1
|
Song DE, Rawal D, Lee WJ, Shim WS. Sphingosylphosphorylcholine induces itch via activation of TRPM3 and TRPA1 in mice. Biochem Pharmacol 2025; 237:116952. [PMID: 40274130 DOI: 10.1016/j.bcp.2025.116952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/19/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Itch is a prevalent symptom in atopic dermatitis (AD), often leading to a strong urge to scratch. Elevated levels of sphingosylphosphorylcholine (SPC) are found in the stratum corneum of AD patients, and while SPC is known to induce itch, its molecular targets are not well understood. This study aims to identify the signaling pathway of SPC-induced itch under AD conditions. We demonstrate that SPC specifically activates the Transient Receptor Potential Melastatin 3 (TRPM3) channel in sensory neurons. In HEK293T cells expressing TRPM3, SPC treatment caused a significant increase in intracellular calcium, which was inhibited by TRPM3 antagonists. Among various TRP channels tested, TRPM3 exhibited the highest reactivity to SPC, followed by TRPA1. Molecular docking analysis also supported interactions between SPC and both TRPM3 and TRPA1. In an AD mouse model, SPC-induced responses were dependent on TRPM3 and TRPA1, and the expression of these channels increased in dorsal root ganglion neurons. SPC-induced scratching behaviors were significantly reduced by TRPM3 and TRPA1 antagonists, with TRPM3 playing a critical role in spontaneous scratching. This study identifies TRPM3 and TRPA1 as key mediators of SPC-induced itch, providing potential therapeutic targets for treating itch in AD patients.
Collapse
Affiliation(s)
- Da Eun Song
- College of Pharmacy, Gachon University, Incheon, South Korea; Gachon Institute of Pharmaceutical Sciences, Incheon, South Korea
| | - Diwas Rawal
- College of Pharmacy, Gachon University, Incheon, South Korea; Gachon Institute of Pharmaceutical Sciences, Incheon, South Korea
| | - Wook-Joo Lee
- College of Pharmacy, Gachon University, Incheon, South Korea; Gachon Institute of Pharmaceutical Sciences, Incheon, South Korea
| | - Won-Sik Shim
- College of Pharmacy, Gachon University, Incheon, South Korea; Gachon Institute of Pharmaceutical Sciences, Incheon, South Korea.
| |
Collapse
|
2
|
Huang Z, Li X, Wang X, Wu J, Gong Z, Kõks S, Wang M. Nuclear paraspeckle assembly transcript 1 promotes photophobia behavior in mice via miR-196a-5p/Trpm3 coupling. J Headache Pain 2025; 26:118. [PMID: 40399770 PMCID: PMC12096706 DOI: 10.1186/s10194-025-02057-5] [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: 03/18/2025] [Accepted: 04/30/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The long noncoding RNA, NEAT1, is recognized as a key regulator of proinflammatory gene expression; Yet, its functional role in migraine remains unexplored, despite the central role of neuroinflammatory mechanisms in migraine pathophysiology. This study examines the implication of NEAT1 in the trigeminal ganglion activation, which underlies photophobia associated with migraine. METHODS Light aversion behavior was induced by intranasal injection of the TRPA1 activator, umbellulone. Male mouse behavior was assessed by the total time the mouse stays in the light between the dark and light compartments. To gain insight to the NEAT1-mediated photophobia mechanism, gene expression of candidate genes and non-coding RNAs interactions were assessed using RNA-sequencing, qPCR analysis, histology and dual-luciferase reporter gene assay. RESULTS NEAT1 was upregulated in the trigeminal ganglion of male photophobia mice; Downregulation of NEAT1 by intravenous injection of shNEAT1 adeno-associated virus vectors attenuated NEAT1 expression and alleviated photophobia-like behavior in mice. The elevated NEAT1 expression in the trigeminal ganglion of photophobia mice corresponds to the downregulation of miR-196a-5p and upregulation Trpm3 RNA level. Predicted analysis suggested NEAT1/miR-196a-5p ceRNA network exists in photophobia mice. Indeed, knocking down NEAT1 upregulated miR-196a-5p, whilst downregulated Trpm3 gene expression level, in the trigeminal ganglion of photophobia mice. Further investigation using dual-luciferase reporter gene assay identified NEAT1 interacting with miR-196a-5p, whilst miR-196a-5p interacting with Trpm3. Similar to knocking down NEAT1, TRPM3 inhibition reduced photophobia-like behavior. CONCLUSION We conclude that NEAT1 is critical for promoting photophobia behavior via miR-196a-5p/Trpm3 coupling.
Collapse
Affiliation(s)
- Zhuoan Huang
- Department of Biosciences and Bioinformatics, School of Science, Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Xingshen Li
- Department of Biosciences and Bioinformatics, School of Science, Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, 215123, China
| | - Xiaolin Wang
- Department of Biosciences and Bioinformatics, School of Science, Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, 215123, China
| | - Jiaqi Wu
- Department of Biosciences and Bioinformatics, School of Science, Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, 215123, China
| | - Ziyang Gong
- Department of Biosciences and Bioinformatics, School of Science, Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Sulev Kõks
- Personalised Medicine Centre, Perron Institute for Neurological and Translational Science, Murdoch University, Perth, WA, 6009, Australia.
| | - Minyan Wang
- Department of Biosciences and Bioinformatics, School of Science, Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, 215123, China.
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK.
| |
Collapse
|
3
|
Krivoshein G, Rivera-Mancilla E, MaassenVanDenBrink A, Giniatullin R, van den Maagdenberg AMJM. Sex difference in TRPM3 channel functioning in nociceptive and vascular systems: an emerging target for migraine therapy in females? J Headache Pain 2025; 26:40. [PMID: 39994546 PMCID: PMC11853570 DOI: 10.1186/s10194-025-01966-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Transient Receptor Potential Melastatin 3 (TRPM3) channels are Ca2+ permeable ion channels that act as polymodal sensors of mechanical, thermal, and various chemical stimuli. TRPM3 channels are highly expressed in the trigeminovascular system, including trigeminal neurons and the vasculature. Their presence in dural afferents suggests that they are potential triggers of migraine pain, which is originating from the meningeal area. This area is densely innervated by autonomous and trigeminal nerves that contain the major migraine mediator calcitonin gene-related peptide (CGRP) in peptidergic nerve fibers. Co-expression of TRPM3 channels and CGRP receptors in meningeal nerves suggests a potential interplay between both signalling systems. Compared to other members of the TRP family, TRPM3 channels have a high sensitivity to sex hormones and to the endogenous neurosteroid pregnenolone sulfate (PregS). The predominantly female sex hormones estrogen and progesterone, of which the levels drop during menses, act as natural inhibitors of TRPM3 channels, while PregS is a known endogenous agonist of these channels. A decrease in sex hormone levels has also been suggested as trigger for attacks of menstrually-related migraine. Notably, there is a remarkable sex difference in TRPM3-mediated effects in trigeminal nociceptive signalling and the vasculature. In line with this, the relaxation of human isolated meningeal arteries induced by the activation of TRPM3 channels is greater in females. Additionally, the sex-dependent vasodilatory responses to CGRP in meningeal arteries seem to be influenced by age-related hormonal changes, which could contribute to sex differences in migraine pathology. Consistent with these observations, activation of TRPM3 channels triggers nociceptive sensory firing much more prominently in female than male mouse meninges, suggesting that pain processing in female patients with migraine may differ. Overall, the combined TRPM3-related neuronal and vascular mechanisms could provide a possible explanation for the higher prevalence and even the more severe quality of migraine attacks in females. This narrative review summarizes recent data on the sex-dependent roles of TRPM3 channels in migraine pathophysiology, the potential interplay between TRPM3 and CGRP signalling, and highlights the prospects for translational therapies targeting TRPM3 channels, which may be of particular relevance for women with migraine.
Collapse
Affiliation(s)
- Georgii Krivoshein
- Departments of Human Genetics and Neurology, Leiden University Medical Center, PO Box 9600 2300 RC, Leiden, The Netherlands
| | - Eduardo Rivera-Mancilla
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rashid Giniatullin
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Arn M J M van den Maagdenberg
- Departments of Human Genetics and Neurology, Leiden University Medical Center, PO Box 9600 2300 RC, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
4
|
Kumar S, Jin F, Park SJ, Choi W, Keuning SI, Massimino RP, Vu S, Lü W, Du J. Convergent Agonist and Heat Activation of Nociceptor TRPM3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634542. [PMID: 39896661 PMCID: PMC11785169 DOI: 10.1101/2025.01.23.634542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Detecting noxious heat is vital for survival, triggering pain responses that protect against harm1,2. The TRPM3 channel is a key nociceptor for sensing noxious heat and a promising therapeutic target for pain treatment and neurological disorders such as epilepsy3-11. Here, we functionally and structurally characterized TRPM3 in response to diverse stimuli: the synthetic superagonist CIM0216 Ref12, the anticonvulsant antagonist primidone13,14, and heat1,10,15. Our findings reveal that TRPM3 is intrinsically dynamic, with its intracellular domain (ICD) sampling both resting and activated states, though strongly favoring the resting state without stimulation. CIM0216 binds to the S1-S4 domain, inducing conformational changes in the ICD and shifting the equilibrium toward activation. Remarkably, heat induces similar ICD rearrangements, revealing a converged activation mechanism driven by chemical compounds and temperature. This mechanism is supported by functional data showing that mutations facilitating the ICD movement markedly increase the sensitivity of TRPM3 to both chemical and thermal signals. These findings establish a critical role of the ICD in temperature sensing in TRPM3, a mechanism likely conserved across the TRPM family. Finally, we show that primidone binds to the same site as CIM0216 but acts as an antagonist. This study provides a framework for understanding the thermal sensing mechanisms of temperature-sensitive ion channels and offers a structural foundation for developing TRPM3-target therapeutics for pain and neurological disorders.
Collapse
Affiliation(s)
- Sushant Kumar
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | - Sung Jin Park
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | - Sarah I. Keuning
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | | | - Wei Lü
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Juan Du
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
5
|
Eaton-Fitch N, Muraki K, Sasso EM, Magawa C, Marshall-Gradisnik S. Analysis of Transient Receptor Potential Ion Channels in ME/CFS. Methods Mol Biol 2025; 2920:83-99. [PMID: 40372679 DOI: 10.1007/978-1-0716-4498-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
This chapter provides a comprehensive overview of methodologies currently employed to study ion channels, particularly transient receptor potential melastatin 3 (TRPM3) in the context of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Sample preparation involves the collection of whole blood, separation of peripheral blood mononuclear cells (PBMCs) via density gradient centrifugation, and isolation of natural killer (NK) cells. Protein expression analysis utilizes flow cytometry, liquid chromatography-mass spectrometry (LC-MS), western blotting, and immunofluorescence techniques. Functional analysis focuses on calcium imaging and electrophysiology techniques to investigate ion channel responses to pharmacological stimuli. The authors highlight that some experimental protocols included within this chapter require specialized training and equipment. In order to replicate these protocols extended training is advised, specifically when attempting electrophysiology experimentation. The use of advanced techniques for detailed analysis provides insights into ion channel function and potential implications in the pathomechanism of ME/CFS offering avenues for further research and therapeutic exploration.
Collapse
Affiliation(s)
- Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia.
- Consortium Health International for ME, Griffith University, Gold Coast, QLD, Australia.
| | - Katsuhiko Muraki
- Consortium Health International for ME, Griffith University, Gold Coast, QLD, Australia
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Etianne Martini Sasso
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
- Consortium Health International for ME, Griffith University, Gold Coast, QLD, Australia
| | - Chandi Magawa
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
- Consortium Health International for ME, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
- Consortium Health International for ME, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
6
|
Pawelak A, Polczyk A, Wolańska E, Kłaniewska M, Biela M, Basiak A, Franaszczyk M, Rydzanicz M, Płoski R, Śmigiel R. Case Report: Expanded delineation of phenotype of TRPM3-related neurodevelopmental disorders. Front Pediatr 2024; 12:1435053. [PMID: 39639951 PMCID: PMC11617146 DOI: 10.3389/fped.2024.1435053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
The TRPM3 gene, part of the transient receptor potential (TRP) cation channel family, plays crucial roles in sensory perception and ion transport. Mutations in TRPM3 are linked to a range of neurological and developmental disorders. The c.2509G>A variant specifically leads to a substitution at position 837 in the protein, which is likely critical for its normal function. This study presents a male pediatric patient with a pathogenic TRPM3 variant c.2509G>A [p.(Val837Met)], contributing to a complex clinical phenotype characterized by developmental delays, significant hypotonia, and neurological abnormalities. The patient demonstrated delayed motor milestones, including the inability to sit independently until 20 months, and abnormal EEG findings without epileptic seizures. Ophthalmologic issues, such as hyperopia and astigmatism, were also identified. Behavioral abnormalities and cognitive impairment aligned with previous reports of TRPM3-related neurodevelopmental disorders. This case highlights the phenotypic variability linked to the p.(Val837Met) variant and emphasizes the need for further research into effective therapeutic strategies for TRPM3-associated conditions.
Collapse
Affiliation(s)
- Agnieszka Pawelak
- Department of Genetics, Medical University of Wroclaw, Wroclaw, Poland
| | - Artur Polczyk
- Medical Education and Simulation Laboratory, University Centre of Physiotherapy and Rehabilitation, Wroclaw Medical University, Wroclaw, Poland
| | - Ewelina Wolańska
- Department of Family and Pediatric Nursing, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Kłaniewska
- Department of Pediatrics, Endocrinology, Diabetology and Metabolic Diseases, Medical University of Wroclaw, Wroclaw, Poland
| | - Mateusz Biela
- Department of Pediatrics, Endocrinology, Diabetology and Metabolic Diseases, Medical University of Wroclaw, Wroclaw, Poland
| | - Aleksander Basiak
- Department of Pediatrics, Endocrinology, Diabetology and Metabolic Diseases, Medical University of Wroclaw, Wroclaw, Poland
| | - Maria Franaszczyk
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | | | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Robert Śmigiel
- Department of Pediatrics, Endocrinology, Diabetology and Metabolic Diseases, Medical University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
7
|
Behrendt M. Implications of TRPM3 and TRPM8 for sensory neuron sensitisation. Biol Chem 2024; 405:583-599. [PMID: 39417661 DOI: 10.1515/hsz-2024-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Sensory neurons serve to receive and transmit a wide range of information about the conditions of the world around us as well as the external and internal state of our body. Sensitisation of these nerve cells, i.e. becoming more sensitive to stimuli or the emergence or intensification of spontaneous activity, for example in the context of inflammation or nerve injury, can lead to chronic diseases such as neuropathic pain. For many of these disorders there are only very limited treatment options and in order to find and establish new therapeutic approaches, research into the exact causes of sensitisation with the elucidation of the underlying mechanisms and the identification of the molecular components is therefore essential. These components include plasma membrane receptors and ion channels that are involved in signal reception and transmission. Members of the transient receptor potential (TRP) channel family are also expressed in sensory neurons and some of them play a crucial role in temperature perception. This review article focuses on the heat-sensitive TRPM3 and the cold-sensitive TRPM8 (and TRPA1) channels and their importance in sensitisation of dorsal root ganglion sensory neurons is discussed based on studies related to inflammation and injury- as well as chemotherapy-induced neuropathy.
Collapse
Affiliation(s)
- Marc Behrendt
- Experimental Pain Research, Medical Faculty Mannheim, Heidelberg University, MCTN, Tridomus, Building C, Ludolf-Krehl-Straße 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
8
|
Gandini MA, Zamponi GW. Navigating the Controversies: Role of TRPM Channels in Pain States. Int J Mol Sci 2024; 25:10284. [PMID: 39408620 PMCID: PMC11476983 DOI: 10.3390/ijms251910284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/17/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Chronic pain is a debilitating condition that affects up to 1.5 billion people worldwide and bears a tremendous socioeconomic burden. The success of pain medicine relies on our understanding of the type of pain experienced by patients and the mechanisms that give rise to it. Ion channels are among the key targets for pharmacological intervention in chronic pain conditions. Therefore, it is important to understand how changes in channel properties, trafficking, and molecular interactions contribute to pain sensation. In this review, we discuss studies that have demonstrated the involvement of transient receptor potential M2, M3, and M8 channels in pain generation and transduction, as well as the controversies surrounding these findings.
Collapse
Affiliation(s)
- Maria A. Gandini
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Clinical Neurosciences, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Gerald W. Zamponi
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Clinical Neurosciences, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
9
|
Löhn M, Wirth KJ. Potential pathophysiological role of the ion channel TRPM3 in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and the therapeutic effect of low-dose naltrexone. J Transl Med 2024; 22:630. [PMID: 38970055 PMCID: PMC11227206 DOI: 10.1186/s12967-024-05412-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/17/2024] [Indexed: 07/07/2024] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating disease with a broad overlap of symptomatology with Post-COVID Syndrome (PCS). Despite the severity of symptoms and various neurological, cardiovascular, microvascular, and skeletal muscular findings, no biomarkers have been identified. The Transient receptor potential melastatin 3 (TRPM3) channel, involved in pain transduction, thermosensation, transmitter and neuropeptide release, mechanoregulation, vasorelaxation, and immune defense, shows altered function in ME/CFS. Dysfunction of TRPM3 in natural killer (NK) cells, characterized by reduced calcium flux, has been observed in ME/CFS and PCS patients, suggesting a role in ineffective pathogen clearance and potential virus persistence and autoimmunity development. TRPM3 dysfunction in NK cells can be improved by naltrexone in vitro and ex vivo, which may explain the moderate clinical efficacy of low-dose naltrexone (LDN) treatment. We propose that TRPM3 dysfunction may have a broader involvement in ME/CFS pathophysiology, affecting other organs. This paper discusses TRPM3's expression in various organs and its potential impact on ME/CFS symptoms, with a focus on small nerve fibers and the brain, where TRPM3 is involved in presynaptic GABA release.
Collapse
Affiliation(s)
- Matthias Löhn
- Institute for General Pharmacology and Toxicology, University Hospital, Goethe University, Frankfurt am Main, Germany.
| | - Klaus Josef Wirth
- Institute for General Pharmacology and Toxicology, University Hospital, Goethe University, Frankfurt am Main, Germany.
- Mitodicure GmbH, D-65830, Kriftel, Germany.
| |
Collapse
|
10
|
Roelens R, Peigneur ANF, Voets T, Vriens J. Neurodevelopmental disorders caused by variants in TRPM3. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119709. [PMID: 38522727 DOI: 10.1016/j.bbamcr.2024.119709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
Developmental and epileptic encephalopathies (DEE) are a broad and varied group of disorders that affect the brain and are characterized by epilepsy and comorbid intellectual disability (ID). These conditions have a broad spectrum of symptoms and can be caused by various underlying factors, including genetic mutations, infections, and other medical conditions. The exact cause of DEE remains largely unknown in the majority of cases. However, in around 25 % of patients, rare nonsynonymous coding variants in genes encoding ion channels, cell-surface receptors, and other neuronally expressed proteins are identified. This review focuses on a subgroup of DEE patients carrying variations in the gene encoding the Transient Receptor Potential Melastatin 3 (TRPM3) ion channel, where recent data indicate that gain-of-function of TRPM3 channel activity underlies a spectrum of dominant neurodevelopmental disorders.
Collapse
Affiliation(s)
- Robbe Roelens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Laboratory of Ion Channel Research, Department of Molecular Medicine, KU Leuven, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Ana Nogueira Freitas Peigneur
- Laboratory of Ion Channel Research, Department of Molecular Medicine, KU Leuven, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Molecular Medicine, KU Leuven, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Laboratory of Ion Channel Research, Department of Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
11
|
Gou R, Liu Y, Gou L, Mi S, Li X, Yang Y, Cheng X, Zhang Y. Transient Receptor Potential Channels in Sensory Mechanisms of the Lower Urinary Tract. Urol Int 2024; 108:464-476. [PMID: 38657590 DOI: 10.1159/000538855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Urine storage and excretion require a network of interactions in the urinary tract and the central nervous system, which is mediated by a reservoir of water in the bladder and the outlet to the bladder neck, urethra, and external urethral sphincter. Through communicating and coordinating each other, micturition system eventually showed a switch-like activity pattern. SUMMARY At cervicothoracic and lumbosacral spine, the spinal reflex pathway of the lower urinary tract (LUT) received mechanosensory input from the urothelium to regulate the bladder contraction activity, thereby controlled urination voluntarily. Impairment of above-mentioned any level could result in lower urinary tract dysfunction, placed a huge burden on patients and society. Specific expression of purinergic receptors and transient receptor potential (TRP) channels are thought to play an important role in urinary excretion in the LUT. KEY MESSAGES This article reviewed the knowledge about the voiding reflex and described the role and function of TRP channels during voiding.
Collapse
Affiliation(s)
- Ruiqiang Gou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China,
| | - Yuanyuan Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Li Gou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Shengyan Mi
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaonan Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yichen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaorong Cheng
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yibao Zhang
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
12
|
Zhou Y, Bennett TM, Ruzycki PA, Guo Z, Cao YQ, Shahidullah M, Delamere NA, Shiels A. A Cataract-Causing Mutation in the TRPM3 Cation Channel Disrupts Calcium Dynamics in the Lens. Cells 2024; 13:257. [PMID: 38334649 PMCID: PMC10854584 DOI: 10.3390/cells13030257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
TRPM3 belongs to the melastatin sub-family of transient receptor potential (TRPM) cation channels and has been shown to function as a steroid-activated, heat-sensitive calcium ion (Ca2+) channel. A missense substitution (p.I65M) in the TRPM3 gene of humans (TRPM3) and mice (Trpm3) has been shown to underlie an inherited form of early-onset, progressive cataract. Here, we model the pathogenetic effects of this cataract-causing mutation using 'knock-in' mutant mice and human cell lines. Trpm3 and its intron-hosted micro-RNA gene (Mir204) were strongly co-expressed in the lens epithelium and other non-pigmented and pigmented ocular epithelia. Homozygous Trpm3-mutant lenses displayed elevated cytosolic Ca2+ levels and an imbalance of sodium (Na+) and potassium (K+) ions coupled with increased water content. Homozygous TRPM3-mutant human lens epithelial (HLE-B3) cell lines and Trpm3-mutant lenses exhibited increased levels of phosphorylated mitogen-activated protein kinase 1/extracellular signal-regulated kinase 2 (MAPK1/ERK2/p42) and MAPK3/ERK1/p44. Mutant TRPM3-M65 channels displayed an increased sensitivity to external Ca2+ concentration and an altered dose response to pregnenolone sulfate (PS) activation. Trpm3-mutant lenses shared the downregulation of genes involved in insulin/peptide secretion and the upregulation of genes involved in Ca2+ dynamics. By contrast, Trpm3-deficient lenses did not replicate the pathophysiological changes observed in Trpm3-mutant lenses. Collectively, our data suggest that a cataract-causing substitution in the TRPM3 cation channel elicits a deleterious gain-of-function rather than a loss-of-function mechanism in the lens.
Collapse
Affiliation(s)
- Yuefang Zhou
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas M. Bennett
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Philip A. Ruzycki
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhaohua Guo
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yu-Qing Cao
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mohammad Shahidullah
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Nicholas A. Delamere
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Alan Shiels
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
13
|
Vydra Bousova K, Zouharova M, Jiraskova K, Vetyskova V. Interaction of Calmodulin with TRPM: An Initiator of Channel Modulation. Int J Mol Sci 2023; 24:15162. [PMID: 37894842 PMCID: PMC10607381 DOI: 10.3390/ijms242015162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Transient receptor potential melastatin (TRPM) channels, a subfamily of the TRP superfamily, constitute a diverse group of ion channels involved in mediating crucial cellular processes like calcium homeostasis. These channels exhibit complex regulation, and one of the key regulatory mechanisms involves their interaction with calmodulin (CaM), a cytosol ubiquitous calcium-binding protein. The association between TRPM channels and CaM relies on the presence of specific CaM-binding domains in the channel structure. Upon CaM binding, the channel undergoes direct and/or allosteric structural changes and triggers down- or up-stream signaling pathways. According to current knowledge, ion channel members TRPM2, TRPM3, TRPM4, and TRPM6 are directly modulated by CaM, resulting in their activation or inhibition. This review specifically focuses on the interplay between TRPM channels and CaM and summarizes the current known effects of CaM interactions and modulations on TRPM channels in cellular physiology.
Collapse
|
14
|
Barnden L, Thapaliya K, Eaton-Fitch N, Barth M, Marshall-Gradisnik S. Altered brain connectivity in Long Covid during cognitive exertion: a pilot study. Front Neurosci 2023; 17:1182607. [PMID: 37425014 PMCID: PMC10323677 DOI: 10.3389/fnins.2023.1182607] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Debilitating Long-Covid symptoms occur frequently after SARS-COVID-19 infection. Methods Functional MRI was acquired in 10 Long Covid (LCov) and 13 healthy controls (HC) with a 7 Tesla scanner during a cognitive (Stroop color-word) task. BOLD time series were computed for 7 salience and 4 default-mode network hubs, 2 hippocampus and 7 brainstem regions (ROIs). Connectivity was characterized by the correlation coefficient between each pair of ROI BOLD time series. We tested for HC versus LCov differences in connectivity between each pair of the 20 regions (ROI-to-ROI) and between each ROI and the rest of the brain (ROI-to-voxel). For LCov, we also performed regressions of ROI-to-ROI connectivity with clinical scores. Results Two ROI-to-ROI connectivities differed between HC and LCov. Both involved the brainstem rostral medulla, one connection to the midbrain, another to a DM network hub. Both were stronger in LCov than HC. ROI-to-voxel analysis detected multiple other regions where LCov connectivity differed from HC located in all major lobes. Most, but not all connections, were weaker in LCov than HC. LCov, but not HC connectivity, was correlated with clinical scores for disability and autonomic function and involved brainstem ROI. Discussion Multiple connectivity differences and clinical correlations involved brainstem ROIs. Stronger connectivity in LCov between the medulla and midbrain may reflect a compensatory response. This brainstem circuit regulates cortical arousal, autonomic function and the sleep-wake cycle. In contrast, this circuit exhibited weaker connectivity in ME/CFS. LCov connectivity regressions with disability and autonomic scores were consistent with altered brainstem connectivity in LCov.
Collapse
Affiliation(s)
- Leighton Barnden
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Kiran Thapaliya
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Markus Barth
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, QLD, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| |
Collapse
|
15
|
Ciaglia T, Vestuto V, Bertamino A, González-Muñiz R, Gómez-Monterrey I. On the modulation of TRPM channels: Current perspectives and anticancer therapeutic implications. Front Oncol 2023; 12:1065935. [PMID: 36844925 PMCID: PMC9948629 DOI: 10.3389/fonc.2022.1065935] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/15/2022] [Indexed: 02/11/2023] Open
Abstract
The transient melastatin receptor potential (TRPM) ion channel subfamily functions as cellular sensors and transducers of critical biological signal pathways by regulating ion homeostasis. Some members of TRPM have been cloned from cancerous tissues, and their abnormal expressions in various solid malignancies have been correlated with cancer cell growth, survival, or death. Recent evidence also highlights the mechanisms underlying the role of TRPMs in tumor epithelial-mesenchymal transition (EMT), autophagy, and cancer metabolic reprogramming. These implications support TRPM channels as potential molecular targets and their modulation as an innovative therapeutic approach against cancer. Here, we discuss the general characteristics of the different TRPMs, focusing on current knowledge about the connection between TRPM channels and critical features of cancer. We also cover TRPM modulators used as pharmaceutical tools in biological trials and an indication of the only clinical trial with a TRPM modulator about cancer. To conclude, the authors describe the prospects for TRPM channels in oncology.
Collapse
Affiliation(s)
- Tania Ciaglia
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Vincenzo Vestuto
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | - Alessia Bertamino
- Dipartimento di Farmacia (DIFARMA), Università degli Studi di Salerno, Fisciano, Italy
| | | | | |
Collapse
|
16
|
Burglen L, Van Hoeymissen E, Qebibo L, Barth M, Belnap N, Boschann F, Depienne C, De Clercq K, Douglas AGL, Fitzgerald MP, Foulds N, Garel C, Helbig I, Held K, Horn D, Janssen A, Kaindl AM, Narayanan V, Prager C, Rupin-Mas M, Afenjar A, Zhao S, Ramaekers VT, Ruggiero SM, Thomas S, Valence S, Van Maldergem L, Rohacs T, Rodriguez D, Dyment D, Voets T, Vriens J. Gain-of-function variants in the ion channel gene TRPM3 underlie a spectrum of neurodevelopmental disorders. eLife 2023; 12:81032. [PMID: 36648066 PMCID: PMC9886277 DOI: 10.7554/elife.81032] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
TRPM3 is a temperature- and neurosteroid-sensitive plasma membrane cation channel expressed in a variety of neuronal and non-neuronal cells. Recently, rare de novo variants in TRPM3 were identified in individuals with developmental and epileptic encephalopathy, but the link between TRPM3 activity and neuronal disease remains poorly understood. We previously reported that two disease-associated variants in TRPM3 lead to a gain of channel function . Here, we report a further 10 patients carrying one of seven additional heterozygous TRPM3 missense variants. These patients present with a broad spectrum of neurodevelopmental symptoms, including global developmental delay, intellectual disability, epilepsy, musculo-skeletal anomalies, and altered pain perception. We describe a cerebellar phenotype with ataxia or severe hypotonia, nystagmus, and cerebellar atrophy in more than half of the patients. All disease-associated variants exhibited a robust gain-of-function phenotype, characterized by increased basal activity leading to cellular calcium overload and by enhanced responses to the neurosteroid ligand pregnenolone sulfate when co-expressed with wild-type TRPM3 in mammalian cells. The antiseizure medication primidone, a known TRPM3 antagonist, reduced the increased basal activity of all mutant channels. These findings establish gain-of-function of TRPM3 as the cause of a spectrum of autosomal dominant neurodevelopmental disorders with frequent cerebellar involvement in humans and provide support for the evaluation of TRPM3 antagonists as a potential therapy.
Collapse
Affiliation(s)
- Lydie Burglen
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Developmental Brain Disorders Laboratory, Imagine InstituteParisFrance
| | - Evelien Van Hoeymissen
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| | - Leila Qebibo
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
| | - Magalie Barth
- Department of Genetics, University Hospital of AngersAngersFrance
| | - Newell Belnap
- Translational Genomics Research Institute (TGen), Neurogenomics Division, Center for Rare Childhood DisordersPhoenixUnited States
| | - Felix Boschann
- Charité – Universitäts medizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Genetics and Human GeneticsBerlinGermany
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University Duisburg-EssenEssenGermany
| | - Katrien De Clercq
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| | - Andrew GL Douglas
- University Hospital Southampton NHS Foundation TrustSouthamptonUnited Kingdom
| | | | - Nicola Foulds
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation TrustSouthamptonUnited Kingdom
| | - Catherine Garel
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Service de Radiologie Pédiatrique, Hôpital Armand-Trousseau, Médecine Sorbonne UniversitéParisFrance
| | - Ingo Helbig
- Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Katharina Held
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| | - Denise Horn
- Charité – Universitäts medizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Genetics and Human GeneticsBerlinGermany
| | - Annelies Janssen
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
| | - Angela M Kaindl
- Institute of Cell Biology and Neurobiology, Charité - Universitäts medizin BerlinBerlinGermany
- Department of Pediatric Neurology, Charité - Universitäts medizin BerlinBerlinGermany
- Charité – Universitäts medizin Berlin, Center for Chronically Sick ChildrenBerlinGermany
| | - Vinodh Narayanan
- Translational Genomics Research Institute (TGen), Neurogenomics Division, Center for Rare Childhood DisordersPhoenixUnited States
| | - Christina Prager
- Department of Pediatric Neurology, Charité - Universitäts medizin BerlinBerlinGermany
- Charité – Universitäts medizin Berlin, Center for Chronically Sick ChildrenBerlinGermany
| | - Mailys Rupin-Mas
- Department of Neuropediatrics, University Hospital of AngersAngersFrance
| | - Alexandra Afenjar
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
| | - Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New JerseyNewarkUnited States
| | | | | | - Simon Thomas
- Wessex Regional Genetics Laboratory, Salisbury District HospitalSalisburyUnited Kingdom
| | - Stéphanie Valence
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Sorbonne Université, Service de Neuropédiatrie, Hôpital Trousseau AP-HPParisFrance
| | - Lionel Van Maldergem
- Centre de Génétique Humaine, Université de Franche-Comté BesançonBesanconFrance
- Center of Clinical Investigation 1431, National Institute of Health and Medical ResearchBesanconFrance
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, The State University of New JerseyNewarkUnited States
| | - Diana Rodriguez
- Centre de référence des malformations et maladies congénitales du cervelet, Départementde Génétique, APHP, Sorbonne UniversityParisFrance
- Sorbonne Université, Service de Neuropédiatrie, Hôpital Trousseau AP-HPParisFrance
| | - David Dyment
- Children's Hospital of Eastern Ontario Research Institute, University of OttawaOttawaCanada
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of cellular and molecular medicine, University of LeuvenLeuvenBelgium
- VIB Center for Brain & Disease ResearchLeuvenBelgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department Development & Regeneration, University of LeuvenLeuvenBelgium
| |
Collapse
|
17
|
Pintér E, Helyes Z, Szőke É, Bölcskei K, Kecskés A, Pethő G. The triple function of the capsaicin-sensitive sensory neurons: In memoriam János Szolcsányi. Temperature (Austin) 2022; 10:13-34. [PMID: 38059854 PMCID: PMC10177685 DOI: 10.1080/23328940.2022.2147388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
This paper is dedicated to the memory of János Szolcsányi (1938-2018), an outstanding Hungarian scientist. Among analgesics that act on pain receptors, he identified capsaicin as a selective lead molecule. He studied the application of capsaicin and revealed several physiological (pain, thermoregulation) and pathophysiological (inflammation, gastric ulcer) mechanisms. He discovered a new neuroregulatory system without sensory efferent reflex and investigated its pharmacology. The authors of this review are his former Ph.D. students who carried out their doctoral work in Szolcsányi's laboratory between 1985 and 2010 and report on the scientific results obtained under his guidance. His research group provided evidence for the triple function of the peptidergic capsaicin-sensitive sensory neurons including classical afferent function, local efferent responses, and remote, hormone-like anti-inflammatory, and antinociceptive actions. They also proposed somatostatin receptor type 4 as a promising drug target for the treatment of pain and inflammation. They revealed that neonatal capsaicin treatment caused no acute neuronal death but instead long-lasting selective ultrastructural and functional changes in B-type sensory neurons, similar to adult treatment. They described that lipid raft disruption diminished the agonist-induced channel opening of the TRPV1, TRPA1, and TRPM8 receptors in native sensory neurons. Szolcsányi's group has developed new devices for noxious heat threshold measurement: an increasing temperature hot plate and water bath. This novel approach proved suitable for assessing the thermal antinociceptive effects of analgesics as well as for analyzing peripheral mechanisms of thermonociception.
Collapse
Affiliation(s)
- Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok krt. 2. H-1117Budapest, Hungary
- Eötvös Lorand Research Network, Chronic Pain Research Group, University of Pécs, H7624, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok krt. 2. H-1117Budapest, Hungary
- Eötvös Lorand Research Network, Chronic Pain Research Group, University of Pécs, H7624, Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- National Laboratory for Drug Research and Development, Magyar tudósok krt. 2. H-1117Budapest, Hungary
- Eötvös Lorand Research Network, Chronic Pain Research Group, University of Pécs, H7624, Pécs, Hungary
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
| | - Angéla Kecskés
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
| | - Gábor Pethő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12, H-7624, Pécs, Hungary
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2, H-7624 , Pécs, Hungary
| |
Collapse
|
18
|
Zhao S, Carnevale V, Gabrielle M, Gianti E, Rohacs T. Computational and functional studies of the PI(4,5)P 2 binding site of the TRPM3 ion channel reveal interactions with other regulators. J Biol Chem 2022; 298:102547. [PMID: 36181791 DOI: 10.1016/j.jbc.2022.102547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 10/14/2022] Open
Abstract
Transient Receptor Potential Melastatin 3 (TRPM3) is a heat-activated ion channel expressed in peripheral sensory neurons and the central nervous system. TRPM3 activity depends on the membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2], but the molecular mechanism of activation by PI(4,5)P2 is not known. As no experimental structure of TRPM3 is available, we built a homology model of the channel in complex with PI(4,5)P2via molecular modeling. We identified putative contact residues for PI(4,5)P2 in the pre-S1 segment, the S4-S5 linker, and the proximal C-terminal TRP-domain. Mutating these residues increased sensitivity to inhibition of TRPM3 by decreasing PI(4,5)P2 levels. Changes in ligand-binding affinities via Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) showed reduced PI(4,5)P2 affinity for the mutants. Mutating PI(4,5)P2 interacting residues also reduced sensitivity for activation by the endogenous ligand pregnenolone sulfate (PregS), pointing to an allosteric interaction between PI(4,5)P2 and PregS. Similarly, mutating residues in the PI(4,5)P2 binding site in TRPM8 resulted in increased sensitivity to PI(4,5)P2 depletion, and reduced sensitivity to menthol. Mutations of most PI(4,5)P2-interacting residues in TRPM3 also increased sensitivity to inhibition by Gβγ, indicating allosteric interaction between Gβγ and PI(4,5)P2. Disease-associated gain-of-function TRPM3 mutations on the other hand, resulted in no change of PI(4,5)P2 sensitivity, indicating that mutations did not increase channel activity via increasing PI(4,5)P2 interactions. Our data provide insight into the mechanism of regulation of TRPM3 by PI(4,5)P2, its relationship to endogenous activators and inhibitors, as well as identify similarities and differences between PI(4,5)P2 regulation of TRPM3 and TRPM8.
Collapse
Affiliation(s)
- Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ 07103
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA 19122; Department of Biology, Temple University, Philadelphia, PA 19122
| | - Matthew Gabrielle
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ 07103
| | - Eleonora Gianti
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA 19122; Department of Chemistry, Temple University, Philadelphia, PA 19122.
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ 07103.
| |
Collapse
|
19
|
Sasajima S, Kondo M, Ohno N, Ujisawa T, Motegi M, Hayami T, Asano S, Asano-Hayami E, Nakai-Shimoda H, Inoue R, Yamada Y, Miura-Yura E, Morishita Y, Himeno T, Tsunekawa S, Kato Y, Nakamura J, Kamiya H, Tominaga M. Thermal gradient ring reveals thermosensory changes in diabetic peripheral neuropathy in mice. Sci Rep 2022; 12:9724. [PMID: 35697861 PMCID: PMC9192750 DOI: 10.1038/s41598-022-14186-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 05/18/2022] [Indexed: 12/03/2022] Open
Abstract
Diabetic peripheral neuropathy (DPN) includes symptoms of thermosensory impairment, which are reported to involve changes in the expression or function, or both, of nociceptive TRPV1 and TRPA1 channels in rodents. In the present study, we did not find changes in the expression or function of TRPV1 or TRPA1 in DPN mice caused by STZ, although thermal hypoalgesia was observed in a murine model of DPN or TRPV1−/− mice with a Plantar test, which specifically detects temperature avoidance. With a Thermal Gradient Ring in which mice can move freely in a temperature gradient, temperature preference can be analyzed, and we clearly discriminated the temperature-dependent phenotype between DPN and TRPV1−/− mice. Accordingly, we propose approaches with multiple behavioral methods to analyze the progression of DPN by response to thermal stimuli. Attention to both thermal avoidance and preference may provide insight into the symptoms of DPN.
Collapse
Affiliation(s)
- Sachiko Sasajima
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.,Division of Cell Signaling, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| | - Masaki Kondo
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.,Division of Ultrastructural Research, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| | - Tomoyo Ujisawa
- Division of Cell Signaling, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan.,Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| | - Mikio Motegi
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Tomohide Hayami
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Saeko Asano
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Emi Asano-Hayami
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Hiromi Nakai-Shimoda
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Rieko Inoue
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Yuichiro Yamada
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Emiri Miura-Yura
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Yoshiaki Morishita
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Tatsuhito Himeno
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Shin Tsunekawa
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Yoshiro Kato
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Jiro Nakamura
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.,Department of Innovative Diabetes Therapy, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hideki Kamiya
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan. .,Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan. .,Department of Physiological Sciences, Sokendai, Okazaki, Japan.
| |
Collapse
|
20
|
Kleene SJ. Regenerative Calcium Currents in Renal Primary Cilia. Front Physiol 2022; 13:894518. [PMID: 35620606 PMCID: PMC9127361 DOI: 10.3389/fphys.2022.894518] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Polycystic kidney disease (PKD) is a leading cause of end-stage renal disease. PKD arises from mutations in proteins, one a Ca2+-conducting channel, expressed in the primary cilia of renal epithelial cells. A common hypothesis is that Ca2+ entering through ciliary ion channels may reduce cystogenesis. The cilia have at least two Ca2+-conducting channels: polycystin-2 (PC2) and TRPV4 (transient receptor potential (TRP) cation channel, subfamily V, member 4), but how substantially they can increase intraciliary Ca2+ is unknown. By recording channel activities in isolated cilia, conditions are identified under which the channels can increase free Ca2+ within the cilium by at least 500-fold through regenerative (positive-feedback) signaling. Ca2+ that has entered through a channel can activate the channel internally, which increases the Ca2+ influx, and so on. Regenerative signaling is favored when the concentration of the Ca2+ buffer is reduced or when a slower buffer is used. Under such conditions, the Ca2+ that enters the cilium through a single PC2 channel is sufficient to almost fully activate that same channel. Regenerative signaling is not detectable with reduced external Ca2+. Reduced buffering also allows regenerative signaling through TRPV4 channels, but not through TRPM4 (TRP subfamily M, member 4) channels, which are activated by Ca2+ but do not conduct it. On a larger scale, Ca2+ that enters through TRPV4 channels can cause secondary activation of PC2 channels. I discuss the likelihood of regenerative ciliary Ca2+ signaling in vivo, a possible mechanism for its activation, and how it might relate to cystogenesis.
Collapse
Affiliation(s)
- Steven J. Kleene
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
21
|
Behrendt M, Solinski HJ, Schmelz M, Carr R. Bradykinin-Induced Sensitization of Transient Receptor Potential Channel Melastatin 3 Calcium Responses in Mouse Nociceptive Neurons. Front Cell Neurosci 2022; 16:843225. [PMID: 35496916 PMCID: PMC9043526 DOI: 10.3389/fncel.2022.843225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
TRPM3 is a calcium-permeable cation channel expressed in a range of sensory neurons that can be activated by heat and the endogenous steroid pregnenolone sulfate (PS). During inflammation, the expression and function of TRPM3 are both augmented in somatosensory nociceptors. However, in isolated dorsal root ganglion (DRG) neurons application of inflammatory mediators like prostaglandins and bradykinin (BK) inhibit TRPM3. Therefore, the aim of this study was to examine the effect of preceding activation of cultured 1 day old mouse DRG neurons by the inflammatory mediator BK on TRPM3-mediated calcium responses. Calcium signals were recorded using the intensity-based dye Fluo-8. We found that TRPM3-mediated calcium responses to PS were enhanced by preceding application of BK in cells that responded to BK with a calcium signal, indicating BK receptor (BKR) expression. The majority of cells that co-expressed TRPM3 and BKRs also expressed TRPV1, however, only a small fraction co-expressed TRPA1, identified by calcium responses to capsaicin and supercinnamaldehyde, respectively. Signaling and trafficking pathways responsible for sensitization of TRPM3 following BK were characterized using inhibitors of second messenger signaling cascades and exocytosis. Pharmacological blockade of protein kinase C, calcium–calmodulin-dependent protein kinase II and diacylglycerol (DAG) lipase did not affect BK-induced sensitization, but inhibition of DAG kinase did. In addition, release of calcium from intracellular stores using thapsigargin also resulted in TRPM3 sensitization. Finally, BK did not sensitize TRPM3 in the presence of exocytosis inhibitors. Collectively, we show that preceding activation of DRG neurons by BK sensitized TRPM3-mediated calcium responses to PS. Our results indicate that BKR-mediated activation of intracellular signaling pathways comprising DAG kinase, calcium and exocytosis may contribute to TRPM3 sensitization during inflammation.
Collapse
|
22
|
Souza Bomfim GH, Niemeyer BA, Lacruz RS, Lis A. On the Connections between TRPM Channels and SOCE. Cells 2022; 11:1190. [PMID: 35406753 PMCID: PMC8997886 DOI: 10.3390/cells11071190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
Plasma membrane protein channels provide a passageway for ions to access the intracellular milieu. Rapid entry of calcium ions into cells is controlled mostly by ion channels, while Ca2+-ATPases and Ca2+ exchangers ensure that cytosolic Ca2+ levels ([Ca2+]cyt) are maintained at low (~100 nM) concentrations. Some channels, such as the Ca2+-release-activated Ca2+ (CRAC) channels and voltage-dependent Ca2+ channels (CACNAs), are highly Ca2+-selective, while others, including the Transient Receptor Potential Melastatin (TRPM) family, have broader selectivity and are mostly permeable to monovalent and divalent cations. Activation of CRAC channels involves the coupling between ORAI1-3 channels with the endoplasmic reticulum (ER) located Ca2+ store sensor, Stromal Interaction Molecules 1-2 (STIM1/2), a pathway also termed store-operated Ca2+ entry (SOCE). The TRPM family is formed by 8 members (TRPM1-8) permeable to Mg2+, Ca2+, Zn2+ and Na+ cations, and is activated by multiple stimuli. Recent studies indicated that SOCE and TRPM structure-function are interlinked in some instances, although the molecular details of this interaction are only emerging. Here we review the role of TRPM and SOCE in Ca2+ handling and highlight the available evidence for this interaction.
Collapse
Affiliation(s)
- Guilherme H. Souza Bomfim
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Barbara A. Niemeyer
- Department of Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany;
| | - Rodrigo S. Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Annette Lis
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
23
|
Behrendt M. TRPM3 in the eye and in the nervous system - from new findings to novel mechanisms. Biol Chem 2022; 403:859-868. [PMID: 35240732 DOI: 10.1515/hsz-2021-0403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/10/2022] [Indexed: 01/13/2023]
Abstract
The calcium-permeable cation channel TRPM3 can be activated by heat and the endogenous steroid pregnenolone sulfate. TRPM3's best understood function is its role as a peripheral noxious heat sensor in mice. However, the channel is expressed in various tissues and cell types including neurons as well as glial and epithelial cells. TRPM3 expression patterns differ between species and change during development. Furthermore, a plethora of TRPM3 variants that result from alternative splicing have been identified and the majority of these isoforms are yet to be characterized. Moreover, the mechanisms underlying regulation of TRPM3 are largely unexplored. In addition, a micro-RNA gene (miR-204) is located within the TRPM3 gene. This complexity makes it difficult to obtain a clear picture of TRPM3 characteristics. However, a clear picture is needed to unravel TRPM3's full potential as experimental tool, diagnostic marker and therapeutic target. Therefore, the newest data related to TRPM3 have to be discussed and to be put in context as soon as possible to be up-to-date and to accelerate the translation from bench to bedside. The aim of this review is to highlight recent results and developments with particular focus on findings from studies involving ocular tissues and cells or peripheral neurons of rodents and humans.
Collapse
Affiliation(s)
- Marc Behrendt
- Department of Experimental Pain Research, Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Str. 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
24
|
Lines MA, Goldenberg P, Wong A, Srivastava S, Bayat A, Hove H, Karstensen HG, Anyane-Yeboa K, Liao J, Jiang N, May A, Guzman E, Morleo M, D'Arrigo S, Ciaccio C, Pantaleoni C, Castello R, McKee S, Ong J, Zibdeh-Lough H, Tran-Mau-Them F, Gerasimenko A, Heron D, Keren B, Margot H, de Sainte Agathe JM, Burglen L, Voets T, Vriens J, Innes AM, Dyment DA. Phenotypic spectrum of the recurrent TRPM3 p.(Val837Met) substitution in seven individuals with global developmental delay and hypotonia. Am J Med Genet A 2022; 188:1667-1675. [PMID: 35146895 DOI: 10.1002/ajmg.a.62673] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/08/2022] [Accepted: 01/13/2022] [Indexed: 11/11/2022]
Abstract
TRPM3 encodes a transient receptor potential cation channel of the melastatin family, expressed in the central nervous system and in peripheral sensory neurons of the dorsal root ganglia. The recurrent substitution in TRPM3: c.2509G>A, p.(Val837Met) has been associated with syndromic intellectual disability and seizures. In this report, we present the clinical and molecular features of seven previously unreported individuals, identified by exome sequencing, with the recurrent p.(Val837Met) variant and global developmental delay. Other shared clinical features included congenital hypotonia, dysmorphic facial features (broad forehead, deep-set eyes, and down turned mouth), exotropia, and musculoskeletal issues (hip dysplasia, hip dislocation, scoliosis). Seizures were observed in two of seven individuals (febrile seizure in one and generalized tonic-clonic seizures with atonic drops in another), and epileptiform activity was observed in an additional two individuals. This report extends the number of affected individuals to 16 who are heterozygous for the de novo recurrent substitution p.(Val837Met). In contrast with the initial report, epilepsy was not a mandatory feature observed in this series. TRPM3 pathogenic variation should be considered in individuals with global developmental delays, moderate-severe intellectual disability with, or without, childhood-onset epilepsy.
Collapse
Affiliation(s)
- Matthew A Lines
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paula Goldenberg
- Medical Genetics Unit, Department of Pediatrics, MassGeneral Hospital for Children, Boston, Massachusetts, USA
| | - Ashley Wong
- Medical Genetics Unit, Department of Pediatrics, MassGeneral Hospital for Children, Boston, Massachusetts, USA
| | | | - Allan Bayat
- Department of Epilepsy Genetics and Personalized Medicine, Filadelfia Epilepsy Hospital, Dianalund, Denmark.,Institute for Regional Health Services, University of Southern Denmark, Odense, Denmark
| | - Hanne Hove
- Department of Pediatrics, Center of Rare Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Helena Gásdal Karstensen
- Department of Genetics, Center of Diagnostics, Copenhagen University Hospital - Rigshospitalet, Rigshospitalet, Denmark
| | - Kwame Anyane-Yeboa
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Jun Liao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Nan Jiang
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Alison May
- Division of Child Neurology, Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Edwin Guzman
- Division of Clinical Genetics, Department of Pediatrics, New York Presbyterian Hospital, Columbia University, New York, New York, USA
| | - Manuela Morleo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Stefano D'Arrigo
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Ciaccio
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Pantaleoni
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaele Castello
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | -
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Shane McKee
- Northern Ireland Regional Genetics Service, Belfast, UK
| | - Jinfon Ong
- Child Neurology Consultants of Austin, Austin, Texas, USA
| | - Hana Zibdeh-Lough
- Department of Pediatrics, Dell Children's Medical Center of Central Texas, Austin, Texas, USA
| | | | - Anna Gerasimenko
- APHP Sorbonne Université, GH Pitié Salpêtriére et Trousseau, Département de Génétique, Centre de référence "déficiences intellectuelles de causes rares", Paris, France
| | - Delphine Heron
- APHP Sorbonne Université, GH Pitié Salpêtriére et Trousseau, Département de Génétique, Centre de référence "déficiences intellectuelles de causes rares", Paris, France
| | - Boris Keren
- APHP Sorbonne Université, GH Pitié Salpêtriére et Trousseau, Département de Génétique, Centre de référence "déficiences intellectuelles de causes rares", Paris, France
| | - Henri Margot
- Universitie Bordeaux, MRGM INSERM U1211, CHU de Bordeaux, Service de Génétique Médicale, Bordeaux, France
| | - Jean-Madeleine de Sainte Agathe
- APHP Sorbonne Université, GH Pitié Salpêtriére et Trousseau, Département de Génétique, Centre de référence "déficiences intellectuelles de causes rares", Paris, France
| | - Lydie Burglen
- APHP, Sorbonne Université, Hôpital TROUSSEAU, Centre de Référence des Malformations et Maladies Congénitales du Cervelet et Département de Génétique, Paris, France
| | - Thomas Voets
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Joris Vriens
- Laboratory of Experimental Gynecology and Obstetrics, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - David A Dyment
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada
| |
Collapse
|
25
|
Persoons E, Kerselaers S, Voets T, Vriens J, Held K. Partial Agonistic Actions of Sex Hormone Steroids on TRPM3 Function. Int J Mol Sci 2021; 22:13652. [PMID: 34948452 PMCID: PMC8708174 DOI: 10.3390/ijms222413652] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022] Open
Abstract
Sex hormone steroidal drugs were reported to have modulating actions on the ion channel TRPM3. Pregnenolone sulphate (PS) presents the most potent known endogenous chemical agonist of TRPM3 and affects several gating modes of the channel. These includes a synergistic action of PS and high temperatures on channel opening and the PS-induced opening of a noncanonical pore in the presence of other TRPM3 modulators. Moreover, human TRPM3 variants associated with neurodevelopmental disease exhibit an increased sensitivity for PS. However, other steroidal sex hormones were reported to influence TRPM3 functions with activating or inhibiting capacity. Here, we aimed to answer how DHEAS, estradiol, progesterone and testosterone act on the various modes of TRPM3 function in the wild-type channel and two-channel variants associated with human disease. By means of calcium imaging and whole-cell patch clamp experiments, we revealed that all four drugs are weak TRPM3 agonists that share a common steroidal interaction site. Furthermore, they exhibit increased activity on TRPM3 at physiological temperatures and in channels that carry disease-associated mutations. Finally, all steroids are able to open the noncanonical pore in wild-type and DHEAS also in mutant TRPM3. Collectively, our data provide new valuable insights in TRPM3 gating, structure-function relationships and ligand sensitivity.
Collapse
Affiliation(s)
- Eleonora Persoons
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 Box 611, 3000 Leuven, Belgium; (E.P.); (K.H.)
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| | - Sara Kerselaers
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 Box 611, 3000 Leuven, Belgium; (E.P.); (K.H.)
| | - Katharina Held
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 Box 611, 3000 Leuven, Belgium; (E.P.); (K.H.)
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| |
Collapse
|
26
|
Becker A, Götz C, Montenarh M, Philipp SE. Control of TRPM3 Ion Channels by Protein Kinase CK2-Mediated Phosphorylation in Pancreatic β-Cells of the Line INS-1. Int J Mol Sci 2021; 22:13133. [PMID: 34884938 PMCID: PMC8658122 DOI: 10.3390/ijms222313133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
In pancreatic β-cells of the line INS-1, glucose uptake and metabolism induce the openings of Ca2+-permeable TRPM3 channels that contribute to the elevation of the intracellular Ca2+ concentration and the fusion of insulin granules with the plasma membrane. Conversely, glucose-induced Ca2+ signals and insulin release are reduced by the activity of the serine/threonine kinase CK2. Therefore, we hypothesized that TRPM3 channels might be regulated by CK2 phosphorylation. We used recombinant TRPM3α2 proteins, native TRPM3 proteins from INS-1 β-cells, and TRPM3-derived oligopeptides to analyze and localize CK2-dependent phosphorylation of TRPM3 channels. The functional consequences of CK2 phosphorylation upon TRPM3-mediated Ca2+ entry were investigated in Fura-2 Ca2+-imaging experiments. Recombinant TRPM3α2 channels expressed in HEK293 cells displayed enhanced Ca2+ entry in the presence of the CK2 inhibitor CX-4945 and their activity was strongly reduced after CK2 overexpression. TRPM3α2 channels were phosphorylated by CK2 in vitro at serine residue 1172. Accordingly, a TRPM3α2 S1172A mutant displayed enhanced Ca2+ entry. The TRPM3-mediated Ca2+ entry in INS-1 β-cells was also strongly increased in the presence of CX-4945 and reduced after overexpression of CK2. Our study shows that CK2-mediated phosphorylation controls TRPM3 channel activity in INS-1 β-cells.
Collapse
Affiliation(s)
- Alexander Becker
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Building 46, D-66424 Homburg, Germany;
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, Building 44, D-66424 Homburg, Germany; (C.G.); (M.M.)
| | - Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, Building 44, D-66424 Homburg, Germany; (C.G.); (M.M.)
| | - Stephan E. Philipp
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Building 46, D-66424 Homburg, Germany;
| |
Collapse
|
27
|
Zhao S, Rohacs T. The newest TRP channelopathy: Gain of function TRPM3 mutations cause epilepsy and intellectual disability. Channels (Austin) 2021; 15:386-397. [PMID: 33853504 PMCID: PMC8057083 DOI: 10.1080/19336950.2021.1908781] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Transient Receptor Potential Melastatin 3 (TRPM3) is a Ca2+ permeable nonselective cation channel, activated by heat and chemical agonists, such as the endogenous neuro-steroid Pregnenolone Sulfate (PregS) and the chemical compound CIM0216. TRPM3 is expressed in peripheral sensory neurons of the dorsal root ganglia (DRG), and its role in noxious heat sensation in mice is well established. TRPM3 is also expressed in a number of other tissues, including the brain, but its role there has been largely unexplored. Recent reports showed that two mutations in TRPM3 are associated with a developmental and epileptic encephalopathy, pointing to an important role of TRPM3 in the human brain. Subsequent reports found that the two disease-associated mutations increased basal channel activity, and sensitivity of the channel to activation by heat and chemical agonists. This review will discuss these mutations in the context of human diseases caused by mutations in other TRP channels, and in the context of the biophysical properties and physiological functions of TRPM3.
Collapse
Affiliation(s)
- Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
28
|
Cabanas H, Muraki K, Eaton-Fitch N, Staines DR, Marshall-Gradisnik S. Potential Therapeutic Benefit of Low Dose Naltrexone in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Role of Transient Receptor Potential Melastatin 3 Ion Channels in Pathophysiology and Treatment. Front Immunol 2021; 12:687806. [PMID: 34326841 PMCID: PMC8313851 DOI: 10.3389/fimmu.2021.687806] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating multi-systemic chronic condition of unknown aetiology classified as an immune dysfunction syndrome and neurological disorder. The discovery of the widely expressed Transient Receptor Potential Melastatin 3 (TRPM3) as a nociceptor channel substantially targeted by certain opioid receptors, and its implication in calcium (Ca2+)-dependent Natural Killer (NK) cell immune functions has raised the possibility that TRPM3 may be pharmacologically targeted to treat characteristic symptoms of ME/CFS. Naltrexone hydrochloride (NTX) acts as an antagonist to the mu (μ)-opioid receptor thus negating its inhibitory function on TRPM3. Based on the benefits reported by patients on their symptoms, low dose NTX (LDN, 3.0-5.0 mg/day) treatment seems to offer some potential benefit suggesting that its effect may be targeted towards the pathomechanism of ME/CFS. As there is no literature confirming the efficacy of LDN for ME/CFS patients in vitro, this study investigates the potential therapeutic effect of LDN in ME/CFS patients. TRPM3 ion channel activity was measured after modulation with Pregnenolone sulfate (PregS) and ononetin in NK cells on 9 ME/CFS patients taking LDN and 9 age- and sex-matched healthy controls using whole-cell patch-clamp technique. We report that ME/CFS patients taking LDN have restored TRPM3-like ionic currents in NK cells. Small ionic currents with a typical TRPM3-like outward rectification were measured after application of PregS, a TRPM3-agonist, in NK cells from patients taking LDN. Additionally, PregS-evoked ionic currents through TRPM3 were significantly modulated by ononetin, a TRPM3-antagonist, in NK cells from ME/CFS patients taking LDN. These data support the hypothesis that LDN may have potential as a treatment for ME/CFS by characterising the underlying regulatory mechanisms of LDN treatment involving TRPM3 and opioid receptors in NK cells. Finally, this study may serve for the repurpose of marketed drugs, as well as support the approval of prospective randomized clinical studies on the role and dose of NTX in treating ME/CFS patients.
Collapse
Affiliation(s)
- Helene Cabanas
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Katsuhiko Muraki
- Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia.,Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Natalie Eaton-Fitch
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Donald Ross Staines
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
29
|
Zhou Y, Bennett TM, Shiels A. Mutation of the TRPM3 cation channel underlies progressive cataract development and lens calcification associated with pro-fibrotic and immune cell responses. FASEB J 2021; 35:e21288. [PMID: 33484482 DOI: 10.1096/fj.202002037r] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/23/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Transient-receptor-potential cation channel, subfamily M, member 3 (TRPM3) serves as a polymodal calcium sensor in diverse mammalian cell-types. Mutation of the human TRPM3 gene (TRPM3) has been linked with inherited forms of early-onset cataract with or without other eye abnormalities. Here, we have characterized the ocular phenotypes of germline "knock-in" mice that harbor a human cataract-associated isoleucine-to-methionine mutation (p.I65M) in TRPM3 (Trpm3-mutant) compared with germline "knock-out" mice that functionally lack TRPM3 (Trpm3-null). Despite strong expression of Trpm3 in lens epithelial cells, neither heterozygous (Trpm3+/- ) nor homozygous (Trpm3-/- ) Trpm3-null mice developed cataract; however, the latter exhibited a mild impairment of lens growth. In contrast, homozygous Trpm3-M/M mutants developed severe, progressive, anterior pyramid-like cataract with microphthalmia, whereas heterozygous Trpm3-I/M and hemizygous Trpm3-M/- mutants developed anterior pyramidal cataract with delayed onset and progression-consistent with a semi-dominant lens phenotype. Histochemical staining revealed abnormal accumulation of calcium phosphate-like deposits and collagen fibrils in Trpm3-mutant lenses and immunoblotting detected increased αII-spectrin cleavage products consistent with calpain hyper-activation. Immunofluorescent confocal microscopy of Trpm3-M/M mutant lenses revealed fiber cell membrane degeneration that was accompanied by accumulation of alpha-smooth muscle actin positive (α-SMA+ve) myofibroblast-like cells and macrosialin positive (CD68+ve) macrophage-like cells. Collectively, our mouse model data support an ocular disease association for TRPM3 in humans and suggest that (1) Trpm3 deficiency impaired lens growth but not lens transparency and (2) Trpm3 dysfunction resulted in progressive lens degeneration and calcification coupled with pro-fibrotic (α-SMA+ve) and immune (CD68+ve) cell responses.
Collapse
Affiliation(s)
- Yuefang Zhou
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Thomas M Bennett
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Alan Shiels
- Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
30
|
Abstract
Already for centuries, humankind is driven to understand the physiological and pathological mechanisms that occur in our brains. Today, we know that ion channels play an essential role in the regulation of neural processes and control many functions of the central nervous system. Ion channels present a diverse group of membrane-spanning proteins that allow ions to penetrate the insulating cell membrane upon opening of their channel pores. This regulated ion permeation results in different electrical and chemical signals that are necessary to maintain physiological excitatory and inhibitory processes in the brain. Therefore, it is no surprise that disturbances in the functions of cerebral ion channels can result in a plethora of neurological disorders, which present a tremendous health care burden for our current society. The identification of ion channel-related brain disorders also fuel the research into the roles of ion channel proteins in various brain states. In the last decade, mounting evidence has been collected that indicates a pivotal role for transient receptor potential (TRP) ion channels in the development and various physiological functions of the central nervous system. For instance, TRP channels modulate neurite growth, synaptic plasticity and integration, and are required for neuronal survival. Moreover, TRP channels are involved in numerous neurological disorders. TRPM3 belongs to the melastatin subfamily of TRP channels and represents a non-selective cation channel that can be activated by several different stimuli, including the neurosteroid pregnenolone sulfate, osmotic pressures and heat. The channel is best known as a peripheral nociceptive ion channel that participates in heat sensation. However, recent research identifies TRPM3 as an emerging new player in the brain. In this review, we summarize the available data regarding the roles of TRPM3 in the brain, and correlate these data with the neuropathological processes in which this ion channel may be involved.
Collapse
Affiliation(s)
- Katharina Held
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine and VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
| | - Balázs István Tóth
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
31
|
Hossain Saad MZ, Xiang L, Liao YS, Reznikov LR, Du J. The Underlying Mechanism of Modulation of Transient Receptor Potential Melastatin 3 by protons. Front Pharmacol 2021; 12:632711. [PMID: 33603674 PMCID: PMC7884864 DOI: 10.3389/fphar.2021.632711] [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: 11/23/2020] [Accepted: 01/04/2021] [Indexed: 12/03/2022] Open
Abstract
Transient receptor potential melastatin 3 channel (TRPM3) is a calcium-permeable nonselective cation channel that plays an important role in modulating glucose homeostasis in the pancreatic beta cells. However, how TRPM3 is regulated under physiological and pathological conditions is poorly understood. In this study, we found that both intracellular and extracellular protons block TRPM3 through its binding sites in the pore region. We demonstrated that external protons block TRPM3 with an inhibitory pH50 of 5.5. whereas internal protons inhibit TRPM3 with an inhibitory pH50 of 6.9. We identified three titratable residues, D1059, D1062, and D1073, at the vestibule of the channel pore that contributes to pH sensitivity. The mutation of D1073Q reduced TRPM3 current by low external pH 5.5 from 62 ± 3% in wildtype to 25 ± 6.0% in D1073Q mutant. These results indicate that D1073 is essential for pH sensitivity. In addition, we found that a single mutation of D1059 or D1062 enhanced pH sensitivity. In summary, our findings identify molecular determinants respionsible for the pH regulation of TRPM3. The inhibition of TRPM3 by protons may indicate an endogenous mechanism governing TRPM3 gating and its physiological/pathological functions.
Collapse
Affiliation(s)
- Md Zubayer Hossain Saad
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Liuruimin Xiang
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Biological Sciences, University of Toledo, Toledo, OH, United States.,Program of Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yan-Shin Liao
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Leah R Reznikov
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Jianyang Du
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States.,Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
32
|
TRPM3 Channels Play Roles in Heat Hypersensitivity and Spontaneous Pain after Nerve Injury. J Neurosci 2021; 41:2457-2474. [PMID: 33478988 DOI: 10.1523/jneurosci.1551-20.2020] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/11/2020] [Accepted: 11/07/2020] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential melastatin 3 (TRPM3) is a heat-activated ion channel in primary sensory neurons of the dorsal root ganglia (DRGs). Pharmacological and genetic studies implicated TRPM3 in various pain modalities, but TRPM3 inhibitors were not validated in TRPM3-/- mice. Here we tested two inhibitors of TRPM3 in male and female wild-type and TRPM3-/- mice in nerve injury-induced neuropathic pain. We found that intraperitoneal injection of either isosakuranetin or primidone reduced heat hypersensitivity induced by chronic constriction injury (CCI) of the sciatic nerve in wild-type, but not in TRPM3-/- mice. Primidone was also effective when injected locally in the hindpaw or intrathecally. Consistently, intrathecal injection of the TRPM3 agonist CIM0216 reduced paw withdrawal latency to radiant heat in wild-type, but not in TRPM3-/- mice. Intraperitoneal injection of 2 mg/kg, but not 0.5 mg/kg isosakuranetin, inhibited cold and mechanical hypersensitivity in CCI, both in wild-type and TRPM3-/- mice, indicating a dose-dependent off-target effect. Primidone had no effect on cold sensitivity, and only a marginal effect on mechanical hypersensitivity. Genetic deletion or inhibitors of TRPM3 reduced the increase in the levels of the early genes c-Fos and pERK in the spinal cord and DRGs in CCI mice, suggesting spontaneous activity of the channel. Intraperitoneal isosakuranetin also inhibited spontaneous pain related behavior in CCI in the conditioned place preference assay, and this effect was eliminated in TRPM3-/- mice. Overall, our data indicate a role of TRPM3 in heat hypersensitivity and in spontaneous pain after nerve injury.SIGNIFICANCE STATEMENT Neuropathic pain is a major unsolved medical problem. The heat-activated TRPM3 ion channel is a potential target for novel pain medications, but the pain modalities in which it plays a role are not clear. Here we used a combination of genetic and pharmacological tools to assess the role of this channel in spontaneous pain, heat, cold, and mechanical hypersensitivity in a nerve injury model of neuropathic pain in mice. Our findings indicate a role for TRPM3 in heat hyperalgesia, and spontaneous pain, but not in cold and mechanical hypersensitivity. We also find that not only TRPM3 located in the peripheral nerve termini, but also TRPM3 in the spinal cord or proximal segments of DRG neurons are important for heat hypersensitivity.
Collapse
|
33
|
Wei H, Chen Z, Koivisto A, Pertovaara A. Spinal mechanisms contributing to the development of pain hypersensitivity induced by sphingolipids in the rat. Pharmacol Rep 2021; 73:672-679. [PMID: 33389723 PMCID: PMC7994220 DOI: 10.1007/s43440-020-00207-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 12/11/2022]
Abstract
Background Earlier studies show that endogenous sphingolipids can induce pain hypersensitivity, activation of spinal astrocytes, release of proinflammatory cytokines and activation of TRPM3 channel. Here we studied whether the development of pain hypersensitivity induced by sphingolipids in the spinal cord can be prevented by pharmacological inhibition of potential downstream mechanisms that we hypothesized to include TRPM3, σ1 and NMDA receptors, gap junctions and D-amino acid oxidase. Methods Experiments were performed in adult male rats with a chronic intrathecal catheter for spinal drug administrations. Mechanical nociception was assessed with monofilaments and heat nociception with radiant heat. N,N-dimethylsphingosine (DMS) was administered to induce pain hypersensitivity. Ononetin, isosakuranetin, naringenin (TRPM3 antagonists), BD-1047 (σ1 receptor antagonist), carbenoxolone (a gap junction decoupler), MK-801 (NMDA receptor antagonist) and AS-057278 (inhibitor of D-amino acid oxidase, DAAO) were used to prevent the DMS-induced hypersensitivity, and pregnenolone sulphate (TRPM3 agonist) to recapitulate hypersensitivity. Results DMS alone produced within 15 min a dose-related mechanical hypersensitivity that lasted at least 24 h, without effect on heat nociception. Preemptive treatments with ononetin, isosakuranetin, naringenin, BD-1047, carbenoxolone, MK-801 or AS-057278 attenuated the development of the DMS-induced hypersensitivity, but had no effects when administered alone. Pregnenolone sulphate (TRPM3 agonist) alone induced a dose-related mechanical hypersensitivity that was prevented by ononetin, isosakuranetin and naringenin. Conclusions Among spinal pronociceptive mechanisms activated by DMS are TRPM3, gap junction coupling, the σ1 and NMDA receptors, and DAAO.
Collapse
Affiliation(s)
- Hong Wei
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, POB 63, 00140, Helsinki, Finland
| | - Zuyue Chen
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, POB 63, 00140, Helsinki, Finland
| | - Ari Koivisto
- Research and Development, Orion Corporation, Orion Pharma, Tengströminkatu 8, POB 425, 20101, Turku, Finland
| | - Antti Pertovaara
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, POB 63, 00140, Helsinki, Finland.
| |
Collapse
|
34
|
The Role of Thermosensitive Ion Channels in Mammalian Thermoregulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:355-370. [DOI: 10.1007/978-981-16-4254-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Ragozzino FJ, Arnold RA, Fenwick AJ, Riley TP, Lindberg JEM, Peterson B, Peters JH. TRPM3 expression and control of glutamate release from primary vagal afferent neurons. J Neurophysiol 2020; 125:199-210. [PMID: 33296617 DOI: 10.1152/jn.00229.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vagal afferent fibers contact neurons in the nucleus of the solitary tract (NTS) and release glutamate via three distinct release pathways: synchronous, asynchronous, and spontaneous. The presence of TRPV1 in vagal afferents is predictive of activity-dependent asynchronous glutamate release along with temperature-sensitive spontaneous vesicle fusion. However, pharmacological blockade or genetic deletion of TRPV1 does not eliminate the asynchronous profile and only attenuates the temperature-dependent spontaneous release at high temperatures (>40°C), indicating additional temperature-sensitive calcium conductance(s) contributing to these release pathways. The transient receptor potential cation channel melastatin subtype 3 (TRPM3) is a calcium-selective channel that functions as a thermosensor (30-37°C) in somatic primary afferent neurons. We predict that TRPM3 is expressed in vagal afferent neurons and contributes to asynchronous and spontaneous glutamate release pathways. We investigated these hypotheses via measurements on cultured nodose neurons and in brainstem slice preparations containing vagal afferent to NTS synaptic contacts. We found histological and genetic evidence that TRPM3 is highly expressed in vagal afferent neurons. The TRPM3-selective agonist, pregnenolone sulfate, rapidly and reversibly activated the majority (∼70%) of nodose neurons; most of which also contained TRPV1. We confirmed the role of TRPM3 with pharmacological blockade and genetic deletion. In the brain, TRPM3 signaling strongly controlled both basal and temperature-driven spontaneous glutamate release. Surprisingly, genetic deletion of TRPM3 did not alter synchronous or asynchronous glutamate release. These results provide convergent evidence that vagal afferents express functional TRPM3 that serves as an additional temperature-sensitive calcium conductance involved in controlling spontaneous glutamate release onto neurons in the NTS.NEW & NOTEWORTHY Vagal afferent signaling coordinates autonomic reflex function and informs associated behaviors. Thermosensitive transient receptor potential (TRP) channels detect temperature and nociceptive stimuli in somatosensory afferent neurons, however their role in vagal signaling remains less well understood. We report that the TRPM3 ion channel provides a major thermosensitive point of control over vagal signaling and synaptic transmission. We conclude that TRPM3 translates physiological changes in temperature to neurophysiological outputs and can serve as a cellular integrator in vagal afferent signaling.
Collapse
Affiliation(s)
- Forrest J Ragozzino
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Rachel A Arnold
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Axel J Fenwick
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Timothy P Riley
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Jonathan E M Lindberg
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - BreeAnne Peterson
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - James H Peters
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
36
|
Behrendt M, Gruss F, Enzeroth R, Dembla S, Zhao S, Crassous PA, Mohr F, Nys M, Louros N, Gallardo R, Zorzini V, Wagner D, Economou A, Rousseau F, Schymkowitz J, Philipp SE, Rohacs T, Ulens C, Oberwinkler J. The structural basis for an on-off switch controlling Gβγ-mediated inhibition of TRPM3 channels. Proc Natl Acad Sci U S A 2020; 117:29090-29100. [PMID: 33122432 PMCID: PMC7682392 DOI: 10.1073/pnas.2001177117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
TRPM3 channels play important roles in the detection of noxious heat and in inflammatory thermal hyperalgesia. The activity of these ion channels in somatosensory neurons is tightly regulated by µ-opioid receptors through the signaling of Gβγ proteins, thereby reducing TRPM3-mediated pain. We show here that Gβγ directly binds to a domain of 10 amino acids in TRPM3 and solve a cocrystal structure of this domain together with Gβγ. Using these data and mutational analysis of full-length proteins, we pinpoint three amino acids in TRPM3 and their interacting partners in Gβ1 that are individually necessary for TRPM3 inhibition by Gβγ. The 10-amino-acid Gβγ-interacting domain in TRPM3 is subject to alternative splicing. Its inclusion in or exclusion from TRPM3 channel proteins therefore provides a mechanism for switching on or off the inhibitory action that Gβγ proteins exert on TRPM3 channels.
Collapse
Affiliation(s)
- Marc Behrendt
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, 35032 Marburg, Germany
| | - Fabian Gruss
- Laboratory of Structural Neurobiology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Raissa Enzeroth
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, 35032 Marburg, Germany
| | - Sandeep Dembla
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, 35032 Marburg, Germany
| | - Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Pierre-Antoine Crassous
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Florian Mohr
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Mieke Nys
- Laboratory of Structural Neurobiology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Nikolaos Louros
- Switch Laboratory, VIB Center for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Rodrigo Gallardo
- Switch Laboratory, VIB Center for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Valentina Zorzini
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Doris Wagner
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Anastassios Economou
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Stephan E Philipp
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Chris Ulens
- Laboratory of Structural Neurobiology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium;
| | - Johannes Oberwinkler
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany;
- Center for Mind, Brain and Behavior (CMBB), Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, 35032 Marburg, Germany
| |
Collapse
|
37
|
Horváth Á, Biró-Sütő T, Kántás B, Payrits M, Skoda-Földes R, Szánti-Pintér E, Helyes Z, Szőke É. Antinociceptive Effects of Lipid Raft Disruptors, a Novel Carboxamido-Steroid and Methyl β-Cyclodextrin, in Mice by Inhibiting Transient Receptor Potential Vanilloid 1 and Ankyrin 1 Channel Activation. Front Physiol 2020; 11:559109. [PMID: 33071817 PMCID: PMC7539994 DOI: 10.3389/fphys.2020.559109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/18/2020] [Indexed: 12/29/2022] Open
Abstract
Transient Receptor Potential Vanilloid 1 and Ankyrin 1 (TRPV1, TRPA1) cation channels are expressed in nociceptive primary sensory neurons, and play an integrative role in pain processing and inflammatory functions. Lipid rafts are liquid-ordered plasma membrane microdomains rich in cholesterol, sphingomyelin, and gangliosides. We earlier proved that lipid raft disintegration by cholesterol depletion using a novel carboxamido-steroid compound (C1) and methyl β-cyclodextrin (MCD) significantly and concentration-dependently inhibit TRPV1 and TRPA1 activation in primary sensory neurons and receptor-expressing cell lines. Here we investigated the effects of C1 compared to MCD in mouse pain models of different mechanisms. Both C1 and MCD significantly decreased the number of the TRPV1 activation (capsaicin)-induced nocifensive eye-wiping movements in the first hour by 45% and 32%, respectively, and C1 also in the second hour by 26%. Furthermore, C1 significantly decreased the TRPV1 stimulation (resiniferatoxin)-evoked mechanical hyperalgesia involving central sensitization processes, while its inhibitory effect on thermal allodynia was not statistically significant. In contrast, MCD did not affect these resiniferatoxin-evoked nocifensive responses. Both C1 and MCD had inhibitory action on TRPA1 activation (formalin)-induced acute nocifensive reactions (paw liftings, lickings, holdings, and shakings) in the second, neurogenic inflammatory phase by 36% and 51%, respectively. These are the first in vivo data showing that our novel lipid raft disruptor carboxamido-steroid compound exerts antinociceptive and antihyperalgesic effects by inhibiting TRPV1 and TRPA1 ion channel activation similarly to MCD, but in 150-fold lower concentrations. It is concluded that C1 is a useful experimental tool to investigate the effects of cholesterol depletion in animal models, and it also might open novel analgesic drug developmental perspectives.
Collapse
Affiliation(s)
- Ádám Horváth
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Tünde Biró-Sütő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Boglárka Kántás
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Maja Payrits
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Rita Skoda-Földes
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Veszprém, Hungary
| | - Eszter Szánti-Pintér
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Veszprém, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- János Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| |
Collapse
|
38
|
Held K, Aloi VD, Freitas ACN, Janssens A, Segal A, Przibilla J, Philipp SE, Wang YT, Voets T, Vriens J. Pharmacological properties of TRPM3 isoforms are determined by the length of the pore loop. Br J Pharmacol 2020; 179:3560-3575. [PMID: 32780479 PMCID: PMC9290681 DOI: 10.1111/bph.15223] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/17/2020] [Accepted: 07/08/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Transient receptor potential melastatin 3 (TRPM3) is a non-selective cation channel that plays a pivotal role in the peripheral nervous system as a transducer of painful heat signals. Alternative splicing gives rise to several TRPM3 variants. The functional consequences of these splice isoforms are poorly understood. Here, the pharmacological properties of TRPM3 variants arising from alternative splicing in the pore-forming region were compared. EXPERIMENTAL APPROACH Calcium microfluorimetry and patch clamp recordings were used to compare the properties of heterologously expressed TRPM3α1 (long pore variant) and TRPM3α2-α6 (short pore variants). Furthermore, site-directed mutagenesis was done to investigate the influence of the length of the pore loop on the channel function. KEY RESULTS All short pore loop TRPM3α variants (TRPM3α2-α6) were activated by the neurosteroid pregnenolone sulphate (PS) and by nifedipine, whereas the long pore loop variant TRPM3α1 was insensitive to either compound. In contrast, TRPM3α1 was robustly activated by clotrimazole, a compound that does not directly activate the short pore variants but potentiates their responses to PS. Clotrimazole-activated TRPM3α1 currents were largely insensitive to established TRPM3α2 antagonists and were only partially inhibited upon activation of the μ opioid receptor. Finally, by creating a set of mutant channels with pore loops of intermediate length, we showed that the length of the pore loop dictates differential channel activation by PS and clotrimazole. CONCLUSION AND IMPLICATIONS Alternative splicing in the pore-forming region of TRPM3 defines the channel's pharmacological properties, which depend critically on the length of the pore-forming loop.
Collapse
Affiliation(s)
- Katharina Held
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Molecular Medicine, KU Leuven, Leuven, Belgium.,DM Centre for Brain Health, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vincenzo Davide Aloi
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ana Cristina Nogueira Freitas
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Annelies Janssens
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Andrei Segal
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Julia Przibilla
- Experimental and Clinical Pharmacology and Toxicology/Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Stephan Ernst Philipp
- Experimental and Clinical Pharmacology and Toxicology/Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Yu Tian Wang
- DM Centre for Brain Health, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Zhao S, Yudin Y, Rohacs T. Disease-associated mutations in the human TRPM3 render the channel overactive via two distinct mechanisms. eLife 2020; 9:e55634. [PMID: 32343227 PMCID: PMC7255801 DOI: 10.7554/elife.55634] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022] Open
Abstract
Transient Receptor Potential Melastatin 3 (TRPM3) is a Ca2+ permeable non-selective cation channel activated by heat and chemical agonists such as pregnenolone sulfate and CIM0216. TRPM3 mutations in humans were recently reported to be associated with intellectual disability and epilepsy; the functional effects of those mutations, however, were not reported. Here, we show that both disease-associated mutations in the human TRPM3 render the channel overactive, but likely via different mechanisms. The Val to Met substitution in the S4-S5 loop induced a larger increase in basal activity and agonist sensitivity at room temperature than the Pro to Gln substitution in the extracellular segment of S6. In contrast, heat activation was increased more by the S6 mutant than by the S4-S5 segment mutant. Both mutants were inhibited by the TRPM3 antagonist primidone, suggesting a potential therapeutic intervention to treat this disease.
Collapse
Affiliation(s)
- Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| | - Yevgen Yudin
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| |
Collapse
|
40
|
Talagas M, Lebonvallet N, Berthod F, Misery L. Lifting the veil on the keratinocyte contribution to cutaneous nociception. Protein Cell 2020; 11:239-250. [PMID: 31907794 PMCID: PMC7093357 DOI: 10.1007/s13238-019-00683-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/16/2019] [Indexed: 12/22/2022] Open
Abstract
Cutaneous nociception is essential to prevent individuals from sustaining injuries. According to the conventional point of view, the responses to noxious stimuli are thought to be exclusively initiated by sensory neurons, whose activity would be at most modulated by keratinocytes. However recent studies have demonstrated that epidermal keratinocytes can also act as primary nociceptive transducers as a supplement to sensory neurons. To enlighten our understanding of cutaneous nociception, this review highlights recent and relevant findings on the cellular and molecular elements that underlie the contribution of epidermal keratinocytes as nociceptive modulators and noxious sensors, both under healthy and pathological conditions.
Collapse
Affiliation(s)
- Matthieu Talagas
- Univ Brest, LIEN, 29200, Brest, France.
- Laboratoire d'Organogenèse Expérimentale (LOEX), University of Laval, Quebec, Canada.
- Department of Dermatology, Brest University Hospital, Brest, France.
- Univ Brest, IBSAM (Institut Brestois de Santé Agro matière), 29200, Brest, France.
| | - Nicolas Lebonvallet
- Univ Brest, LIEN, 29200, Brest, France
- Univ Brest, IBSAM (Institut Brestois de Santé Agro matière), 29200, Brest, France
| | - François Berthod
- Laboratoire d'Organogenèse Expérimentale (LOEX), University of Laval, Quebec, Canada
| | - Laurent Misery
- Univ Brest, LIEN, 29200, Brest, France
- Department of Dermatology, Brest University Hospital, Brest, France
- Univ Brest, IBSAM (Institut Brestois de Santé Agro matière), 29200, Brest, France
| |
Collapse
|
41
|
Shiels A. TRPM3_miR-204: a complex locus for eye development and disease. Hum Genomics 2020; 14:7. [PMID: 32070426 PMCID: PMC7027284 DOI: 10.1186/s40246-020-00258-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
First discovered in a light-sensitive retinal mutant of Drosophila, the transient receptor potential (TRP) superfamily of non-selective cation channels serve as polymodal cellular sensors that participate in diverse physiological processes across the animal kingdom including the perception of light, temperature, pressure, and pain. TRPM3 belongs to the melastatin sub-family of TRP channels and has been shown to function as a spontaneous calcium channel, with permeability to other cations influenced by alternative splicing and/or non-canonical channel activity. Activators of TRPM3 channels include the neurosteroid pregnenolone sulfate, calmodulin, phosphoinositides, and heat, whereas inhibitors include certain drugs, plant-derived metabolites, and G-protein subunits. Activation of TRPM3 channels at the cell membrane elicits a signal transduction cascade of mitogen-activated kinases and stimulus response transcription factors. The mammalian TRPM3 gene hosts a non-coding microRNA gene specifying miR-204 that serves as both a tumor suppressor and a negative regulator of post-transcriptional gene expression during eye development in vertebrates. Ocular co-expression of TRPM3 and miR-204 is upregulated by the paired box 6 transcription factor (PAX6) and mutations in all three corresponding genes underlie inherited forms of eye disease in humans including early-onset cataract, retinal dystrophy, and coloboma. This review outlines the genomic and functional complexity of the TRPM3_miR-204 locus in mammalian eye development and disease.
Collapse
Affiliation(s)
- Alan Shiels
- Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Ave., Box 8096, St. Louis, MO, 63110, USA.
| |
Collapse
|
42
|
Behrendt M. Transient receptor potential channels in the context of nociception and pain - recent insights into TRPM3 properties and function. Biol Chem 2020; 400:917-926. [PMID: 30844758 DOI: 10.1515/hsz-2018-0455] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/25/2019] [Indexed: 01/09/2023]
Abstract
Potential harmful stimuli like heat, mechanical pressure or chemicals are detected by specialized cutaneous nerve fiber endings of nociceptor neurons in a process called nociception. Acute stimulation results in immediate protective reflexes and pain sensation as a normal, physiological behavior. However, ongoing (chronic) pain is a severe pathophysiological condition with diverse pathogeneses that is clinically challenging because of limited therapeutic options. Therefore, an urgent need exists for new potent and specific analgesics without afflicting adverse effects. Recently, TRPM3, a member of the superfamily of transient receptor potential (TRP) ion channels, has been shown to be expressed in nociceptors and to be involved in the detection of noxious heat (acute pain) as well as inflammatory hyperalgesia (acute and chronic pain). Current results in TRPM3 research indicate that this ion channel might not only be part of yet unraveled mechanisms underlying chronic pain but also has the potential to become a clinically relevant pharmacological target of future analgesic strategies. The aim of this review is to summarize and present the basic features of TRPM3 proteins and channels, to highlight recent findings and developments and to provide an outlook on emerging directions of TRPM3 research in the field of chronic pain.
Collapse
Affiliation(s)
- Marc Behrendt
- Experimental Pain Research, Heidelberg University, Medical Faculty Mannheim, CBTM, Tridomus, Building C, Ludolf-Krehl-Straße 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
43
|
Kelemen B, Lisztes E, Vladár A, Hanyicska M, Almássy J, Oláh A, Szöllősi AG, Pénzes Z, Posta J, Voets T, Bíró T, Tóth BI. Volatile anaesthetics inhibit the thermosensitive nociceptor ion channel transient receptor potential melastatin 3 (TRPM3). Biochem Pharmacol 2020; 174:113826. [PMID: 31987857 DOI: 10.1016/j.bcp.2020.113826] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/22/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Volatile anaesthetics (VAs) are the most widely used compounds to induce reversible loss of consciousness and maintain general anaesthesia during surgical interventions. Although the mechanism of their action is not yet fully understood, it is generally believed, that VAs depress central nervous system functions mainly through modulation of ion channels in the neuronal membrane, including 2-pore-domain K+ channels, GABA and NMDA receptors. Recent research also reported their action on nociceptive and thermosensitive TRP channels expressed in the peripheral nervous system, including TRPV1, TRPA1, and TRPM8. Here, we investigated the effect of VAs on TRPM3, a less characterized member of the thermosensitive TRP channels playing a central role in noxious heat sensation. METHODS We investigated the effect of VAs on the activity of recombinant and native TRPM3, by monitoring changes in the intracellular Ca2+ concentration and measuring TRPM3-mediated transmembrane currents. RESULTS All the investigated VAs (chloroform, halothane, isoflurane, sevoflurane) inhibited both the agonist-induced (pregnenolone sulfate, CIM0216) and heat-activated Ca2+ signals and transmembrane currents in a concentration dependent way in HEK293T cells overexpressing recombinant TRPM3. Among the tested VAs, halothane was the most potent blocker (IC50 = 0.52 ± 0.05 mM). We also investigated the effect of VAs on native TRPM3 channels expressed in sensory neurons of the dorsal root ganglia. While VAs activated certain sensory neurons independently of TRPM3, they strongly and reversibly inhibited the agonist-induced TRPM3 activity. CONCLUSIONS These data provide a better insight into the molecular mechanism beyond the analgesic effect of VAs and propose novel strategies to attenuate TRPM3 dependent nociception.
Collapse
Affiliation(s)
- Balázs Kelemen
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Erika Lisztes
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Vladár
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Martin Hanyicska
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - János Almássy
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Gábor Szöllősi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pénzes
- Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary; Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Posta
- Laboratory of Toxicology, Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; Department of Cellular and Molecular Medicine and TRP Research Platform Leuven (TRPLe), KU Leuven, Leuven, Belgium
| | - Tamás Bíró
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Hungarian Center of Excellence for Molecular Medicine, Szeged, Hungary
| | - Balázs István Tóth
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
44
|
Cabanas H, Muraki K, Staines D, Marshall-Gradisnik S. Naltrexone Restores Impaired Transient Receptor Potential Melastatin 3 Ion Channel Function in Natural Killer Cells From Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Patients. Front Immunol 2019; 10:2545. [PMID: 31736966 PMCID: PMC6834647 DOI: 10.3389/fimmu.2019.02545] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a seriously long-term and debilitating illness of unknown cause hallmarked by chronic pain and fatigue, memory and concentration impairment, and inflammation. ME/CFS hypothesis involves impaired Transient receptor potential melastatin 3 (TRPM3) ion channel function, affecting calcium signaling and Natural killer (NK) cell functions. Currently, substances called opioids, agonists of mu (μ)-opioid receptors (μOR), are the strongest painkillers clinically available for people suffering from strong or long-lasting pain characteristic of ME/CFS. μOR have been reported to specifically inhibit TRPM3 and to be expressed in immune cells where they play an immunomodulatory and immunosuppressive role. Naltrexone hydrochloride (NTX) acts as an antagonist to the μOR thus negating the inhibitory function of this opioid receptor on TRPM3. Therefore, understanding the mechanism of action for NTX in regulating and modulating TRPM3 channel function in NK cells will provide important information for the development of effective therapeutic interventions for ME/CFS. Whole-cell patch-clamp technique was used to measure TRPM3 activity in Interleukin-2 (IL-2) stimulated and NTX-treated NK cells for 24 h on eight ME/CFS patients and 8 age- and sex-matched healthy controls, after modulation with a TRPM3-agonist, pregnenolone sulfate (PregS), NTX and a TRPM3-antagonist, ononetin. We confirmed impaired TRPM3 function in ME/CFS patients through electrophysiological investigations in IL-2 stimulated NK cells after modulation with PregS and ononetin. Importantly, TRPM3 channel activity was restored in IL-2 stimulated NK cells isolated from ME/CFS patients after incubation for 24 h with NTX. Moreover, we demonstrated that NTX does not act as an agonist by directly coupling on the TRPM3 ion channel gating. The opioid antagonist NTX has the potential to negate the inhibitory function of opioid receptors on TRPM3 in NK cells from ME/CFS patients, resulting in calcium signals remodeling, which will in turn affect cell functions, supporting the hypothesis that NTX may have potential for use as a treatment for ME/CFS. Our results demonstrate, for the first time, and based on novel patch clamp electrophysiology, potential pharmaco-therapeutic interventions in ME/CFS.
Collapse
Affiliation(s)
- Helene Cabanas
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Katsuhiko Muraki
- Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia.,Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Donald Staines
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
45
|
Yudin Y, Rohacs T. The G-protein-biased agents PZM21 and TRV130 are partial agonists of μ-opioid receptor-mediated signalling to ion channels. Br J Pharmacol 2019; 176:3110-3125. [PMID: 31074038 DOI: 10.1111/bph.14702] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/10/2019] [Accepted: 04/13/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Opioids remain the most efficient medications against severe pain; they act on receptors that couple to heterotrimeric G-proteins in the Gαi/o family. Opioids exert many of their acute effects through modulating ion channels via Gβγ subunits. Many of their side effects are attributed to β-arrestin recruitment. Several biased agonists that do not recruit β-arrestins, but activate G-protein-dependent pathways, have recently been developed. While these compounds have been proposed to be full agonists of G-protein signalling in several high throughput pharmacological assays, their effects were not studied on ion channel targets. EXPERIMENTAL APPROACH Here, we used patch-clamp electrophysiology and Ca2+ imaging to test the effects of TRV130, PZM21, and herkinorin, three G-protein-biased agonists of μ-opioid receptors, on ion channel targets of Gαi/o /Gβγ signalling. We also studied G-protein dissociation using a FRET-based assay. KEY RESULTS All three biased agonists induced smaller activation of G-protein-coupled inwardly rectifying K+ channels (Kir 3.2) and smaller inhibition of transient receptor potential melastatin (TRPM3) channels than the full μ receptor agonist DAMGO. Co-application of TRV130 or PZM21, but not herkinorin, alleviated the effects of DAMGO on both channels. PZM21 and TRV130 also decreased the effect of morphine on Kir 3.2 channels. The CaV 2.2 channel was also inhibited less by PZM21 and TRV130 than by DAMGO. We also found that TRV130, PZM21, and herkinorin were less effective than DAMGO at inducing dissociation of the Gαi /Gβγ complex. CONCLUSION AND IMPLICATIONS TRV130, PZM21, and potentially herkinorin are partial agonists of μ receptors.
Collapse
Affiliation(s)
- Yevgen Yudin
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, New Jersey
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
46
|
Uchida K, Fukuta N, Yamazaki J, Tominaga M. Identification and classification of a new TRPM3 variant (γ subtype). J Physiol Sci 2019; 69:623-634. [PMID: 31011981 PMCID: PMC6583685 DOI: 10.1007/s12576-019-00677-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/10/2019] [Indexed: 01/16/2023]
Abstract
TRPM3 is a non-selective cation channel that is activated by neural steroids such as pregnenolone sulfate, nifedipine, and clotrimazole. Despite the number of TRPM3 variants, few reports have described functional analyses of these different TRPM3 types. Here we identified a new TRPM variant from mouse dorsal root ganglion, termed TRPM3γ3. We classified TRPM3γ3 and another known variant (variant 6) into the γ subtype, and analyzed the TRPM3γ variants. mRNA expression of TRPM3γ was higher than that of TRPM3α variants in the mouse dorsal root ganglion. In Ca2+-imaging of HEK293 cells expressing either the TRPM3γ variants or TRPM3α2, increases in cytosolic Ca2+ concentrations ([Ca2+]i) induced by pregnenolone sulfate or nifedipine were smaller in cells expressing the TRPM3γ variants compared to those expressing TRPM3α2. On the other hand, co-expression of TRPM3γ variants had no effect on [Ca2+]i increases induced by pregnenolone sulfate or nifedipine treatment of HEK293 cells expressing TRPM3α2. In Xenopus oocytes, small responses of TRPM3γ variants to chemical agonists compared to TRPM3α2 were also observed. Interestingly, Xenopus oocytes expressing TRPM3α2 displayed heat-evoked currents with clear thresholds of about 40 °C that were larger than those evoked in oocytes expressing TRPM3γ variants. Overall, these findings indicate that TRPM3γ variants have low channel activity compared to TRPM3α.
Collapse
Affiliation(s)
- Kunitoshi Uchida
- Departments of Physiological Science and Molecular Biology and Morphological Biology, Fukuoka Dental College, Sawara-ku, Fukuoka, 814-0193, Japan.
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki Institute for Integrative Bioscience, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi, 444-8787, Japan.
| | - Naomi Fukuta
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki Institute for Integrative Bioscience, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi, 444-8787, Japan
| | - Jun Yamazaki
- Departments of Physiological Science and Molecular Biology and Morphological Biology, Fukuoka Dental College, Sawara-ku, Fukuoka, 814-0193, Japan
- Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, Kanagawa, 252-0880, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki Institute for Integrative Bioscience, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi, 444-8787, Japan.
- Department of Physiological Sciences, SOKENDAI (The Graduated University for Advanced Studies), Okazaki, Aichi, 444-8585, Japan.
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan.
- Institute for Environmental and Gender-Specific Medicine, Juntendo University, Chiba, 279-0021, Japan.
| |
Collapse
|
47
|
Cabanas H, Muraki K, Balinas C, Eaton-Fitch N, Staines D, Marshall-Gradisnik S. Validation of impaired Transient Receptor Potential Melastatin 3 ion channel activity in natural killer cells from Chronic Fatigue Syndrome/ Myalgic Encephalomyelitis patients. Mol Med 2019; 25:14. [PMID: 31014226 PMCID: PMC6480905 DOI: 10.1186/s10020-019-0083-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background Chronic Fatigue Syndrome/ Myalgic Encephalomyelitis (CFS/ME) is a complex multifactorial disorder of unknown cause having multi-system manifestations. Although the aetiology of CFS/ME remains elusive, immunological dysfunction and more particularly reduced cytotoxic activity in natural killer (NK) cells is the most consistent laboratory finding. The Transient Receptor Potential (TRP) superfamily of cation channels play a pivotal role in the pathophysiology of immune diseases and are therefore potential therapeutic targets. We have previously identified single nucleotide polymorphisms in TRP genes in peripheral NK cells from CFS/ME patients. We have also described biochemical pathway changes and calcium signaling perturbations in NK cells from CFS/ME patients. Notably, we have previously reported a decrease of TRP cation channel subfamily melastatin member 3 (TRPM3) function in NK cells isolated from CFS/ME patients compared with healthy controls after modulation with pregnenolone sulfate and ononetin using a patch-clamp technique. In the present study, we aim to confirm the previous results describing an impaired TRPM3 activity in a new cohort of CFS/ME patients using a whole cell patch-clamp technique after modulation with reversible TRPM3 agonists, pregnenolone sulfate and nifedipine, and an effective TRPM3 antagonist, ononetin. Indeed, no formal research has commented on using pregnenolone sulfate or nifedipine to treat CFS/ME patients while there is evidence that clinicians prescribe calcium channel blockers to improve different symptoms. Methods Whole-cell patch-clamp technique was used to measure TRPM3 activity in isolated NK cells from twelve age- and sex-matched healthy controls and CFS/ME patients, after activation with pregnenolone sulfate and nifedipine and inhibition with ononetin. Results We confirmed a significant reduction in amplitude of TRPM3 currents after pregnenolone sulfate stimulation in isolated NK cells from another cohort of CFS/ME patients compared with healthy controls. The pregnenolone sulfate-evoked ionic currents through TRPM3 channels were again significantly modulated by ononetin in isolated NK cells from healthy controls compared with CFS/ME patients. In addition, we used nifedipine, another reversible TRPM3 agonist to support the previous findings and found similar results confirming a significant loss of the TRPM3 channel activity in CFS/ME patients. Conclusions Impaired TRPM3 activity was validated in NK cells isolated from CFS/ME patients using different pharmacological tools and whole-cell patch-clamp technique as the gold standard for ion channel research. This investigation further helps to establish TRPM3 channels as a prognostic marker and/ or a potential therapeutic target for CFS/ME.
Collapse
Affiliation(s)
- H Cabanas
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia. .,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4222, Australia. .,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia.
| | - K Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Chikusa, Nagoya, Japan.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - C Balinas
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4222, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - N Eaton-Fitch
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4222, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - D Staines
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4222, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - S Marshall-Gradisnik
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4222, Australia.,Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
48
|
Kleene SJ, Siroky BJ, Landero-Figueroa JA, Dixon BP, Pachciarz NW, Lu L, Kleene NK. The TRPP2-dependent channel of renal primary cilia also requires TRPM3. PLoS One 2019; 14:e0214053. [PMID: 30883612 PMCID: PMC6422334 DOI: 10.1371/journal.pone.0214053] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 03/06/2019] [Indexed: 12/21/2022] Open
Abstract
Primary cilia of renal epithelial cells express several members of the transient receptor potential (TRP) class of cation-conducting channel, including TRPC1, TRPM3, TRPM4, TRPP2, and TRPV4. Some cases of autosomal dominant polycystic kidney disease (ADPKD) are caused by defects in TRPP2 (also called polycystin-2, PC2, or PKD2). A large-conductance, TRPP2-dependent channel in renal cilia has been well described, but it is not known whether this channel includes any other protein subunits. To study this question, we investigated the pharmacology of the TRPP2-dependent channel through electrical recordings from the cilia of mIMCD-3 cells, a murine cell line of renal epithelial origin. The pharmacology was found to match that of TRPM3 channels. The ciliary TRPP2-dependent channel is known to be activated by depolarization and by increasing cytoplasmic Ca2+. This activation was greatly enhanced by external pregnenolone sulfate, an agonist of TRPM3 channels. Pregnenolone sulfate did not change the single-channel current-voltage relation. The channels were effectively blocked by isosakuranetin, a specific inhibitor of TRPM3 channels. Both pregnenolone sulfate and isosakuranetin were effective at concentrations as low as 1 μM. Knocking out TRPM3 by CRISPR/Cas9 genome editing eliminated the ciliary channel. Thus the channel is both TRPM3-dependent and TRPP2-dependent, suggesting that it may include both types of subunit. Knocking out TRPM3 did not change the level of TRPP2 protein in the cilia, so it is unlikely that the absence of functional ciliary channels results from a failure of trafficking.
Collapse
Affiliation(s)
- Steven J. Kleene
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail:
| | - Brian J. Siroky
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | | | - Bradley P. Dixon
- Renal Section, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Nolan W. Pachciarz
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Lu Lu
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Nancy K. Kleene
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, United States of America
| |
Collapse
|
49
|
Alonso-Carbajo L, Alpizar YA, Startek JB, López-López JR, Pérez-García MT, Talavera K. Activation of the cation channel TRPM3 in perivascular nerves induces vasodilation of resistance arteries. J Mol Cell Cardiol 2019; 129:219-230. [PMID: 30853321 DOI: 10.1016/j.yjmcc.2019.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
The Transient Receptor Potential Melastatin 3 (TRPM3) is a Ca2+-permeable non-selective cation channel activated by the neurosteroid pregnenolone sulfate (PS). This compound was previously shown to contract mouse aorta by activating TRPM3 in vascular smooth muscle cells (VSMC), and proposed as therapeutic modulator of vascular functions. However, PS effects and the role of TRPM3 in resistance arteries remain unknown. Thus, we aimed at determining the localization and physiological role of TRPM3 in mouse mesenteric arteries. Real-time qPCR experiments, anatomical localization using immunofluorescence microscopy and patch-clamp recordings in isolated VSMC showed that TRPM3 expression in mesenteric arteries is restricted to perivascular nerves. Pressure myography experiments in wild type (WT) mouse arteries showed that PS vasodilates with a concentration-dependence that was best fit by two Hill components (effective concentrations, EC50, of 14 and 100 μM). The low EC50 component was absent in preparations from Trpm3 knockout (KO) mice and in WT arteries in the presence of the CGRP receptor antagonist BIBN 4096. TRPM3-dependent vasodilation was partially inhibited by a cocktail of K+ channel blockers, and not mediated by β-adrenergic signaling. We conclude that, contrary to what was found in aorta, PS dilates mesenteric arteries, partly via an activation of TRPM3 that triggers CGRP release from perivascular nerve endings and a subsequent activation of K+ channels in VSMC. We propose that TRPM3 is implicated in the regulation of the tone of resistance arteries and that its activation by yet unidentified endogenous damage-associated molecules lead to protective vasodilation responses in mesenteric arteries.
Collapse
Affiliation(s)
- Lucía Alonso-Carbajo
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium; Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Universidad de Valladolid y CSIC, Sanz y Forés 3, 47003 Valladolid, Spain
| | - Yeranddy A Alpizar
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium
| | - Justyna B Startek
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium
| | - José Ramón López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Universidad de Valladolid y CSIC, Sanz y Forés 3, 47003 Valladolid, Spain
| | - María Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Universidad de Valladolid y CSIC, Sanz y Forés 3, 47003 Valladolid, Spain
| | - Karel Talavera
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium.
| |
Collapse
|
50
|
Cabanas H, Muraki K, Eaton N, Balinas C, Staines D, Marshall-Gradisnik S. Loss of Transient Receptor Potential Melastatin 3 ion channel function in natural killer cells from Chronic Fatigue Syndrome/Myalgic Encephalomyelitis patients. Mol Med 2018; 24:44. [PMID: 30134818 PMCID: PMC6092868 DOI: 10.1186/s10020-018-0046-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/01/2018] [Indexed: 02/05/2023] Open
Abstract
Background Chronic Fatigue Syndrome (CFS)/ Myalgic Encephalomyelitis (ME) is a debilitating disorder that is accompanied by reduced cytotoxic activity in natural killer (NK) cells. NK cells are an essential innate immune cell, responsible for recognising and inducing apoptosis of tumour and virus infected cells. Calcium is an essential component in mediating this cellular function. Transient Receptor Potential Melastatin 3 (TRPM3) cation channels have an important regulatory role in mediating calcium influx to help maintain cellular homeostasis. Several single nucleotide polymorphisms have been reported in TRPM3 genes from isolated peripheral blood mononuclear cells, NK and B cells in patients with CFS/ME and have been proposed to correlate with illness presentation. Moreover, a significant reduction in both TRPM3 surface expression and intracellular calcium mobilisation in NK cells has been found in CFS/ME patients compared with healthy controls. Despite the functional importance of TRPM3, little is known about the ion channel function in NK cells and the epiphenomenon of CFS/ME. The objective of the present study was to characterise the TRPM3 ion channel function in NK cells from CFS/ME patients in comparison with healthy controls using whole cell patch-clamp techniques. Methods NK cells were isolated from 12 age- and sex-matched healthy controls and CFS patients. Whole cell electrophysiology recording has been used to assess TRPM3 ion channel activity after modulation with pregnenolone sulfate and ononetin. Results We report a significant reduction in amplitude of TRPM3 current after pregnenolone sulfate stimulation in isolated NK cells from CFS/ME patients compared with healthy controls. In addition, we found pregnenolone sulfate-evoked ionic currents through TRPM3 channels were significantly modulated by ononetin in isolated NK cells from healthy controls compared with CFS/ME patients. Conclusions TRPM3 activity is impaired in CFS/ME patients suggesting changes in intracellular Ca2+ concentration, which may impact NK cellular functions. This investigation further helps to understand the intracellular-mediated roles in NK cells and confirm the potential role of TRPM3 ion channels in the aetiology and pathomechanism of CFS/ME.
Collapse
Affiliation(s)
- Hélène Cabanas
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia. .,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Chikusa, Nagoya, Japan
| | - Natalie Eaton
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Cassandra Balinas
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Donald Staines
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- School of Medical Science, Griffith University, Gold Coast, QLD, Australia.,The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|