition to working via Ca2+ influx, TRPA1 channels trigger vesicle release in sensory neurons by releasing Ca2+ from lysosome-like organelles.
Collapse
Affiliation(s)
- Shujiang Shang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.,Laboratory Animal Center, Peking University, Beijing 100871, China.,School of Life Science, Peking University, Beijing 100871, China
| | - Feipeng Zhu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Bin Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Zuying Chai
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Qihui Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Meiqin Hu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Yuan Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Rong Huang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Xiaoyu Zhang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Xi Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Lei Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Yeshi Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Li Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Huadong Xu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Sasa Teng
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Bing Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Lianghong Zheng
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Chen Zhang
- School of Life Science, Peking University, Beijing 100871, China
| | - Fukang Zhang
- Institute for Biomedical Science of Pain, Capital Medical University, Beijing 100069, China
| | - Xinghua Feng
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Desheng Zhu
- Laboratory Animal Center, Peking University, Beijing 100871, China
| | - Changhe Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Tao Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| |
Collapse
|
69
|
Rixecker T, Mathar I, Medert R, Mannebach S, Pfeifer A, Lipp P, Tsvilovskyy V, Freichel M. TRPM4-mediated control of FcεRI-evoked Ca(2+) elevation comprises enhanced plasmalemmal trafficking of TRPM4 channels in connective tissue type mast cells. Sci Rep 2016; 6:32981. [PMID: 27624684 PMCID: PMC5021962 DOI: 10.1038/srep32981] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 08/17/2016] [Indexed: 11/09/2022] Open
Abstract
TRPM4 proteins form Ca2+-activated non selective cation (CAN) channels that affect transmembrane Ca2+-influx by determining the membrane potential. Tight control of the intracellular Ca2+ concentration is essential for mast cell responses. In this study, we analyzed the expression of TRPM4 in peritoneal mast cells (PCMC) as a model for connective tissue type mast cells with respect to FcεRI-evoked calcium changes and the subcellular localization of fluorescently labeled TRPM4 using two viral transduction systems before and following antigen stimulation. Our results show that TRPM4 is expressed in PCMCs, is an essential constituent of the endogenous CAN channels in PCMCs and regulates antigen-evoked increases in intracellular calcium that are significantly enhanced in TRPM4-deficient PCMCs. Compared to PCMCs analyzed before antigen stimulation, the cells depict a substantially increased localization of TRPM4 proteins towards the plasma membrane after FcεRI stimulation. Thus, TRPM4 functions as a limiting factor for antigen evoked calcium rise in connective tissue type mast cells and concurrent translocation of TRPM4 into the plasma membrane is part of this mechanism.
Collapse
Affiliation(s)
- Torben Rixecker
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Ilka Mathar
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Rebekka Medert
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Stefanie Mannebach
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Peter Lipp
- Institut für Molekulare Zellbiologie Universität des Saarlandes, 66421 Homburg, Germany
| | - Volodymyr Tsvilovskyy
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
70
|
Govea RM, Zhou S, Carlton SM. Group III mGluR8 negatively modulates TRPA1. Neuroscience 2016; 334:134-147. [PMID: 27497709 DOI: 10.1016/j.neuroscience.2016.07.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 01/01/2023]
Abstract
Several lines of evidence indicate group III metabotropic glutamate receptors (mGluRs) have systemic anti-hyperalgesic effects. We hypothesized this could occur through modulation of TRP channels on nociceptors. This study used a multifaceted approach to examine the interaction between group III mGluRs (mGluR8) and transient receptor potential ankyrin 1 (TRPA1) on cutaneous nociceptors in rats. Ca2+ imaging studies demonstrated co-localization and functional coupling of TRPA1 and mGluR8, since 1μM (S)-3,4-dicarboxyphenylglycine (DCPG) (mGluR8 agonist) significantly reduced Ca2+ mobilization produced by 30μM mustard oil (MO), a TRPA1 agonist. Behavioral studies demonstrated that 10mM MO produced mechanical hypersensitivity when topically applied to the hind paw, significantly decreasing paw withdrawal threshold (PWT) from 15g to 6g. However, administration of 30μM DCPG prior to 10mM MO reversed this hypersensitivity such that PWT was not significantly different from baseline. At the single-fiber level, compared to vehicle, 30μM MO significantly increased nociceptor activity and decreased mechanical threshold. However, 30μM DCPG reversed both of these MO-induced effects. Furthermore, DCPG significantly reduced the number of MO-induced mechanically sensitive fibers. Inhibition of protein kinase A (PKA) using Rp-cyclic 3',5'-hydrogen phosphorothioate adenosine triethylammonium salt (RpCAMPS) (PKA inhibitor, 1 and 10μM) significantly reduced MO-induced Ca2+ mobilization. Taken together, these results show that group III mGluRs negatively modulate TRPA1 activity on cutaneous nociceptors. Furthermore, it is likely that this modulation occurs intracellularly at the level of the cAMP/PKA pathway. This study demonstrates that group III agonists may be effective in the treatment of mechanical hypersensitivity which can develop as a result of inflammation, nerve injury, chemotherapy and other disease states.
Collapse
Affiliation(s)
- R M Govea
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1069, United States
| | - S Zhou
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1069, United States
| | - S M Carlton
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1069, United States.
| |
Collapse
|
71
|
Meents JE, Fischer MJM, McNaughton PA. Agonist-induced sensitisation of the irritant receptor ion channel TRPA1. J Physiol 2016; 594:6643-6660. [PMID: 27307078 DOI: 10.1113/jp272237] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/08/2016] [Indexed: 12/23/2022] Open
Abstract
KEY POINTS The transient receptor potential ankyrin 1 (TRPA1) ion channel is expressed in nociceptive neurons and its activation causes ongoing pain and inflammation; TRPA1 is thought to play an important role in inflammation in the airways. TRPA1 is sensitised by repeated stimulation with chemical agonists in a calcium-free environment and this sensitisation is very long lasting following agonist removal. We show that agonist-induced sensitisation is independent of the agonist's binding site and is also independent of ion channel trafficking or of other typical signalling pathways. We find that sensitisation is intrinsic to the TRPA1 protein and is accompanied by a slowly developing shift in the voltage dependence of TRPA1 towards more negative membrane potentials. Agonist-induced sensitisation may provide an explanation for sensitisation following long-term exposure to harmful irritants and pollutants, particularly in the airways. ABSTRACT The TRPA1 ion channel is expressed in nociceptive (pain-sensitive) neurons and responds to a wide variety of chemical irritants, such as acrolein in smoke or isothiocyanates in mustard. Here we show that in the absence of extracellular calcium the current passing through TRPA1 gradually increases (sensitises) during prolonged application of agonists. Activation by an agonist is essential, because activation of TRPA1 by membrane depolarisation did not cause sensitisation. Sensitisation is independent of the site of action of the agonist, because covalent and non-covalent agonists were equally effective, and is long lasting following agonist removal. Mutating N-terminal cysteines, the target of covalent agonists, did not affect sensitisation by the non-covalent agonist carvacrol, which activates by binding to a different site. Sensitisation is unaffected by agents blocking ion channel trafficking or by block of signalling pathways involving ATP, protein kinase A or the formation of lipid rafts, and does not require ion flux through the channel. Examination of the voltage dependence of TRPA1 activation shows that sensitisation is accompanied by a slowly developing shift in the voltage dependence of TRPA1 towards more negative membrane potentials, and is therefore intrinsic to the TRPA1 channel. Sensitisation may play a role in exacerbating the pain caused by prolonged activation of TRPA1.
Collapse
Affiliation(s)
- Jannis E Meents
- Department of Pharmacology, University of Cambridge, CB2 1PD, UK.,Institute of Physiology, Uniklinik RWTH Aachen, 52074, Germany
| | - Michael J M Fischer
- Department of Pharmacology, University of Cambridge, CB2 1PD, UK.,Institute of Physiology and Pathophysiology, University of Erlangen-Nuremberg, 91054, Germany
| | - Peter A McNaughton
- Department of Pharmacology, University of Cambridge, CB2 1PD, UK.,Wolfson Centre for Age-Related Diseases, King's College London, SE1 1UL, UK
| |
Collapse
|
72
|
Janssens A, Gees M, Toth BI, Ghosh D, Mulier M, Vennekens R, Vriens J, Talavera K, Voets T. Definition of two agonist types at the mammalian cold-activated channel TRPM8. eLife 2016; 5. [PMID: 27449282 PMCID: PMC4985286 DOI: 10.7554/elife.17240] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/22/2016] [Indexed: 11/13/2022] Open
Abstract
Various TRP channels act as polymodal sensors of thermal and chemical stimuli, but the mechanisms whereby chemical ligands impact on TRP channel gating are poorly understood. Here we show that AITC (allyl isothiocyanate; mustard oil) and menthol represent two distinct types of ligands at the mammalian cold sensor TRPM8. Kinetic analysis of channel gating revealed that AITC acts by destabilizing the closed channel, whereas menthol stabilizes the open channel, relative to the transition state. Based on these differences, we classify agonists as either type I (menthol-like) or type II (AITC-like), and provide a kinetic model that faithfully reproduces their differential effects. We further demonstrate that type I and type II agonists have a distinct impact on TRPM8 currents and TRPM8-mediated calcium signals in excitable cells. These findings provide a theoretical framework for understanding the differential actions of TRP channel ligands, with important ramifications for TRP channel structure-function analysis and pharmacology. DOI:http://dx.doi.org/10.7554/eLife.17240.001 Sensory neurons in our skin detect cues from the environment – such as temperature and touch – and pass the information onto other cells in the nervous system. A protein called TRPM8 in sensory neurons is responsible for our ability to detect cool temperatures. TRPM8 sits in the membrane that surrounds the cell and forms a channel that can allow sodium and calcium ions to enter the cell. Cold temperatures activate TRPM8, which opens the channel and triggers electrical activity in the sensory neurons. Chemicals that cause a cold sensation – such as menthol, the refreshing substance found in mint plants – can also open the TRPM8 channel. Janssens, Gees, Toth et al. investigated how menthol, and another natural compound called mustard oil, influence the opening of TRPM8. The experiments show that menthol and mustard oil both stimulate sensory neurons by opening the TRPM8 ion channel, but using different mechanisms. Mustard oil forces the channel to open faster than it normally would, whereas menthol prevents the channel from closing. Further experiments show that these mechanisms explain why some compounds stimulate sensory neurons more strongly than others. The findings of Janssens, Gees, Toth et al. will help to understand how chemicals act on this class of ion channels, and how this affects the roles of the ion channels in cells. Altering the activity of TRPM8 and related ion channels may help to reduce pain in humans so a future challenge is to use these new insights to develop drugs that target these channels more efficiently. DOI:http://dx.doi.org/10.7554/eLife.17240.002
Collapse
Affiliation(s)
- Annelies Janssens
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Maarten Gees
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Balazs Istvan Toth
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Debapriya Ghosh
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Marie Mulier
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Rudi Vennekens
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Joris Vriens
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium.,Laboratory of Experimental Gynaecology, University of Leuven, Leuven, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research and TRP channel Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| |
Collapse
|
73
|
Kim S, Barry DM, Liu XY, Yin S, Munanairi A, Meng QT, Cheng W, Mo P, Wan L, Liu SB, Ratnayake K, Zhao ZQ, Gautam N, Zheng J, Karunarathne WKA, Chen ZF. Facilitation of TRPV4 by TRPV1 is required for itch transmission in some sensory neuron populations. Sci Signal 2016; 9:ra71. [PMID: 27436359 DOI: 10.1126/scisignal.aaf1047] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The transient receptor potential channels (TRPs) respond to chemical irritants and temperature. TRPV1 responds to the itch-inducing endogenous signal histamine, and TRPA1 responds to the itch-inducing chemical chloroquine. We showed that, in sensory neurons, TRPV4 is important for both chloroquine- and histamine-induced itch and that TRPV1 has a role in chloroquine-induced itch. Chloroquine-induced scratching was reduced in mice in which TRPV1 was knocked down or pharmacologically inhibited. Both TRPV4 and TRPV1 were present in some sensory neurons. Pharmacological blockade of either TRPV4 or TRPV1 significantly attenuated the Ca(2+) response of sensory neurons exposed to histamine or chloroquine. Knockout of Trpv1 impaired Ca(2+) responses and reduced scratching behavior evoked by a TRPV4 agonist, whereas knockout of Trpv4 did not alter TRPV1-mediated capsaicin responses. Electrophysiological analysis of human embryonic kidney (HEK) 293 cells coexpressing TRPV4 and TRPV1 revealed that the presence of both channels enhanced the activation kinetics of TRPV4 but not of TRPV1. Biochemical and biophysical studies suggested a close proximity between TRPV4 and TRPV1 in dorsal root ganglion neurons and in cultured cells. Thus, our studies identified TRPV4 as a channel that contributes to both histamine- and chloroquine-induced itch and indicated that the function of TRPV4 in itch signaling involves TRPV1-mediated facilitation. TRP facilitation through the formation of heteromeric complexes could be a prevalent mechanism by which the vast array of somatosensory information is encoded in sensory neurons.
Collapse
Affiliation(s)
- Seungil Kim
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Devin M Barry
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xian-Yu Liu
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shijin Yin
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Admire Munanairi
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Qing-Tao Meng
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Cheng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, P.R. China
| | - Ping Mo
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Nanhai Hospital of Southern Medical University, Foshan 528000, P.R. China
| | - Li Wan
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shen-Bin Liu
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kasun Ratnayake
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Zhong-Qiu Zhao
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Narasimhan Gautam
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Zheng
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, CA 95616, USA
| | | | - Zhou-Feng Chen
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA. Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
74
|
N-terminal tetrapeptide T/SPLH motifs contribute to multimodal activation of human TRPA1 channel. Sci Rep 2016; 6:28700. [PMID: 27345869 PMCID: PMC4922051 DOI: 10.1038/srep28700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/08/2016] [Indexed: 01/05/2023] Open
Abstract
Human transient receptor potential ankyrin channel 1 (TRPA1) is a polymodal sensor implicated in pain, inflammation and itching. An important locus for TRPA1 regulation is the cytoplasmic N-terminal domain, through which various exogenous electrophilic compounds such as allyl-isothiocyanate from mustard oil or cinnamaldehyde from cinnamon activate primary afferent nociceptors. This major region is comprised of a tandem set of 17 ankyrin repeats (AR1-AR17), five of them contain a strictly conserved T/SPLH tetrapeptide motif, a hallmark of an important and evolutionarily conserved contribution to conformational stability. Here, we characterize the functional consequences of putatively stabilizing and destabilizing mutations in these important structural units and identify AR2, AR6, and AR11-13 to be distinctly involved in the allosteric activation of TRPA1 by chemical irritants, cytoplasmic calcium, and membrane voltage. Considering the potential involvement of the T/SP motifs as putative phosphorylation sites, we also show that proline-directed Ser/Thr kinase CDK5 modulates the activity of TRPA1, and that T673 outside the AR-domain is its only possible target. Our data suggest that the most strictly conserved N-terminal ARs define the energetics of the TRPA1 channel gate and contribute to chemical-, calcium- and voltage-dependence.
Collapse
|
75
|
Rouwette T, Sondermann J, Avenali L, Gomez-Varela D, Schmidt M. Standardized Profiling of The Membrane-Enriched Proteome of Mouse Dorsal Root Ganglia (DRG) Provides Novel Insights Into Chronic Pain. Mol Cell Proteomics 2016; 15:2152-68. [PMID: 27103637 DOI: 10.1074/mcp.m116.058966] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 01/08/2023] Open
Abstract
Chronic pain is a complex disease with limited treatment options. Several profiling efforts have been employed with the aim to dissect its molecular underpinnings. However, generated results are often inconsistent and nonoverlapping, which is largely because of inherent technical constraints. Emerging data-independent acquisition (DIA)-mass spectrometry (MS) has the potential to provide unbiased, reproducible and quantitative proteome maps - a prerequisite for standardization among experiments. Here, we designed a DIA-based proteomics workflow to profile changes in the abundance of dorsal root ganglia (DRG) proteins in two mouse models of chronic pain, inflammatory and neuropathic. We generated a DRG-specific spectral library containing 3067 DRG proteins, which enables their standardized quantification by means of DIA-MS in any laboratory. Using this resource, we profiled 2526 DRG proteins in each biological replicate of both chronic pain models and respective controls with unprecedented reproducibility. We detected numerous differentially regulated proteins, the majority of which exhibited pain model-specificity. Our approach recapitulates known biology and discovers dozens of proteins that have not been characterized in the somatosensory system before. Functional validation experiments and analysis of mouse pain behaviors demonstrate that indeed meaningful protein alterations were discovered. These results illustrate how the application of DIA-MS can open new avenues to achieve the long-awaited standardization in the molecular dissection of pathologies of the somatosensory system. Therefore, our findings provide a valuable framework to qualitatively extend our understanding of chronic pain and somatosensation.
Collapse
Affiliation(s)
- Tom Rouwette
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - Julia Sondermann
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - Luca Avenali
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - David Gomez-Varela
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - Manuela Schmidt
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| |
Collapse
|
76
|
Trevisan G, Benemei S, Materazzi S, De Logu F, De Siena G, Fusi C, Fortes Rossato M, Coppi E, Marone IM, Ferreira J, Geppetti P, Nassini R. TRPA1 mediates trigeminal neuropathic pain in mice downstream of monocytes/macrophages and oxidative stress. Brain 2016; 139:1361-77. [PMID: 26984186 DOI: 10.1093/brain/aww038] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/24/2016] [Indexed: 02/04/2023] Open
Abstract
Despite intense investigation, the mechanisms of the different forms of trigeminal neuropathic pain remain substantially unidentified. The transient receptor potential ankyrin 1 channel (encoded by TRPA1) has been reported to contribute to allodynia or hyperalgesia in some neuropathic pain models, including those produced by sciatic nerve constriction. However, the role of TRPA1 and the processes that cause trigeminal pain-like behaviours from nerve insult are poorly understood. The role of TRPA1, monocytes and macrophages, and oxidative stress in pain-like behaviour evoked by the constriction of the infraorbital nerve in mice were explored. C57BL/6 and wild-type (Trpa1(+/+)) mice that underwent constriction of the infraorbital nerve exhibited prolonged (20 days) non-evoked nociceptive behaviour and mechanical, cold and chemical hypersensitivity in comparison to sham-operated mice (P < 0.05-P < 0.001). Both genetic deletion of Trpa1 (Trpa1(-/-)) and pharmacological blockade (HC-030031 and A-967079) abrogated pain-like behaviours (both P < 0.001), which were abated by the antioxidant, α-lipoic acid, and the nicotinamide adenine dinucleotide phosphate oxidase inhibitor, apocynin (both P < 0.001). Nociception and hypersensitivity evoked by constriction of the infraorbital nerve was associated with intra- and perineural monocytic and macrophagic invasion and increased levels of oxidative stress by-products (hydrogen peroxide and 4-hydroxynonenal). Attenuation of monocyte/macrophage increase by systemic treatment with an antibody against the monocyte chemoattractant chemokine (C-C motif) ligand 2 (CCL2) or the macrophage-depleting agent, clodronate (both P < 0.05), was associated with reduced hydrogen peroxide and 4-hydroxynonenal perineural levels and pain-like behaviours (all P < 0.01), which were abated by perineural administration of HC-030031, α-lipoic acid or the anti-CCL2 antibody (all P < 0.001). The present findings propose that, in the constriction of the infraorbital nerve model of trigeminal neuropathic pain, pain-like behaviours are entirely mediated by the TRPA1 channel, targeted by increased oxidative stress by-products released from monocytes and macrophages clumping at the site of nerve injury.
Collapse
Affiliation(s)
- Gabriela Trevisan
- Laboratory of Cellular and Molecular Biology, Graduate Program in Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma 88806-000, SC, Brazil
| | - Silvia Benemei
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Serena Materazzi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Gaetano De Siena
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Camilla Fusi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Mateus Fortes Rossato
- Department of Pharmacology, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, SC, Brazil
| | - Elisabetta Coppi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Ilaria Maddalena Marone
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Juliano Ferreira
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy Department of Pharmacology, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, SC, Brazil
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| |
Collapse
|
77
|
Zhang X, Strassman AM, Novack V, Brin MF, Burstein R. Extracranial injections of botulinum neurotoxin type A inhibit intracranial meningeal nociceptors' responses to stimulation of TRPV1 and TRPA1 channels: Are we getting closer to solving this puzzle? Cephalalgia 2016; 36:875-86. [PMID: 26984967 PMCID: PMC4959034 DOI: 10.1177/0333102416636843] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/09/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Administration of onabotulinumtoxinA (BoNT-A) to peripheral tissues outside the calvaria reduces the number of days chronic migraine patients experience headache. Because the headache phase of a migraine attack, especially those preceded by aura, is thought to involve activation of meningeal nociceptors by endogenous stimuli such as changes in intracranial pressure (i.e. mechanical) or chemical irritants that appear in the meninges as a result of a yet-to-be-discovered sequence of molecular/cellular events triggered by the aura, we sought to determine whether extracranial injections of BoNT-A alter the chemosensitivity of meningeal nociceptors to stimulation of their intracranial receptive fields. MATERIAL AND METHODS Using electrophysiological techniques, we identified 161 C- and 135 Aδ-meningeal nociceptors in rats and determined their mechanical response threshold and responsiveness to chemical stimulation of their dural receptive fields with TRPV1 and TRPA1 agonists seven days after BoNT-A administration to different extracranial sites. Two paradigms were compared: distribution of 5 U BoNT-A to the lambdoid and sagittal sutures alone, and 1.25 U to the sutures and 3.75 U to the temporalis and trapezius muscles. RESULTS Seven days after it was administered to tissues outside the calvaria, BoNT-A inhibited responses of C-type meningeal nociceptors to stimulation of their intracranial dural receptive fields with the TRPV1 agonist capsaicin and the TRPA1 agonist mustard oil. BoNT-A inhibition of responses to capsaicin was more effective when the entire dose was injected along the suture lines than when it was injected into muscles and sutures. As in our previous study, BoNT-A had no effect on non-noxious mechanosensitivity of C-fibers or on responsiveness of Aδ-fibers to mechanical and chemical stimulation. DISCUSSION This study demonstrates that extracranial administration of BoNT-A suppresses meningeal nociceptors' responses to stimulation of their intracranial dural receptive fields with capsaicin and mustard oil. The findings suggest that surface expression of TRPV1 and TRPA1 channels in dural nerve endings of meningeal nociceptors is reduced seven days after extracranial administration of BoNT-A. In the context of chronic migraine, reduced sensitivity to molecules that activate meningeal nociceptors through the TRPV1 and TRPA1 channels can be important for BoNT-A's ability to act as a prophylactic.
Collapse
Affiliation(s)
- XiChun Zhang
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, USA Harvard Medical School, USA
| | - Andrew M Strassman
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, USA Harvard Medical School, USA
| | - Victor Novack
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, USA Clinical Research Center, Soroka University Medical Center, Israel Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel
| | | | - Rami Burstein
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, USA Harvard Medical School, USA
| |
Collapse
|
78
|
TNFα induces co-trafficking of TRPV1/TRPA1 in VAMP1-containing vesicles to the plasmalemma via Munc18-1/syntaxin1/SNAP-25 mediated fusion. Sci Rep 2016; 6:21226. [PMID: 26888187 PMCID: PMC4758037 DOI: 10.1038/srep21226] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/16/2015] [Indexed: 11/08/2022] Open
Abstract
Transient receptor potential (TRP) A1 and V1 channels relay sensory signals, yet little is known about their transport to the plasmalemma during inflammation. Herein, TRPA1 and TRPV1 were found on vesicles containing calcitonin gene-related peptide (CGRP), accumulated at sites of exo- and endo-cytosis, and co-localised on fibres and cell bodies of cultured sensory neurons expressing both. A proinflammatory cytokine, TNFα, elevated their surface content, and both resided in close proximity, indicating co-trafficking. Syntaxin 1-interacting protein, Munc18-1, proved necessary for the response to TNFα, and for TRPV1-triggered CGRP release. TNFα-induced surface trafficking of TRPV1 and TRPA1 required a synaptic vesicle membrane protein VAMP1 (but not 2/3), which is essential for CGRP exocytosis from large dense-core vesicles. Inactivation of two proteins on the presynaptic plasma membrane, syntaxin-1 or SNAP-25, by botulinum neurotoxin (BoNT)/C1 or /A inhibited the TNFα-elevated delivery. Accordingly, enhancement by TNFα of Ca(2+) influx through the upregulated surface-expressed TRPV1 and TRPA1 channels was abolished by BoNT/A. Thus, in addition, the neurotoxins' known inhibition of the release of pain transmitters, their therapeutic potential is augmented by lowering the exocytotic delivery of transducing channels and the resultant hyper-sensitisation in inflammation.
Collapse
|
79
|
Ohya S, Kito H, Hatano N, Muraki K. Recent advances in therapeutic strategies that focus on the regulation of ion channel expression. Pharmacol Ther 2016; 160:11-43. [PMID: 26896566 DOI: 10.1016/j.pharmthera.2016.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A number of different ion channel types are involved in cell signaling networks, and homeostatic regulatory mechanisms contribute to the control of ion channel expression. Profiling of global gene expression using microarray technology has recently provided novel insights into the molecular mechanisms underlying the homeostatic and pathological control of ion channel expression. It has demonstrated that the dysregulation of ion channel expression is associated with the pathogenesis of neural, cardiovascular, and immune diseases as well as cancers. In addition to the transcriptional, translational, and post-translational regulation of ion channels, potentially important evidence on the mechanisms controlling ion channel expression has recently been accumulated. The regulation of alternative pre-mRNA splicing is therefore a novel therapeutic strategy for the treatment of dominant-negative splicing disorders. Epigenetic modification plays a key role in various pathological conditions through the regulation of pluripotency genes. Inhibitors of pre-mRNA splicing and histone deacetyalase/methyltransferase have potential as potent therapeutic drugs for cancers and autoimmune and inflammatory diseases. Moreover, membrane-anchoring proteins, lysosomal and proteasomal degradation-related molecules, auxiliary subunits, and pharmacological agents alter the protein folding, membrane trafficking, and post-translational modifications of ion channels, and are linked to expression-defect channelopathies. In this review, we focused on recent insights into the transcriptional, spliceosomal, epigenetic, and proteasomal regulation of ion channel expression: Ca(2+) channels (TRPC/TRPV/TRPM/TRPA/Orai), K(+) channels (voltage-gated, KV/Ca(2+)-activated, KCa/two-pore domain, K2P/inward-rectifier, Kir), and Ca(2+)-activated Cl(-) channels (TMEM16A/TMEM16B). Furthermore, this review highlights expression of these ion channels in expression-defect channelopathies.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan.
| |
Collapse
|
80
|
Abstract
The detection of temperature is one of the most fundamental sensory functions across all species, and is critical for animal survival. Animals have thus evolved a diversity of thermosensory mechanisms allowing them to sense and respond to temperature changes (thermoreception). A key process underlying thermoreception is the translation of thermal energy into electrical signals, a process mediated by thermal sensors (thermoreceptors) that are sensitive to a specific range of temperatures. In disease conditions, the temperature sensitivity of thermoreceptors is altered, leading to abnormal temperature sensation such as heat hyperalgesia. Therefore, the identification of thermal sensors and understanding their functions and regulation hold great potential for developing novel therapeutics against many medical conditions such as pain.
Collapse
Affiliation(s)
- Xuming Zhang
- a Rowett Institute of Nutrition and Health & Institute of Medical Sciences ; University of Aberdeen , Foresterhill , Aberdeen , UK
| |
Collapse
|
81
|
Ghosh D, Pinto S, Danglot L, Vandewauw I, Segal A, Van Ranst N, Benoit M, Janssens A, Vennekens R, Vanden Berghe P, Galli T, Vriens J, Voets T. VAMP7 regulates constitutive membrane incorporation of the cold-activated channel TRPM8. Nat Commun 2016; 7:10489. [PMID: 26843440 PMCID: PMC4742910 DOI: 10.1038/ncomms10489] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
The cation channel TRPM8 plays a central role in the somatosensory system, as a key sensor of innocuously cold temperatures and cooling agents. Although increased functional expression of TRPM8 has been implicated in various forms of pathological cold hypersensitivity, little is known about the cellular and molecular mechanisms that determine TRPM8 abundance at the plasma membrane. Here we demonstrate constitutive transport of TRPM8 towards the plasma membrane in atypical, non-acidic transport vesicles that contain lysosomal-associated membrane protein 1 (LAMP1), and provide evidence that vesicle-associated membrane protein 7 (VAMP7) mediates fusion of these vesicles with the plasma membrane. In line herewith, VAMP7-deficient mice exhibit reduced functional expression of TRPM8 in sensory neurons and concomitant deficits in cold avoidance and icilin-induced cold hypersensitivity. Our results uncover a cellular pathway that controls functional plasma membrane incorporation of a temperature-sensitive TRP channel, and thus regulates thermosensitivity in vivo.
Collapse
Affiliation(s)
- Debapriya Ghosh
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Silvia Pinto
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Lydia Danglot
- Institut Jacques Monod, CNRS UMR 7592, University of Paris Diderot, F-75013 Paris, France
- INSERM ERL U950, Membrane Traffic in Health & disease Group, F-75013 Paris, France
| | - Ine Vandewauw
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Andrei Segal
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Nele Van Ranst
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Melissa Benoit
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Annelies Janssens
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Rudi Vennekens
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience, Department of Clinical and Experimental Medicine, University of Leuven, B-3000 Leuven, Belgium
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Herestraat 49 box 701, B-3000 Leuven, Belgium
| | - Thierry Galli
- Institut Jacques Monod, CNRS UMR 7592, University of Paris Diderot, F-75013 Paris, France
- INSERM ERL U950, Membrane Traffic in Health & disease Group, F-75013 Paris, France
| | - Joris Vriens
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
- Laboratory of Experimental Gynaecology, Department of Development and Regeneration, University of Leuven, Herestraat 49 box 611, B-3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research and TRP Channel Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Herestraat 49 box 802, B-3000 Leuven, Belgium
| |
Collapse
|
82
|
Calcium Entry Through Thermosensory Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:265-304. [PMID: 27161233 DOI: 10.1007/978-3-319-26974-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ThermoTRPs are unique channels that mediate Na(+) and Ca(2+) currents in response to changes in ambient temperature. In combination with their activation by other physical and chemical stimuli, they are considered key integrators of environmental cues into neuronal excitability. Furthermore, roles of thermoTRPs in non-neuronal tissues are currently emerging such as insulin secretion in pancreatic β-cells, and links to cancer. Calcium permeability through thermoTRPs appears a central hallmark for their physiological and pathological activities. Moreover, it is currently being proposed that beyond working as a second messenger, Ca(2+) can function locally by acting on protein complexes near the membrane. Interestingly, thermoTRPs can enhance and expand the inherent plasticity of signalplexes by conferring them temperature, pH and lipid regulation through Ca(2+) signalling. Thus, unveiling the local role of Ca(2+) fluxes induced by thermoTRPs on the dynamics of membrane-attached signalling complexes as well as their significance in cellular processes, are central issues that will expand the opportunities for therapeutic intervention in disorders involving dysfunction of thermoTRP channels.
Collapse
|
83
|
Hui L, Geiger NH, Bloor-Young D, Churchill GC, Geiger JD, Chen X. Release of calcium from endolysosomes increases calcium influx through N-type calcium channels: Evidence for acidic store-operated calcium entry in neurons. Cell Calcium 2015; 58:617-27. [PMID: 26475051 DOI: 10.1016/j.ceca.2015.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/02/2015] [Accepted: 10/04/2015] [Indexed: 01/22/2023]
Abstract
Neurons possess an elaborate system of endolysosomes. Recently, endolysosomes were found to have readily releasable stores of intracellular calcium; however, relatively little is known about how such 'acidic calcium stores' affect calcium signaling in neurons. Here we demonstrated in primary cultured neurons that calcium released from acidic calcium stores triggered calcium influx across the plasma membrane, a phenomenon we have termed "acidic store-operated calcium entry (aSOCE)". aSOCE was functionally distinct from store-operated calcium release and calcium entry involving endoplasmic reticulum. aSOCE appeared to be governed by N-type calcium channels (NTCCs) because aSOCE was attenuated significantly by selectively blocking NTCCs or by siRNA knockdown of NTCCs. Furthermore, we demonstrated that NTCCs co-immunoprecipitated with the lysosome associated membrane protein 1 (LAMP1), and that aSOCE is accompanied by increased cell-surface expression levels of NTCC and LAMP1 proteins. Moreover, we demonstrated that siRNA knockdown of LAMP1 or Rab27a, both of which are key proteins involved in lysosome exocytosis, attenuated significantly aSOCE. Taken together our data suggest that aSOCE occurs in neurons, that aSOCE plays an important role in regulating the levels and actions of intraneuronal calcium, and that aSOCE is regulated at least in part by exocytotic insertion of N-type calcium channels into plasma membranes through LAMP1-dependent lysosome exocytosis.
Collapse
Affiliation(s)
- Liang Hui
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Nicholas H Geiger
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Duncan Bloor-Young
- Department of Pharmacology, University of Oxford, Mansfield Rd., Oxford OX1 3QT, UK
| | - Grant C Churchill
- Department of Pharmacology, University of Oxford, Mansfield Rd., Oxford OX1 3QT, UK
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58203, USA.
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| |
Collapse
|
84
|
Asgar J, Zhang Y, Saloman JL, Wang S, Chung MK, Ro JY. The role of TRPA1 in muscle pain and mechanical hypersensitivity under inflammatory conditions in rats. Neuroscience 2015; 310:206-15. [PMID: 26393428 DOI: 10.1016/j.neuroscience.2015.09.042] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 09/08/2015] [Accepted: 09/15/2015] [Indexed: 12/30/2022]
Abstract
Transient receptor potential cation channel, subfamily A, member 1 (TRPA1) is expressed in muscle afferents and direct activation of these receptors induces acute mechanical hypersensitivity. However, the functional role of TRPA1 under pathological muscle pain conditions and mechanisms by which TRPA1 mediate muscle pain and hyperalgesia are not clearly understood. Two rodent behavioral models validated to assess craniofacial muscle pain conditions were used to study ATP- and N-Methyl-D-aspartate (NMDA)-induced acute mechanical hypersensitivity and complete Freund's adjuvant (CFA)-induced persistent mechanical hypersensitivity. The rat grimace scale (RGS) was utilized to assess inflammation-induced spontaneous muscle pain. Behavioral pharmacology experiments were performed to assess the effects of AP18, a selective TRPA1 antagonist under these conditions. TRPA1 expression levels in trigeminal ganglia (TG) were examined before and after CFA treatment in the rat masseter muscle. Pre-treatment of the muscle with AP18 dose-dependently blocked the development of acute mechanical hypersensitivity induced by NMDA and α,β-methylene adenosine triphosphate (αβmeATP), a specific agonist for NMDA and P2X3 receptor, respectively. CFA-induced mechanical hypersensitivity and spontaneous muscle pain responses were significantly reversed by post-treatment of the muscle with AP18 when CFA effects were most prominent. CFA-induced myositis was accompanied by significant up-regulation of TRPA1 expression in TG. Our findings showed that TRPA1 in muscle afferents plays an important role in the development of acute mechanical hypersensitivity and in the maintenance of persistent muscle pain and hypersensitivity. Our data suggested that TRPA1 may serve as a downstream target of pro-nociceptive ion channels, such as P2X3 and NMDA receptors in masseter afferents, and that increased TRPA1 expression under inflammatory conditions may contribute to the maintenance of persistent muscle pain and mechanical hyperalgesia. Mechanistic studies elucidating transcriptional or post-translational regulation of TRPA1 expression under pathological pain conditions should provide important basic information to further advance the treatment of craniofacial muscle pain conditions.
Collapse
Affiliation(s)
- J Asgar
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Y Zhang
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - J L Saloman
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - S Wang
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - M-K Chung
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - J Y Ro
- University of Maryland School of Dentistry, Department of Neural and Pain Sciences, 650 W. Baltimore Street, Baltimore, MD 21201, USA; Kyung Hee University, School of Dentistry, Department of Oral Medicine, 1 Hoegi Dong, Dongdaemun Gu, Seoul, Republic of Korea.
| |
Collapse
|
85
|
Xing J, Lu J, Li J. TRPA1 mediates amplified sympathetic responsiveness to activation of metabolically sensitive muscle afferents in rats with femoral artery occlusion. Front Physiol 2015; 6:249. [PMID: 26441669 PMCID: PMC4569976 DOI: 10.3389/fphys.2015.00249] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/21/2015] [Indexed: 11/13/2022] Open
Abstract
Autonomic responses to activation of mechanically and metabolically sensitive muscle afferent nerves during static contraction are augmented in rats with femoral artery occlusion. Moreover, metabolically sensitive transient receptor potential cation channel subfamily A, member 1 (TRPA1) has been reported to contribute to sympathetic nerve activity (SNA) and arterial blood pressure (BP) responses evoked by static muscle contraction. Thus, in the present study, we examined the mechanisms by which afferent nerves' TRPA1 plays a role in regulating amplified sympathetic responsiveness due to a restriction of blood flow directed to the hindlimb muscles. Our data show that 24-72 h of femoral artery occlusion (1) upregulates the protein levels of TRPA1 in dorsal root ganglion (DRG) tissues; (2) selectively increases expression of TRPA1 in DRG neurons supplying metabolically sensitive afferent nerves of C-fiber (group IV); and (3) enhances renal SNA and BP responses to AITC (a TRPA1 agonist) injected into the hindlimb muscles. In addition, our data demonstrate that blocking TRPA1 attenuates SNA and BP responses during muscle contraction to a greater degree in ligated rats than those responses in control rats. In contrast, blocking TRPA1 fails to attenuate SNA and BP responses during passive tendon stretch in both groups. Overall, results of this study indicate that alternations in muscle afferent nerves' TRPA1 likely contribute to enhanced sympathetically mediated autonomic responses via the metabolic component of the muscle reflex under circumstances of chronic muscle ischemia.
Collapse
Affiliation(s)
- Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University Changchun, Jilin, China
| | - Jian Lu
- Pennsylvania State Heart and Vascular Institute, The Pennsylvania State University College of Medicine Hershey, PA, USA
| | - Jianhua Li
- Pennsylvania State Heart and Vascular Institute, The Pennsylvania State University College of Medicine Hershey, PA, USA
| |
Collapse
|
86
|
|
87
|
Lee LY, Hsu CC, Lin YJ, Lin RL, Khosravi M. Interaction between TRPA1 and TRPV1: Synergy on pulmonary sensory nerves. Pulm Pharmacol Ther 2015; 35:87-93. [PMID: 26283426 DOI: 10.1016/j.pupt.2015.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/11/2015] [Indexed: 12/15/2022]
Abstract
Transient receptor potential ankyrin type 1 (TRPA1) and vanilloid type 1 (TRPV1) receptors are co-expressed in vagal pulmonary C-fiber sensory nerves. Because both these ligand-gated non-selective cation channels are sensitive to a number of endogenous inflammatory mediators, it is highly probable that they can be activated simultaneously during airway inflammation. Studies were carried out to investigate whether there is an interaction between these two polymodal transducers upon simultaneous activation, and how it modulates the activity of vagal pulmonary C-fiber sensory nerves. Our studies showed a distinct potentiating effect induced abruptly by simultaneous activations of TRPA1 and TRPV1 by their respective selective agonists, allyl isothiocyanate (AITC) and capsaicin (Cap), at near-threshold concentrations. This synergistic effect was demonstrated in the studies of single-unit recording of vagal bronchopulmonary C-fiber afferents and the reflex responses elicited by activation of these afferents in intact animals, as well as in the isolated nodose and jugular bronchopulmonary sensory neurons. This potentiating effect was absent when either AITC or Cap was replaced by non-TRPA1 and non-TRPV1 chemical activators of these neurons, demonstrating the selectivity of the interaction between these two TRP channels. Furthermore, the synergism was dependent upon the extracellular Ca(2+), and the rapid onset of the action further suggests that the interaction probably occurred locally at the sites of these channels. These findings suggest that the TRPA1-TRPV1 interaction may play an important role in regulating the function and excitability of pulmonary sensory neurons during airway inflammation, but the mechanism underlying this positive interaction is not yet fully understood.
Collapse
Affiliation(s)
- Lu-Yuan Lee
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY 40536-0298, USA.
| | - Chun-Chun Hsu
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY 40536-0298, USA
| | - Yu-Jung Lin
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY 40536-0298, USA
| | - Ruei-Lung Lin
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY 40536-0298, USA
| | - Mehdi Khosravi
- Department of Internal Medicine, University of Kentucky Medical Center, Lexington, KY 40536-0298, USA
| |
Collapse
|
88
|
Morita T, McClain SP, Batia LM, Pellegrino M, Wilson SR, Kienzler MA, Lyman K, Olsen ASB, Wong JF, Stucky CL, Brem RB, Bautista DM. HTR7 Mediates Serotonergic Acute and Chronic Itch. Neuron 2015; 87:124-38. [PMID: 26074006 DOI: 10.1016/j.neuron.2015.05.044] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/31/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022]
Abstract
Chronic itch is a prevalent and debilitating condition for which few effective therapies are available. We harnessed the natural variation across genetically distinct mouse strains to identify transcripts co-regulated with itch behavior. This survey led to the discovery of the serotonin receptor HTR7 as a key mediator of serotonergic itch. Activation of HTR7 promoted opening of the ion channel TRPA1, which in turn triggered itch behaviors. In addition, acute itch triggered by serotonin or a selective serotonin reuptake inhibitor required both HTR7 and TRPA1. Aberrant serotonin signaling has long been linked to a variety of human chronic itch conditions, including atopic dermatitis. In a mouse model of atopic dermatitis, mice lacking HTR7 or TRPA1 displayed reduced scratching and skin lesion severity. These data highlight a role for HTR7 in acute and chronic itch and suggest that HTR7 antagonists may be useful for treating a variety of pathological itch conditions.
Collapse
Affiliation(s)
- Takeshi Morita
- Department of Molecular & Cell Biology, 142 Life Sciences Addition, University of California, Berkeley, Berkeley, CA 94720-3200, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shannan P McClain
- Department of Molecular & Cell Biology, 142 Life Sciences Addition, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Lyn M Batia
- Department of Molecular & Cell Biology, 142 Life Sciences Addition, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Maurizio Pellegrino
- Department of Molecular & Cell Biology, 142 Life Sciences Addition, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Sarah R Wilson
- Department of Molecular & Cell Biology, 142 Life Sciences Addition, University of California, Berkeley, Berkeley, CA 94720-3200, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael A Kienzler
- Neurobiology Course, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Kyle Lyman
- Neurobiology Course, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | | | - Justin F Wong
- Department of Molecular & Cell Biology, 142 Life Sciences Addition, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Cheryl L Stucky
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rachel B Brem
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA; Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Diana M Bautista
- Department of Molecular & Cell Biology, 142 Life Sciences Addition, University of California, Berkeley, Berkeley, CA 94720-3200, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
89
|
Coste B, Murthy SE, Mathur J, Schmidt M, Mechioukhi Y, Delmas P, Patapoutian A. Piezo1 ion channel pore properties are dictated by C-terminal region. Nat Commun 2015; 6:7223. [PMID: 26008989 PMCID: PMC4445471 DOI: 10.1038/ncomms8223] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 04/20/2015] [Indexed: 02/06/2023] Open
Abstract
Piezo1 and Piezo2 encode mechanically activated cation channels that function as mechanotransducers involved in vascular system development and touch sensing, respectively. Structural features of Piezos remain unknown. Mouse Piezo1 is bioinformatically predicted to have 30–40 transmembrane (TM) domains. Here, we find that nine of the putative inter-transmembrane regions are accessible from the extracellular side. We use chimeras between mPiezo1 and dPiezo to show that ion-permeation properties are conferred by C-terminal region. We further identify a glutamate residue within a conserved region adjacent to the last two putative TM domains of the protein, that when mutated, affects unitary conductance and ion selectivity, and modulates pore block. We propose that this amino acid is either in the pore or closely associates with the pore. Our results describe important structural motifs of this channel family and lay the groundwork for a mechanistic understanding of how Piezos are mechanically gated and conduct ions. Piezo ion channels function as mechanotransducers involved in vascular development and touch sensing, but their structural features remain unknown. Here the authors find that the C-terminal region of Piezo protein encompasses the pore and identify a glutamate residue within this region involved in ion conduction properties.
Collapse
Affiliation(s)
- Bertrand Coste
- 1] Aix Marseille Université, CNRS, CRN2M-UMR7286, 13344 Marseille, France [2] Howard Hughes Medical Institute, Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Swetha E Murthy
- Howard Hughes Medical Institute, Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jayanti Mathur
- Genomics Institute of the Novartis Research Foundation, San Diego, California 92121, USA
| | - Manuela Schmidt
- Howard Hughes Medical Institute, Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | - Patrick Delmas
- Aix Marseille Université, CNRS, CRN2M-UMR7286, 13344 Marseille, France
| | - Ardem Patapoutian
- 1] Howard Hughes Medical Institute, Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA [2] Genomics Institute of the Novartis Research Foundation, San Diego, California 92121, USA
| |
Collapse
|
90
|
Abstract
The spatial and temporal distribution of receptors constitutes an important mechanism for controlling the magnitude of cellular responses. Several members of the transient receptor potential (TRP) ion channel family can regulate their function by modulating their expression at the plasma membrane (PM) through rapid vesicular translocation and fusion. The mechanisms underlying this regulation are not completely understood, and the contribution of vesicular trafficking to physiological function is unknown. TRPM8 receptors are expressed in mammalian peripheral sensory neurons and are essential for the detection of cold temperatures. Previously, we showed that TRPM8-containing vesicles are segregated into three main pools, immobile at the PM, simple diffusive and corralled-hopping. Here, we show that channel expression at the PM is modulated by TRPM8 agonists in F11 and HEK293T cells. Our results support a model in which the activation of TRPM8 channels, located at the PM, induces a short-lived recruitment of a TRPM8-containing vesicular pool to the cell surface causing a transitory increase in the number of functional channels, affecting intrinsic properties of cold receptor responses. We further demonstrate the requirement of intact vesicular trafficking to support sustained cold responses in the skin of mice.
Collapse
|
91
|
Kunkler PE, Zhang L, Pellman JJ, Oxford GS, Hurley JH. Sensitization of the trigeminovascular system following environmental irritant exposure. Cephalalgia 2015; 35:1192-201. [PMID: 25724913 DOI: 10.1177/0333102415574845] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Air pollution is linked to increased emergency room visits for headache, and migraine patients frequently cite chemicals or odors as headache triggers, but the association between air pollutants and headache is not well understood. We previously reported that nasal administration of environmental irritants acutely increases meningeal blood flow via a TRPA1-dependent mechanism involving the trigeminovascular system. Here, we examine whether chronic environmental irritant exposure sensitizes the trigeminovascular system. METHODS Male rats were exposed to acrolein, a TRPA1 agonist, or room air by inhalation for four days prior to meningeal blood flow measurements. Some animals were injected daily with a TRPA1 antagonist, AP-18, or vehicle prior to inhalation exposure. Trigeminal ganglia were isolated following blood flow measurements for immunocytochemistry and/or qPCR determination of TRPV1, TRPA1 and CGRP levels. RESULTS Acrolein inhalation exposure potentiated blood flow responses both to TRPA1 and TRPV1 agonists compared to room air. Acrolein exposure did not alter TRPV1 or TRPA1 mRNA levels or TRPV1 or CGRP immunoreactive cell counts in the trigeminal ganglion. Acrolein sensitization of trigeminovascular responses to a TRPA1 agonist was attenuated by pre-treatment with AP-18. INTERPRETATION These results suggest trigeminovascular sensitization as a mechanism for enhanced headache susceptibility after chemical exposure.
Collapse
Affiliation(s)
- Phillip Edward Kunkler
- The Department of Biochemistry and Molecular Biology, Stark Neurosciences Research Institute, Indiana University School of Medicine, USA
| | - LuJuan Zhang
- The Department of Biochemistry and Molecular Biology, Stark Neurosciences Research Institute, Indiana University School of Medicine, USA
| | - Jessica Joan Pellman
- The Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, Indiana University School of Medicine, USA
| | - Gerry Stephen Oxford
- The Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, Indiana University School of Medicine, USA
| | - Joyce Harts Hurley
- The Department of Biochemistry and Molecular Biology, Stark Neurosciences Research Institute, Indiana University School of Medicine, USA
| |
Collapse
|
92
|
|
93
|
Aggarwal S, Kheirkhah A, Cavalcanti BM, Cruzat A, Colon C, Brown E, Borsook D, Prüss H, Hamrah P. Autologous Serum Tears for Treatment of Photoallodynia in Patients with Corneal Neuropathy: Efficacy and Evaluation with In Vivo Confocal Microscopy. Ocul Surf 2015; 13:250-62. [PMID: 26045233 DOI: 10.1016/j.jtos.2015.01.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/21/2015] [Accepted: 01/01/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Patients suffering from corneal neuropathy may present with photoallodynia; i.e., increased light sensitivity, frequently with a normal slit-lamp examination. This study aimed to evaluate the efficacy of autologous serum tears (AST) for treatment of severe photoallodynia in corneal neuropathy and to correlate clinical findings with corneal subbasal nerve alterations by in vivo confocal microscopy (IVCM). METHODS Retrospective case control study with 16 patients with neuropathy-induced severe photoallodynia compared to 16 normal controls. Symptom severity, clinical examination and bilateral corneal IVCM scans were recorded. RESULTS All patients suffered from extreme photoallodynia (8.8±1.1) with no concurrent ocular surface disease. Subbasal nerves were significantly decreased at baseline in patients compared to controls; total nerve length (9208±1264 vs 24714±1056 μm/mm(2); P<.0001) and total nerve number (9.6±1.4 vs 28.6±2.0; P<.0001), respectively. Morphologically, significantly increased reflectivity (2.9±0.2 vs 1.8±0.1; P<.0001), beading (in 93.7%), and neuromas (in 62.5%) were seen. AST (3.6±2.1 months) resulted in significantly decreased symptom severity (1.6±1.7; P=.02). IVCM demonstrated significantly improved nerve parameters (P<.005), total nerve length (15451±1595 μm/mm(2)), number (13.9±2.1), and reflectivity (1.9±0.1). Beading and neuromas were seen in only 56.2% and 7.6% of patients. CONCLUSION Patients with corneal neuropathy-induced photoallodynia show profound alterations in corneal nerves. AST restores nerve topography through nerve regeneration, and this correlated with improvement in patient-reported photoallodynia. The data support the notion that corneal nerve damage results in alterations in afferent trigeminal pathways to produce photoallodynia.
Collapse
Affiliation(s)
- Shruti Aggarwal
- Ocular Surface Imaging Center, Cornea and Refractive Surgery Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Ahmad Kheirkhah
- Ocular Surface Imaging Center, Cornea and Refractive Surgery Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Bernardo M Cavalcanti
- Ocular Surface Imaging Center, Cornea and Refractive Surgery Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Andrea Cruzat
- Ocular Surface Imaging Center, Cornea and Refractive Surgery Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Clara Colon
- Ocular Surface Imaging Center, Cornea and Refractive Surgery Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Emma Brown
- Ocular Surface Imaging Center, Cornea and Refractive Surgery Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - David Borsook
- Pain/Analgesia Imaging Neuroscience (P.A.I.N.) Group, Department of Anesthesia, Boston Children's Hospital, Center for Pain and the Brain, Harvard Medical School, Waltham, MA, USA
| | - Harald Prüss
- Department of Neurology, Charité University Medicine Berlin, Germany
| | - Pedram Hamrah
- Ocular Surface Imaging Center, Cornea and Refractive Surgery Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
94
|
Role of transient receptor potential channels in intestinal inflammation and visceral pain: novel targets in inflammatory bowel diseases. Inflamm Bowel Dis 2015; 21:419-27. [PMID: 25437822 DOI: 10.1097/mib.0000000000000234] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transient receptor potential (TRP) channels are a large group of ion channels that are prevalent in mammalian tissues. They are widely distributed in the central and peripheral nervous systems, and in nonneuronal cells, where they are implicated in sensing temperature, noxious substances, and pain. TRPs play an important role in immune response and nociception and, therefore, may be involved in the pathogenesis of inflammatory bowel diseases, whose major symptoms include chronic inflammatory state and abdominal pain. In this review, we summarize what is known on TRP channels in inflammatory bowel disease and visceral pain; we focus in particular on TRPV1, TRPV4, TRPA1, and TRPM. We also analyze scientific reports that evidence potential use of TRP regulators in future inflammatory bowel disease treatment.
Collapse
|
95
|
Valtcheva MV, Samineni VK, Golden JP, Gereau RW, Davidson S. Enhanced nonpeptidergic intraepidermal fiber density and an expanded subset of chloroquine-responsive trigeminal neurons in a mouse model of dry skin itch. THE JOURNAL OF PAIN 2015; 16:346-56. [PMID: 25640289 DOI: 10.1016/j.jpain.2015.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 01/08/2015] [Accepted: 01/16/2015] [Indexed: 12/13/2022]
Abstract
UNLABELLED Chronic pruritic conditions are often associated with dry skin and loss of epidermal barrier integrity. In this study, repeated application of acetone and ether followed by water (AEW) to the cheek skin of mice produced persistent scratching behavior with no increase in pain-related forelimb wiping, indicating the generation of itch without pain. Cheek skin immunohistochemistry showed a 64.5% increase in total epidermal innervation in AEW-treated mice compared to water-treated controls. This increase was independent of scratching, because mice prevented from scratching by Elizabethan collars showed similar hyperinnervation. To determine the effects of dry skin treatment on specific subsets of peripheral fibers, we examined Ret-positive, calcitonin gene-related peptide (CGRP)-positive, and glial cell line-derived neurotrophic factor family receptor α3 (GFRα3)-positive intraepidermal fiber density. AEW treatment increased Ret-positive fibers but not CGRP-positive or GFRα3-positive fibers, suggesting that a specific subset of nonpeptidergic fibers could contribute to dry skin itch. To test whether trigeminal ganglion neurons innervating the cheek exhibited altered excitability after AEW treatment, primary cultures of retrogradely labeled neurons were examined using whole-cell patch clamp electrophysiology. AEW treatment produced no differences in measures of excitability compared to water-treated controls. In contrast, a significantly higher proportion of trigeminal ganglion neurons was responsive to the nonhistaminergic pruritogen chloroquine after AEW treatment. We conclude that nonpeptidergic, Ret-positive fibers and chloroquine-sensitive neurons may contribute to dry skin pruritus. PERSPECTIVE This study examines the underlying neurobiological mechanisms of persistent dry skin itch. Our results indicate that nonpeptidergic epidermal hyperinnervation and nonhistaminergic pruritic receptors are potential targets for chronic pruritus.
Collapse
Affiliation(s)
- Manouela V Valtcheva
- Washington University Pain Center and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri; Medical Scientist Training Program, Washington University in St. Louis, St. Louis, Missouri
| | - Vijay K Samineni
- Washington University Pain Center and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri
| | - Judith P Golden
- Washington University Pain Center and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri
| | - Robert W Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri
| | - Steve Davidson
- Washington University Pain Center and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri.
| |
Collapse
|
96
|
Weng HJ, Patel KN, Jeske NA, Bierbower SM, Zou W, Tiwari V, Zheng Q, Tang Z, Mo GCH, Wang Y, Geng Y, Zhang J, Guan Y, Akopian AN, Dong X. Tmem100 Is a Regulator of TRPA1-TRPV1 Complex and Contributes to Persistent Pain. Neuron 2015; 85:833-46. [PMID: 25640077 DOI: 10.1016/j.neuron.2014.12.065] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/03/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022]
Abstract
TRPA1 and TRPV1 are crucial pain mediators, but how their interaction contributes to persistent pain is unknown. Here, we identify Tmem100 as a potentiating modulator of TRPA1-V1 complexes. Tmem100 is coexpressed and forms a complex with TRPA1 and TRPV1 in DRG neurons. Tmem100-deficient mice show a reduction in inflammatory mechanical hyperalgesia and TRPA1- but not TRPV1-mediated pain. Single-channel recording in a heterologous system reveals that Tmem100 selectively potentiates TRPA1 activity in a TRPV1-dependent manner. Mechanistically, Tmem100 weakens the association of TRPA1 and TRPV1, thereby releasing the inhibition of TRPA1 by TRPV1. A Tmem100 mutant, Tmem100-3Q, exerts the opposite effect; i.e., it enhances the association of TRPA1 and TRPV1 and strongly inhibits TRPA1. Strikingly, a cell-permeable peptide (CPP) containing the C-terminal sequence of Tmem100-3Q mimics its effect and inhibits persistent pain. Our study unveils a context-dependent modulation of the TRPA1-V1 complex, and Tmem100-3Q CPP is a promising pain therapy.
Collapse
Affiliation(s)
- Hao-Jui Weng
- Departments of Neuroscience and Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Dermatology, National Taiwan University Hospital, Taipei City 100, Taiwan
| | - Kush N Patel
- Departments of Neuroscience and Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Nathaniel A Jeske
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Sonya M Bierbower
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Vinod Tiwari
- Department of Anesthesiology and Critical Care, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Qin Zheng
- Departments of Neuroscience and Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Zongxiang Tang
- Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Gary C H Mo
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yan Wang
- Departments of Neuroscience and Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yixun Geng
- Departments of Neuroscience and Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jin Zhang
- Departments of Neuroscience and Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Armen N Akopian
- Department of Endodontics, University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | - Xinzhong Dong
- Departments of Neuroscience and Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
97
|
Abstract
The transient receptor potential A1 (TRPA1) channel is essential for vertebrate pain. Even though TRPA1 activation by ligands has been studied extensively, the molecular machinery regulating TRPA1 is only poorly understood. Using an unbiased proteomics-based approach we uncovered the physical association of Annexin A2 (AnxA2) with native TRPA1 in mouse sensory neurons. AnxA2 is enriched in a subpopulation of sensory neurons and coexpressed with TRPA1. Furthermore, we observe an increase of TRPA1 membrane levels in cultured sensory neurons from AnxA2-deficient mice. This is reflected by our calcium imaging experiments revealing higher responsiveness upon TRPA1 activation in AnxA2-deficient neurons. In vivo these findings are associated with enhanced nocifensive behaviors specifically in TRPA1-dependent paradigms of acute and inflammatory pain, while heat and mechanical sensitivity as well as TRPV1-mediated pain are preserved in AnxA2-deficient mice. Our results support a model whereby AnxA2 limits the availability of TRPA1 channels to regulate nociceptive signaling in vertebrates.
Collapse
|
98
|
Sousa-Valente J, Andreou AP, Urban L, Nagy I. Transient receptor potential ion channels in primary sensory neurons as targets for novel analgesics. Br J Pharmacol 2014; 171:2508-27. [PMID: 24283624 DOI: 10.1111/bph.12532] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/11/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed an explosion in novel findings relating to the molecules involved in mediating the sensation of pain in humans. Transient receptor potential (TRP) ion channels emerged as the greatest group of molecules involved in the transduction of various physical stimuli into neuronal signals in primary sensory neurons, as well as, in the development of pain. Here, we review the role of TRP ion channels in primary sensory neurons in the development of pain associated with peripheral pathologies and possible strategies to translate preclinical data into the development of effective new analgesics. Based on available evidence, we argue that nociception-related TRP channels on primary sensory neurons provide highly valuable targets for the development of novel analgesics and that, in order to reduce possible undesirable side effects, novel analgesics should prevent the translocation from the cytoplasm to the cell membrane and the sensitization of the channels rather than blocking the channel pore or binding sites for exogenous or endogenous activators.
Collapse
Affiliation(s)
- J Sousa-Valente
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | | | | | | |
Collapse
|
99
|
αCGRP is essential for algesic exocytotic mobilization of TRPV1 channels in peptidergic nociceptors. Proc Natl Acad Sci U S A 2014; 111:18345-50. [PMID: 25489075 DOI: 10.1073/pnas.1420252111] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Proalgesic sensitization of peripheral nociceptors in painful syndromes is a complex molecular process poorly understood that involves mobilization of thermosensory receptors to the neuronal surface. However, whether recruitment of vesicular thermoTRP channels is a general mechanism underlying sensitization of all nociceptor types or is subtype-specific remains controversial. We report that sensitization-induced Ca(2+)-dependent exocytotic insertion of transient receptor potential vanilloid 1 (TRPV1) receptors to the neuronal plasma membrane is a mechanism specifically used by peptidergic nociceptors to potentiate their excitability. Notably, we found that TRPV1 is present in large dense-core vesicles (LDCVs) that were mobilized to the neuronal surface in response to a sensitizing insult. Deletion or silencing of calcitonin-gene-related peptide alpha (αCGRP) gene expression drastically reduced proalgesic TRPV1 potentiation in peptidergic nociceptors by abrogating its Ca(2+)-dependent exocytotic recruitment. These findings uncover a context-dependent molecular mechanism of TRPV1 algesic sensitization and a previously unrecognized role of αCGRP in LDCV mobilization in peptidergic nociceptors. Furthermore, these results imply that concurrent secretion of neuropeptides and channels in peptidergic C-type nociceptors facilitates a rapid modulation of pain signaling.
Collapse
|
100
|
Lin YJ, Lin RL, Ruan T, Khosravi M, Lee LY. A synergistic effect of simultaneous TRPA1 and TRPV1 activations on vagal pulmonary C-fiber afferents. J Appl Physiol (1985) 2014; 118:273-81. [PMID: 25414245 DOI: 10.1152/japplphysiol.00805.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transient receptor potential ankyrin type 1 (TRPA1) and vanilloid type 1 (TRPV1) receptors are coexpressed in vagal pulmonary C-fiber sensory nerves. Because both these receptors are sensitive to a number of endogenous inflammatory mediators, it is conceivable that they can be activated simultaneously during airway inflammation. This study aimed to determine whether there is an interaction between these two polymodal transducers upon simultaneous activation, and how it modulates the activity of vagal pulmonary C-fiber sensory nerves. In anesthetized, spontaneously breathing rats, the reflex-mediated apneic response to intravenous injection of a combined dose of allyl isothiocyanate (AITC, a TRPA1 activator) and capsaicin (Cap, a TRPV1 activator) was ∼202% greater than the mathematical sum of the responses to AITC and Cap when they were administered individually. Similar results were also observed in anesthetized mice. In addition, the synergistic effect was clearly demonstrated when the afferent activity of single vagal pulmonary C-fiber afferents were recorded in anesthetized, artificially ventilated rats; C-fiber responses to AITC, Cap and AITC + Cap (in combination) were 0.6 ± 0.1, 0.8 ± 0.1, and 4.8 ± 0.6 impulses/s (n = 24), respectively. This synergism was absent when either AITC or Cap was replaced by other chemical activators of pulmonary C-fiber afferents. The pronounced potentiating effect was further demonstrated in isolated vagal pulmonary sensory neurons using the Ca(2+) imaging technique. In summary, this study showed a distinct positive interaction between TRPA1 and TRPV1 when they were activated simultaneously in pulmonary C-fiber sensory nerves.
Collapse
Affiliation(s)
- Yu-Jung Lin
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky
| | - Ruei-Lung Lin
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky
| | - Ting Ruan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; and
| | - Mehdi Khosravi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| | - Lu-Yuan Lee
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky;
| |
Collapse
|