1
|
Potapenko IV, Ishikawa T, Okuda H, Hori K, Ozaki N. Chemogenetic inhibition of pain-related neurons in the posterior insula cortex reduces mechanical hyperalgesia and anxiety-like behavior during acute pain. Biochem Biophys Res Commun 2024; 734:150625. [PMID: 39236586 DOI: 10.1016/j.bbrc.2024.150625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/09/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Pain is a complex phenomenon that involves sensory, emotional, and cognitive components. The posterior insula cortex (pIC) has been shown to integrate multisensory experience with emotional and cognitive states. However, the involvement of the pIC in the regulation of affective behavior in pain remains unclear. Here, we investigate the role of pain-related pIC neurons in the regulation of anxiety-like behavior during acute pain. We combined a chemogenetic approach with targeted recombination in active populations (TRAP) in mice. Global chemogenetic inhibition of pIC neurons attenuates chemically-induced mechanical hypersensitivity without affecting pain-related anxiety-like behavior. In contrast, inhibition of pain-related pIC neurons reduces both mechanical hypersensitivity and pain-related anxiety-like behavior. The present study provides important insights into the role of pIC neurons in the regulation of sensory and affective pain-related behavior.
Collapse
Affiliation(s)
- Ilia Viktorovich Potapenko
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Takara-machi, Kanazawa, 920-8640, Japan; Department of Medical and Biological Physics, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Partizan Zheleznyak st. 1, 660022, Krasnoyarsk, Russia
| | - Tatsuya Ishikawa
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Takara-machi, Kanazawa, 920-8640, Japan.
| | - Hiroaki Okuda
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Takara-machi, Kanazawa, 920-8640, Japan
| | - Kiyomi Hori
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Takara-machi, Kanazawa, 920-8640, Japan
| | - Noriyuki Ozaki
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Takara-machi, Kanazawa, 920-8640, Japan.
| |
Collapse
|
2
|
Belinskaia M, Wang J, Kaza SK, Antoniazzi C, Zurawski T, Dolly JO, Lawrence GW. Bipartite Activation of Sensory Neurons by a TRPA1 Agonist Allyl Isothiocyanate Is Reflected by Complex Ca 2+ Influx and CGRP Release Patterns: Enhancement by NGF and Inhibition with VAMP and SNAP-25 Cleaving Botulinum Neurotoxins. Int J Mol Sci 2023; 24:ijms24021338. [PMID: 36674850 PMCID: PMC9865456 DOI: 10.3390/ijms24021338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
The trafficking of transient receptor potential (TRP) channels to the plasma membrane and the release of calcitonin gene-related peptide (CGRP) from trigeminal ganglion neurons (TGNs) are implicated in some aspects of chronic migraines. These exocytotic processes are inhibited by cleavage of SNAREs with botulinum neurotoxins (BoNTs); moreover, type A toxin (/A) clinically reduces the frequency and severity of migraine attacks but not in all patients for unknown reasons. Herein, neonatal rat TGNs were stimulated with allyl isothiocyanate (AITC), a TRPA1 agonist, and dose relationships were established to link the resultant exocytosis of CGRP with Ca2+ influx. The CGRP release, quantified by ELISA, was best fit by a two-site model (EC50 of 6 and 93 µM) that correlates with elevations in intracellular Ca2+ [Ca2+]i revealed by time-lapse confocal microscopy of fluo-4-acetoxymethyl ester (Fluo-4 AM) loaded cells. These signals were all blocked by two TRPA1 antagonists, HC-030031 and A967079. At low [AITC], [Ca2+]i was limited because of desensitisation to the agonist but rose for concentrations > 0.1 mM due to a deduced non-desensitising second phase of Ca2+ influx. A recombinant BoNT chimera (/DA), which cleaves VAMP1/2/3, inhibited AITC-elicited CGRP release to a greater extent than SNAP-25-cleaving BoNT/A. /DA also proved more efficacious against CGRP efflux evoked by a TRPV1 agonist, capsaicin. Nerve growth factor (NGF), a pain-inducing sensitiser of TGNs, enhanced the CGRP exocytosis induced by low [AITC] only. Both toxins blocked NGF-induced neuropeptide secretion and its enhancement of the response to AITC. In conclusion, NGF sensitisation of sensory neurons involves TRPA1, elevated Ca2+ influx, and CGRP exocytosis, mediated by VAMP1/2/3 and SNAP-25 which can be attenuated by the BoNTs.
Collapse
|
3
|
Jennings EM, Sullivan LC, Jamshidi RJ, LoCoco PM, Smith HR, Chavera TS, Berg KA, Clarke WP. Age-related changes in peripheral nociceptor function. Neuropharmacology 2022; 216:109187. [PMID: 35835212 DOI: 10.1016/j.neuropharm.2022.109187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/21/2022] [Accepted: 07/03/2022] [Indexed: 01/05/2023]
Abstract
Pain and pain management in the elderly population is a significant social and medical problem. Pain sensation is a complex phenomenon that typically involves activation of peripheral pain-sensing neurons (nociceptors) which send signals to the spinal cord and brain that are interpreted as pain, an unpleasant sensory experience. In this work, young (4-5 months) and aged (26-27 months) Fischer 344 x Brown Norway (F344xBN) rats were examined for nociceptor sensitivity to activation by thermal (cold and heat) and mechanical stimulation following treatment with inflammatory mediators and activators of transient receptor potential (TRP) channels. Unlike other senses that decrease in sensitivity with age, sensitivity of hindpaw nociceptors to thermal and mechanical stimulation was not different between young and aged F344xBN rats. Intraplantar injection of bradykinin (BK) produced greater thermal and mechanical allodynia in aged versus young rats, whereas only mechanical allodynia was greater in aged rats following injection of prostaglandin E2 (PGE2). Intraplantar injection of TRP channel activators, capsaicin (TRPV1), mustard oil (TRPA1) and menthol (TRPM8) each resulted in greater mechanical allodynia in aged versus young rats and capsaicin-induced heat allodynia was also greater in aged rats. A treatment-induced allodynia that was greater in young rats was never observed. The anti-allodynic effects of intraplantar injection of kappa and delta opioid receptor agonists, salvinorin-A and D-Pen2,D-Pen5]enkephalin (DPDPE), respectively, were greater in aged than young rats, whereas mu opioid receptor agonists, [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) and morphine, were not effective in aged rats. Consistent with these observations, in primary cultures of peripheral sensory neurons, inhibition of cAMP signaling in response to delta and kappa receptor agonists was greater in cultures derived from aged rats. By contrast, mu receptor agonists did not inhibit cAMP signaling in aged rats. Thus, age-related changes in nociceptors generally favor increased pain signaling in aged versus young rats, suggesting that changes in nociceptor sensitivity may play a role in the increased incidence of pain in the elderly population. These results also suggest that development of peripherally-restricted kappa or delta opioid receptor agonists may provide safer and effective pain relief for the elderly.
Collapse
Affiliation(s)
- Elaine M Jennings
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Laura C Sullivan
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Raehannah J Jamshidi
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Peter M LoCoco
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Hudson R Smith
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Teresa S Chavera
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Kelly A Berg
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - William P Clarke
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
4
|
Jorge CO, Melo-Aquino BD, Santos DFDSD, Oliveira MCGD. Muscle pain induced by static contraction is modulated by transient receptor potential vanilloid 1 and ankyrin 1 receptors. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
5
|
Hovhannisyan AH, Son H, Mecklenburg J, Barba-Escobedo PA, Tram M, Gomez R, Shannonhouse J, Zou Y, Weldon K, Ruparel S, Lai Z, Tumanov AV, Kim YS, Akopian AN. Pituitary hormones are specifically expressed in trigeminal sensory neurons and contribute to pain responses in the trigeminal system. Sci Rep 2021; 11:17813. [PMID: 34497285 PMCID: PMC8426369 DOI: 10.1038/s41598-021-97084-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/20/2021] [Indexed: 11/29/2022] Open
Abstract
Trigeminal (TG), dorsal root (DRG), and nodose/jugular (NG/JG) ganglia each possess specialized and distinct functions. We used RNA sequencing of two-cycle sorted Pirt-positive neurons to identify genes exclusively expressing in L3-L5 DRG, T10-L1 DRG, NG/JG, and TG mouse ganglion neurons. Transcription factor Phox2b and Efcab6 are specifically expressed in NG/JG while Hoxa7 is exclusively present in both T10-L1 and L3-L5 DRG neurons. Cyp2f2, Krt18, and Ptgds, along with pituitary hormone prolactin (Prl), growth hormone (Gh), and proopiomelanocortin (Pomc) encoding genes are almost exclusively in TG neurons. Immunohistochemistry confirmed selective expression of these hormones in TG neurons and dural nerves; and showed GH expression in subsets of TRPV1+ and CGRP+ TG neurons. We next examined GH roles in hypersensitivity in the spinal versus trigeminal systems. Exogenous GH produced mechanical hypersensitivity when injected intrathecally, but not intraplantarly. GH-induced thermal hypersensitivity was not detected in the spinal system. GH dose-dependently generated orofacial and headache-like periorbital mechanical hypersensitivity after administration into masseter muscle and dura, respectively. Periorbital mechanical hypersensitivity was reversed by a GH receptor antagonist, pegvisomant. Overall, pituitary hormone genes are selective for TG versus other ganglia somatotypes; and GH has distinctive functional significance in the trigeminal versus spinal systems.
Collapse
Affiliation(s)
- Anahit H Hovhannisyan
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Hyeonwi Son
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Jennifer Mecklenburg
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Priscilla Ann Barba-Escobedo
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Meilinn Tram
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Ruben Gomez
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - John Shannonhouse
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Yi Zou
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Korri Weldon
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Shivani Ruparel
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Zhao Lai
- Departments of Molecular Medicine, Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Alexei V Tumanov
- Departments of Microbiology, Immunology and Molecular Genetics, Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Yu Shin Kim
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Armen N Akopian
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA.
| |
Collapse
|
6
|
Lindquist KA, Belugin S, Hovhannisyan AH, Corey TM, Salmon A, Akopian AN. Identification of Trigeminal Sensory Neuronal Types Innervating Masseter Muscle. eNeuro 2021; 8:ENEURO.0176-21.2021. [PMID: 34580157 PMCID: PMC8513531 DOI: 10.1523/eneuro.0176-21.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/18/2021] [Accepted: 09/01/2021] [Indexed: 11/21/2022] Open
Abstract
Understanding masseter muscle (MM) innervation is critical for the study of cell-specific mechanisms of pain induced by temporomandibular disorder (TMDs) or after facial surgery. Here, we identified trigeminal (TG) sensory neuronal subtypes (MM TG neurons) innervating MM fibers, masseteric fascia, tendons, and adjusted tissues. A combination of patch clamp electrophysiology and immunohistochemistry (IHC) on TG neurons back-traced from reporter mouse MM found nine distinct subtypes of MM TG neurons. Of these neurons, 24% belonged to non-peptidergic IB-4+/TRPA1- or IB-4+/TRPA1+ groups, while two TRPV1+ small-sized neuronal groups were classified as peptidergic/CGRP+ One small-sized CGRP+ neuronal group had a unique electrophysiological profile and were recorded from Nav1.8- or trkC+ neurons. The remaining CGRP+ neurons were medium-sized, could be divided into Nav1.8-/trkC- and Nav1.8low/trkC+ clusters, and showed large 5HT-induced current. The final two MM TG neuronal groups were trkC+ and had no Nav1.8 and CGRP. Among MM TG neurons, TRPV1+/CGRP- (somatostatin+), tyrosine hydroxylase (TH)+ (C-LTMR), TRPM8+, MrgprA3+, or trkB+ (Aδ-LTMR) subtypes have not been detected. Masseteric muscle fibers, tendons and masseteric fascia in mice and the common marmoset, a new world monkey, were exclusively innervated by either CGRP+/NFH+ or CGRP-/NFH+ medium-to-large neurons, which we found using a Nav1.8-YFP reporter, and labeling with CGRP, TRPV1, neurofilament heavy chain (NFH) and pgp9.5 antibodies. These nerves were mainly distributed in tendon and at junctions of deep-middle-superficial parts of MM. Overall, the data presented here demonstrates that MM is innervated by a distinct subset of TG neurons, which have unique characteristics and innervation patterns.
Collapse
Affiliation(s)
- Karen A Lindquist
- Integrated Biomedical Sciences (IBMS) Program, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Sergei Belugin
- Endodontics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Anahit H Hovhannisyan
- Endodontics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Tatiana M Corey
- Laboratory Animal Resources Departments, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Adam Salmon
- Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- Sam and Ann Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- South Texas Veterans Health Care System, Geriatric Research Education and Clinical Center San Antonio, TX 78229
| | - Armen N Akopian
- Integrated Biomedical Sciences (IBMS) Program, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- Endodontics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| |
Collapse
|
7
|
Wilzopolski J, Kietzmann M, Mishra SK, Stark H, Bäumer W, Rossbach K. TRPV1 and TRPA1 Channels Are Both Involved Downstream of Histamine-Induced Itch. Biomolecules 2021; 11:1166. [PMID: 34439832 PMCID: PMC8391774 DOI: 10.3390/biom11081166] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 01/17/2023] Open
Abstract
Two histamine receptor subtypes (HR), namely H1R and H4R, are involved in the transmission of histamine-induced itch as key components. Although exact downstream signaling mechanisms are still elusive, transient receptor potential (TRP) ion channels play important roles in the sensation of histaminergic and non-histaminergic itch. The aim of this study was to investigate the involvement of TRPV1 and TRPA1 channels in the transmission of histaminergic itch. The potential of TRPV1 and TRPA1 inhibitors to modulate H1R- and H4R-induced signal transmission was tested in a scratching assay in mice in vivo as well as via Ca2+ imaging of murine sensory dorsal root ganglia (DRG) neurons in vitro. TRPV1 inhibition led to a reduction of H1R- and H4R- induced itch, whereas TRPA1 inhibition reduced H4R- but not H1R-induced itch. TRPV1 and TRPA1 inhibition resulted in a reduced Ca2+ influx into sensory neurons in vitro. In conclusion, these results indicate that both channels, TRPV1 and TRPA1, are involved in the transmission of histamine-induced pruritus.
Collapse
Affiliation(s)
- Jenny Wilzopolski
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (M.K.); (K.R.)
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA; (S.K.M.); (W.B.)
- Department of Veterinary Medicine, Institute of Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Manfred Kietzmann
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (M.K.); (K.R.)
| | - Santosh K. Mishra
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA; (S.K.M.); (W.B.)
| | - Holger Stark
- Institute of Pharmaceutical and Medical Chemistry, Heinrich Heine University Düsseldorf, 40225 Duesseldorf, Germany;
| | - Wolfgang Bäumer
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA; (S.K.M.); (W.B.)
- Department of Veterinary Medicine, Institute of Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Kristine Rossbach
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (M.K.); (K.R.)
| |
Collapse
|
8
|
Lou Y, Liu Y, Zhao J, Tian W, Xu N, Zang C, Que K. Activation of Transient Receptor Potential Ankyrin 1 and Vanilloid 1 Channels Promotes Odontogenic Differentiation of Human Dental Pulp Cells. J Endod 2021; 47:1409-1416. [PMID: 34126160 DOI: 10.1016/j.joen.2021.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/20/2021] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Transient receptor potential ankyrin 1 (TRPA1) and vanilloid 1 (TRPV1) are thermosensitive channels that play an important role in thermal sensation or tooth pain by regulating intracellular Ca2+ concentration that is essential for pulp tissue repair. The aim of this study was to evaluate the role of TRPA1 and TRPV1 channels in the odontogenic differentiation of human dental pulp cells (HDPCs). METHODS HDPCs were isolated from healthy human intact third molars and cultured in odontogenic differentiation medium. Gene and protein expression levels of TRPA1 and TRPV1 channels during the odontogenic differentiation of HDPCs were evaluated by real-time quantitative polymerase chain reaction and Western blot analysis. HDPCs were then treated with channel agonists or antagonists, and the expression levels of odontogenic markers dentin sialophosphoprotein (DSPP) and osteopontin (OPN) were examined. Alkaline phosphatase activity and alizarin red staining were also conducted to detect mineralization levels. RESULTS Consistent with the mineralization degree and DSPP and OPN expression, messenger RNA and protein expression of TRPA1 and TRPV1 channels was up-regulated during the odontogenic differentiation of HDPCs. The application of TRPA1 or TRPV1 agonists increased the mineralized nodules of alizarin red staining and alkaline phosphatase activity and up-regulated the messenger RNA and protein expression of DSPP and OPN, respectively, with the highest values reached on the seventh day (P < .05). On the contrary, the mineralization level and DSPP and OPN expression could be suppressed by using the antagonists of these 2 channels. CONCLUSIONS TRPA1 and TRPV1 channels not only showed up-regulated expression along with the odontogenic differentiation of HDPCs but also could affect the odontogenic differentiation by regulating intracellular Ca2+ concentration.
Collapse
Affiliation(s)
- Yaxin Lou
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Yangqiu Liu
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Jiange Zhao
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China
| | - Weiping Tian
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Na Xu
- Department of Paediatric Dentistry, Hospital of Stomatology, NanKai University, Tianjin, China
| | - Chengcheng Zang
- Department of Prosthodontics, College of Stomatology, Tianjin Medical University, Tianjin, China.
| | - Kehua Que
- Department of Endodontics, College of Stomatology, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
9
|
Avona A, Mason BN, Burgos-Vega C, Hovhannisyan AH, Belugin SN, Mecklenburg J, Goffin V, Wajahat N, Price TJ, Akopian AN, Dussor G. Meningeal CGRP-Prolactin Interaction Evokes Female-Specific Migraine Behavior. Ann Neurol 2021; 89:1129-1144. [PMID: 33749851 PMCID: PMC8195469 DOI: 10.1002/ana.26070] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Migraine is three times more common in women. CGRP plays a critical role in migraine pathology and causes female-specific behavioral responses upon meningeal application. These effects are likely mediated through interactions of CGRP with signaling systems specific to females. Prolactin (PRL) levels have been correlated with migraine attacks. Here, we explore a potential interaction between CGRP and PRL in the meninges. METHODS Prolactin, CGRP, and receptor antagonists CGRP8-37 or Δ1-9-G129R-hPRL were administered onto the dura of rodents followed by behavioral testing. Immunohistochemistry was used to examine PRL, CGRP and Prolactin receptor (Prlr) expression within the dura. Electrophysiology on cultured and back-labeled trigeminal ganglia (TG) neurons was used to assess PRL-induced excitability. Finally, the effects of PRL on evoked CGRP release from ex vivo dura were measured. RESULTS We found that dural PRL produced sustained and long-lasting migraine-like behavior in cycling and ovariectomized female, but not male rodents. Prlr was expressed on dural afferent nerves in females with little-to-no presence in males. Consistent with this, PRL increased excitability only in female TG neurons innervating the dura and selectively sensitized CGRP release from female ex vivo dura. We demonstrate crosstalk between PRL and CGRP systems as CGRP8-37 decreases migraine-like responses to dural PRL. Reciprocally, Δ1-9-G129R-hPRL attenuates dural CGRP-induced migraine behaviors. Similarly, Prlr deletion from sensory neurons significantly reduced migraine-like responses to dural CGRP. INTERPRETATION This CGRP-PRL interaction in the meninges is a mechanism by which these peptides could produce female-selective responses and increase the prevalence of migraine in women. ANN NEUROL 2021;89:1129-1144.
Collapse
Affiliation(s)
- Amanda Avona
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX
| | - Bianca N. Mason
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX
| | - Carolina Burgos-Vega
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX
| | - Anahit H. Hovhannisyan
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Sergei N. Belugin
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Jennifer Mecklenburg
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | | | - Naureen Wajahat
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX
| | - Theodore J. Price
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX
| | - Armen N. Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Gregory Dussor
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX
| |
Collapse
|
10
|
Pavlenko D, Funahashi H, Sakai K, Hashimoto T, Lozada T, Yosipovitch G, Akiyama T. IL-23 modulates histamine-evoked itch and responses of pruriceptors in mice. Exp Dermatol 2020; 29:1209-1215. [PMID: 33010057 DOI: 10.1111/exd.14206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2023]
Abstract
Accumulating evidence has highlighted the essential roles of cytokines in itch processing. Although IL-23 and Th17 cytokines are elevated in inflammatory skin disorders, their role in itch is unknown. Here, we investigated the role of IL-23 and IL-17A in itch response using an in vitro calcium imaging of mouse dorsal root ganglion (DRG) neurons and an in vivo behaviour test. Calcium imaging studies revealed that a few DRG neurons (~5%) responded to either IL-23 or IL-17A. Pretreatment cells with IL-23 significantly reduced calcium responses to histamine and capsaicin but not chloroquine. Behaviour experiments showed neither IL-23 nor IL-17A evoked scratching. IL-23 significantly decreased histamine-evoked scratching without affecting chloroquine-evoked scratching. There was no difference in scratching between IL-17A- and vehicle-treated groups. These results indicate that IL-23 might play a role in regulating histaminergic itch via modulation of TRPV1 activity.
Collapse
Affiliation(s)
- Darya Pavlenko
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hideki Funahashi
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Psychiatry, University of Miyazaki, Kiyotake, Japan
| | - Kent Sakai
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Takashi Hashimoto
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Taisa Lozada
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gil Yosipovitch
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tasuku Akiyama
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
11
|
Zhang X, He Y. The Role of Nociceptive Neurons in the Pathogenesis of Psoriasis. Front Immunol 2020; 11:1984. [PMID: 33133059 PMCID: PMC7550422 DOI: 10.3389/fimmu.2020.01984] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease. Emerging evidence shows that neurogenic inflammation, induced by nociceptive neurons and T helper 17 cell (Th17) responses, has a fundamental role in maintaining the changes in the immune system due to psoriasis. Nociceptive neurons, specific primary sensory nerves, have a multi-faceted role in detecting noxious stimuli, maintaining homeostasis, and regulating the immunity responses in the skin. Therefore, it is critical to understand the connections and interplay between the nociceptive neurons and the immune system in psoriasis. Here, we review works on the altered innervation that occurs in psoriasis. We examine how these distinct sensory neurons and their signal transducers participate in regulating inflammation. Numerous clinical studies report the dysfunction of nociceptive neurons in psoriasis. We discuss the mechanism behind the inconsistent activation of nociceptive neurons. Moreover, we review how neuropeptides, involved in regulating Th17 responses and the role of nociceptive neurons, regulate immunity in psoriasis. Understanding how nociceptive neurons regulate immune responses enhances our knowledge of the neuroimmunity involved in the pathogenesis of psoriasis and may form the basis for new approaches to treat it.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yanling He
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Khalil M, Zhang Z, Abdel-Aziz H, Rabini S, Ammar R, Reeh P, Engel M. Dual opposing actions of STW 5 on TRP receptors mediate neuronal desensitisation in vitro. Life Sci 2020; 257:118112. [DOI: 10.1016/j.lfs.2020.118112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
|
13
|
Upregulation of the TRPA1 Ion Channel in the Gastric Mucosa after Iodoacetamide-Induced Gastritis in Rats: A Potential New Therapeutic Target. Int J Mol Sci 2020; 21:ijms21165591. [PMID: 32764237 PMCID: PMC7460663 DOI: 10.3390/ijms21165591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 02/08/2023] Open
Abstract
Acute gastritis is often untreatable by acid secretion-inhibiting drugs. Understanding the protective mechanisms including the role of Transient Receptor Potential Ankyrin1 (TRPA1) and Vanilloid1 (TRPV1) channels localized on capsaicin-sensitive afferents and non-neuronal structures might identify novel therapeutic approaches. Therefore, we characterized a translational gastritis model using iodoacetamide (IAA) and investigated TRPA1/V1 expressions. Wistar rats and CD1, C57Bl/6J mice were exposed to IAA-containing (0.05, 0.1, 0.2, 0.3, 0.5%) drinking water for 7 or 14 days. Body weight and water consumption were recorded daily. Macroscopic lesions were scored, qualitative histopathologic investigation was performed, TRPA1/V1 immunopositivity and mRNA expressions were measured. IAA induced a concentration-dependent weight loss and reduced water intake in both species. Hyperemia, submucosal edema, inflammatory infiltration and hemorrhagic erosions developed after 7 days, while ulcers after 14 days in rats. Trpa1 mRNA/protein expressions were upregulated at both timepoints. Meanwhile, TRPV1 immunopositivity was upregulated in the gastric corpus after 0.05% IAA ingestion, but downregulated after 0.2%, whereas Trpv1 mRNA did not change. Interestingly, no macroscopic/microscopic changes were observed in mice. These are the first data for the concentration- and duration-dependent changes in the IAA-induced gastritis in rats accompanied by TRPA1 upregulation, therefore, its therapeutic potential in gastritis should further be investigated.
Collapse
|
14
|
TRPA1 and TRPV1 channels participate in atmospheric-pressure plasma-induced [Ca 2+] i response. Sci Rep 2020; 10:9687. [PMID: 32546738 PMCID: PMC7297720 DOI: 10.1038/s41598-020-66510-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/18/2020] [Indexed: 11/08/2022] Open
Abstract
Despite successful clinical application of non-equilibrium atmospheric pressure plasma (APP), the details of the molecular mechanisms underlying APP-inducible biological responses remain ill-defined. We previously reported that exposure of 3T3L1 cells to APP-irradiated buffer raised the cytoplasmic free Ca2+ ([Ca2+]i) concentration by eliciting Ca2+ influx in a manner sensitive to transient receptor potential (TRP) channel inhibitors. However, the precise identity of the APP-responsive channel molecule(s) remains unclear. In the present study, we aimed to clarify channel molecule(s) responsible for indirect APP-responsive [Ca2+]i rises. siRNA-mediated silencing experiments revealed that TRPA1 and TRPV1 serve as the major APP-responsive Ca2+ channels in 3T3L1 cells. Conversely, ectopic expression of either TRPA1 or TRPV1 in APP-unresponsive C2C12 cells actually triggered [Ca2+]i elevation in response to indirect APP exposure. Desensitization experiments using 3T3L1 cells revealed APP responsiveness to be markedly suppressed after pretreatment with allyl isothiocyanate or capsaicin, TRPA1 and TRPV1 agonists, respectively. APP exposure also desensitized the cells to these chemical agonists, indicating the existence of a bi-directional heterologous desensitization property of APP-responsive [Ca2+]i transients mediated through these TRP channels. Mutational analyses of key cysteine residues in TRPA1 (Cys421, Cys621, Cys641, and Cys665) and in TRPV1 (Cys258, Cys363, and Cys742) have suggested that multiple reactive oxygen and nitrogen species are intricately involved in activation of the channels via a broad range of modifications involving these cysteine residues. Taken together, these observations allow us to conclude that both TRPA1 and TRPV1 channels play a pivotal role in evoking indirect APP-dependent [Ca2+]i responses.
Collapse
|
15
|
Becker AK, Auditore A, Pischetsrieder M, Messlinger K, Fleming T, Reeh PW, Sauer SK. Reactive dicarbonyl compounds cause Calcitonin Gene-Related Peptide release and synergize with inflammatory conditions in mouse skin and peritoneum. J Biol Chem 2020; 295:6330-6343. [PMID: 32198181 DOI: 10.1074/jbc.ra120.012890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/18/2020] [Indexed: 02/02/2023] Open
Abstract
The plasmas of diabetic or uremic patients and of those receiving peritoneal dialysis treatment have increased levels of the glucose-derived dicarbonyl metabolites like methylglyoxal (MGO), glyoxal (GO), and 3-deoxyglucosone (3-DG). The elevated dicarbonyl levels can contribute to the development of painful neuropathies. Here, we used stimulated immunoreactive Calcitonin Gene-Related Peptide (iCGRP) release as a measure of nociceptor activation, and we found that each dicarbonyl metabolite induces a concentration-, TRPA1-, and Ca2+-dependent iCGRP release. MGO, GO, and 3-DG were about equally potent in the millimolar range. We hypothesized that another dicarbonyl, 3,4-dideoxyglucosone-3-ene (3,4-DGE), which is present in peritoneal dialysis (PD) solutions after heat sterilization, activates nociceptors. We also showed that at body temperatures 3,4-DGE is formed from 3-DG and that concentrations of 3,4-DGE in the micromolar range effectively induced iCGRP release from isolated murine skin. In a novel preparation of the isolated parietal peritoneum PD fluid or 3,4-DGE alone, at concentrations found in PD solutions, stimulated iCGRP release. We also tested whether inflammatory tissue conditions synergize with dicarbonyls to induce iCGRP release from isolated skin. Application of MGO together with bradykinin or prostaglandin E2 resulted in an overadditive effect on iCGRP release, whereas MGO applied at a pH of 5.2 resulted in reduced release, probably due to an MGO-mediated inhibition of transient receptor potential (TRP) V1 receptors. These results indicate that several reactive dicarbonyls activate nociceptors and potentiate inflammatory mediators. Our findings underline the roles of dicarbonyls and TRPA1 receptors in causing pain during diabetes or renal disease.
Collapse
Affiliation(s)
- Anna K Becker
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsstrasse 17, 91054 Erlangen, Germany
| | - Andrea Auditore
- Department of Chemistry and Pharmacy, Food Chemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Monika Pischetsrieder
- Department of Chemistry and Pharmacy, Food Chemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Strasse 10, 91058 Erlangen, Germany
| | - Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsstrasse 17, 91054 Erlangen, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry and Pharmacology, University of Heidelberg, INF 410, 69120 Heidelberg, Germany.,German Center for Diabetes Research (DZD), Eberhard-Karls-University of Tuebingen, Otfried-Müller-Strasse 10, 72076 Tuebingen, Germany
| | - Peter W Reeh
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsstrasse 17, 91054 Erlangen, Germany
| | - Susanne K Sauer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsstrasse 17, 91054 Erlangen, Germany
| |
Collapse
|
16
|
Borgonovo G, De Petrocellis L, Schiano Moriello A, Bertoli S, Leone A, Battezzati A, Mazzini S, Bassoli A. Moringin, A Stable Isothiocyanate from Moringa oleifera, Activates the Somatosensory and Pain Receptor TRPA1 Channel In Vitro. Molecules 2020; 25:molecules25040976. [PMID: 32098328 PMCID: PMC7070407 DOI: 10.3390/molecules25040976] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Moringa oleifera Lam. is a tropical plant widely used in traditional medicines and as a food supplement. It is characterized by the presence of glucosinolates and isothiocyanates; the stable isothiocyanate 4-[(α-l-rhamnosyloxy)benzyl]isothiocyanate (moringin) has been widely studied for its bioactivity as hypoglycemic, antimicrobial, anticancer and in particular for its involvement in nociception and neurogenic pain. Moringa extracts and pure moringin were submitted to in vitro assays with the somatosensory TRPA1 ion channel, proving that moringin is a potent and effective agonist of this receptor involved in nociceptive function and pain states. Moringin do not activate or activates very weakly the vanilloids somatosensory channels TRPV1,2,3 and 4, and the melastatin cooling receptor TRPM8. The comparison of moringin’s activity with other known agonists of natural origin is also discussed.
Collapse
Affiliation(s)
- Gigliola Borgonovo
- Department of Food, Environment and Nutrition-DeFENS, University of Milan, Via Celoria 2, I-20133 Milano, Italy; (G.B.); (S.B.); (A.L.); (A.B.); (S.M.)
| | - Luciano De Petrocellis
- Endocannabinoid Research Group-Institute of Biomolecular Chemistry-CNR, Pozzuoli, I-87078 Napoli, Italy; (L.D.P.); (A.S.M.)
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group-Institute of Biomolecular Chemistry-CNR, Pozzuoli, I-87078 Napoli, Italy; (L.D.P.); (A.S.M.)
- Epitech Group SpA, Saccolongo, 35030 Padova, Italy
| | - Simona Bertoli
- Department of Food, Environment and Nutrition-DeFENS, University of Milan, Via Celoria 2, I-20133 Milano, Italy; (G.B.); (S.B.); (A.L.); (A.B.); (S.M.)
| | - Alessandro Leone
- Department of Food, Environment and Nutrition-DeFENS, University of Milan, Via Celoria 2, I-20133 Milano, Italy; (G.B.); (S.B.); (A.L.); (A.B.); (S.M.)
| | - Alberto Battezzati
- Department of Food, Environment and Nutrition-DeFENS, University of Milan, Via Celoria 2, I-20133 Milano, Italy; (G.B.); (S.B.); (A.L.); (A.B.); (S.M.)
| | - Stefania Mazzini
- Department of Food, Environment and Nutrition-DeFENS, University of Milan, Via Celoria 2, I-20133 Milano, Italy; (G.B.); (S.B.); (A.L.); (A.B.); (S.M.)
| | - Angela Bassoli
- Department of Food, Environment and Nutrition-DeFENS, University of Milan, Via Celoria 2, I-20133 Milano, Italy; (G.B.); (S.B.); (A.L.); (A.B.); (S.M.)
- Correspondence: ; Tel.: +39-0250316815
| |
Collapse
|
17
|
Patil MJ, Salas M, Bialuhin S, Boyd JT, Jeske NA, Akopian AN. Sensitization of small-diameter sensory neurons is controlled by TRPV1 and TRPA1 association. FASEB J 2020; 34:287-302. [PMID: 31914619 PMCID: PMC7539696 DOI: 10.1096/fj.201902026r] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/13/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023]
Abstract
Unique features of sensory neuron subtypes are manifest by their distinct physiological and pathophysiological functions. Using patch-clamp electrophysiology, Ca2+ imaging, calcitonin gene-related peptide release assay from tissues, protein biochemistry approaches, and behavioral physiology on pain models, this study demonstrates the diversity of sensory neuron pathophysiology is due in part to subtype-dependent sensitization of TRPV1 and TRPA1. Differential sensitization is influenced by distinct expression of inflammatory mediators, such as prostaglandin E2 (PGE2), bradykinin (BK), and nerve growth factor (NGF) as well as multiple kinases, including protein kinase A (PKA) and C (PKC). However, the co-expression and interaction of TRPA1 with TRPV1 proved to be the most critical for differential sensitization of sensory neurons. We identified N- and C-terminal domains on TRPV1 responsible for TRPA1-TRPV1 (A1-V1) complex formation. Ablation of A1-V1 complex with dominant-negative peptides against these domains substantially reduced the sensitization of TRPA1, as well as BK- and CFA-induced hypersensitivity. These data indicate that often occurring TRP channel complexes regulate diversity in neuronal sensitization and may provide a therapeutic target for many neuroinflammatory pain conditions.
Collapse
Affiliation(s)
- Mayur J. Patil
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
- The Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224
| | - Margaux Salas
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
- United States Army Institute of Surgical Research, Air Force- 59th Medical Wing, San Antonio, TX 78234
| | - Siarhei Bialuhin
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Jacob T. Boyd
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
- Department of Pharmcology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Nathaniel A. Jeske
- Department of Oral and Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
- Department of Pharmcology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Armen N. Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
- Department of Pharmcology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| |
Collapse
|
18
|
Ostrow KL, Donaldson KJ, Caterina MJ, Belzberg A, Hoke A. The Secretomes of Painful Versus Nonpainful Human Schwannomatosis Tumor Cells Differentially Influence Sensory Neuron Gene Expression and Sensitivity. Sci Rep 2019; 9:13098. [PMID: 31511601 PMCID: PMC6739480 DOI: 10.1038/s41598-019-49705-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/27/2019] [Indexed: 01/08/2023] Open
Abstract
Schwannomatosis is a multiple tumor syndrome in which patients develop benign tumors along peripheral nerves throughout the body. The first symptom with which schwannomatosis patients often present, prior to discovery of tumors, is pain. This pain can be debilitating and is often inadequately alleviated by pharmacological approaches. Schwannomatosis-associated pain can be localized to the area of a tumor, or widespread. Moreover, not all tumors are painful, and the occurrence of pain is often unrelated to tumor size or location. We speculate that some individual tumors, but not others, secrete factors that act on nearby nerves to augment nociception by producing neuronal sensitization or spontaneous neuronal firing. We created cell lines from human SWN tumors with varying degrees of pain. We have found that conditioned medium (CM) collected from painful SWN tumors, but not that from nonpainful SWN tumors, sensitized DRG neurons, causing increased sensitivity to depolarization by KCl, increased response to noxious TRPV1 and TRPA1 agonists and also upregulated the expression of pain-associated genes in DRG cultures. Multiple cytokines were also detected at higher levels in CM from painful tumors. Taken together our data demonstrate a differential ability of painful versus non-painful human schwannomatosis tumor cells to secrete factors that augment sensory neuron responsiveness, and thus identify a potential determinant of pain heterogeneity in schwannomatosis.
Collapse
Affiliation(s)
- Kimberly Laskie Ostrow
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Neurosurgery Pain Research Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Katelyn J Donaldson
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Michael J Caterina
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Neurosurgery Pain Research Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Allan Belzberg
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Neurosurgery Pain Research Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ahmet Hoke
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
19
|
Csekő K, Beckers B, Keszthelyi D, Helyes Z. Role of TRPV1 and TRPA1 Ion Channels in Inflammatory Bowel Diseases: Potential Therapeutic Targets? Pharmaceuticals (Basel) 2019; 12:E48. [PMID: 30935063 PMCID: PMC6630403 DOI: 10.3390/ph12020048] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
Inflammatory bowel diseases (IBD) have long been recognized to be accompanied by pain resulting in high morbidity. Transient receptor potential vanilloid 1 (TRPV1) and ankyrin 1 (TRPA1) ion channels located predominantly on the capsaicin-sensitive sensory neurons play a complex role in hyperalgesia and neurogenic inflammation. This review provides an overview of their expression and role in intestinal inflammation, in particular colitis, that appears to be virtually inconsistent based on the thorough investigations of the last twenty years. However, preclinical results with pharmacological interventions, as well as scarcely available human studies, more convincingly point out the potential therapeutic value of TRPV1 and TRPA1 antagonists in colitis and visceral hypersensitivity providing future therapeutical perspectives through a complex, unique mechanism of action for drug development in IBD.
Collapse
Affiliation(s)
- Kata Csekő
- Department of Pharmacology and Pharmacotherapy, Medical School and Molecular Pharmacology Research Group, Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary.
| | - Bram Beckers
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands.
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University, 6202 AZ Maastricht, The Netherlands.
| | - Daniel Keszthelyi
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center (MUMC+), 6202 AZ Maastricht, The Netherlands.
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University, 6202 AZ Maastricht, The Netherlands.
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School and Molecular Pharmacology Research Group, Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary.
- PharmInVivo Ltd., H-7629 Pécs, Hungary.
| |
Collapse
|
20
|
Long-Term Diabetic Microenvironment Augments the Decay Rate of Capsaicin-Induced Currents in Mouse Dorsal Root Ganglion Neurons. Molecules 2019; 24:775. [PMID: 30795543 PMCID: PMC6412516 DOI: 10.3390/molecules24040775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/16/2019] [Accepted: 02/19/2019] [Indexed: 12/04/2022] Open
Abstract
Individuals with end-stage diabetic peripheral neuropathy present with decreased pain sensation. Transient receptor potential vanilloid type 1 (TRPV1) is implicated in pain signaling and resides on sensory dorsal root ganglion (DRG) neurons. We investigated the expression and functional activity of TRPV1 in DRG neurons of the Ins2+/Akita mouse at 9 months of diabetes using immunohistochemistry, live single cell calcium imaging, and whole-cell patch-clamp electrophysiology. 2′,7′-Dichlorodihydrofluorescein diacetate (DCFH-DA) fluorescence assay was used to determine the level of Reactive Oxygen Species (ROS) in DRGs. Although TRPV1 expressing neuron percentage was increased in Ins2+/Akita DRGs at 9 months of diabetes compared to control, capsaicin-induced Ca2+ influx was smaller in isolated Ins2+/Akita DRG neurons, indicating impaired TRPV1 function. Consistently, capsaicin-induced Ca2+ influx was decreased in control DRG neurons cultured in the presence of 25 mM glucose for seven days versus those cultured with 5.5 mM glucose. The high glucose environment increased cytoplasmic ROS accumulation in cultured DRG neurons. Patch-clamp recordings revealed that capsaicin-activated currents decayed faster in isolated Ins2+/Akita DRG neurons as compared to those in control neurons. We propose that in poorly controlled diabetes, the accelerated rate of capsaicin-sensitive TRPV1 current decay in DRG neurons decreases overall TRPV1 activity and contributes to peripheral neuropathy.
Collapse
|
21
|
Abstract
The transient receptor potential ankyrin 1 (TRPA1) ion channel is expressed in pain-sensing neurons and other tissues and has become a major target in the development of novel pharmaceuticals. A remarkable feature of the channel is its long list of activators, many of which we are exposed to in daily life. Many of these agonists induce pain and inflammation, making TRPA1 a major target for anti-inflammatory and analgesic therapies. Studies in human patients and in experimental animals have confirmed an important role for TRPA1 in a number of pain conditions. Over the recent years, much progress has been made in elucidating the molecular structure of TRPA1 and in discovering binding sites and modulatory sites of the channel. Because the list of published mutations and important molecular sites is steadily growing and because it has become difficult to see the forest for the trees, this review aims at summarizing the current knowledge about TRPA1, with a special focus on the molecular structure and the known binding or gating sites of the channel.
Collapse
Affiliation(s)
- Jannis E Meents
- Institute of Physiology, University Hospital RWTH Aachen , Aachen , Germany
| | - Cosmin I Ciotu
- Center for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Michael J M Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
22
|
Characterization of sensory neuronal subtypes innervating mouse tongue. PLoS One 2018; 13:e0207069. [PMID: 30408082 PMCID: PMC6224080 DOI: 10.1371/journal.pone.0207069] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/24/2018] [Indexed: 12/14/2022] Open
Abstract
The tongue is uniquely exposed to water-soluble environmental chemicals that may lead to injury or tumorigenesis. However, comparatively little research has focused on the molecular and functional organization of trigeminal ganglia (TG) afferent neurons innervating the tongue. The current study identified and characterized lingual sensory neurons based on a neuronal subtype classification previously characterized in the dorsal root ganglion (DRG) neurons. We employed immunohistochemistry on transgenic reporter mouse lines as well as single-cell PCR of known markers of neuronal subtypes to characterize neuronal subtypes innervating the tongue. Markers expressed in retrogradely labeled TG neurons were evaluated for the proportion of neurons expressing each marker, intensity of expression, and overlapping genes. We found that tongue-innervating sensory neurons primarily expressed CGRP, TRPV1, TrkC, 5HT3A and Parvalbumin. These markers correspond to peptidergic and a subgroup of non-peptidergic C-nociceptors, peptidergic A nociceptors, proprioceptors and myelinated low-threshold mechanoreceptors (LTMRs). Interestingly, as reported previously, we also found several differences between TG and DRG neurons indicating the need for single-cell sequencing of neuronal types based on tissue type within all TG as well as DRG neurons.
Collapse
|
23
|
Galindo T, Reyna J, Weyer A. Evidence for Transient Receptor Potential (TRP) Channel Contribution to Arthritis Pain and Pathogenesis. Pharmaceuticals (Basel) 2018; 11:E105. [PMID: 30326593 PMCID: PMC6315622 DOI: 10.3390/ph11040105] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023] Open
Abstract
Based on clinical and preclinical evidence, Transient Receptor Potential (TRP) channels have emerged as potential drug targets for the treatment of osteoarthritis, rheumatoid arthritis, and gout. This review summarizes the relevant data supporting a role for various TRP channels in arthritis pain and pathogenesis, as well as the current state of pharmacological efforts to ameliorate arthritis symptoms in patient populations.
Collapse
Affiliation(s)
- Tabitha Galindo
- School of Physical Therapy and Athletic Training, Pacific University, Hillsboro, OR 97116, USA.
| | - Jose Reyna
- School of Physical Therapy and Athletic Training, Pacific University, Hillsboro, OR 97116, USA.
| | - Andy Weyer
- Biological Sciences Department, City College of San Francisco, San Francisco, CA 94112, USA.
| |
Collapse
|
24
|
Xia Y, Xia L, Lou L, Jin R, Shen H, Li W. Transient Receptor Potential Channels and Chronic Airway Inflammatory Diseases: A Comprehensive Review. Lung 2018; 196:505-516. [PMID: 30094794 DOI: 10.1007/s00408-018-0145-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/04/2018] [Indexed: 12/22/2022]
Abstract
Chronic airway inflammatory diseases remain a major problem worldwide, such that there is a need for additional therapeutic targets and novel drugs. Transient receptor potential (TRP) channels are a group of non-selective cation channels expressed throughout the body that are regulated by various stimuli. TRP channels have been identified in numerous cell types in the respiratory tract, including sensory neurons, airway epithelial cells, airway smooth muscle cells, and fibroblasts. Different types of TRP channels induce cough in sensory neurons via the vagus nerve. Permeability and cytokine production are also regulated by TRP channels in airway epithelial cells, and these channels also contribute to the modulation of bronchoconstriction. TRP channels may cooperate with other TRP channels, or act in concert with calcium-dependent potassium channels and calcium-activated chloride channel. Hence, TRP channels could be the potential therapeutic targets for chronic airway inflammatory diseases. In this review, we aim to discuss the expression profiles and physiological functions of TRP channels in the airway, and the roles they play in chronic airway inflammatory diseases.
Collapse
Affiliation(s)
- Yang Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| | - Lexin Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Lingyun Lou
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Rui Jin
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
25
|
Wang S, Brigoli B, Lim J, Karley A, Chung MK. Roles of TRPV1 and TRPA1 in Spontaneous Pain from Inflamed Masseter Muscle. Neuroscience 2018; 384:290-299. [PMID: 29890293 DOI: 10.1016/j.neuroscience.2018.05.048] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 01/26/2023]
Abstract
Craniofacial muscle pain, such as spontaneous pain and bite-evoked pain, are major symptoms in patients with temporomandibular disorders and infection. However, the underlying mechanisms of muscle pain, especially mechanisms of highly prevalent spontaneous pain, are poorly understood. Recently, we reported that transient receptor potential vanilloid 1 (TRPV1) contributes to spontaneous pain but only marginally contributes to bite-evoked pain during masseter inflammation. Here, we investigated the role of transient receptor potential ankyrin 1 (TRPA1) in spontaneous and bite-evoked pain during masseter inflammation, and dissected the relative contributions of TRPA1 and TRPV1. Masseter inflammation increased mouse grimace scale (MGS) scores and face wiping behaviors. Pharmacological or genetic inhibition of TRPA1 significantly attenuated MGS but not face wiping behaviors. MGS scores were also attenuated by scavenging putative endogenous ligands for TRPV1 or TRPA1. Simultaneous inhibition of TRPA1 by AP18 and TRPV1 by AMG9810 in masseter muscle resulted in robust inhibition of both MGS and face wiping behaviors. Administration of AP18 or AMG9810 to masseter muscle induced conditioned place preference (CPP). The extent of CPP following simultaneous administration of AP18 and AMG9810 was greater than that induced by the individual antagonists. In contrast, inflammation-induced reduction of bite force was not affected by the inhibition of TRPA1 alone or in combination with TRPV1. These results suggest that simultaneous inhibition of TRPV1 and TRPA1 produces additive relief of spontaneous pain, but does not ameliorate bite-evoked pain during masseter inflammation. Our results provide further evidence that distinct mechanisms underlie spontaneous and bite-evoked pain from inflamed masseter muscle.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Benjamin Brigoli
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Jongseuk Lim
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Alisha Karley
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA.
| |
Collapse
|
26
|
Patil MJ, Hovhannisyan AH, Akopian AN. Characteristics of sensory neuronal groups in CGRP-cre-ER reporter mice: Comparison to Nav1.8-cre, TRPV1-cre and TRPV1-GFP mouse lines. PLoS One 2018; 13:e0198601. [PMID: 29864146 PMCID: PMC5986144 DOI: 10.1371/journal.pone.0198601] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/22/2018] [Indexed: 12/19/2022] Open
Abstract
Peptidergic sensory neurons play a critical role in nociceptive pathways. To precisely define the function and plasticity of sensory neurons in detail, new tools such as transgenic mouse models are needed. We employed electrophysiology and immunohistochemistry to characterize in detail dorsal root ganglion (DRG) neurons expressing an inducible CGRPcre-ER (CGRP-cre+); and compared them to DRG neurons expressing Nav1.8cre (Nav1.8-cre+), TRPV1cre (TRPV1-cre+) and TRPV1-GFP (V1-GFP+). Tamoxifen effectively induced CGRPcre-ER production in DRG. ≈87% of CGRPcre-ER-expressing neurons were co-labeled CGRP antibody. Three small and two medium-large-sized (5HT3a+/NPY2R- and NPY2R+) neuronal groups with unique electrophysiological profiles were CGRP-cre+. Nav1.8-cre+ neurons were detected in all CGRP-cre+ groups, as well as in 5 additional neuronal groups: MrgprD+/TRPA1-, MrgprD+/TRPA1+, TRPV1+/CGRP-, vGLUT3+ and ≈30% of trkC+ neurons. Differences between TRPV1cre and Nav1.8cre reporters were that unlike TRPV1-cre+, Nav1.8-cre+ expression was detected in non-nociceptive vGLUT3+ and trkC+ populations. Many TRPV1-cre+ neurons did not respond to capsaicin. In contrast, V1-GFP+ neurons were in 4 groups, each of which was capsaicin-sensitive. Finally, none of the analyzed reporter lines showed cre-recombination in trkB+, calbindin+, 70% of trkC+ or parvalbumin+ neurons, which together encompassed ≈20% of Nav1.8-cre- DRG neurons. The data presented here increases our knowledge of peptidergic sensory neuron characteristics, while showing the efficiency and specificity manipulation of peptidergic neurons by the CGRPcre-ER reporter. We also demonstrate that manipulation of all C- and A-nociceptors is better achieved with TRPV1-cre reporter. Finally, the described approach for detailed characterization of sensory neuronal groups can be applied to a variety of reporter mice.
Collapse
Affiliation(s)
- Mayur J. Patil
- Departments of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Anahit H. Hovhannisyan
- Departments of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Armen N. Akopian
- Departments of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Departments of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
27
|
Beckel JM, de Groat WC. The effect of the electrophilic fatty acid nitro-oleic acid on TRP channel function in sensory neurons. Nitric Oxide 2018; 78:S1089-8603(17)30289-6. [PMID: 29578059 PMCID: PMC6151181 DOI: 10.1016/j.niox.2018.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 01/28/2023]
Abstract
Nitro-oleic acid (NO2-OA) and related nitroalkenes are electrophilic fatty acid derivatives that are present in normal tissues at nanomolar concentrations and can increase significantly during inflammation. These substances can suppress multiple intracellular signaling pathways contributing to inflammation by reversible Michael addition reactions with nucleophilic residues such as cysteine and histidine leading to post-translational modification of proteins. NO2-OA also can influence inflammation and pain by acting on transient receptor potential (TRP) channels in primary sensory neurons. TRPV1, TRPA1 and TRPC can respond to electrophilic fatty acids because they have ankyrin-like repeats in their N terminus that are rich in cysteine residues that react with electrophiles and other thiol modifying species. NO2-OA acts on TRP channels to initially depolarize and induce firing in sensory neurons followed by desensitization and suppression of firing. In vivo experiments revealed that pretreatment with NO2-OA reduces nociceptive behavior evoked by local administration of a TRPA1 agonist (AITC) to the rat hind paw. These results raise the possibility that NO2-OA might be useful clinically to reduce neurogenic inflammation and certain types of painful sensations by desensitizing TRPA1 expressing nociceptive afferents.
Collapse
Affiliation(s)
- Jonathan M Beckel
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | - William C de Groat
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
28
|
Thom G, Burrell M, Haqqani AS, Yogi A, Lessard E, Brunette E, Delaney C, Baumann E, Callaghan D, Rodrigo N, Webster CI, Stanimirovic DB. Enhanced Delivery of Galanin Conjugates to the Brain through Bioengineering of the Anti-Transferrin Receptor Antibody OX26. Mol Pharm 2018; 15:1420-1431. [PMID: 29485883 DOI: 10.1021/acs.molpharmaceut.7b00937] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) is a formidable obstacle for brain delivery of therapeutic antibodies. However, antibodies against the transferrin receptor (TfR), enriched in brain endothelial cells, have been developed as delivery carriers of therapeutic cargoes into the brain via a receptor-mediated transcytosis pathway. In vitro and in vivo studies demonstrated that either a low-affinity or monovalent binding of these antibodies to the TfR improves their release on the abluminal side of the BBB and target engagement in brain parenchyma. However, these studies have been performed with mouse-selective TfR antibodies that recognize different TfR epitopes and have varied binding characteristics. In this study, we evaluated serum pharmacokinetics and brain and CSF exposure of the rat TfR-binding antibody OX26 affinity variants, having KDs of 5 nM, 76 nM, 108 nM, and 174 nM, all binding the same epitope in bivalent format. Pharmacodynamic responses were tested in the Hargreaves chronic pain model after conjugation of OX26 affinity variants with the analgesic and antiepileptic peptide, galanin. OX26 variants with affinities of 76 nM and 108 nM showed enhanced brain and cerebrospinal fluid (CSF) exposure and higher potency in the Hargreaves model, compared to a 5 nM affinity variant; lowering affinity to 174 nM resulted in prolonged serum pharmacokinetics, but reduced brain and CSF exposure. The study demonstrates that binding affinity optimization of TfR-binding antibodies could improve their brain and CSF exposure even in the absence of monovalent TfR engagement.
Collapse
Affiliation(s)
- George Thom
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Matthew Burrell
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Arsalan S Haqqani
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Alvaro Yogi
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Etienne Lessard
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Eric Brunette
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Christie Delaney
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Ewa Baumann
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Deborah Callaghan
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| | - Natalia Rodrigo
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Carl I Webster
- Antibody Discovery and Protein Engineering , MedImmune , Milstein Building, Granta Park, Cambridge CB21 6GH , U.K
| | - Danica B Stanimirovic
- Human Health Therapeutics Portfolio , National Research Council of Canada , Ottawa , Ontario K1A0R6 , Canada
| |
Collapse
|
29
|
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: 2.3] [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
|
30
|
Kichko TI, Neuhuber W, Kobal G, Reeh PW. The roles of TRPV1, TRPA1 and TRPM8 channels in chemical and thermal sensitivity of the mouse oral mucosa. Eur J Neurosci 2018; 47:201-210. [PMID: 29247491 DOI: 10.1111/ejn.13799] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 12/24/2022]
Abstract
Spices in food and beverages and compounds in tobacco smoke interact with sensory irritant receptors of the transient receptor potential (TRP) cation channel family. TRPV1 (vanilloid type 1), TRPA1 (ankyrin 1) and TRPM8 (melastatin 8) not only elicit action potential signaling through trigeminal nerves, eventually evoking pungent or cooling sensations, but by their calcium conductance they also stimulate the release of calcitonin gene-related peptide (CGRP). This is measured as an index of neuronal activation to elucidate the chemo- and thermosensory transduction in the isolated mouse buccal mucosa of wild types and pertinent knockouts. We found that the lipophilic capsaicin, mustard oil and menthol effectively get access to the nerve endings below the multilayered squamous epithelium, while cigarette smoke and its gaseous phase were weakly effective releasing CGRP. The hydrophilic nicotine was ineffective unless applied unprotonated in alkaline (pH9) solution, activating TRPA1 and TRPV1. Also, mustard oil activated both these irritant receptors in millimolar but only TRPA1 in micromolar concentrations; in combination (1 mm) with heat (45 °C), it showed supraadditive, that is heat sensitizing, effects in TRPV1 and TRPA1 knockouts, suggesting action on an unknown heat-activated channel and mustard oil receptor. Menthol caused little CGRP release by itself, but in subliminal concentration (2 mm), it enabled a robust cold response that was absent in TRPM8-/- but retained in TRPA1-/- and strongly reduced by TRPM8 inhibitors. In conclusion, all three relevant irritant receptors are functionally expressed in the oral mucosa and play their specific roles in inducing neurogenic inflammation and sensitization to heat and cold.
Collapse
Affiliation(s)
- Tatjana I Kichko
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstrasse 17, Erlangen, 91056, Germany
| | - Winfried Neuhuber
- Institute of Anatomy I, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gerd Kobal
- Altria Client Services Inc., Richmond, VA, USA
| | - Peter W Reeh
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstrasse 17, Erlangen, 91056, Germany
| |
Collapse
|
31
|
Boiko N, Medrano G, Montano E, Jiang N, Williams CR, Madungwe NB, Bopassa JC, Kim CC, Parrish JZ, Hargreaves KM, Stockand JD, Eaton BA. TrpA1 activation in peripheral sensory neurons underlies the ionic basis of pain hypersensitivity in response to vinca alkaloids. PLoS One 2017; 12:e0186888. [PMID: 29084244 PMCID: PMC5662086 DOI: 10.1371/journal.pone.0186888] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 10/09/2017] [Indexed: 12/30/2022] Open
Abstract
Chemotherapy induced peripheral neuropathy (CIPN), a side effect of many anti-cancer drugs including the vinca alkaloids, is characterized by a severe pain syndrome that compromises treatment in many patients. Currently there are no effective treatments for this pain syndrome except for the reduction of anti-cancer drug dose. Existing data supports the model that the pain associated with CIPN is the result of anti-cancer drugs augmenting the function of the peripheral sensory nociceptors but the cellular mechanisms underlying the effects of anti-cancer drugs on sensory neuron function are not well described. Studies from animal models have suggested a number of disease etiologies including mitotoxicity, axonal degeneration, immune signaling, and reduced sensory innervations but these outcomes are the result of prolonged treatment paradigms and do not necessarily represent the early formative events associated with CIPN. Here we show that acute exposure to vinca alkaloids results in an immediate pain syndrome in both flies and mice. Furthermore, we demonstrate that exposure of isolated sensory neurons to vinca alkaloids results in the generation of an inward sodium current capable of depolarizing these neurons to threshold resulting in neuronal firing. These neuronal effects of vinca alkaloids require the transient receptor potential ankyrin-1 (TrpA1) channel, and the hypersensitization to painful stimuli in response to the acute exposure to vinca alkaloids is reduced in TrpA1 mutant flies and mice. These findings demonstrate the direct excitation of sensory neurons by CIPN-causing chemotherapy drugs, and identify TrpA1 as an important target during the pathogenesis of CIPN.
Collapse
Affiliation(s)
- Nina Boiko
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| | - Geraldo Medrano
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| | - Elizabeth Montano
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| | - Nan Jiang
- Department of Biology, University of Washington, Seattle, Washington, United States of America
| | - Claire R. Williams
- Department of Biology, University of Washington, Seattle, Washington, United States of America
| | - Ngonidzashe B. Madungwe
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| | - Jean C. Bopassa
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| | - Charles C. Kim
- Verily, South San Francisco, California, United States of America
| | - Jay Z. Parrish
- Department of Biology, University of Washington, Seattle, Washington, United States of America
| | - Kenneth M. Hargreaves
- Department of Endodontics, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| | - James D. Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| | - Benjamin A. Eaton
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
32
|
Benemei S, De Logu F, Li Puma S, Marone IM, Coppi E, Ugolini F, Liedtke W, Pollastro F, Appendino G, Geppetti P, Materazzi S, Nassini R. The anti-migraine component of butterbur extracts, isopetasin, desensitizes peptidergic nociceptors by acting on TRPA1 cation channel. Br J Pharmacol 2017. [PMID: 28622417 DOI: 10.1111/bph.13917] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE The mechanism of the anti-migraine action of extracts of butterbur [Petasites hybridus (L.) Gaertn.] is unknown. Here, we investigated the ability of isopetasin, a major constituent of these extracts, to specifically target TRPA1 channel and to affect functional responses relevant to migraine. EXPERIMENTAL APPROACH Single-cell calcium imaging and patch-clamp recordings in human and rodent TRPA1-expressing cells, neurogenic motor responses in rodent isolated urinary bladder, release of CGRP from mouse spinal cord in vitro and facial rubbing in mice and meningeal blood flow in rats were examined. KEY RESULTS Isopetasin induced (i) calcium responses and currents in rat/mouse trigeminal ganglion (TG) neurons and in cells expressing the human TRPA1, (ii) substance P-mediated contractions of rat isolated urinary bladders and (iii) CGRP release from mouse dorsal spinal cord, responses that were selectively abolished by genetic deletion or pharmacological antagonism of TRPA1 channels. Pre-exposure to isopetasin produced marked desensitization of allyl isothiocyanate (AITC, TRPA1 channel agonist)- or capsaicin (TRPV1 channel agonist)-evoked currents in rat TG neurons, contractions of rat or mouse bladder and CGRP release from mouse central terminals of primary sensory neurons. Repeated intragastric administration of isopetasin attenuated mouse facial rubbing, evoked by local AITC or capsaicin, and dilation of rat meningeal arteries by acrolein or ethanol (TRPA1 and TRPV1 channel agonists respectively). CONCLUSION AND IMPLICATIONS Activation of TRPA1 channels by isopetasin results in excitation of neuropeptide-containing nociceptors, followed by marked heterologous neuronal desensitization. Such atten uation in pain and neurogenic inflammation may account for the anti-migraine action of butterbur.
Collapse
Affiliation(s)
- Silvia Benemei
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Simone Li Puma
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Ilaria Maddalena Marone
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Elisabetta Coppi
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Wolfgang Liedtke
- Departments of Neurology, Anesthesiology and Neurobiology, Clinics for Headache, Head-Pain and Trigeminal Sensory Disorders, Duke University, Durham, NC, USA
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Novara, Italy
| | - Giovanni Appendino
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Novara, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Serena Materazzi
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| |
Collapse
|
33
|
Oehler B, Kistner K, Martin C, Schiller J, Mayer R, Mohammadi M, Sauer RS, Filipovic MR, Nieto FR, Kloka J, Pflücke D, Hill K, Schaefer M, Malcangio M, Reeh PW, Brack A, Blum R, Rittner HL. Inflammatory pain control by blocking oxidized phospholipid-mediated TRP channel activation. Sci Rep 2017; 7:5447. [PMID: 28710476 PMCID: PMC5511297 DOI: 10.1038/s41598-017-05348-3] [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: 02/10/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022] Open
Abstract
Phospholipids occurring in cell membranes and lipoproteins are converted into oxidized phospholipids (OxPL) by oxidative stress promoting atherosclerotic plaque formation. Here, OxPL were characterized as novel targets in acute and chronic inflammatory pain. Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (OxPAPC) and its derivatives were identified in inflamed tissue by mass spectrometry and binding assays. They elicited calcium influx, hyperalgesia and induced pro-nociceptive peptide release. Genetic, pharmacological and mass spectrometric evidence in vivo as well as in vitro confirmed the role of transient receptor potential channels (TRPA1 and TRPV1) as OxPAPC targets. Treatment with the monoclonal antibody E06 or with apolipoprotein A-I mimetic peptide D-4F, capturing OxPAPC in atherosclerosis, prevented inflammatory hyperalgesia, and in vitro TRPA1 activation. Administration of D-4F or E06 to rats profoundly ameliorated mechanical hyperalgesia and inflammation in collagen-induced arthritis. These data reveal a clinically relevant role for OxPAPC in inflammation offering therapy for acute and chronic inflammatory pain treatment by scavenging OxPAPC.
Collapse
Affiliation(s)
- Beatrice Oehler
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Katrin Kistner
- Institute for Physiology and Pathophysiology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Corinna Martin
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Jürgen Schiller
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Rafaela Mayer
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Milad Mohammadi
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Reine-Solange Sauer
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Milos R Filipovic
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany.,University of Bordeaux, IBGC, UMR 5095, Bordeaux, France
| | - Francisco R Nieto
- Wolfson CARD, King's College London, Guys' Campus, London, United Kingdom.,University of Granada, Department of Pharmacology, Granada, Spain
| | - Jan Kloka
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Diana Pflücke
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Kerstin Hill
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Michael Schaefer
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Marzia Malcangio
- Wolfson CARD, King's College London, Guys' Campus, London, United Kingdom
| | - Peter W Reeh
- Institute for Physiology and Pathophysiology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexander Brack
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Heike L Rittner
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
34
|
Actions and Regulation of Ionotropic Cannabinoid Receptors. ADVANCES IN PHARMACOLOGY 2017; 80:249-289. [PMID: 28826537 DOI: 10.1016/bs.apha.2017.04.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Almost three decades have passed since the identification of the two specific metabotropic receptors mediating cannabinoid pharmacology. Thereafter, many cannabinoid effects, both at central and peripheral levels, have been well documented and characterized. However, numerous evidences demonstrated that these pharmacological actions could not be attributable solely to the activation of CB1 and CB2 receptors since several important cannabimimetic actions have been found in biological systems lacking CB1 or CB2 gene such as in specific cell lines or transgenic mice. It is now well accepted that, beyond their receptor-mediated effects, these molecules can act also via CB1/CB2-receptor-independent mechanism. Cannabinoids have been demonstrated to modulate several voltage-gated channels (including Ca2+, Na+, and various type of K+ channels), ligand-gated ion channels (i.e., GABA, glycine), and ion-transporting membranes proteins such as transient potential receptor class (TRP) channels. The first direct, cannabinoid receptor-independent interaction was reported on the function of serotonin 5-HT3 receptor-ion channel complex. Similar effects were reported also on the other above mentioned ion channels. In the early ninety, studies searching for endogenous modulators of L-type Ca2+ channels identified anandamide as ligand for L-type Ca2+ channel. Later investigations indicated that other types of Ca2+ currents are also affected by endocannabinoids, and, in the late ninety, it was discovered that endocannabinoids activate the vanilloid receptor subtype 1 (TRPV1), and nowadays, it is known that (endo)cannabinoids gate at least five distinct TRP channels. This chapter focuses on cannabinoid regulation of ion channels and lays special emphasis on their action at transient receptor channels.
Collapse
|
35
|
Jardín I, López JJ, Diez R, Sánchez-Collado J, Cantonero C, Albarrán L, Woodard GE, Redondo PC, Salido GM, Smani T, Rosado JA. TRPs in Pain Sensation. Front Physiol 2017. [PMID: 28649203 PMCID: PMC5465271 DOI: 10.3389/fphys.2017.00392] [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] [Indexed: 12/11/2022] Open
Abstract
According to the International Association for the Study of Pain (IASP) pain is characterized as an "unpleasant sensory and emotional experience associated with actual or potential tissue damage". The TRP super-family, compressing up to 28 isoforms in mammals, mediates a myriad of physiological and pathophysiological processes, pain among them. TRP channel might be constituted by similar or different TRP subunits, which will result in the formation of homomeric or heteromeric channels with distinct properties and functions. In this review we will discuss about the function of TRPs in pain, focusing on TRP channles that participate in the transduction of noxious sensation, especially TRPV1 and TRPA1, their expression in nociceptors and their sensitivity to a large number of physical and chemical stimuli.
Collapse
Affiliation(s)
- Isaac Jardín
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - José J López
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Raquel Diez
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - José Sánchez-Collado
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Carlos Cantonero
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Letizia Albarrán
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health SciencesBethesda, MD, United States
| | - Pedro C Redondo
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Ginés M Salido
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Sevilla, University of SevilleSevilla, Spain
| | - Juan A Rosado
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| |
Collapse
|
36
|
Logashina YA, Mosharova IV, Korolkova YV, Shelukhina IV, Dyachenko IA, Palikov VA, Palikova YA, Murashev AN, Kozlov SA, Stensvåg K, Andreev YA. Peptide from Sea Anemone Metridium senile Affects Transient Receptor Potential Ankyrin-repeat 1 (TRPA1) Function and Produces Analgesic Effect. J Biol Chem 2017; 292:2992-3004. [PMID: 28077580 DOI: 10.1074/jbc.m116.757369] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/03/2017] [Indexed: 11/06/2022] Open
Abstract
The transient receptor potential ankyrin-repeat 1 (TRPA1) is an important player in pain and inflammatory pathways. It is a promising target for novel drug development for the treatment of a number of pathological states. A novel peptide producing a significant potentiating effect on allyl isothiocyanate- and diclofenac-induced currents of TRPA1 was isolated from the venom of sea anemone Metridium senile. It is a 35-amino acid peptide cross-linked by two disulfide bridges named τ-AnmTX Ms 9a-1 (short name Ms 9a-1) according to a structure similar to other sea anemone peptides belonging to structural group 9a. The structures of the two genes encoding the different precursor proteins of Ms 9a-1 were determined. Peptide Ms 9a-1 acted as a positive modulator of TRPA1 in vitro but did not cause pain or thermal hyperalgesia when injected into the hind paw of mice. Intravenous injection of Ms 9a-1 (0.3 mg/kg) produced a significant decrease in the nociceptive and inflammatory response to allyl isothiocyanate (the agonist of TRPA1) and reversed CFA (Complete Freund's Adjuvant)-induced inflammation and thermal hyperalgesia. Taken together these data support the hypothesis that Ms 9a-1 potentiates the response of TRPA1 to endogenous agonists followed by persistent functional loss of TRPA1-expressing neurons. We can conclude that TRPA1 potentiating may be useful as a therapeutic approach as Ms 9a-1 produces significant analgesic and anti-inflammatory effects in mice models of pain.
Collapse
Affiliation(s)
- Yulia A Logashina
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10,117997 Moscow, Russia.,the Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Trubetskaya St. 8, Bldg. 2, 119991 Moscow, Russia
| | - Irina V Mosharova
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10,117997 Moscow, Russia
| | - Yulia V Korolkova
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10,117997 Moscow, Russia
| | - Irina V Shelukhina
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10,117997 Moscow, Russia
| | - Igor A Dyachenko
- the Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290 Pushchino, Moscow, Russia, and
| | - Victor A Palikov
- the Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290 Pushchino, Moscow, Russia, and
| | - Yulia A Palikova
- the Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290 Pushchino, Moscow, Russia, and
| | - Arkadii N Murashev
- the Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290 Pushchino, Moscow, Russia, and
| | - Sergey A Kozlov
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10,117997 Moscow, Russia
| | - Klara Stensvåg
- the Norwegian College of Fishery Science, University of Tromsø, N9037 Tromsø, Norway
| | - Yaroslav A Andreev
- From the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10,117997 Moscow, Russia, .,the Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Trubetskaya St. 8, Bldg. 2, 119991 Moscow, Russia
| |
Collapse
|
37
|
Aghazadeh Tabrizi M, Baraldi PG, Baraldi S, Gessi S, Merighi S, Borea PA. Medicinal Chemistry, Pharmacology, and Clinical Implications of TRPV1 Receptor Antagonists. Med Res Rev 2016; 37:936-983. [PMID: 27976413 DOI: 10.1002/med.21427] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/28/2022]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is an ion channel expressed on sensory neurons triggering an influx of cations. TRPV1 receptors function as homotetramers responsive to heat, proinflammatory substances, lipoxygenase products, resiniferatoxin, endocannabinoids, protons, and peptide toxins. Its phosphorylation increases sensitivity to both chemical and thermal stimuli, while desensitization involves a calcium-dependent mechanism resulting in receptor dephosphorylation. TRPV1 functions as a sensor of noxious stimuli and may represent a target to avoid pain and injury. TRPV1 activation has been associated to chronic inflammatory pain and peripheral neuropathy. Its expression is also detected in nonneuronal areas such as bladder, lungs, and cochlea where TRPV1 activation is responsible for pathology development of cystitis, asthma, and hearing loss. This review offers a comprehensive overview about TRPV1 receptor in the pathophysiology of chronic pain, epilepsy, cough, bladder disorders, diabetes, obesity, and hearing loss, highlighting how drug development targeting this channel could have a clinical therapeutic potential. Furthermore, it summarizes the advances of medicinal chemistry research leading to the identification of highly selective TRPV1 antagonists and their analysis of structure-activity relationships (SARs) focusing on new strategies to target this channel.
Collapse
Affiliation(s)
- Mojgan Aghazadeh Tabrizi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Pier Giovanni Baraldi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Baraldi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Gessi
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Merighi
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Pier Andrea Borea
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| |
Collapse
|
38
|
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: 4.8] [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
|
39
|
De Logu F, Tonello R, Materazzi S, Nassini R, Fusi C, Coppi E, Li Puma S, Marone IM, Sadofsky LR, Morice AH, Susini T, Terreni A, Moneti G, Di Tommaso M, Geppetti P, Benemei S. TRPA1 Mediates Aromatase Inhibitor–Evoked Pain by the Aromatase Substrate Androstenedione. Cancer Res 2016; 76:7024-7035. [DOI: 10.1158/0008-5472.can-16-1492] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/13/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
|
40
|
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: 9.7] [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
|
41
|
Abstract
Sensory nerve endings within the airway epithelial cells and the solitary chemoreceptor cells, synapsing with sensory nerves, respond to airborne irritants. Transient receptor potential (TRP) channels (A1 and V1 subtypes, specifically) on these nerve endings initiate local antidromic reflexes resulting in the release of neuropeptides such as substance P and calcitonin G-related peptides. These neuropeptides dilate epithelial submucosal blood vessels and may therefore increase transudation across these vessels resulting in submucosal edema, congestion, and rhinitis. Altered expression or activity of these TRP channels can therefore influence responsiveness to irritants. Besides these pathogenic mechanisms, additional mechanisms such as dysautonomia resulting in diminished sympathetic activity and comparative parasympathetic overactivity have also been suggested as a probable mechanism. Therapeutic effectiveness for this condition has been demonstrated through desensitization of TRPV1 channels with typical agonists such as capsaicin. Other agents effective in treating nonallergic rhinitis (NAR) such as azelastine have been demonstrated to exhibit TRPV1 channel activity through the modulation of Ca(2+) signaling on sensory neurons and in nasal epithelial cells. Roles of antimuscarinic agents such as tiotropium in NAR have been suggested by associations of muscarinic cholinergic receptors with TRPV1. The associations between these channels have also been suggested as mechanisms of airway hyperreactivity in asthma. The concept of the united airway disease hypothesis suggests a significant association between rhinitis and asthma. This concept is supported by the development of late-onset asthma in about 10-40 % of NAR patients who also exhibit a greater severity in their asthma. The factors and mechanisms associating NAR with nonallergic asthma are currently unknown. Nonetheless, free immunoglobulin light chains and microRNA alteration as mediators of these inflammatory conditions may play key roles in this association.
Collapse
Affiliation(s)
- Jonathan A Bernstein
- Division of Immunology/Allergy Section, Department of Internal Medicine, University of Cincinnati College of Medicine, 3255 Eden Ave., ML#563 Suite 350, Cincinnati, OH, 45267, USA,
| | | |
Collapse
|
42
|
Kichko TI, Kobal G, Reeh PW. Cigarette smoke has sensory effects through nicotinic and TRPA1 but not TRPV1 receptors on the isolated mouse trachea and larynx. Am J Physiol Lung Cell Mol Physiol 2015; 309:L812-20. [PMID: 26472811 PMCID: PMC4609941 DOI: 10.1152/ajplung.00164.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/16/2015] [Indexed: 02/08/2023] Open
Abstract
Cigarette smoke (CS) exposes chemosensory nerves in the airways to a multitude of chemicals, some acting through the irritant receptors TRPV1 and TRPA1 but potentially also through nicotinic acetylcholine receptors (nAChR). Our aim was to characterize the differences in sensory neuronal effects of CS, gas phase, and particulate matter as well as of typical constituents, such as nicotine and reactive carbonyls. Isolated mouse trachea and larynx were employed to measure release of calcitonin gene-related peptide (CGRP) as an index of sensory neuron activation evoked by CS, by filtered CS gas phase essentially free of nicotine, and by dilute total particulate matter (TPM) containing defined nicotine concentrations. With CS stimulation of the superfused trachea, TRPV1 null mutants showed about the same large responses as wild-type mice, whereas both TRPA1(-/-) and double knockouts exhibited 80% reduction; the retained 20% response was abolished by mecamylamine (10 μM), indicating a distinct contribution of nAChRs. These phenotypes were accentuated by using TPM to stimulate the immersed trachea; 50% of response was retained in TRPA1(-/-) and abolished by mecamylamine. In contrast, the gas phase acted like a sheer TRPA1 agonist, consistent with its composition, among other compounds, of volatile reactive carbonyls like formaldehyde and acrolein. In the trachea, the gas phase and CS were equally effective in releasing CGRP, whereas the larynx showed much larger CS than gas phase responses. Thus nicotinic receptors contribute to the sensory effects of cigarette smoke on the trachea, which are dominated by TRPA1. How this translates to human perception affords future research.
Collapse
Affiliation(s)
- Tatjana I Kichko
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany; and
| | - Gerd Kobal
- Altria Client Services Inc., Richmond, Virginia
| | - Peter W Reeh
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany; and
| |
Collapse
|
43
|
Persistent Nociception Triggered by Nerve Growth Factor (NGF) Is Mediated by TRPV1 and Oxidative Mechanisms. J Neurosci 2015; 35:8593-603. [PMID: 26041925 DOI: 10.1523/jneurosci.3993-14.2015] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nerve growth factor (NGF) is elevated in certain chronic pain conditions and is a sufficient stimulus to cause lasting pain in humans, but the actual mechanisms underlying the persistent effects of NGF remain incompletely understood. We developed a rat model of NGF-induced persistent thermal hyperalgesia and mechanical allodynia to determine the role of transient receptor potential vanilloid 1 (TRPV1) and oxidative mechanisms in the persistent effects of NGF. Persistent thermal hypersensitivity and mechanical allodynia require de novo protein translation and are mediated by TRPV1 and oxidative mechanisms. By comparing effects after systemic (subcutaneous), spinal (intrathecal) or hindpaw (intraplantar) injections of test compounds, we determined that TRPV1 and oxidation mediate persistent thermal hypersensitivity via peripheral and spinal sites of action and mechanical allodynia via only a spinal site of action. Therefore, NGF-evoked thermal and mechanical allodynia are mediated by spatially distinct mechanisms. NGF treatment evoked sustained increases in peripheral and central TRPV1 activity, as demonstrated by increased capsaicin-evoked nocifensive responses, increased calcitonin gene-related peptide release from hindpaw skin biopsies, and increased capsaicin-evoked inward current and membrane expression of TRPV1 protein in dorsal root ganglia neurons. Finally, we showed that NGF treatment increased concentrations of linoleic and arachidonic-acid-derived oxidized TRPV1 agonists in spinal cord and skin biopsies. Furthermore, increases in oxidized TRPV1-active lipids were reduced by peripheral and spinal injections of compounds that completely blocked persistent nociception. Collectively, these data indicate that NGF evokes a persistent nociceptive state mediated by increased TRPV1 activity and oxidative mechanisms, including increased production of oxidized lipid TRPV1 agonists.
Collapse
|
44
|
Sinharoy P, Zhang H, Sinha S, Prudner BC, Bratz IN, Damron DS. Propofol restores TRPV1 sensitivity via a TRPA1-, nitric oxide synthase-dependent activation of PKCε. Pharmacol Res Perspect 2015; 3:e00153. [PMID: 26171233 PMCID: PMC4492729 DOI: 10.1002/prp2.153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/07/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022] Open
Abstract
We previously demonstrated that the intravenous anesthetic, propofol, restores the sensitivity of transient receptor potential vanilloid channel subtype-1 (TRPV1) receptors via a protein kinase C epsilon (PKCε)-dependent and transient receptor potential ankyrin channel subtype-1 (TRPA1)-dependent pathway in sensory neurons. The extent to which the two pathways are directly linked or operating in parallel has not been determined. Using a molecular approach, our objectives of the current study were to confirm that TRPA1 activation directly results in PKCε activation and to elucidate the cellular mechanism by which this occurs. F-11 cells were transfected with complimentary DNA (cDNA) for TRPV1 only or both TRPV1 and TRPA1. Intracellular Ca(2+) concentration was measured in individual cells via fluorescence microscopy. An immunoblot analysis of the total and phosphorylated forms of PKCε, nitric oxide synthase (nNOS), and TRPV1 was also performed. In F-11 cells containing both channels, PKCε inhibition prevented the propofol- and allyl isothiocyanate (AITC)-induced restoration of TRPV1 sensitivity to agonist stimulation as well as increased phosphorylation of PKCε and TRPV1. In cells containing TRPV1 only, neither agonist induced PKCε or TRPV1 phosphorylation. Moreover, NOS inhibition blocked propofol-and AITC-induced restoration of TRPV1 sensitivity and PKCε phosphorylation, and PKCε inhibition prevented the nitric oxide donor, SNAP, from restoring TRPV1 sensitivity. Also, propofol-and AITC-induced phosphorylation of nNOS and nitric oxide (NO) production were blocked with the TRPA1-antagonist, HC-030031. These data indicate that the AITC- and propofol-induced restoration of TRPV1 sensitivity is mediated by a TRPA1-dependent, nitric oxide synthase-dependent activation of PKCε.
Collapse
Affiliation(s)
- Pritam Sinharoy
- Department of Biological Sciences, Kent State University Kent, Ohio, 44242
| | | | - Sayantani Sinha
- Department of Biological Sciences, Kent State University Kent, Ohio, 44242
| | - Bethany C Prudner
- Department of Biological Sciences, Kent State University Kent, Ohio, 44242
| | - Ian N Bratz
- Department of Integrated Medical Sciences, Northeast Ohio Medical University Rootstown, Ohio
| | - Derek S Damron
- Department of Biological Sciences, Kent State University Kent, Ohio, 44242
| |
Collapse
|
45
|
Rouwette T, Avenali L, Sondermann J, Narayanan P, Gomez-Varela D, Schmidt M. Modulation of nociceptive ion channels and receptors via protein-protein interactions: implications for pain relief. Channels (Austin) 2015; 9:175-85. [PMID: 26039491 DOI: 10.1080/19336950.2015.1051270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In the last 2 decades biomedical research has provided great insights into the molecular signatures underlying painful conditions. However, chronic pain still imposes substantial challenges to researchers, clinicians and patients alike. Under pathological conditions, pain therapeutics often lack efficacy and exhibit only minimal safety profiles, which can be largely attributed to the targeting of molecules with key physiological functions throughout the body. In light of these difficulties, the identification of molecules and associated protein complexes specifically involved in chronic pain states is of paramount importance for designing selective interventions. Ion channels and receptors represent primary targets, as they critically shape nociceptive signaling from the periphery to the brain. Moreover, their function requires tight control, which is usually implemented by protein-protein interactions (PPIs). Indeed, manipulation of such PPIs entails the modulation of ion channel activity with widespread implications for influencing nociceptive signaling in a more specific way. In this review, we highlight recent advances in modulating ion channels and receptors via their PPI networks in the pursuit of relieving chronic pain. Moreover, we critically discuss the potential of targeting PPIs for developing novel pain therapies exhibiting higher efficacy and improved safety profiles.
Collapse
Affiliation(s)
- Tom Rouwette
- a Max Planck Institute for Experimental Medicine. Somatosensory Signaling Group ; Goettingen , Germany
| | | | | | | | | | | |
Collapse
|
46
|
Sinha S, Sinharoy P, Bratz IN, Damron DS. Propofol causes vasodilation in vivo via TRPA1 ion channels: role of nitric oxide and BKCa channels. PLoS One 2015; 10:e0122189. [PMID: 25830814 PMCID: PMC4382130 DOI: 10.1371/journal.pone.0122189] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 02/10/2015] [Indexed: 11/18/2022] Open
Abstract
Background Transient receptor potential (TRP) ion channels of the A1 (TRPA1) and V1 (TRPV1) subtypes are key regulators of vasomotor tone. Propofol is an intravenous anesthetic known to cause vasorelaxation. Our objectives were to examine the extent to which TRPA1 and/or TRPV1 ion channels mediate propofol-induced depressor responses in vivo and to delineate the signaling pathway(s) involved. Methods Mice were subjected to surgery under 1.5–2.5% sevoflurane gas with supplemental oxygen. After a stable baseline in mean arterial pressure (MAP) was achieved propofol (2.5, 5.0, 10.0 mg/kg/min) was administered to assess the hemodynamic actions of the intravenous anesthetic. The effect of nitric oxide synthase (NOS) inhibition with L-NAME and/or calcium-gated K+ channel (BKCa) inhibition with Penetrim A (Pen A), alone and in combination, on propofol-induced decreases in mean arterial pressure were assessed in control C57Bl/6J, TRPA1-/-, TRPV1-/- and double-knockout mice (TRPAV-/-). Results Propofol decreased MAP in control mice and this effect was markedly attenuated in TRPA1-/- and TRPAV-/- mice but unaffected in TRPV1-/-mice. Moreover, pretreatment with L-NAME or Pen A attenuated the decrease in MAP in control and TRPV1-/- mice, and combined inhibition abolished the depressor response. In contrast, the markedly attenuated propofol-induced depressor response observed in TRPA1-/- and TRPAV-/- mice was unaffected by pre-treatment with Pen A or L-NAME when used either alone or in combination. Conclusion These data demonstrate for the first time that propofol-induced depressor responses in vivo are predominantly mediated by TRPA1 ion channels with no involvement of TRPV1 ion channels and includes activation of both NOS and BKCa channels.
Collapse
Affiliation(s)
- Sayantani Sinha
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Pritam Sinharoy
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Ian N. Bratz
- Department of Integrative Medical Sciences, Northeast Ohio Medical College, Rootstown, Ohio, United States of America
| | - Derek S. Damron
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
- * E-mail:
| |
Collapse
|
47
|
Mihara S, Shibamoto T. The role of flavor and fragrance chemicals in TRPA1 (transient receptor potential cation channel, member A1) activity associated with allergies. Allergy Asthma Clin Immunol 2015; 11:11. [PMID: 25897313 PMCID: PMC4404258 DOI: 10.1186/s13223-015-0074-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/18/2015] [Indexed: 01/07/2023] Open
Abstract
TRPA1 has been proposed to be associated with diverse sensory allergic reactions, including thermal (cold) nociception, hearing and allergic inflammatory conditions. Some naturally occurring compounds are known to activate TRPA1 by forming a Michael addition product with a cysteine residue of TRPA1 through covalent protein modification and, in consequence, to cause allergic reactions. The anti-allergic property of TRPA1 agonists may be due to the activation and subsequent desensitization of TRPA1 expressed in sensory neurons. In this review, naturally occurring TRPA1 antagonists, such as camphor, 1,8-cineole, menthol, borneol, fenchyl alcohol and 2-methylisoborneol, and TRPA1 agonists, including thymol, carvacrol, 1'S-1'- acetoxychavicol acetate, cinnamaldehyde, α-n-hexyl cinnamic aldehyde and thymoquinone as well as isothiocyanates and sulfides are discussed.
Collapse
Affiliation(s)
- Satoru Mihara
- 2-10-12 Nishinippori, Arakawa-ku, Tokyo, 116-0013 Japan
| | - Takayuki Shibamoto
- Department of Environmental Toxicology, University of California Davis, Davis, CA 95616 USA
| |
Collapse
|
48
|
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: 142] [Impact Index Per Article: 14.2] [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
|
49
|
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
|
50
|
Abstract
Objectives:Resiniferatoxin, the most potent agonist of inflammatory pain/vanilloid receptor/cation channel (TRPV1) can be used for neuron subtype specific ablation of pain generating cells at the level of the peripheral nervous system by Ca2+-excytotoxicity. Molecular neurosurgery is an emerging technology either to alleviate severe pain in cancer or treat/prevent different local neuropathies. Our aim was determining sensory modalities that may be lost after resiniferatoxin treatment.Methods:Newborn or adult mice were treated with resiniferatoxin, then changes in chemical and heat sensitivity were correlated with alterations of the cell composition of sensory ganglions.Results:Only mice treated at adult age became less sensitive to heat stimuli, while both treatment groups lost sensitivity to specific vanilloid agonists of TRPV1 and, interestingly, to allyl-isothiocyanate, a selective agonist of TRPA1. Our in vivo and post mortem analytical results confirmed that TRPV1 and TRPA1 function together and resiniferatoxin-mediated neurosurgery removes both sensor moleculesDiscussion:In adult mice resiniferatoxin causes: i) desensitization to heat and ii) sensitization to cold. Cold hyperalgesia, an imbalance in thermosensation, might be conferred by a prominent cold receptor that is expressed in surviving resiniferatoxin-resistant sensory neurons and compensates for pain signals lost with TRPA1 and TRPV1 double positive cells in the peripheral nervous system.
Collapse
|