1
|
Liu TT, Chen PY, Tseng CY, Chen YN, Chen JB, Ni TH, Wang SJ, Chen SP, Yen JC. Activation of central and peripheral transient receptor potential melastatin 8 increases susceptibility to spreading depolarization and facilitates trigeminal neuroinflammation. J Headache Pain 2025; 26:55. [PMID: 40087597 PMCID: PMC11907788 DOI: 10.1186/s10194-025-01997-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/09/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Transient receptor potential melastatin 8 (TRPM8), a gene encoding a nonselective cation channel responsive to cold stimuli, has been implicated in migraine susceptibility. Despite this association, the role of TRPM8 to migraine pathogenesis remains elusive. This study aims to elucidate the potential role of TRPM8 in migraine pathophysiology. METHODS TRPM8 expression in the cortex and primary trigeminal ganglion (TG) cells was analyzed via immunostaining. The central role of TRPM8 was assessed using a spreading depolarization (SD) model, where intracerebroventricular injections or topical applications of TRPM8 agonists and antagonists were administered to rats to investigate their effects on KCl-evoked SD and SD-induced cortical inflammation. The peripheral role of TRPM8 in migraine was evaluated using primary cultures of rat TG cells by analyzing the effects of TRPM8 activation on calcitonin gene-related peptide (CGRP) expression, release, and trigeminal neuroinflammation. RESULTS TRPM8 was homogeneously distributed in the cerebral cortex, predominantly co-localizing with cortical neurons. Activation of cortical TRPM8 increased the frequency of KCl-evoked SD and exacerbated SD-induced cortical inflammation. Interestingly. Interestingly, inhibition of cerebral TRPM8 had negligible effects. In TG primary cultures, TRPM8 activation upregulated CGRP expression and release and induced cyclooxygenase-2 (Cox2) upregulation via a calmodulin kinase II (CaMKII)-dependent mechanism. CONCLUSIONS TRPM8 activation increased susceptibility to SD and facilitated the effects of CGRP and trigeminal neuroinflammation, implicating that TRPM8 may contribute to migraine pathophysiology through central and peripheral mechanisms.
Collapse
Affiliation(s)
- Tzu-Ting Liu
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Pin-Yu Chen
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chyun-Yea Tseng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Yun-Ning Chen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Jian-Bang Chen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Tz-Han Ni
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Shuu-Jiun Wang
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
| | - Shih-Pin Chen
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
- Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Jiin-Cherng Yen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan.
| |
Collapse
|
2
|
Lewis CM, Griffith TN. Ion channels of cold transduction and transmission. J Gen Physiol 2024; 156:e202313529. [PMID: 39051992 PMCID: PMC11273221 DOI: 10.1085/jgp.202313529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/04/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Thermosensation requires the activation of a unique collection of ion channels and receptors that work in concert to transmit thermal information. It is widely accepted that transient receptor potential melastatin 8 (TRPM8) activation is required for normal cold sensing; however, recent studies have illuminated major roles for other ion channels in this important somatic sensation. In addition to TRPM8, other TRP channels have been reported to contribute to cold transduction mechanisms in diverse sensory neuron populations, with both leak- and voltage-gated channels being identified for their role in the transmission of cold signals. Whether the same channels that contribute to physiological cold sensing also mediate noxious cold signaling remains unclear; however, recent work has found a conserved role for the kainite receptor, GluK2, in noxious cold sensing across species. Additionally, cold-sensing neurons likely engage in functional crosstalk with nociceptors to give rise to cold pain. This Review will provide an update on our understanding of the relationship between various ion channels in the transduction and transmission of cold and highlight areas where further investigation is required.
Collapse
Affiliation(s)
- Cheyanne M Lewis
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Theanne N Griffith
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
3
|
Stinson RJ, Morice AH, Ahmad B, Sadofsky LR. Ingredients of Vicks VapoRub inhibit rhinovirus-induced ATP release. Drugs Context 2023; 12:2023-3-2. [PMID: 37849655 PMCID: PMC10578958 DOI: 10.7573/dic.2023-3-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/28/2023] [Indexed: 10/19/2023] Open
Abstract
Background Over-the-counter therapies, such as Vicks VapoRub, are frequently used in the management of upper respiratory tract infection symptoms. Of these, acute cough is the most bothersome; however, the mechanisms involved have not been fully elucidated. The temperature-sensitive transient receptor potential (TRP) channels, including TRPA1, TRPV1, TRPM8 and TRPV4, are potential candidates. TRPV4 is also thought to be involved in cough through the TRPV4-ATP-P2X3 pathway. Here, we hypothesise that Vicks VapoRub ingredients (VVRIs) modulate the TRP cough channels. Methods Stably transfected HEK cells expressing TRP channels were challenged with VVRIs, individually or in combination, and the agonist and antagonist effects were measured using calcium signalling responses. In addition, rhinovirus serotype-16 (RV16)-infected A549 airway epithelial cells were pre-incubated with individual or combinations of VVRIs prior to hypotonic challenge and extracellular ATP release analysis. Results Calcium signalling reconfirmed some previously defined activation of TRP channels by specific VVRIs. The combined VVRIs containing menthol, camphor and eucalyptus oil activated TRPV1, TRPV4, TRPM8 and untransfected wild-type HEK293 cells. However, pre-incubation with VVRIs did not significantly inhibit any of the channels compared with the standard agonist responses. Pre-incubation of RV16-infected A549 cells with individual or combined VVRIs, except menthol, resulted in a 0.45-0.55-fold reduction in total ATP release following hypotonic stimulation, compared with infected cells not treated with VVRIs. Conclusion These findings suggest that some VVRIs may reduce symptoms associated with upper respiratory tract infection by modulating specific TRP receptors and by reducing RV16-induced ATP release.
Collapse
Affiliation(s)
- Rebecca J Stinson
- Centre for Biomedicine, Hull York Medical School, The University of Hull, Hull, UK
| | - Alyn H Morice
- Clinical Sciences Centre, Hull York Medical School, Castle Hill Hospital, Hull, UK
| | - Basir Ahmad
- Centre for Biomedicine, Hull York Medical School, The University of Hull, Hull, UK
| | - Laura R Sadofsky
- Centre for Biomedicine, Hull York Medical School, The University of Hull, Hull, UK
| |
Collapse
|
4
|
Yang C, Yamaki S, Jung T, Kim B, Huyhn R, McKemy DD. Endogenous Inflammatory Mediators Produced by Injury Activate TRPV1 and TRPA1 Nociceptors to Induce Sexually Dimorphic Cold Pain That Is Dependent on TRPM8 and GFRα3. J Neurosci 2023; 43:2803-2814. [PMID: 36898840 PMCID: PMC10089246 DOI: 10.1523/jneurosci.2303-22.2023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/06/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
The detection of environmental temperatures is critical for survival, yet inappropriate responses to thermal stimuli can have a negative impact on overall health. The physiological effect of cold is distinct among somatosensory modalities in that it is soothing and analgesic, but also agonizing in the context of tissue damage. Inflammatory mediators produced during injury activate nociceptors to release neuropeptides, such as calcitonin gene-related peptide (CGRP) and substance P, inducing neurogenic inflammation, which further exasperates pain. Many inflammatory mediators induce sensitization to heat and mechanical stimuli but, conversely, inhibit cold responsiveness, and the identity of molecules inducing cold pain peripherally is enigmatic, as are the cellular and molecular mechanisms altering cold sensitivity. Here, we asked whether inflammatory mediators that induce neurogenic inflammation via the nociceptive ion channels TRPV1 (vanilloid subfamily of transient receptor potential channel) and TRPA1 (transient receptor potential ankyrin 1) lead to cold pain in mice. Specifically, we tested cold sensitivity in mice after intraplantar injection of lysophosphatidic acid or 4-hydroxy-2-nonenal, finding that each induces cold pain that is dependent on the cold-gated channel transient receptor potential melastatin 8 (TRPM8). Inhibition of CGRP, substance P, or toll-like receptor 4 (TLR4) signaling attenuates this phenotype, and each neuropeptide produces TRPM8-dependent cold pain directly. Further, the inhibition of CGRP or TLR4 signaling alleviates cold allodynia differentially by sex. Last, cold pain induced by both inflammatory mediators and neuropeptides requires TRPM8, as well as the neurotrophin artemin and its receptor GDNF receptor α3 (GFRα3). These results are consistent with artemin-induced cold allodynia requiring TRPM8, demonstrating that neurogenic inflammation alters cold sensitivity via localized artemin release that induces cold pain via GFRα3 and TRPM8.SIGNIFICANCE STATEMENT The cellular and molecular mechanisms that generate pain are complex with a diverse array of pain-producing molecules generated during injury that act to sensitize peripheral sensory neurons, thereby inducing pain. Here we identify a specific neuroinflammatory pathway involving the ion channel TRPM8 (transient receptor potential cation channel subfamily M member 8) and the neurotrophin receptor GFRα3 (GDNF receptor α3) that leads to cold pain, providing select targets for potential therapies for this pain modality.
Collapse
Affiliation(s)
- Chenyu Yang
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, California 90089
| | - Shanni Yamaki
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, California 90089
| | - Tyler Jung
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - Brian Kim
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - Ryan Huyhn
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - David D McKemy
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
5
|
Herbert J, Kelty JS, Laskin JD, Laskin DL, Gow AJ. Menthol flavoring in e-cigarette condensate causes pulmonary dysfunction and cytotoxicity in precision cut lung slices. Am J Physiol Lung Cell Mol Physiol 2023; 324:L345-L357. [PMID: 36692165 PMCID: PMC10026991 DOI: 10.1152/ajplung.00222.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 01/05/2023] [Accepted: 01/14/2023] [Indexed: 01/25/2023] Open
Abstract
E-cigarette consumption is under scrutiny by regulatory authorities due to concerns about product toxicity, lack of manufacturing standards, and increasing reports of e-cigarette- or vaping-associated acute lung injury. In vitro studies have demonstrated cytotoxicity, mitochondrial dysfunction, and oxidative stress induced by unflavored e-cigarette aerosols and flavoring additives. However, e-cigarette effects on the complex lung parenchyma remain unclear. Herein, the impact of e-cigarette condensates with or without menthol flavoring on functional, structural, and cellular responses was investigated using mouse precision cut lung slices (PCLS). PCLS were exposed to e-cigarette condensates prepared from aerosolized vehicle, nicotine, nicotine + menthol, and menthol e-fluids at doses from 50 to 500 mM. Doses were normalized to the glycerin content of vehicle. Video-microscopy of PCLS revealed impaired contractile responsiveness of airways to methacholine and dampened ciliary beating following exposure to menthol-containing condensates at concentrations greater than 300 mM. Following 500 mM menthol-containing condensate exposure, epithelial exfoliation in intrabronchial airways was identified in histological sections of PCLS. Measurement of lactate dehydrogenase release, mitochondrial water-soluble-tetrazolium salt-1 conversion, and glutathione content supported earlier findings of nicotine or nicotine + menthol e-cigarette-induced dose-dependent cytotoxicity and oxidative stress responses. Evaluation of PCLS metabolic activity revealed dose-related impairment of mitochondrial oxidative phosphorylation and glycolysis after exposure to menthol-containing condensates. Taken together, these data demonstrate prominent menthol-induced pulmonary toxicity and impairment of essential physiological functions in the lung, which warrants concerns about e-cigarette consumer safety and emphasizes the need for further investigations of molecular mechanisms of toxicity and menthol effects in an experimental model of disease.
Collapse
Affiliation(s)
- Julia Herbert
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, United States
| | - Jacklyn S Kelty
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, United States
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, New Jersey, United States
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, United States
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, United States
| |
Collapse
|
6
|
Huang Y, Li S, Liu Q, Wang Z, Li S, Liu L, Zhao W, Wang K, Zhang R, Wang L, Wang M, William Ali D, Michalak M, Chen XZ, Zhou C, Tang J. The LCK-14-3-3ζ-TRPM8 axis regulates TRPM8 function/assembly and promotes pancreatic cancer malignancy. Cell Death Dis 2022; 13:524. [PMID: 35665750 PMCID: PMC9167300 DOI: 10.1038/s41419-022-04977-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 01/21/2023]
Abstract
Transient receptor potential melastatin 8 (TRPM8) functions as a Ca2+-permeable channel in the plasma membrane (PM). Dysfunction of TRPM8 is associated with human pancreatic cancer and several other diseases in clinical patients, but the underlying mechanisms are unclear. Here, we found that lymphocyte-specific protein tyrosine kinase (LCK) directly interacts with TRPM8 and potentiates TRPM8 phosphorylation at Y1022. LCK positively regulated channel function characterized by increased TRPM8 current densities by enhancing TRPM8 multimerization. Furthermore, 14-3-3ζ interacted with TRPM8 and positively modulated channel multimerization. LCK significantly enhanced the binding of 14-3-3ζ and TRPM8, whereas mutant TRPM8-Y1022F impaired TRPM8 multimerization and the binding of TRPM8 and 14-3-3ζ. Knockdown of 14-3-3ζ impaired the regulation of TRPM8 multimerization by LCK. In addition, TRPM8 phosphotyrosine at Y1022 feedback regulated LCK activity by inhibiting Tyr505 phosphorylation and modulating LCK ubiquitination. Finally, we revealed the importance of TRPM8 phosphorylation at Y1022 in the proliferation, migration, and tumorigenesis of pancreatic cancer cells. Our findings demonstrate that the LCK-14-3-3ζ-TRPM8 axis for regulates TRPM8 assembly, channel function, and LCK activity and maybe provide potential therapeutic targets for pancreatic cancer.
Collapse
Affiliation(s)
- Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Shi Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Qinfeng Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Zhijie Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Shunyao Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Lei Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Weiwei Zhao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Kai Wang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Longfei Wang
- Children's Hospital Affiliated to Zhengzhou University, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Ming Wang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| |
Collapse
|
7
|
Constitutive Phosphorylation as a Key Regulator of TRPM8 Channel Function. J Neurosci 2021; 41:8475-8493. [PMID: 34446569 DOI: 10.1523/jneurosci.0345-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/04/2021] [Accepted: 08/13/2021] [Indexed: 11/21/2022] Open
Abstract
In mammals, environmental cold sensing conducted by peripheral cold thermoreceptor neurons mostly depends on TRPM8, an ion channel that has evolved to become the main molecular cold transducer. This TRP channel is activated by cold, cooling compounds, such as menthol, voltage, and rises in osmolality. TRPM8 function is regulated by kinase activity that phosphorylates the channel under resting conditions. However, which specific residues, how this post-translational modification modulates TRPM8 activity, and its influence on cold sensing are still poorly understood. By mass spectrometry, we identified four serine residues within the N-terminus (S26, S29, S541, and S542) constitutively phosphorylated in the mouse ortholog. TRPM8 function was examined by Ca2+ imaging and patch-clamp recordings, revealing that treatment with staurosporine, a kinase inhibitor, augmented its cold- and menthol-evoked responses. S29A mutation is sufficient to increase TRPM8 activity, suggesting that phosphorylation of this residue is a central molecular determinant of this negative regulation. Biophysical and total internal reflection fluorescence-based analysis revealed a dual mechanism in the potentiated responses of unphosphorylated TRPM8: a shift in the voltage activation curve toward more negative potentials and an increase in the number of active channels at the plasma membrane. Importantly, basal kinase activity negatively modulates TRPM8 function at cold thermoreceptors from male and female mice, an observation accounted for by mathematical modeling. Overall, our findings suggest that cold temperature detection could be rapidly and reversibly fine-tuned by controlling the TRPM8 basal phosphorylation state, a mechanism that acts as a dynamic molecular brake of this thermo-TRP channel function in primary sensory neurons.SIGNIFICANCE STATEMENT Post-translational modifications are one of the main molecular mechanisms involved in adjusting the sensitivity of sensory ion channels to changing environmental conditions. Here we show, for the first time, that constitutive phosphorylation of the well-conserved serine 29 within the N-terminal domain negatively modulates TRPM8 channel activity, reducing its activation by agonists and decreasing the number of active channels at the plasma membrane. Basal phosphorylation of TRPM8 acts as a key regulator of its function as the main cold-transduction channel, significantly contributing to the net response of primary sensory neurons to temperature reductions. This reversible and dynamic modulatory mechanism opens new opportunities to regulate TRPM8 function in pathologic conditions where this thermo-TRP channel plays a critical role.
Collapse
|
8
|
Abstract
Transient receptor potential melastatin 8 (TRPM8) channels play a central role in the detection of environmental cold temperatures in the somatosensory system. TRPM8 is found in a subset of unmyelinated (C-type) afferents located in the dorsal root (DRG) and trigeminal ganglion (TG). Cold hypersensitivity is a common symptom of neuropathic pain conditions caused by cancer therapy, spinal cord injury, viral infection, multiple sclerosis, diabetes, or withdrawal symptoms associated with chronic morphine treatment. Therefore, TRPM8 has received great attention as a therapeutic target. However, as the activity of TRPM8 is unique in sensing cool temperature as well as warming, it is critical to understand the signaling transduction pathways that control modality-specific activity of TRPM8 in healthy versus pathological settings. This review summarizes recent advances in our understanding of the mechanisms involved in the regulation of the TRPM8 activity.
Collapse
Affiliation(s)
- Mircea Iftinca
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary , Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary , Calgary, Alberta, Canada
| |
Collapse
|
9
|
Rimola V, Osthues T, Königs V, Geißlinger G, Sisignano M. Oxaliplatin Causes Transient Changes in TRPM8 Channel Activity. Int J Mol Sci 2021; 22:4962. [PMID: 34066977 PMCID: PMC8125753 DOI: 10.3390/ijms22094962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023] Open
Abstract
Oxaliplatin is a third-generation platinum-based anticancer drug that is widely used as first-line treatment for colorectal carcinoma. Patients treated with oxaliplatin develop an acute peripheral pain several hours after treatment, mostly characterized by cold allodynia as well as a long-term chronic neuropathy. These two phenomena seem to be causally connected. However, the underlying mechanisms that trigger the acute peripheral pain are still poorly understood. Here we show that the activity of the transient receptor potential melastatin 8 (TRPM8) channel but not the activity of any other member of the TRP channel family is transiently increased 1 h after oxaliplatin treatment and decreased 24 h after oxaliplatin treatment. Mechanistically, this is connected with activation of the phospholipase C (PLC) pathway and depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) after oxaliplatin treatment. Inhibition of the PLC pathway can reverse the decreased TRPM8 activity as well as the decreased PIP2-concentrations after oxaliplatin treatment. In summary, these results point out transient changes in TRPM8 activity early after oxaliplatin treatment and a later occurring TRPM8 channel desensitization in primary sensory neurons. These mechanisms may explain the transient cold allodynia after oxaliplatin treatment and highlight an important role of TRPM8 in oxaliplatin-induced acute and neuropathic pain.
Collapse
Affiliation(s)
- Vittoria Rimola
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (V.R.); (G.G.)
| | - Tabea Osthues
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (T.O.); (V.K.)
| | - Vanessa Königs
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (T.O.); (V.K.)
| | - Gerd Geißlinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (V.R.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (T.O.); (V.K.)
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (V.R.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; (T.O.); (V.K.)
| |
Collapse
|
10
|
Negative Modulation of TRPM8 Channel Function by Protein Kinase C in Trigeminal Cold Thermoreceptor Neurons. Int J Mol Sci 2020; 21:ijms21124420. [PMID: 32580281 PMCID: PMC7352406 DOI: 10.3390/ijms21124420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/30/2020] [Accepted: 06/12/2020] [Indexed: 01/19/2023] Open
Abstract
TRPM8 is the main molecular entity responsible for cold sensing. This polymodal ion channel is activated by cold, cooling compounds such as menthol, voltage, and rises in osmolality. In corneal cold thermoreceptor neurons (CTNs), TRPM8 expression determines not only their sensitivity to cold, but also their role as neural detectors of ocular surface wetness. Several reports suggest that Protein Kinase C (PKC) activation impacts on TRPM8 function; however, the molecular bases of this functional modulation are still poorly understood. We explored PKC-dependent regulation of TRPM8 using Phorbol 12-Myristate 13-Acetate to activate this kinase. Consistently, recombinant TRPM8 channels, cultured trigeminal neurons, and free nerve endings of corneal CTNs revealed a robust reduction of TRPM8-dependent responses under PKC activation. In corneal CTNs, PKC activation decreased ongoing activity, a key parameter in the role of TRPM8-expressing neurons as humidity detectors, and also the maximal cold-evoked response, which were validated by mathematical modeling. Biophysical analysis indicated that PKC-dependent downregulation of TRPM8 is mainly due to a decreased maximal conductance value, and complementary noise analysis revealed a reduced number of functional channels at the cell surface, providing important clues to understanding the molecular mechanisms of how PKC activity modulates TRPM8 channels in CTNs.
Collapse
|
11
|
Iftinca M, Basso L, Flynn R, Kwok C, Roland C, Hassan A, Defaye M, Ramachandran R, Trang T, Altier C. Chronic morphine regulates TRPM8 channels via MOR-PKCβ signaling. Mol Brain 2020; 13:61. [PMID: 32290846 PMCID: PMC7155267 DOI: 10.1186/s13041-020-00599-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/02/2020] [Indexed: 02/05/2023] Open
Abstract
Postoperative shivering and cold hypersensitivity are major side effects of acute and chronic opioid treatments respectively. TRPM8 is a cold and menthol-sensitive channel found in a subset of dorsal root ganglion (DRG) nociceptors. Deletion or inhibition of the TRPM8 channel was found to prevent the cold hyperalgesia induced by chronic administration of morphine. Here, we examined the mechanisms by which morphine was able to promote cold hypersensitivity in DRG neurons and transfected HEK cells. Mice daily injected with morphine for 5 days developed cold hyperalgesia. Treatment with morphine did not alter the expressions of cold sensitive TREK-1, TRAAK and TRPM8 in DRGs. However, TRPM8-expressing DRG neurons isolated from morphine-treated mice exhibited hyperexcitability. Sustained morphine treatment in vitro sensitized TRPM8 responsiveness to cold or menthol and reduced activation-evoked desensitization of the channel. Blocking phospholipase C (PLC) as well as protein kinase C beta (PKCβ), but not protein kinase A (PKA) or Rho-associated protein kinase (ROCK), restored channel desensitization. Identification of two PKC phosphorylation consensus sites, S1040 and S1041, in the TRPM8 and their site-directed mutation were able to prevent the MOR-induced reduction in TRPM8 desensitization. Our results show that activation of MOR by morphine 1) promotes hyperexcitability of TRPM8-expressing neurons and 2) induces a PKCβ-mediated reduction of TRPM8 desensitization. This MOR-PKCβ dependent modulation of TRPM8 may underlie the onset of cold hyperalgesia caused by repeated administration of morphine. Our findings point to TRPM8 channel and PKCβ as important targets for opioid-induced cold hypersensitivity.
Collapse
Affiliation(s)
- Mircea Iftinca
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Lilian Basso
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Robyn Flynn
- Hotchkiss Brain Institute. Cumming School of Medicine. University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Charlie Kwok
- Hotchkiss Brain Institute. Cumming School of Medicine. University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Corinne Roland
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Ahmed Hassan
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology. Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Tuan Trang
- Hotchkiss Brain Institute. Cumming School of Medicine. University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
12
|
He Y, Wang ZJ. Spinal and afferent PKC signaling mechanisms that mediate chronic pain in sickle cell disease. Neurosci Lett 2019; 706:56-60. [PMID: 31051220 DOI: 10.1016/j.neulet.2019.04.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/22/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
Pain is the most characteristic feature of sickle cell disease (SCD). Patients with SCD live with unpredictable, recurrent episodes of acute painful crisis, as well as chronic unremitting pain throughout their lifetime. While most of the research and medical efforts have focused on treating vaso-occlusion crisis and acute pain, chronic pain remains a significant challenge faced by patients and physicians. Emerging evidence from human and animal studies has suggested the presence of a neuropathic component in SCD pain. New knowledge on the neurobiology of chronic pain in SCD has significant implications in unraveling the underlying mechanisms. This review focuses on the recent advances on the role of protein kinase C or PKC in promoting and maintaining chronic pain conditions. With a highlight of a specific PKC isoform, PKCδ, we aim to propose PKC as an essential regulator of chronic pain in SCD, which may ultimately lead to innovative therapeutic strategies for treating this devastating life-long problem in patients with SCD.
Collapse
Affiliation(s)
- Ying He
- Department of Biopharmaceutical Sciences and Center for Biomolecular Sciences, University of Illinois, Chicago, IL 60612, United States.
| | - Zaijie Jim Wang
- Department of Biopharmaceutical Sciences and Center for Biomolecular Sciences, University of Illinois, Chicago, IL 60612, United States.
| |
Collapse
|
13
|
Hatta A, Kurose M, Sullivan C, Okamoto K, Fujii N, Yamamura K, Meng ID. Dry eye sensitizes cool cells to capsaicin-induced changes in activity via TRPV1. J Neurophysiol 2019; 121:2191-2201. [PMID: 30969886 DOI: 10.1152/jn.00126.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Corneal cool cells are sensitive to the ocular fluid status of the corneal surface and may be responsible for the regulation of basal tear production. Previously, we have shown that dry eye, induced by lacrimal gland excision (LGE) in rats, sensitized corneal cool cells to the transient receptor potential melastatin 8 (TRPM8) agonist menthol and to cool stimulation. In the present study, we examined the effect of dry eye on the sensitivity of cool cells to the transient receptor potential vanilloid 1 (TRPV1) agonist capsaicin. Single-unit recordings in the trigeminal ganglion were performed 7-10 days after LGE. At a concentration of 0.3 μM, capsaicin did not affect ongoing or cool-evoked activity in control animals yet facilitated ongoing activity and suppressed cool-evoked activity in LGE animals. At higher concentrations (3 μM), capsaicin continued to facilitate ongoing activity in LGE animals but suppressed ongoing activity in control animals. Higher concentrations of capsaicin also suppressed cool-evoked activity in both groups of animals, with an overall greater effect in LGE animals. In addition to altering cool-evoked activity, capsaicin enhanced the sensitivity of cool cells to heat in LGE animals. Capsaicin-induced changes were prevented by the application of the TRPV1 antagonist capsazepine. With the use of fluorescent in situ hybridization, TRPV1 and TRPM8 expression was examined in retrograde tracer-identified corneal neurons. The coexpression of TRPV1 and TRPM8 in corneal neurons was significantly greater in LGE-treated animals when compared with sham controls. These results indicate that LGE-induced dry eye increases TRPV1-mediated responses in corneal cool cells at least in part through the increased expression of TRPV1. NEW & NOTEWORTHY Corneal cool cells are known to detect drying of the ocular surface. Our study is the first to report that dry eye induced alterations in cool cell response properties, including the increased responsiveness to noxious heat and activation by capsaicin. Along with the changes in cell response properties, it is possible these neurons also function differently in dry eye, relaying information related to the perception of ocular irritation in addition to regulating tearing and blinking.
Collapse
Affiliation(s)
- Azusa Hatta
- Division of Oral Physiology, Department of Oral Biological Sciences, Niigata University, Graduate School of Medical and Dental Sciences , Niigata , Japan.,General Dentistry and Clinical Education Unit, Niigata University Medical and Dental Hospital , Niigata , Japan
| | - Masayuki Kurose
- Division of Oral Physiology, Department of Oral Biological Sciences, Niigata University, Graduate School of Medical and Dental Sciences , Niigata , Japan
| | - Cara Sullivan
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine.,Graduate Studies in Biomedical Sciences and Engineering, University of Maine , Orono, Maine
| | - Keiichiro Okamoto
- Division of Oral Physiology, Department of Oral Biological Sciences, Niigata University, Graduate School of Medical and Dental Sciences , Niigata , Japan
| | - Noritaka Fujii
- General Dentistry and Clinical Education Unit, Niigata University Medical and Dental Hospital , Niigata , Japan
| | - Kensuke Yamamura
- Division of Oral Physiology, Department of Oral Biological Sciences, Niigata University, Graduate School of Medical and Dental Sciences , Niigata , Japan
| | - Ian D Meng
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine
| |
Collapse
|
14
|
Sensitization of TRPV1 and TRPA1 via peripheral mGluR5 signaling contributes to thermal and mechanical hypersensitivity. Pain 2018. [PMID: 28621704 DOI: 10.1097/j.pain.0000000000000973] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peripheral tissue inflammation or injury causes glutamate release from nociceptive axons, keratinocytes, and Schwann cells, resulting in thermal hypersensitivity. However, the detailed molecular mechanisms underlying glutamate-induced thermal hypersensitivity are unknown. The aim of this study was to clarify the involvement of peripheral transient receptor potential (TRP) TRP vanilloid 1 (TRPV1), TRP ankyrin 1 (TRPA1), and protein kinase C epsilon (PKCε) in glutamate-induced pain hypersensitivity. The amount of glutamate in the facial tissue was significantly increased 3 days after facial Complete Freund's adjuvant injection. The head-withdrawal reflex threshold to heat, cold, or mechanical stimulation was significantly decreased on day 7 after continuous glutamate or metabotropic glutamate receptor 5 (mGluR5) agonist (CHPG) injection into the facial skin compared with vehicle-injected rats, and glutamate-induced hypersensitivity was significantly recovered by mGluR5 antagonist MTEP, TRPA1 antagonist HC-030031, TRPV1 antagonist SB366791, or PKCε translocation inhibitor administration into the facial skin. TRPV1 and TRPA1 were expressed in mGluR5-immunoreactive (IR) trigeminal ganglion (TG) neurons innervating the facial skin, and mGluR5-IR TG neurons expressed PKCε. There was no significant difference in the number of GluR5-IR TG neurons among glutamate-injected, saline-injected, and naive rats, whereas that of TRPV1- or TRPA1-IR TG neurons was significantly increased 7 days after continuous glutamate injection into the facial skin compared with vehicle injection. PKCε phosphorylation in TG was significantly enhanced following glutamate injection into the facial skin. Moreover, neuronal activity of TG neurons was significantly increased following facial glutamate treatment. The present findings suggest that sensitization of TRPA1 and/or TRPV1 through mGluR5 signaling via PKCε is involved in facial thermal and mechanical hypersensitivity.
Collapse
|
15
|
Signaling Pathways Relevant to Nerve Growth Factor-induced Upregulation of Transient Receptor Potential M8 Expression. Neuroscience 2017; 367:178-188. [DOI: 10.1016/j.neuroscience.2017.10.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/06/2017] [Accepted: 10/24/2017] [Indexed: 12/29/2022]
|
16
|
Rittich AB, Ellrich J, Said Yekta-Michael S. Assessment of lingual nerve functions after smoking cessation. Acta Odontol Scand 2017; 75:338-344. [PMID: 28372503 DOI: 10.1080/00016357.2017.1308551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Cigarette smoking is associated with a variety of oral diseases. A previous study showed a reduction of thermal sensitivity in the innervation area of the lingual nerve in smokers possibly caused by a degeneration of thermosensitive receptors as a consequence of smoking. The current study investigates somatosensory changes in ex-smokers. MATERIALS AND METHODS Sensory functions in innervation areas of lingual nerve were investigated in 40 ex-smokers by psychophysical means. Functions of lingual nerve in 40 ex-smokers were compared to those in 40 smokers and 40 non-smokers. Subjects were investigated using quantitative sensory testing (QST, cold and warm detection, thermal sensory limen, heat and cold pain, and mechanical detection). RESULTS Significant differences were found in both groups, ex-smokers and smokers compared to non-smokers. Cold (p < .001), warm (ex-smokers: p < .01; smokers: p < .001) detection thresholds and thermal sensory limen (p < .001) showed significantly lower sensitivity in ex-smokers and smokers in comparison to non-smokers. CONCLUSIONS The lower temperature sensitivity of ex-smokers compared to that in non-smokers indicates a reduction of somatosensory function of the tongue, possibly caused by irreversible nerve degeneration associated with smoking. Influencing factors leading to sensory changes could be modulation of thermo-receptors, demyelination as well as a change of the epithelial structure.
Collapse
Affiliation(s)
- Anne Barbara Rittich
- Department of Prosthodontics and Biomaterials, Centre of Implantology, University Hospital Aachen, Aachen, Germany,
| | - Jens Ellrich
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Sareh Said Yekta-Michael
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, University Hospital Aachen, Aachen, Germany
- Interdisciplinary Centre for Clinical Research, University Hospital Aachen, Aachen, Germany
| |
Collapse
|
17
|
Balemans D, Boeckxstaens GE, Talavera K, Wouters MM. Transient receptor potential ion channel function in sensory transduction and cellular signaling cascades underlying visceral hypersensitivity. Am J Physiol Gastrointest Liver Physiol 2017; 312:G635-G648. [PMID: 28385695 DOI: 10.1152/ajpgi.00401.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/21/2017] [Accepted: 04/01/2017] [Indexed: 01/31/2023]
Abstract
Visceral hypersensitivity is an important mechanism underlying increased abdominal pain perception in functional gastrointestinal disorders including functional dyspepsia, irritable bowel syndrome, and inflammatory bowel disease in remission. Although the exact pathophysiological mechanisms are poorly understood, recent studies described upregulation and altered functions of nociceptors and their signaling pathways in aberrant visceral nociception, in particular the transient receptor potential (TRP) channel family. A variety of TRP channels are present in the gastrointestinal tract (TRPV1, TRPV3, TRPV4, TRPA1, TRPM2, TRPM5, and TRPM8), and modulation of their function by increased activation or sensitization (decreased activation threshold) or altered expression in visceral afferents have been reported in visceral hypersensitivity. TRP channels directly detect or transduce osmotic, mechanical, thermal, and chemosensory stimuli. In addition, pro-inflammatory mediators released in tissue damage or inflammation can activate receptors of the G protein-coupled receptor superfamily leading to TRP channel sensitization and activation, which amplify pain and neurogenic inflammation. In this review, we highlight the present knowledge on the functional roles of neuronal TRP channels in visceral hypersensitivity and discuss the signaling pathways that underlie TRP channel modulation. We propose that a better understanding of TRP channels and their modulators may facilitate the development of more selective and effective therapies to treat visceral hypersensitivity.
Collapse
Affiliation(s)
- Dafne Balemans
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Guy E Boeckxstaens
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Karel Talavera
- Laboratory of Ion Channel Research and TRP Research Platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven Belgium
| | - Mira M Wouters
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| |
Collapse
|
18
|
Mickle AD, Shepherd AJ, Mohapatra DP. Nociceptive TRP Channels: Sensory Detectors and Transducers in Multiple Pain Pathologies. Pharmaceuticals (Basel) 2016; 9:ph9040072. [PMID: 27854251 PMCID: PMC5198047 DOI: 10.3390/ph9040072] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 02/07/2023] Open
Abstract
Specialized receptors belonging to the transient receptor potential (TRP) family of ligand-gated ion channels constitute the critical detectors and transducers of pain-causing stimuli. Nociceptive TRP channels are predominantly expressed by distinct subsets of sensory neurons of the peripheral nervous system. Several of these TRP channels are also expressed in neurons of the central nervous system, and in non-neuronal cells that communicate with sensory nerves. Nociceptive TRPs are activated by specific physico-chemical stimuli to provide the excitatory trigger in neurons. In addition, decades of research has identified a large number of immune and neuromodulators as mediators of nociceptive TRP channel activation during injury, inflammatory and other pathological conditions. These findings have led to aggressive targeting of TRP channels for the development of new-generation analgesics. This review summarizes the complex activation and/or modulation of nociceptive TRP channels under pathophysiological conditions, and how these changes underlie acute and chronic pain conditions. Furthermore, development of small-molecule antagonists for several TRP channels as analgesics, and the positive and negative outcomes of these drugs in clinical trials are discussed. Understanding the diverse functional and modulatory properties of nociceptive TRP channels is critical to function-based drug targeting for the development of evidence-based and efficacious new generation analgesics.
Collapse
Affiliation(s)
- Aaron D Mickle
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | - Andrew J Shepherd
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | - Durga P Mohapatra
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
19
|
Pérez de Vega MJ, Gómez-Monterrey I, Ferrer-Montiel A, González-Muñiz R. Transient Receptor Potential Melastatin 8 Channel (TRPM8) Modulation: Cool Entryway for Treating Pain and Cancer. J Med Chem 2016; 59:10006-10029. [PMID: 27437828 DOI: 10.1021/acs.jmedchem.6b00305] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
TRPM8 ion channels, the primary cold sensors in humans, are activated by innocuous cooling (<28 °C) and cooling compounds (menthol, icilin) and are implicated in sensing unpleasant cold stimuli as well as in mammalian thermoregulation. Overexpression of these thermoregulators in prostate cancer and in other life-threatening tumors, along with their contribution to an increasing number of pathological conditions, opens a plethora of medicinal chemistry opportunities to develop receptor modulators. This Perspective seeks to describe current known modulators for this ion channel because both agonists and antagonists may be useful for the treatment of most TRPM8-mediated pathologies. We primarily focus on SAR data for the different families of compounds and the pharmacological properties of the most promising ligands. Furthermore, we also address the knowledge about the channel structure, although still in its infancy, and the role of the TRPM8 protein signalplex to channel function and dysfunction. We finally outline the potential future prospects of the challenging TRPM8 drug discovery field.
Collapse
Affiliation(s)
| | - Isabel Gómez-Monterrey
- Dipartimento di Farmacia, Università "Federico II" de Napoli , Via D. Montesano 49, 80131, Naples, Italy
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular. Universitas Miguel Hernández . 03202 Alicante, Spain
| | | |
Collapse
|
20
|
Cao S, Li Y, Wang L, Cui J, Jia N, Li R, Zhao C, Wang C, Wu Y, Wen A. Synergistic analgesic effect of propofol-alfentanil combination through detecting the inhibition of cAMP signal pathway. ACTA ACUST UNITED AC 2016; 68:1170-6. [PMID: 27334358 DOI: 10.1111/jphp.12578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 04/30/2016] [Indexed: 12/26/2022]
Abstract
OBJECTIVE The study aims to investigate the possible mechanism of the synergistic analgesic effect of propofol-alfentanil combination. METHODS The synergistic analgesic effects of propofol-alfentanil combination in Sprague-Dawley (SD) rats were analysed through the von Frey test. Then, we examined the activity of phospholipase C (PLC) and the intracellular levels of Ca(2+) and adenosine 3', 5'cyclic monophosphate (cAMP) in primary neuronal cells of fetal SD rats. We detected the intracellular Ca(2+) concentration by fluorescence and flow cytometry. The PLC activity of the primary neuronal cells was assayed using the EnzChek(®) Direct Phospholipase C Assay Kit. The cAMP content of the cells was assayed using the cAMP Direct Immunoassay Kit (Fluorometric). KEY FINDINGS Both propofol and alfentanil treatments depressed cAMP levels and PLC activity, but propofol-alfentanil combination decreased these parameters to a greater extent than alfentanil treatment alone. Propofol and alfentanil both inhibited Ca(2+) channel, but propofol-alfentanil combination suppressed this channel to a greater extent than alfentanil treatment alone. Fluorescent image analysis revealed that both propofol and alfentanil reduced the intracellular levels of Ca(2+) , and propofol-alfentanil combination showed weaker signals than alfentanil alone. Propofol-alfentanil combination significantly reduced intracellular Ca(2+) level, cAMP level and PLC activity. CONCLUSION Propofol and alfentanil exert synergistic analgesic effects through the adenylyl cyclase pathway.
Collapse
Affiliation(s)
- Shanshan Cao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuwen Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lu Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jia Cui
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Na Jia
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ruili Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yin Wu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
21
|
Calcium Entry Through Thermosensory Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:265-304. [PMID: 27161233 DOI: 10.1007/978-3-319-26974-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ThermoTRPs are unique channels that mediate Na(+) and Ca(2+) currents in response to changes in ambient temperature. In combination with their activation by other physical and chemical stimuli, they are considered key integrators of environmental cues into neuronal excitability. Furthermore, roles of thermoTRPs in non-neuronal tissues are currently emerging such as insulin secretion in pancreatic β-cells, and links to cancer. Calcium permeability through thermoTRPs appears a central hallmark for their physiological and pathological activities. Moreover, it is currently being proposed that beyond working as a second messenger, Ca(2+) can function locally by acting on protein complexes near the membrane. Interestingly, thermoTRPs can enhance and expand the inherent plasticity of signalplexes by conferring them temperature, pH and lipid regulation through Ca(2+) signalling. Thus, unveiling the local role of Ca(2+) fluxes induced by thermoTRPs on the dynamics of membrane-attached signalling complexes as well as their significance in cellular processes, are central issues that will expand the opportunities for therapeutic intervention in disorders involving dysfunction of thermoTRP channels.
Collapse
|
22
|
Abstract
UNLABELLED Extracellular signal-regulated kinases 1 and 2 (ERK1/2) are highly homologous yet distinct components of signal transduction pathways known to regulate cell survival and function. Recent evidence indicates an isoform-specific role for ERK2 in pain processing and peripheral sensitization. However, the function of ERK2 in primary sensory neurons has not been directly tested. To dissect the isoform-specific function of ERK2 in sensory neurons, we used mice with Cre-loxP-mediated deletion of ERK2 in Nav1.8(+) sensory neurons that are predominantly nociceptors. We find that ERK2, unlike ERK1, is required for peripheral sensitization and cold sensation. We also demonstrate that ERK2, but not ERK1, is required to preserve epidermal innervation in a subset of peptidergic neurons. Additionally, deletion of both ERK isoforms in Nav1.8(+) sensory neurons leads to neuron loss not observed with deletion of either isoform alone, demonstrating functional redundancy in the maintenance of sensory neuron survival. Thus, ERK1 and ERK2 exhibit both functionally distinct and redundant roles in sensory neurons. SIGNIFICANCE STATEMENT ERK1/2 signaling affects sensory neuron function and survival. However, it was not clear whether ERK isoform-specific roles exist in these processes postnatally. Previous work from our laboratory suggested either functional redundancy of ERK isoforms or a predominant role for ERK2 in pain; however, the tools to discriminate between these possibilities were not available at the time. In the present study, we use new genetic knock-out lines to demonstrate that ERK2 in sensory neurons is necessary for development of inflammatory pain and for postnatal maintenance of peptidergic epidermal innervation. Interestingly, postnatal loss of both ERK isoforms leads to a profound loss of sensory neurons. Therefore, ERK1 and ERK2 display both functionally distinct and redundant roles in sensory neurons.
Collapse
|
23
|
Finch PM, Drummond PD. Topical treatment in pain medicine: from ancient remedies to modern usage. Pain Manag 2015. [DOI: 10.2217/pmt.15.23] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over several millennia, substances have been applied to the skin for treatment of pain. Some ingredients are in current use; others have been discontinued. Mechanisms of action include interactions with nociceptive neural networks and inflammatory processes. Substances must penetrate the stratum corneum barrier and vehicles that enhance penetration have been developed. Topical drugs with links to the past include menthol, capsaicin, some opioids, local anesthetic agents and NSAIDs. Mandragora is also described as an example of a herbal remedy that has been discontinued due to its toxicity. The future for topical drugs is promising, with the advent of new drugs tailored for specific pain mechanisms and the development of both penetration enhancers and sterile preparation methods.
Collapse
Affiliation(s)
- Philip M Finch
- Perth Pain Management Centre, Perth, Western Australia, Australia
| | - Peter D Drummond
- Centre for Research on Chronic Pain & Inflammatory Diseases, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
24
|
Thermosensitive transient receptor potential (TRP) channel agonists and their role in mechanical, thermal and nociceptive sensations as assessed using animal models. CHEMOSENS PERCEPT 2015; 8:96-108. [PMID: 26388966 DOI: 10.1007/s12078-015-9176-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The present paper summarizes research using animal models to investigate the roles of thermosensitive transient receptor potential (TRP) channels in somatosensory functions including touch, temperature and pain. We present new data assessing the effects of eugenol and carvacrol, agonists of the warmth-sensitive TRPV3, on thermal, mechanical and pain sensitivity in rats. METHODS Thermal sensitivity was assessed using a thermal preference test, which measured the amount of time the animal occupied one of two adjacent thermoelectric plates set at different temperatures. Pain sensitivity was assessed as an increase in latency of hindpaw withdrawal away from a noxious thermal stimulus directed to the plantar hindpaw (Hargreaves test). Mechanical sensitivity was assessed by measuring the force exerted by an electronic von Frey filament pressed against the plantar surface that elicited withdrawal. RESULTS Topical application of eugenol and carvacrol did not significantly affect thermal preference, although there was a trend toward avoidance of the hotter surface in a 30 vs. 45°C preference test for rats treated with 1 or 10% eugenol and carvacrol. Both eugenol and carvacrol induced a concentration-dependent increase in thermal withdrawal latency (analgesia), with no significant effect on mechanosensitivity. CONCLUSIONS The analgesic effect of eugenol and carvacrol is consistent with previous studies. The tendency for these chemicals to increase the avoidance of warmer temperatures suggests a possible role for TRPV3 in warmth detection, also consistent with previous studies. Additional roles of other thermosensitive TRP channels (TRPM8 TRPV1, TRPV2, TRPV4, TRPM3, TRPM8, TRPA1, TRPC5) in touch, temperature and pain are reviewed.
Collapse
|
25
|
Mickle AD, Shepherd AJ, Mohapatra DP. Sensory TRP channels: the key transducers of nociception and pain. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:73-118. [PMID: 25744671 DOI: 10.1016/bs.pmbts.2015.01.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Peripheral detection of nociceptive and painful stimuli by sensory neurons involves a complex repertoire of molecular detectors and/or transducers on distinct subsets of nerve fibers. The majority of such molecular detectors/transducers belong to the transient receptor potential (TRP) family of cation channels, which comprise both specific receptors for distinct nociceptive stimuli, as well as for multiple stimuli. This chapter discusses the classification, distribution, and functional properties of individual TRP channel types that have been implicated in various nociceptive and/or painful conditions.
Collapse
Affiliation(s)
- Aaron D Mickle
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrew J Shepherd
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Durga P Mohapatra
- Department of Pharmacology, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa, USA; Department of Anesthesia, The University of Iowa Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
26
|
Abstract
The ability of the body to perceive noxious stimuli lies in a heterogeneous group of primary somatosensory neurons termed nociceptors. The molecular receptors of noxious mechanical, temperature, or chemical stimuli are expressed in these neurons and have drawn considerable attention as possible targets for analgesic development to improve treatment for the millions who suffer from chronic pain conditions. A number of thermoTRPs, a subset of the transient receptor potential family of ion channels, are activated by a wide range on noxious stimuli. In this review, we review the function of these channels and examine the evidence that thermoTRPs play a vital role in acute, inflammatory and neuropathic nociception.
Collapse
Affiliation(s)
- Robyn J Laing
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Ajay Dhaka
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| |
Collapse
|
27
|
Kozyreva TV, Tkachenko EY, Khramova GM. Effects of pharmacological activation of TRPM8 ion channels on the thermoregulatory responses during warming. Bull Exp Biol Med 2015; 155:335-8. [PMID: 24137597 DOI: 10.1007/s10517-013-2147-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Preliminary non-thermal activation of cold-sensitive TRPM8 ion channel facilitates initiation of the heat-defense responses in homoiothermal animals by decreasing temperature threshold of the vasodilatory response. TRPM8 activation leads to earlier heat-initiated increase in oxygen consumption, but reduces its magnitude. Warming inhibits the lipolytic effect of menthol activation of TRPM8 observed under thermoneutral conditions. Thus, modulation of the skin temperature afferent signal by ion channel agonist TRPM8 changes not only cold-defense, but also heat-defense responses of the body.
Collapse
Affiliation(s)
- T V Kozyreva
- Laboratory of Thermophysiology, Institute of Physiology, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk, Russia.
| | | | | |
Collapse
|
28
|
Sousa-Valente J, Andreou AP, Urban L, Nagy I. Transient receptor potential ion channels in primary sensory neurons as targets for novel analgesics. Br J Pharmacol 2014; 171:2508-27. [PMID: 24283624 DOI: 10.1111/bph.12532] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/11/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed an explosion in novel findings relating to the molecules involved in mediating the sensation of pain in humans. Transient receptor potential (TRP) ion channels emerged as the greatest group of molecules involved in the transduction of various physical stimuli into neuronal signals in primary sensory neurons, as well as, in the development of pain. Here, we review the role of TRP ion channels in primary sensory neurons in the development of pain associated with peripheral pathologies and possible strategies to translate preclinical data into the development of effective new analgesics. Based on available evidence, we argue that nociception-related TRP channels on primary sensory neurons provide highly valuable targets for the development of novel analgesics and that, in order to reduce possible undesirable side effects, novel analgesics should prevent the translocation from the cytoplasm to the cell membrane and the sensitization of the channels rather than blocking the channel pore or binding sites for exogenous or endogenous activators.
Collapse
Affiliation(s)
- J Sousa-Valente
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | | | | | | |
Collapse
|
29
|
Ferrandiz-Huertas C, Mathivanan S, Wolf CJ, Devesa I, Ferrer-Montiel A. Trafficking of ThermoTRP Channels. MEMBRANES 2014; 4:525-64. [PMID: 25257900 PMCID: PMC4194048 DOI: 10.3390/membranes4030525] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/11/2014] [Accepted: 08/08/2014] [Indexed: 12/19/2022]
Abstract
ThermoTRP channels (thermoTRPs) define a subfamily of the transient receptor potential (TRP) channels that are activated by changes in the environmental temperature, from noxious cold to injurious heat. Acting as integrators of several stimuli and signalling pathways, dysfunction of these channels contributes to several pathological states. The surface expression of thermoTRPs is controlled by both, the constitutive and regulated vesicular trafficking. Modulation of receptor surface density during pathological processes is nowadays considered as an interesting therapeutic approach for management of diseases, such as chronic pain, in which an increased trafficking is associated with the pathological state. This review will focus on the recent advances trafficking of the thermoTRP channels, TRPV1, TRPV2, TRPV4, TRPM3, TRPM8 and TRPA1, into/from the plasma membrane. Particularly, regulated membrane insertion of thermoTRPs channels contributes to a fine tuning of final channel activity, and indeed, it has resulted in the development of novel therapeutic approaches with successful clinical results such as disruption of SNARE-dependent exocytosis by botulinum toxin or botulinomimetic peptides.
Collapse
Affiliation(s)
| | - Sakthikumar Mathivanan
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Christoph Jakob Wolf
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Isabel Devesa
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| |
Collapse
|
30
|
Straub RH. TRPV1, TRPA1, and TRPM8 channels in inflammation, energy redirection, and water retention: role in chronic inflammatory diseases with an evolutionary perspective. J Mol Med (Berl) 2014; 92:925-37. [DOI: 10.1007/s00109-014-1175-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/17/2014] [Accepted: 05/20/2014] [Indexed: 01/04/2023]
|
31
|
Icilin reduces voltage-gated calcium channel currents in naïve and injured DRG neurons in the rat spinal nerve ligation model. Brain Res 2014; 1557:171-9. [PMID: 24560602 DOI: 10.1016/j.brainres.2014.02.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 02/07/2014] [Accepted: 02/11/2014] [Indexed: 11/21/2022]
Abstract
Recently, the transient receptor potential (TRP) channels TRPM8 and TRPA1 have been identified as molecular sensors for cold, and it has been suggested that they play a crucial role in allodynia by modulating voltage-gated calcium channel currents (ICa(V)). The aim of this study was to analyze the modulation of ICa(V) by the TRPM8-agonist icilin in vitro and to investigate the analgesic effect of icilin in a neuropathic pain model in vivo. Whole cell patch-clamp recordings were performed on isolated naïve and injured rat dorsal root ganglia (DRG) neurons, and the analgesic efficacy of icilin applied topically to the paws or intrathecally was tested in rats after spinal nerve ligation (SNL). ICa(V) (depolarization from -80 to 0mV) in naïve DRG neurons was reduced dose dependently (0.002-200µM) by icilin (18-80%). Subtype isolation of calcium channels show a marked reduction of L-type channel currents compared to N-type channel currents. The effects of icilin on ICa(V) were not significantly different in non-injured and SNL-injured DRG neurons. In vivo, neither topical (10-200µM) nor intrathecal application of icilin (0.1nM to 1µM) affected tactile allodynia or thermal hyperalgesia after SNL, but it increases cold allodynia 6h after application. We conclude that the icilin-induced modulation of ICa(V) in DRG neurons is unlikely to mediate analgesic effects or contribute directly to the pathogenesis of cold allodynia in the rat SNL model, but it is a potential mechanism for the analgesic effects of icilin in other pain models.
Collapse
|
32
|
Madrid R, Pertusa M. Intimacies and physiological role of the polymodal cold-sensitive ion channel TRPM8. CURRENT TOPICS IN MEMBRANES 2014; 74:293-324. [PMID: 25366241 DOI: 10.1016/b978-0-12-800181-3.00011-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The detection of environmental temperature is critical for the survival of the most diverse organisms. Thermosensitive transient receptor potential (thermoTRP) channels have evolved as a class of ion channels activated by a wide range of temperatures. These molecular thermal sensors are spread through the different TRP channel subfamilies. Among the Melastatin subfamily of TRP channels, the eighth member, TRPM8, is a calcium-permeable cationic ion channel activated by cold, by substances that evoke cold sensation such as menthol, and by voltage. This channel is considered the main molecular entity responsible for the sensitivity to cold of primary sensory neurons of the somatosensory system. Here we present to the readers a summary of some the most relevant biophysical properties, physiological role, and molecular intimacies of this polymodal thermoTRP channel.
Collapse
Affiliation(s)
- Rodolfo Madrid
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - María Pertusa
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
33
|
Abstract
Transient receptor potential melastatin 8 (TRPM8) was originally cloned from prostate tissue. Shortly thereafter, the protein was identified as a cold- and menthol-activated ion channel in peripheral sensory neurons, where it plays a critical role in cold temperature detection. In this chapter, we review our current understanding of the molecular and biophysical properties, the pharmacology, and the modulation by signaling molecules of this TRP channel. Finally, we examine the physiological role of TRPM8 and its emerging link to various human diseases, including pain, prostate cancer, dry eye disease, and metabolic disorders.
Collapse
Affiliation(s)
- Laura Almaraz
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Avenida S. Ramón y Cajal s.n., San Juan de Alicante, 03550, Spain
| | | | | | | |
Collapse
|
34
|
Tsumura M, Sobhan U, Sato M, Shimada M, Nishiyama A, Kawaguchi A, Soya M, Kuroda H, Tazaki M, Shibukawa Y. Functional expression of TRPM8 and TRPA1 channels in rat odontoblasts. PLoS One 2013; 8:e82233. [PMID: 24358160 PMCID: PMC3864925 DOI: 10.1371/journal.pone.0082233] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/22/2013] [Indexed: 11/18/2022] Open
Abstract
Odontoblasts produce dentin during development, throughout life, and in response to pathological conditions by sensing stimulation of exposed dentin. The functional properties and localization patterns of transient receptor potential (TRP) melastatin subfamily member 8 (TRPM8) and ankyrin subfamily member 1 (TRPA1) channels in odontoblasts remain to be clarified. We investigated the localization and the pharmacological, biophysical, and mechano-sensitive properties of TRPM8 and TRPA1 channels in rat odontoblasts. Menthol and icilin increased the intracellular free Ca(2+) concentration ([Ca(2+)]i). Icilin-, WS3-, or WS12-induced [Ca(2+)]i increases were inhibited by capsazepine or 5-benzyloxytriptamine. The increase in [Ca(2+)]i elicited by allyl isothiocyanate (AITC) was inhibited by HC030031. WS12 and AITC exerted a desensitizing effect on [Ca(2+)]i increase. Low-temperature stimuli elicited [Ca(2+)]i increases that are sensitive to both 5-benzyloxytriptamine and HC030031. Hypotonic stimulation-induced membrane stretch increased [Ca(2+)]i; HC030031 but not 5-benzyloxytriptamine inhibited the effect. The results suggest that TRPM8 channels in rat odontoblasts play a role in detecting low-temperature stimulation of the dentin surface and that TRPA1 channels are involved in sensing membrane stretching and low-temperature stimulation. The results also indicate that odontoblasts act as mechanical and thermal receptor cells, detecting the stimulation of exposed dentin to drive multiple cellular functions, such as sensory transduction.
Collapse
Affiliation(s)
- Maki Tsumura
- Department of Physiology, Tokyo Dental College, Chiba, Japan
| | - Ubaidus Sobhan
- Department of Physiology, Tokyo Dental College, Chiba, Japan
- Clinical Research Center, National Center for Child Health and Development, Tokyo, Japan
| | - Masaki Sato
- Department of Physiology, Tokyo Dental College, Chiba, Japan
| | - Miyuki Shimada
- Department of Clinical Oral Health Science, Tokyo Dental College, Tokyo, Japan
| | - Akihiro Nishiyama
- Department of Oral Medicine, Oral and Maxillofacial Surgery, Tokyo Dental College, Chiba, Japan
| | - Aya Kawaguchi
- Department of Dental Anesthesiology, Tokyo Dental College, Chiba, Japan
| | - Manabu Soya
- Department of Dental Anesthesiology, Tokyo Dental College, Chiba, Japan
| | - Hidetaka Kuroda
- Department of Anesthesiology and Pain Relief Center, University of Tokyo Hospital, Tokyo, Japan
| | - Masakazu Tazaki
- Department of Physiology, Tokyo Dental College, Chiba, Japan
| | | |
Collapse
|
35
|
Li MC, Yang G, Zhou XD, Tselluyko S, Perelman JM. The pathophysiological mechanisms underlying mucus hypersecretion induced by cold temperatures in cigarette smoke-exposed rats. Int J Mol Med 2013; 33:83-90. [PMID: 24154796 DOI: 10.3892/ijmm.2013.1535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/11/2013] [Indexed: 11/05/2022] Open
Abstract
In a recent study, we demonstrated that transient receptor potential melastatin 8 (TRPM8), a calcium-permeable cation channel that is activated by cold temperatures, is localized in the bronchial epithelium and is upregulated in subjects with chronic obstructive pulmonary disease, which causes them to be more sensitive to cold air. In the present study, we found that exposure to cold temperatures induced ciliary ultrastructural anomalies and mucus accumulation on the epithelial surface. Male Sprague-Dawley rats were exposed to cold temperatures to determine the effects of cold air on ultrastructural changes in cilia and the airway epithelial surface. The rats were also exposed to cigarette smoke and/or cold temperatures to determine the effects of smoke and cold air on TRPM8 expression and the role of cold air in cigarette smoke-induced mucus hypersecretion. Following real-time RT-PCR and western blot analysis, we observed a high expression of TRPM8 mRNA and protein in the bronchial tissue following cigarette smoke inhalation. As shown by ELISA, concurrent cold air enhanced the levels of mucin 5AC (MUC5AC) protein, as well as those of inflammatory factors [tumor necrosis factor (TNF)-α and interleukin (IL)-8] that were induced by cigarette smoke inhalation to a greater extent than stimulation with separate stimuli (cold air and cigarette smoke separately). The results suggest that cold air stimuli are responsible for the ultrastructural abnormalities of bronchial cilia, which contribute to abnormal mucus clearance. In addition, cold air synergistically amplifies cigarette smoke-induced mucus hypersecretion and the production of inflammatory factors through the elevated expression of the TRPM8 channel that is initiated by cigarette smoke inhalation.
Collapse
Affiliation(s)
- Min-Chao Li
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | | | | | | | | |
Collapse
|
36
|
Alpizar YA, Sanchez A, Radwan A, Radwan I, Voets T, Talavera K. Lack of correlation between the amplitudes of TRP channel-mediated responses to weak and strong stimuli in intracellular Ca(2+) imaging experiments. Cell Calcium 2013; 54:362-74. [PMID: 24079971 DOI: 10.1016/j.ceca.2013.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 08/29/2013] [Accepted: 08/31/2013] [Indexed: 01/18/2023]
Abstract
It is often observed in intracellular Ca(2+) imaging experiments that the amplitudes of the Ca(2+) signals elicited by newly characterized TRP agonists do not correlate with the amplitudes of the responses evoked subsequently by a specific potent agonist. We investigated this rather controversial phenomenon by first testing whether it is inherent to the comparison of the effects of weak and strong stimuli. Using five well-characterized TRP channel agonists in commonly used heterologous expression systems we found that the correlation between the amplitudes of the Ca(2+) signals triggered by two sequentially applied stimuli is only high when both stimuli are strong. Using mathematical simulations of intracellular Ca(2+) dynamics we illustrate that the innate heterogeneity in expression and functional properties of Ca(2+) extrusion (e.g. plasma membrane Ca(2+) ATPase) and influx (TRP channels) pathways across a cellular population is a sufficient condition for low correlation between the amplitude of Ca(2+) signals elicited by weak and strong stimuli. Taken together, our data demonstrate that this phenomenon is an expected outcome of intracellular Ca(2+) imaging experiments that cannot be taken as evidence for lack of specificity of low-efficacy stimuli, or as an indicator of the need of other cellular components for channel stimulation.
Collapse
Affiliation(s)
- Yeranddy A Alpizar
- Laboratory for Ion Channel Research and TRP Research Platform Leuven (TRPLe), Department of Molecular Cell Biology, KU Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
37
|
Li Q, Zhou XD, Kolosov VP, Perelman JM. Salidroside reduces cold-induced mucin production by inhibiting TRPM8 activation. Int J Mol Med 2013; 32:637-46. [PMID: 23835496 DOI: 10.3892/ijmm.2013.1434] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/28/2013] [Indexed: 11/06/2022] Open
Abstract
Salidroside is an effective component of the traditional Chinese herb, Rhodiola rosea, that is known to have the ability to protect individuals from cold attacks. In the present study, we investigated the effects of salidroside on respiratory epithelial cells exposed to cold temperatures. We wished to determine whether salidroside exerts any effect on cold-induced mucin (MUC) production and the possible mechanisms involved in this process. We incubated HBE16 cells with salidroside, exposed them to a cold stimulus (18˚C), and assayed the following endpoints: MUC production (the expression of MUC5AC), concentration intracellular of free calcium ([Ca2+]i), the activation of the transient receptor potential melastatin 8 (TRPM8) channel and the cAMP response element-binding protein (CREB). Our results revealed a significant increase in the [Ca2+]i concentration, as well as in TRPM8 and CREB expression in the cold-stimulated cells. MUC5AC expression was also increased. Treatment of the cells with salidroside at concentrations of 50 and 100 µM decreased the [Ca2+]i concentration, with a maximal effect detected in the cells treated with 100 µM salidroside. The expression of TRPM8 and TRPM8 channel conductivity were also repressed by salidroside; salidroside decreased the high levels of CREB activity and phosphorylation observed in the cold-stimulated cells. Furthermore, we transfected the cold-stimulated cells with CREB small interfering RNA (siRNA) to analyze TRPM8 gene expression in the absence of CREB activity. The results revealed that the cells treated with either CREB siRNA or salidroside expressed low levels of TRPM8 mRNA and protein. These results indicate that salidroside reduces MUC overproduction induced by cold stimuli and that salidroside exerts its protective effects by inhibiting TRPM8 activation, mainly by decreasing CREB activity.
Collapse
Affiliation(s)
- Qi Li
- Department of Respiratory Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | | | | | | |
Collapse
|
38
|
Liu XR, Liu Q, Chen GY, Hu Y, Sham JSK, Lin MJ. Down-regulation of TRPM8 in pulmonary arteries of pulmonary hypertensive rats. Cell Physiol Biochem 2013; 31:892-904. [PMID: 23817166 DOI: 10.1159/000350107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2013] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is characterized by profound vascular remodeling and alterations in Ca(2+) homeostasis in pulmonary arterial smooth muscle cells (PASMCs). Multiple transient receptor potential melastatin-related (TRPM) subtypes have been identified in vascular tissue. However, the changes in the expression and function of TRPM channels in pulmonary hypertension have not been characterized in detail. METHODS We examined the expression of TRPM channels and characterized the functions of the altered TRPM channels in two widely used rat models of chronic hypoxia (CH)- and monocrotaline (MCT)-induced PH. RESULTS CH-exposed and MCT-treated rats developed severe PH and right ventricular hypertrophy, with a significant decrease in TRPM8 mRNA and protein expression in pulmonary arteries (PAs). The downregulation of TRPM8 was associated with significant reduction in menthol-induced cation-influx. Time-profiles showed that TRPM8 down-regulation occurred prior to the increase of right ventricular systolic pressure (RVSP) and right ventricular mass index (RVMI) in CH-exposed rats, but these changes were delayed in MCT-treated rats. The TRPM8 agonist menthol induced vasorelaxation in phenylephrine-precontracted PAs, and the vasorelaxing effects were significantly attenuated in PAs of both PH rat models, consistent with decreased TRPM8 expression. CONCLUSION Downregulation of TRPM8 may contribute to the enhanced vasoreactivity in PH.
Collapse
Affiliation(s)
- Xiao-Ru Liu
- Department of Physiology and Pathophysiology, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | | | | | | | | | | |
Collapse
|
39
|
Kurose M, Meng ID. Dry eye modifies the thermal and menthol responses in rat corneal primary afferent cool cells. J Neurophysiol 2013; 110:495-504. [PMID: 23636717 DOI: 10.1152/jn.00222.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Dry eye syndrome is a painful condition caused by inadequate or altered tear film on the ocular surface. Primary afferent cool cells innervating the cornea regulate the ocular fluid status by increasing reflex tearing in response to evaporative cooling and hyperosmicity. It has been proposed that activation of corneal cool cells via a transient receptor potential melastatin 8 (TRPM8) channel agonist may represent a potential therapeutic intervention to treat dry eye. This study examined the effect of dry eye on the response properties of corneal cool cells and the ability of the TRPM8 agonist menthol to modify these properties. A unilateral dry eye condition was created in rats by removing the left lacrimal gland. Lacrimal gland removal reduced tears in the dry eye to 35% compared with the contralateral eye and increased the number of spontaneous blinks in the dry eye by over 300%. Extracellular single-unit recordings were performed 8-10 wk following surgery in the trigeminal ganglion of dry eye animals and age-matched controls. Responses of corneal cool cells to cooling were examined after the application of menthol (10 μM-1.0 mM) to the ocular surface. The peak frequency of discharge to cooling was higher and the cooling threshold was warmer in dry eye animals compared with controls. The dry condition also altered the neuronal sensitivity to menthol, causing desensitization to cold-evoked responses at concentrations that produced facilitation in control animals. The menthol-induced desensitization of corneal cool cells would likely result in reduced tearing, a deleterious effect in individuals with dry eye.
Collapse
Affiliation(s)
- Masayuki Kurose
- Division of Oral Physiology, Department of Oral Biological Sciences, Niigata University, Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | |
Collapse
|
40
|
Mandadi S, Armati PJ, Roufogalis BD. Protein kinase C modulation of thermo-sensitive transient receptor potential channels: Implications for pain signaling. J Nat Sci Biol Med 2012; 2:13-25. [PMID: 22470230 PMCID: PMC3312694 DOI: 10.4103/0976-9668.82311] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
A variety of molecules are reported to be involved in chronic pain. This review outlines the specifics of protein kinase C (PKC), its isoforms and their role in modulating thermo-sensitive transient receptor potential (TRP) channels TRPV1-4, TRPM8, and TRPA1. Anatomically, PKC and thermo-sensitive TRPs are co-expressed in cell bodies of nociceptive dorsal root ganglion (DRG) neurons, which are used as physiological correlates of peripheral and central projections involved in pain transmission. In the past decade, modulation of painful heat-sensitive TRPV1 by PKC has received the most attention. Recently, PKC modulation of other newly discovered thermo-sensitive pain-mediating TRPs has come into focus. Such modulation may occur under conditions of chronic pain resulting from nerve damage or inflammation. Since thermo-TRPs are primary detectors of acute pain stimuli, their modulation by PKC can severely alter their function, resulting in chronic pain. Comprehensive knowledge of pain signaling involving interaction of specific isoforms of PKC with specific thermo-sensitive TRP channels is incomplete. Such information is necessary to dissect out modality specific mechanisms to better manage the complex polymodal nature of chronic pain. This review is an attempt to update the readers on current knowledge of PKC modulation of thermo-sensitive TRPs and highlight implications of such modulation for pain signaling
Collapse
Affiliation(s)
- Sravan Mandadi
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | | | | |
Collapse
|
41
|
Klein AH, Sawyer CM, Takechi K, Davoodi A, Ivanov MA, Carstens MI, Carstens E. Topical hindpaw application of L-menthol decreases responsiveness to heat with biphasic effects on cold sensitivity of rat lumbar dorsal horn neurons. Neuroscience 2012; 219:234-42. [PMID: 22687951 PMCID: PMC3402706 DOI: 10.1016/j.neuroscience.2012.05.061] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/25/2012] [Accepted: 05/25/2012] [Indexed: 11/17/2022]
Abstract
Menthol is used in pharmaceutical applications because of its desired cooling and analgesic properties. The neural mechanism by which topical application of menthol decreases heat pain is not fully understood. We investigated the effects of topical menthol application on lumbar dorsal horn wide dynamic range and nociceptive-specific neuronal responses to noxious heat and cooling of glabrous hindpaw cutaneous receptive fields. Menthol increased thresholds for responses to noxious heat in a concentration-dependent manner. Menthol had a biphasic effect on cold-evoked responses, reducing the threshold (to warmer temperatures) at a low (1%) concentration and increasing threshold and reducing response magnitude at high (10%, 40%) concentrations. Menthol had little effect on responses to innocuous or noxious mechanical stimuli, ruling out a local anesthetic action. Application of 40% menthol to the contralateral hindpaw tended to reduce responses to cooling and noxious heat, suggesting a weak heterosegmental inhibitory effect. These results indicate that menthol has an analgesic effect on heat sensitivity of nociceptive dorsal horn neurons, as well as biphasic effects on cold sensitivity, consistent with previous behavioral observations.
Collapse
Affiliation(s)
- Amanda H. Klein
- Department of Neurobiology, Physiology and Behavior University of California, Davis 1 Shields Avenue Davis, CA 95616
| | - Carolyn M. Sawyer
- Department of Neurobiology, Physiology and Behavior University of California, Davis 1 Shields Avenue Davis, CA 95616
| | - Kenichi Takechi
- Department of Neurobiology, Physiology and Behavior University of California, Davis 1 Shields Avenue Davis, CA 95616
| | - Auva Davoodi
- Department of Neurobiology, Physiology and Behavior University of California, Davis 1 Shields Avenue Davis, CA 95616
| | - Margaret A. Ivanov
- Department of Neurobiology, Physiology and Behavior University of California, Davis 1 Shields Avenue Davis, CA 95616
| | - Mirela Iodi Carstens
- Department of Neurobiology, Physiology and Behavior University of California, Davis 1 Shields Avenue Davis, CA 95616
| | - E Carstens
- Department of Neurobiology, Physiology and Behavior University of California, Davis 1 Shields Avenue Davis, CA 95616
| |
Collapse
|
42
|
Premkumar LS, Abooj M. TRP channels and analgesia. Life Sci 2012; 92:415-24. [PMID: 22910182 DOI: 10.1016/j.lfs.2012.08.010] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 07/25/2012] [Accepted: 08/02/2012] [Indexed: 01/07/2023]
Abstract
Since cloning and characterizing the first nociceptive ion channel Transient Receptor Potential (TRP) Vanilloid 1 (TRPV1), other TRP channels involved in nociception have been cloned and characterized, which include TRP Vanilloid 2 (TRPV2), TRP Vanilloid 3 (TRPV3), TRP Vanilloid 4 (TRPV4), TRP Ankyrin 1 (TRPA1) and TRP Melastatin 8 (TRPM8), more recently TRP Canonical 1, 5, 6 (TRPC1, 5, 6), TRP Melastatin 2 (TRPM2) and TRP Melastatin 3 (TRPM3). These channels are predominantly expressed in C and Aδ nociceptors and transmit noxious thermal, mechanical and chemical sensitivities. TRP channels are modulated by pro-inflammatory mediators, neuropeptides and cytokines. Significant advances have been made targeting these receptors either by antagonists or agonists to treat painful conditions. In this review, we will discuss TRP channels as targets for next generation analgesics and the side effects that may ensue as a result of blocking/activating these receptors, because they are also involved in physiological functions such as release of vasoactive neuropeptides and regulation of vascular tone, maintenance of the body temperature, gastrointestinal motility, urinary bladder control, etc.
Collapse
Affiliation(s)
- Louis S Premkumar
- Department of Pharmacology, Southern Illinois University School of Medicine Springfield, IL 62702, USA.
| | | |
Collapse
|
43
|
Yudin Y, Rohacs T. Regulation of TRPM8 channel activity. Mol Cell Endocrinol 2012; 353:68-74. [PMID: 22061619 PMCID: PMC3295897 DOI: 10.1016/j.mce.2011.10.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 10/21/2011] [Accepted: 10/22/2011] [Indexed: 11/28/2022]
Abstract
Transient Receptor Potential Melastatin 8 (TRPM8) is a Ca(2+) permeable non-selective cation channel directly activated by cold temperatures and chemical agonists such as menthol. It is a well established sensor of environmental cold temperatures, found in peripheral sensory neurons, where its activation evokes depolarization and action potentials. The activity of TRPM8 is regulated by a number of cellular signaling pathways, most notably by phosphoinositides and the activation of phospholipase C. This review will summarize current knowledge on the physiological and pathophysiological roles of TRPM8 and its regulation by various intracellular messenger molecules and signaling pathways.
Collapse
Affiliation(s)
- Yevgen Yudin
- Department of Pharmacology and Physiology, UMDNJ - New Jersey Medical School, Newark, NJ 07103, USA
| | | |
Collapse
|
44
|
Matthews JM, Qin N, Colburn RW, Dax SL, Hawkins M, McNally JJ, Reany L, Youngman MA, Baker J, Hutchinson T, Liu Y, Lubin ML, Neeper M, Brandt MR, Stone DJ, Flores CM. The design and synthesis of novel, phosphonate-containing transient receptor potential melastatin 8 (TRPM8) antagonists. Bioorg Med Chem Lett 2012; 22:2922-6. [PMID: 22421018 DOI: 10.1016/j.bmcl.2012.02.060] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 11/17/2022]
Abstract
A series of benzothiophene-based phosphonates was synthesized and many analogs within the series were shown to be potent antagonists of the TRPM8 channel. The compounds were obtained as a racemic mixture in 5 synthetic steps, and were tested for TRPM8 antagonist activity in a recombinant, canine TRPM8-expressing cell line using a fluorometric imaging plate reader (FLIPR) assay. Structure-activity relationships were developed initially by modification of the core structure and subsequently by variation of the aromatic substituents and the phosphonate ester. Compound 9l was administered intraperitoneally to rats and demonstrated engagement of the TRPM8 target in both prevention and reversal-modes in an icilin-induced 'wet-dog' shake model.
Collapse
Affiliation(s)
- Jay M Matthews
- Janssen Pharmaceuticals, Spring House, PA 19477-0776, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Latorre R, Brauchi S, Madrid R, Orio P. A cool channel in cold transduction. Physiology (Bethesda) 2012; 26:273-85. [PMID: 21841075 DOI: 10.1152/physiol.00004.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Transient receptor potential melastatin 8 (TRPM8), a calcium-permeable cation channel activated by cold, cooling compounds and voltage, is the main molecular entity responsible for detection of cold temperatures in the somatosensory system. Here, we review the biophysical properties, physiological role, and near-membrane trafficking of this exciting polymodal ion channel.
Collapse
Affiliation(s)
- Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | | | | |
Collapse
|
46
|
Behavioral Testing of the Effects of Thermosensitive TRP Channel Agonists on Touch, Temperature, and Pain Sensations. NEUROPHYSIOLOGY+ 2011. [DOI: 10.1007/s11062-011-9222-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Yudin Y, Lukacs V, Cao C, Rohacs T. Decrease in phosphatidylinositol 4,5-bisphosphate levels mediates desensitization of the cold sensor TRPM8 channels. J Physiol 2011; 589:6007-27. [PMID: 22005680 DOI: 10.1113/jphysiol.2011.220228] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The activity of the cold- and menthol-activated transient receptor potential melastatin 8 (TRPM8) channels diminishes over time in the presence of extracellular Ca(2+), a phenomenon referred to as desensitization or adaptation. Here we show that activation of TRPM8 by cold or menthol evokes a decrease in cellular phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] levels. The decrease in PtdIns(4,5)P(2) levels was accompanied by increased inositol 1,4,5 trisphosphate (InsP(3)) production, and was inhibited by loading the cells with the Ca(2+) chelator BAPTA-AM, showing that it was the consequence of the activation of phospholipase C (PLC) by increased intracellular Ca(2+) concentrations. PtdIns(4,5)P(2) hydrolysis showed excellent temporal correlation with current desensitization in simultaneous patch clamp and fluorescence-based PtdIns(4,5)P(2) level measurements. Intracellular dialysis of PtdIns(4,5)P(2) inhibited desensitization both in native neuronal and recombinant TRPM8 channels. PtdIns(4)P, the precursor of PtdIns(4,5)P(2), did not inhibit desensitization, consistent with its minimal effect in excised patches. Omission of MgATP from the intracellular solution accelerated desensitization, and MgATP reactivated TRPM8 channels in excised patches in a phosphatidylinositol 4-kinase (PI4K)-dependent manner. PLC-independent depletion of PtdIns(4,5)P(2) using a voltage-sensitive phosphatase (ci-VSP) inhibited TRPM8 currents, and omission of ATP from the intracellular solution inhibited recovery from this inhibition. Inhibitors of PKC had no effect on the kinetics of desensitization. We conclude that Ca(2+) influx through TRPM8 activates a Ca(2+)-sensitive PLC isoform, and the resulting depletion of PtdIns(4,5)P(2) plays a major role in desensitization of both cold and menthol responses.
Collapse
Affiliation(s)
- Yevgen Yudin
- Department of Pharmacology and Physiology, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, NJ 07103, USA
| | | | | | | |
Collapse
|
48
|
Darszon A, Nishigaki T, Beltran C, Treviño CL. Calcium Channels in the Development, Maturation, and Function of Spermatozoa. Physiol Rev 2011; 91:1305-55. [DOI: 10.1152/physrev.00028.2010] [Citation(s) in RCA: 269] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A proper dialogue between spermatozoa and the egg is essential for conception of a new individual in sexually reproducing animals. Ca2+ is crucial in orchestrating this unique event leading to a new life. No wonder that nature has devised different Ca2+-permeable channels and located them at distinct sites in spermatozoa so that they can help fertilize the egg. New tools to study sperm ionic currents, and image intracellular Ca2+ with better spatial and temporal resolution even in swimming spermatozoa, are revealing how sperm ion channels participate in fertilization. This review critically examines the involvement of Ca2+ channels in multiple signaling processes needed for spermatozoa to mature, travel towards the egg, and fertilize it. Remarkably, these tiny specialized cells can express exclusive channels like CatSper for Ca2+ and SLO3 for K+, which are attractive targets for contraception and for the discovery of novel signaling complexes. Learning more about fertilization is a matter of capital importance; societies face growing pressure to counteract rising male infertility rates, provide safe male gamete-based contraceptives, and preserve biodiversity through improved captive breeding and assisted conception initiatives.
Collapse
Affiliation(s)
- Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Takuya Nishigaki
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Carmen Beltran
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Claudia L. Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
49
|
Ciuman RR. Phytotherapeutic and naturopathic adjuvant therapies in otorhinolaryngology. Eur Arch Otorhinolaryngol 2011; 269:389-97. [PMID: 21922427 PMCID: PMC3259400 DOI: 10.1007/s00405-011-1755-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 08/23/2011] [Indexed: 11/26/2022]
Abstract
Phytotherapeutic pharmaceuticals and herbal medicinal products with its roots in classical phytotherapeutic medicine have a well-established role in otolaryngological therapy, especially for diseases of the upper airways and acute and chronic infections. A thorough selection and application could mean huge benefit for the patient, in particular in cases with contraindications, chemo- and antibiotic resistance or patient request. Besides, it might spare other medications. Phytotherapeutic pharmaceuticals must fulfil the same criteria of quality, effectiveness and harmlessness of evidence-based medicine like chemical pharmaceuticals, although they are often prescribed due to its well established or traditional based use. This review focuses on phytotherapeutic therapies well established within the European Community for otolaryngologic disease patterns by referring to clinical studies or meta-analysis.
Collapse
Affiliation(s)
- Raphael Richard Ciuman
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Bochum, St. Elisabeth-Hospital, Bleichstr. 15, 44787, Bochum, Germany.
| |
Collapse
|
50
|
Sarria I, Ling J, Zhu MX, Gu JG. TRPM8 acute desensitization is mediated by calmodulin and requires PIP(2): distinction from tachyphylaxis. J Neurophysiol 2011; 106:3056-66. [PMID: 21900509 DOI: 10.1152/jn.00544.2011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cold-sensing channel transient receptor potential melastatin 8 (TRPM8) features Ca(2+)-dependent downregulation, a cellular process underlying somatosensory accommodation in cold environments. The Ca(2+)-dependent functional downregulation of TRPM8 is manifested with two distinctive phases, acute desensitization and tachyphylaxis. Here we show in rat dorsal root ganglion neurons that TRPM8 acute desensitization critically depends on phosphatidylinositol 4,5-bisphosphate (PIP(2)) availability rather than PIP(2) hydrolysis and is triggered by calmodulin activation. Tachyphylaxis, on the other hand, is mediated by phospholipase hydrolysis of PIP(2) and protein kinase C/phosphatase 1,2A. We further demonstrate that PIP(2) switches TRPM8 channel gating to a high-open probability state with short closed times. Ca(2+)-calmodulin reverses the effect of PIP(2), switching channel gating to a low-open probability state with long closed times. Thus, through gating modulation, Ca(2+)-calmodulin provides a mechanism to rapidly regulate TRPM8 functions in the somatosensory system.
Collapse
Affiliation(s)
- Ignacio Sarria
- Department of Anesthesiology and Graduate Program in Neuroscience, The University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | | |
Collapse
|