1
|
Luo Y, Sun L, Peng Y. The structural basis of the G protein-coupled receptor and ion channel axis. Curr Res Struct Biol 2025; 9:100165. [PMID: 40083915 PMCID: PMC11904507 DOI: 10.1016/j.crstbi.2025.100165] [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: 09/26/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Sensory neurons play an essential role in recognizing and responding to detrimental, irritating, and inflammatory stimuli from our surroundings, such as pain, itch, cough, and neurogenic inflammation. The transduction of these physiological signals is chiefly mediated by G protein-coupled receptors (GPCRs) and ion channels. The binding of ligands to GPCRs triggers a signaling cascade, recruiting G proteins or β-arrestins, which subsequently interact with ion channels (e.g., GIRK and TRP channels). This interaction leads to the sensitization and activation of these channels, initiating the neuron's protective mechanisms. This review delves into the complex interplay between GPCRs and ion channels that underpin these physiological processes, with a particular focus on the role of structural biology in enhancing our comprehension. Through unraveling the intricacies of the GPCR-ion channel axis, we aim to shed light on the sophisticated intermolecular dynamics within these pivotal membrane protein families, ultimately guiding the development of precise therapeutic interventions.
Collapse
Affiliation(s)
- Yulin Luo
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, L Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Liping Sun
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| |
Collapse
|
2
|
Juen Z, Lu Z, Yu R, Chang AN, Wang B, Fitzpatrick AWP, Zuker CS. The structure of human sweetness. Cell 2025:S0092-8674(25)00456-8. [PMID: 40339580 DOI: 10.1016/j.cell.2025.04.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/03/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025]
Abstract
In humans, the detection and ultimately the perception of sweetness begin in the oral cavity, where taste receptor cells (TRCs) dedicated to sweet-sensing interact with sugars, artificial sweeteners, and other sweet-tasting chemicals. Human sweet TRCs express on their cell surface a sweet receptor that initiates the cascade of signaling events responsible for our strong attraction to sweet stimuli. Here, we describe the cryo-electron microscopy (cryo-EM) structure of the human sweet receptor bound to two of the most widely used artificial sweeteners-sucralose and aspartame. Our results reveal the structural basis for sweet detection, provide insights into how a single receptor mediates all our responses to such a wide range of sweet-tasting compounds, and open up unique possibilities for designing a generation of taste modulators informed by the structure of the human receptor.
Collapse
Affiliation(s)
- Zhang Juen
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Zhengyuan Lu
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ruihuan Yu
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Andrew N Chang
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Brian Wang
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anthony W P Fitzpatrick
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Charles S Zuker
- Zuckerman Mind Brain Behavior Institute and Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Howard Hughes Medical Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
3
|
Zhang Y, Zhuang H, Ren X, Zhou P. Implications of mechanosensitive ion channels in the pathogenesis of osteoarthritis: a comprehensive review. Front Cell Dev Biol 2025; 13:1549812. [PMID: 40376614 PMCID: PMC12078208 DOI: 10.3389/fcell.2025.1549812] [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: 12/22/2024] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Osteoarthritis (OA) is the predominant cause of joint pain and limited mobility in older people, and its prevalence is increasing as the population ages. Given the lack of effective therapeutic interventions, the disability rate associated with OA is a staggering 53%, which significantly affects the wellbeing of those affected and represents a significant social and family financial burden. Consequently, OA has emerged as a pressing social and public health concern globally. Various forms of mechanical strain, such as dynamic compression, fluid shear, tissue shear, and hydrostatic pressure, serve as crucial physical stimuli perceived by chondrocytes. Recent studies indicate that aberrant mechanical loading represents a fundamental risk factor for OA. Upon exposure to mechanical loading, chondrocytes translate mechanical cues into chemical signals primarily via mechanosensitive ion channels, resulting in alterations in cartilage metabolism. Numerous studies have demonstrated the significance of mechanosensitive ion channels in the pathogenesis of OA, suggesting that therapeutic interventions targeting these channels on chondrocytes may offer potential benefits.
Collapse
Affiliation(s)
| | | | | | - Panghu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Yin Y, Park CG, Feng S, Guan Z, Lee HJ, Zhang F, Sharma K, Borgnia MJ, Im W, Lee SY. Molecular basis of neurosteroid and anticonvulsant regulation of TRPM3. Nat Struct Mol Biol 2025; 32:828-840. [PMID: 39809942 DOI: 10.1038/s41594-024-01463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Transient receptor potential channel subfamily M member 3 (TRPM3) is a Ca2+-permeable cation channel activated by the neurosteroid pregnenolone sulfate (PregS) or heat, serving as a nociceptor in the peripheral sensory system. Recent discoveries of autosomal dominant neurodevelopmental disorders caused by gain-of-function mutations in TRPM3 highlight its role in the central nervous system. Notably, the TRPM3 inhibitor primidone, an anticonvulsant, has proven effective in treating patients with TRPM3-linked neurological disorders and in mouse models of thermal nociception. However, our understanding of neurosteroids, inhibitors and disease mutations on TRPM3 is limited. Here we present cryogenic electron microscopy structures of the mouse TRPM3 in complex with cholesteryl hemisuccinate, primidone and PregS with the synthetic agonist CIM 0216. Our studies identify the binding sites for the neurosteroid, synthetic agonist and inhibitor and offer insights into their effects and disease mutations on TRPM3 gating, aiding future drug development.
Collapse
Affiliation(s)
- Ying Yin
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Cheon-Gyu Park
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Shasha Feng
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Hyuk-Joon Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Feng Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Kedar Sharma
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Mario J Borgnia
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
5
|
Kulkarni K, Walton RD, Chaigne S. Unlocking the potential of cardiac TRP channels using knockout mice models. Front Physiol 2025; 16:1585356. [PMID: 40313873 PMCID: PMC12043714 DOI: 10.3389/fphys.2025.1585356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Affiliation(s)
- Kanchan Kulkarni
- IHU Liryc, INSERM, U1045, CRCTB, University Bordeaux, Bordeaux, France
| | - Richard D. Walton
- IHU Liryc, INSERM, U1045, CRCTB, University Bordeaux, Bordeaux, France
| | - Sebastien Chaigne
- IHU Liryc, INSERM, U1045, CRCTB, University Bordeaux, Bordeaux, France
- CHU de Bordeaux, Cardiology, INSERM, U1045, CRCTB, Bordeaux, France
| |
Collapse
|
6
|
Thiel G, Rössler OG. Stimulus-Transcription Coupling of TRPM3 Channels: A Signaling Pathway from the Plasma Membrane to the Nucleus. Biomolecules 2025; 15:521. [PMID: 40305282 PMCID: PMC12025076 DOI: 10.3390/biom15040521] [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: 01/19/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Transient receptor potential melastatin-3 (TRPM3) channels are cation channels activated by heat and chemical ligands. TRPM3 regulates heat sensation, secretion, neurotransmitter release, iris constriction, and tumor promotion. Stimulation of TRPM3 triggers an influx of Ca2+ ions into the cells and the initiation of an intracellular signaling cascade. TRPM3 channels are regulated by phosphatidylinositol 4,5-bisphosphate, the βγ subunit of G-protein-coupled receptors, phospholipase C, and calmodulin. Extracellular signal-regulated protein kinase ERK1/2 and c-Jun N-terminal protein kinase (JNK) function as signal transducers. The signaling cascade is negatively regulated by the protein phosphatases MKP-1 and calcineurin and increased concentrations of Zn2+. Stimulation of TRPM3 leads to the activation of stimulus-responsive transcription factors controlled by epigenetic regulators. Potential delayed response genes encoding the pro-inflammatory regulators interleukin-8, calcitonin gene-related peptide, and the prostaglandin-synthesizing enzyme prostaglandin endoperoxide synthase-2 have been identified. Elucidating the TRPM3-induced signaling cascade provides insights into how TRPM3 stimulation alters numerous biochemical and physiological parameters within the cell and throughout the organism and offers intervention points for manipulating TRPM3 signaling and function.
Collapse
Affiliation(s)
- Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Medical Faculty, Saarland University, Building 44, 66421 Homburg, Germany;
| | | |
Collapse
|
7
|
Ovechkina VS, Andrianova SK, Shimanskaia IO, Suvorova PS, Ryabinina AY, Blagonravov ML, Belousov VV, Mozhaev AA. Advances in Optogenetics and Thermogenetics for Control of Non-Neuronal Cells and Tissues in Biomedical Research. ACS Chem Biol 2025; 20:553-572. [PMID: 40056098 DOI: 10.1021/acschembio.4c00842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Optogenetics and chemogenetics are relatively new biomedical technologies that emerged 20 years ago and have been evolving rapidly since then. This has been made possible by the combined use of genetic engineering, optics, and electrophysiology. With the development of optogenetics and thermogenetics, the molecular tools for cellular control are continuously being optimized, studied, and modified, expanding both their applications and their biomedical uses. The most notable changes have occurred in the basic life sciences, especially in neurobiology and the activation of neurons to control behavior. Currently, these methods of activation have gone far beyond neurobiology and are being used in cardiovascular research, for potential cancer therapy, to control metabolism, etc. In this review, we provide brief information on the types of molecular tools for optogenetic and thermogenetic methods─microbial rhodopsins and proteins of the TRP superfamily─and also consider their applications in the field of activation of non-neuronal tissues and mammalian cells. We also consider the potential of these technologies and the prospects for the use of optogenetics and thermogenetics in biomedical research.
Collapse
Affiliation(s)
- Vera S Ovechkina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Sofya K Andrianova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Iana O Shimanskaia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Polina S Suvorova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Anna Y Ryabinina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Mikhail L Blagonravov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Vsevolod V Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117513, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, 121205, Russia
| | - Andrey A Mozhaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| |
Collapse
|
8
|
Jolitz L, Helbig I, Fitzgerald MP, McKeown Ruggiero S, Cohen S, Angelini C, Vallespin E, Michaud V, Gerasimenko A, Cogne B, Isidor B, Keren B, Dyment D, Heron D, Karstensen HG, Cuppen I, Christodoulou J, Wilson M, Lake NJ, Biskup S, Syrbe S, Mori T, Becker L, Kaindl AM. Phenotype Spectrum of TRPM3-Associated Disorders. Ann Neurol 2025; 97:561-570. [PMID: 39749750 PMCID: PMC11831877 DOI: 10.1002/ana.27141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE Monoallelic variants in the transient receptor potential melastatin-related type 3 gene (TRPM3) have been associated with neurodevelopmental manifestations, but knowledge on the clinical manifestations and treatment options is limited. We characterized the clinical spectrum, highlighting particularly the epilepsy phenotype, and the effect of treatments. METHODS We analyzed retrospectively the phenotypes and genotypes of 43 individuals with TRPM3 variants, acquired from GeneMatcher and collaborations (n = 21), and through a systematic literature search (n = 22). We included all patients with a pathogenic TRPM3 variant. RESULTS The median age at the time of the study was 10 years, with a preponderance of girls (60%) versus boys (40%). Frequent findings were developmental delay and/or intellectual disability (93%), global or axial hypotonia (77%), ocular involvement (70%), musculoskeletal anomalies (65%), and dysmorphic features (58%). Epilepsy was diagnosed in 31 patients (72%), classified in all as developmental and epileptic encephalopathy with or without spike wave activation in sleep (DEE/DEE-SWAS). Patients with the variant p.Val1002Met (n = 24) significantly more often had developmental delay and epilepsy. The most effective anti-seizure medication was primidone. All treated patients showed an improvement in seizure frequency, motor and speech development, and/or learning capability with this drug. INTERPRETATION Developmental delay/intellectual disability and epilepsy are dominant phenotypic features in patients with TRPM3 variants. Given that epilepsy can negatively impact development, screening for awake and sleep electroencephalogram abnormalities and other manifestations are essential to offer early intervention. The TRPM3 channel blocker primidone has shown promising effects and should be considered in every child with a TRPM3 gain-of-function variant. ANN NEUROL 2025;97:561-570.
Collapse
Affiliation(s)
- Laura Jolitz
- Department of Pediatric NeurologyCharité–Universitätsmedizin BerlinBerlinGermany
- Center for Chronically Sick ChildrenCharité–Universitätsmedizin BerlinBerlinGermany
- Institute for Cell Biology and NeurobiologyCharité–Universitätsmedizin BerlinBerlinGermany
- Section CNS Development and Neurologic DiseaseGerman Center for Child and Adolescent Health (DZKJ)partner site BerlinGermany
| | - Ingo Helbig
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Mark P. Fitzgerald
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
- Epilepsy Neurogenetics InitiativeChildren's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | | | - Stacey Cohen
- Department of Medicine, Division of Translational Medicine and Human GeneticsUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Chloe Angelini
- Department of Medical GeneticsGroupe Hospitalier Pellegrin, CHU BordeauxBordeaux CedexFrance
| | - Elena Vallespin
- Medical and Molecular Genetics Institute (INGEMM) IdiPaz, CIBERERHospital Universitario La PazMadridSpain
| | - Vincent Michaud
- Service de Génétique MédicaleCHU de BordeauxBordeauxFrance
- INSERM U1211, Maladies Rares, Génétique et MétabolismeUniversité de BordeauxBordeauxFrance
| | - Anna Gerasimenko
- Institut du Cerveau – Paris Brain Institute – ICM, Inserm, CNRSSorbonne UniversitéParisFrance
- Département de Génétique, Centre de référence “déficiences intellectuelles de causes rares”APHP Sorbonne Université, GH Pitié Salpêtrière et TrousseauParisFrance
| | - Benjamin Cogne
- Service de Génétique MédicaleNantes Université, CHU de NantesNantesFrance
- Laboratoire de Biologie Médicale Multi‐Sites SeqOIA (laboratoire‐seqoia.fr)ParisFrance
| | - Bertrand Isidor
- Service de Génétique MédicaleNantes Université, CHU de NantesNantesFrance
| | - Boris Keren
- Département de génétique, Hôpital Pitié‐SalpêtrièreAssistance Publique‐Hôpitaux de ParisParisFrance
| | - David Dyment
- Children's Hospital of Eastern Ontario Research InstituteOttawaCanada
| | - Delphine Heron
- Département de Génétique, Centre de référence “déficiences intellectuelles de causes rares”APHP Sorbonne Université, GH Pitié Salpêtrière et TrousseauParisFrance
| | - Helena Gásdal Karstensen
- Dept. of Genetics, Center of DiagnosticsCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
| | - Inge Cuppen
- Department of Child NeurologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - John Christodoulou
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of MelbourneParkvilleAustralia
| | - Meredith Wilson
- Department of Clinical GeneticsThe Children's Hospital at WestmeadSydneyAustralia
- Discipline of Genomic MedicineUniversity of SydneySydneyAustralia
| | - Nicole J. Lake
- Department of GeneticsYale School of MedicineNew HavenCTUSA
| | - Saskia Biskup
- Center for Genomics and Transcriptomics (CeGaT)TübingenGermany
| | - Steffen Syrbe
- Heidelberg University, Medical Faculty of Heidelberg, Center for Child and Adolescent MedicineDivision of Pediatric EpileptologyHeidelbergGermany
| | - Takayasu Mori
- Department of NephrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Lena‐Luise Becker
- Department of Pediatric NeurologyCharité–Universitätsmedizin BerlinBerlinGermany
- Center for Chronically Sick ChildrenCharité–Universitätsmedizin BerlinBerlinGermany
- Institute for Cell Biology and NeurobiologyCharité–Universitätsmedizin BerlinBerlinGermany
- Section CNS Development and Neurologic DiseaseGerman Center for Child and Adolescent Health (DZKJ)partner site BerlinGermany
| | - Angela M. Kaindl
- Department of Pediatric NeurologyCharité–Universitätsmedizin BerlinBerlinGermany
- Center for Chronically Sick ChildrenCharité–Universitätsmedizin BerlinBerlinGermany
- Institute for Cell Biology and NeurobiologyCharité–Universitätsmedizin BerlinBerlinGermany
- Section CNS Development and Neurologic DiseaseGerman Center for Child and Adolescent Health (DZKJ)partner site BerlinGermany
| |
Collapse
|
9
|
Deng Z, Chen X, Zhang R, Kong L, Fang Y, Guo J, Shen B, Zhang L. Delta opioid peptide [D-ala2, D-leu5]-Enkephalin's ability to enhance mitophagy via TRPV4 to relieve ischemia/reperfusion injury in brain microvascular endothelial cells. Stroke Vasc Neurol 2025; 10:32-44. [PMID: 38697767 PMCID: PMC11877439 DOI: 10.1136/svn-2023-003080] [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: 01/03/2024] [Accepted: 04/20/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Local brain tissue can suffer from ischaemia/reperfusion (I/R) injury, which lead to vascular endothelial damage. The peptide δ opioid receptor (δOR) agonist [D-ala2, D-leu5]-Enkephalin (DADLE) can reduce apoptosis caused by acute I/R injury in brain microvascular endothelial cells (BMECs). OBJECTIVE This study aims to explore the mechanism by which DADLE enhances the level of mitophagy in BMECs by upregulating the expression of transient receptor potential vanilloid subtype 4 (TRPV4). METHODS BMECs were extracted and made to undergo oxygen-glucose deprivation/reoxygenation (OGD/R) accompanied by DADLE. RNA-seq analysis revealed that DADLE induced increased TRPV4 expression. The CCK-8 method was used to assess the cellular viability; quantitative PCR (qPCR) was used to determine the mRNA expression of Drp1; western blot was used to determine the expression of TRPV4 and autophagy-related proteins; and calcium imaging was used to detect the calcium influx. Autophagosomes in in the cells' mitochondria were observed by using transmission electron microscopy. ELISA was used to measure ATP content, and a JC-1 fluorescent probe was used to detect mitochondrial membrane potential. RESULTS When compared with the OGD/R group, OGD/R+DADLE group showed significantly enhanced cellular viability; increased expression of TRPV4, Beclin-1, LC3-II/I, PINK1 and Parkin; decreased p62 expression; a marked rise in calcium influx; further increases in mitophagy, an increase in ATP synthesis and an elevation of mitochondrial membrane potential. These protective effects of DADLE can be blocked by a TRPV4 inhibitor HC067047 or RNAi of TRPV4. CONCLUSION DADLE can promote mitophagy in BMECs through TRPV4, improving mitochondrial function and relieving I/R injury.
Collapse
Affiliation(s)
- Zhongfang Deng
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyu Chen
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Ran Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Lingchao Kong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Fang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| | - Jizheng Guo
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui, China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao
| | - Lesha Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
10
|
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
|
11
|
Talyzina IA, Nadezhdin KD, Sobolevsky AI. Forty sites of TRP channel regulation. Curr Opin Chem Biol 2025; 84:102550. [PMID: 39615427 PMCID: PMC11788071 DOI: 10.1016/j.cbpa.2024.102550] [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: 09/16/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 12/14/2024]
Abstract
Transient receptor potential (TRP) channels are polymodal molecular sensors that integrate chemical, thermal, mechanical and electrical stimuli and convert them into ionic currents that regulate senses of taste, smell, vision, hearing, touch and contribute to perception of temperature and pain. TRP channels are implicated in the pathogenesis of numerous human diseases, including cancers, and represent one of the most ardently pursued drug targets. Recent advances in structural biology, particularly associated with the cryo-EM "resolution revolution", yielded numerous TRP channel structures in complex with ligands that might have therapeutic potential. In this review, we describe the recent progress in TRP channel structural biology, focusing on the description of identified binding sites for small molecules, their relationship to membrane lipids, and interaction of TRP channels with other proteins. The characterized binding sites and interfaces create a diversity of druggable targets and provide a roadmap to aid in the design of new molecules for tuning TRP channel function in disease conditions.
Collapse
Affiliation(s)
- Irina A Talyzina
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
12
|
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
|
13
|
Kumamoto E. Anesthetic- and Analgesic-Related Drugs Modulating Both Voltage-Gated Na + and TRP Channels. Biomolecules 2024; 14:1619. [PMID: 39766326 PMCID: PMC11727300 DOI: 10.3390/biom14121619] [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: 11/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Nociceptive information is transmitted by action potentials (APs) through primary afferent neurons from the periphery to the central nervous system. Voltage-gated Na+ channels are involved in this AP production, while transient receptor potential (TRP) channels, which are non-selective cation channels, are involved in receiving and transmitting nociceptive stimuli in the peripheral and central terminals of the primary afferent neurons. Peripheral terminal TRP vanilloid-1 (TRPV1), ankylin-1 (TRPA1) and melastatin-8 (TRPM8) activation produces APs, while central terminal TRP activation enhances the spontaneous release of L-glutamate from the terminal to spinal cord and brain stem lamina II neurons that play a pivotal role in modulating nociceptive transmission. There is much evidence demonstrating that chemical compounds involved in Na+ channel (or nerve AP conduction) inhibition modify TRP channel functions. Among these compounds are local anesthetics, anti-epileptics, α2-adrenoceptor agonists, antidepressants (all of which are used as analgesic adjuvants), general anesthetics, opioids, non-steroidal anti-inflammatory drugs and plant-derived compounds, many of which are involved in antinociception. This review mentions the modulation of Na+ channels and TRP channels including TRPV1, TRPA1 and TRPM8, both of which modulations are produced by pain-related compounds.
Collapse
Affiliation(s)
- Eiichi Kumamoto
- Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
14
|
Payrits M, Zsidó BZ, Nehr-Majoros AK, Börzsei R, Helyes Z, Hetényi C, Szőke É. Lipid raft disruption inhibits the activation of Transient Receptor Potential Vanilloid 1, but not TRP Melastatin 3 and the voltage-gated L-type calcium channels in sensory neurons. Front Cell Dev Biol 2024; 12:1452306. [PMID: 39676793 PMCID: PMC11638188 DOI: 10.3389/fcell.2024.1452306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
Transient Receptor Potential (TRP) ion channels like Vanilloid 1 (TRPV1) and Melastatin 3 (TRPM3) are nonselective cation channels expressed in primary sensory neurons and peripheral nerve endings, which are located in cholesterol- and sphingolipid-rich membrane lipid raft regions and have important roles in pain processing. Besides TRP ion channels a wide variety of voltage-gated ion channels were also described in the membrane raft regions of neuronal cells. Here we investigated the effects of lipid raft disruption by methyl-beta-cyclodextrin (MCD) and sphingomyelinase (SMase) on TRPV1, TRPM3 and voltage-gated L-type Ca2+ channel activation in cultured trigeminal neurons and sensory nerve terminals of the trachea. We also examined the mechanism of action of MCD by in silico modeling. Disruption of lipid rafts by MCD or SMase did not alter CIM0216-induced TRPM3 cation channel activation and the voltage-gated L-type Ca2+ channel activation by FPL 64126 or veratridine neither on trigeminal sensory neurons nor sensory nerve terminals. We provided the first structural explanation with in silico modeling that the activation of TRPV1, TRPM3 and voltage-gated L-type Ca2+ channels is affected differently by the cholesterol content surrounding them in the plasma membrane. It is concluded that modifying the hydrophobic interactions between lipid rafts and ion channels might provide a selective novel mechanism for peripheral analgesia.
Collapse
Affiliation(s)
- Maja Payrits
- Department of Pharmacology and Pharmacotherapy, Medical School and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group, Pécs, Hungary
| | - Balázs Zoltán Zsidó
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Pharmacoinformatics Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Andrea Kinga Nehr-Majoros
- Department of Pharmacology and Pharmacotherapy, Medical School and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group, Pécs, Hungary
| | - Rita Börzsei
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Pharmacoinformatics Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group, Pécs, Hungary
| | - Csaba Hetényi
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Pharmacoinformatics Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group, Pécs, Hungary
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Uchida K. TRPM3, TRPM4, and TRPM5 as thermo-sensitive channels. J Physiol Sci 2024; 74:43. [PMID: 39294615 PMCID: PMC11409758 DOI: 10.1186/s12576-024-00937-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 09/01/2024] [Indexed: 09/20/2024]
Abstract
Temperature detection is essential for the survival and perpetuation of any species. Thermoreceptors in the skin sense body temperature as well as the temperatures of ambient air and objects. Since Dr. David Julius and his colleagues discovered that TRPV1 is expressed in small-diameter primary sensory neurons, and activated by temperatures above 42 °C, 11 of thermo-sensitive TRP channels have been identified. TRPM3 expressed in sensory neurons acts as a sensor for noxious heat. TRPM4 and TRPM5 are Ca2⁺-activated monovalent cation channels, and their activity is drastically potentiated by temperature increase. This review aims to summarize the expression patterns, electrophysiological properties, and physiological roles of TRPM3, TRPM4, and TRPM5 associated with thermosensation.
Collapse
Affiliation(s)
- Kunitoshi Uchida
- Laboratory of Functional Physiology, Department of Environmental and Life Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Yada 52-1, Suruga-Ku, Shizuoka, Shizuoka, 422-8526, Japan.
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
18
|
Dai P, Chen C, Yu J, Ma C, Zhang X. New insights into sperm physiology regulation: Enlightenment from G-protein-coupled receptors. Andrology 2024; 12:1253-1271. [PMID: 38225815 DOI: 10.1111/andr.13593] [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: 10/13/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND G-protein-coupled receptors are critical in many physiological and pathological processes in various organs. Serving as the control panel for sensing extracellular stimuli, G-protein-coupled receptors recognise various ligands, including light, temperature, odours, pheromones, hormones, neurotransmitters, chemokines, etc. Most recently, G-protein-coupled receptors residing in spermatozoa have been found to be indispensable for sperm function. OBJECTIVE Here, we have summarised cutting-edge findings on the functional mechanisms of G-protein-coupled receptors that are known to be associated with sperm functions and the activation of their downstream effectors, providing new insights into the roles of G-protein-coupled receptors in sperm physiology. RESULTS Emerging studies hint that alterations in G-protein-coupled receptors could affect sperm function, implicating their role in fertility, but solid evidence needs to be continuing excavated with various means. Several members of the G-protein-coupled receptor superfamily, including olfactory receptors, opsins, orphan G-protein-coupled receptors, CXC chemokine receptor 4, CC chemokine receptor 5 and CC chemokine receptor 6 as well as their downstream effector β-arrestins, etc., were suggested to be essential for sperm motility, capacitation, thermotaxis, chemotaxis, Ca2+ influx through CatSper channel and fertilisation capacity. CONCLUSION The present review provides a comprehensive overview of studies describing G-protein-coupled receptors and their potential action in sperm function. We also present a critical discussion of these issues, and a possible framework for future investigations on the diverse ligands, biological functions and cell signalling of G-protein-coupled receptors in spermatozoa. Here, the G-protein-coupled receptors and their related G proteins that specifically were identified in spermatozoa were summarised, and provided references valuable for further illumination, despite the evidence that is not overwhelming in most cases.
Collapse
Affiliation(s)
- Pengyuan Dai
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Chen Chen
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Jingyan Yu
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Chaoye Ma
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| |
Collapse
|
19
|
Karuppan S, Schrag LG, Pastrano CM, Jara-Oseguera A, Zubcevic L. Structural dynamics at cytosolic interprotomer interfaces control gating of a mammalian TRPM5 channel. Proc Natl Acad Sci U S A 2024; 121:e2403333121. [PMID: 38923985 PMCID: PMC11228501 DOI: 10.1073/pnas.2403333121] [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: 02/20/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
The transient receptor potential melastatin (TRPM) tetrameric cation channels are involved in a wide range of biological functions, from temperature sensing and taste transduction to regulation of cardiac function, inflammatory pain, and insulin secretion. The structurally conserved TRPM cytoplasmic domains make up >70 % of the total protein. To investigate the mechanism by which the TRPM cytoplasmic domains contribute to gating, we employed electrophysiology and cryo-EM to study TRPM5-a channel that primarily relies on activation via intracellular Ca2+. Here, we show that activation of mammalian TRPM5 channels is strongly altered by Ca2+-dependent desensitization. Structures of rat TRPM5 identify a series of conformational transitions triggered by Ca2+ binding, whereby formation and dissolution of cytoplasmic interprotomer interfaces appear to control activation and desensitization of the channel. This study shows the importance of the cytoplasmic assembly in TRPM5 channel function and sets the stage for future investigations of other members of the TRPM family.
Collapse
Affiliation(s)
- Sebastian Karuppan
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, Kansas City, KS66160
| | - Lynn Goss Schrag
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, Kansas City, KS66160
| | - Caroline M. Pastrano
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX78712
| | - Andrés Jara-Oseguera
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX78712
| | - Lejla Zubcevic
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
20
|
Thiel G, Rössler OG. Signal Transduction of Transient Receptor Potential TRPM8 Channels: Role of PIP5K, Gq-Proteins, and c-Jun. Molecules 2024; 29:2602. [PMID: 38893478 PMCID: PMC11174004 DOI: 10.3390/molecules29112602] [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: 04/30/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Transient receptor potential melastatin-8 (TRPM8) is a cation channel that is activated by cold and "cooling agents" such as menthol and icilin, which induce a cold sensation. The stimulation of TRPM8 activates an intracellular signaling cascade that ultimately leads to a change in the gene expression pattern of the cells. Here, we investigate the TRPM8-induced signaling pathway that links TRPM8 channel activation to gene transcription. Using a pharmacological approach, we show that the inhibition of phosphatidylinositol 4-phosphate 5 kinase α (PIP5K), an enzyme essential for the biosynthesis of phosphatidylinositol 4,5-bisphosphate, attenuates TRPM8-induced gene transcription. Analyzing the link between TRPM8 and Gq proteins, we show that the pharmacological inhibition of the βγ subunits impairs TRPM8 signaling. In addition, genetic studies show that TRPM8 requires an activated Gα subunit for signaling. In the nucleus, the TRPM8-induced signaling cascade triggers the activation of the transcription factor AP-1, a complex consisting of a dimer of basic region leucine zipper (bZIP) transcription factors. Here, we identify the bZIP protein c-Jun as an essential component of AP-1 within the TRPM8-induced signaling cascade. In summary, with PIP5K, Gq subunits, and c-Jun, we identified key molecules in TRPM8-induced signaling from the plasma membrane to the nucleus.
Collapse
Affiliation(s)
- Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Medical Faculty, Saarland University, 66421 Homburg, Germany;
| | | |
Collapse
|
21
|
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
|
22
|
Sasso EM, Muraki K, Eaton-Fitch N, Smith P, Jeremijenko A, Griffin P, Marshall-Gradisnik S. Investigation into the restoration of TRPM3 ion channel activity in post-COVID-19 condition: a potential pharmacotherapeutic target. Front Immunol 2024; 15:1264702. [PMID: 38765011 PMCID: PMC11099221 DOI: 10.3389/fimmu.2024.1264702] [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: 07/21/2023] [Accepted: 04/09/2024] [Indexed: 05/21/2024] Open
Abstract
Introduction Recently, we reported that post COVID-19 condition patients also have Transient Receptor Potential Melastatin 3 (TRPM3) ion channel dysfunction, a potential biomarker reported in natural killer (NK) cells from Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) patients. As there is no universal treatment for post COVID-19 condition, knowledge of ME/CFS may provide advances to investigate therapeutic targets. Naltrexone hydrochloride (NTX) has been demonstrated to be beneficial as a pharmacological intervention for ME/CFS patients and experimental investigations have shown NTX restored TRPM3 function in NK cells. This research aimed to: i) validate impaired TRPM3 ion channel function in post COVID-19 condition patients compared with ME/CFS; and ii) investigate NTX effects on TRPM3 ion channel activity in post COVID-19 condition patients. Methods Whole-cell patch-clamp was performed to characterize TRPM3 ion channel activity in freshly isolated NK cells of post COVID-19 condition (N = 9; 40.56 ± 11.26 years), ME/CFS (N = 9; 39.33 ± 9.80 years) and healthy controls (HC) (N = 9; 45.22 ± 9.67 years). NTX effects were assessed on post COVID-19 condition (N = 9; 40.56 ± 11.26 years) and HC (N = 7; 45.43 ± 10.50 years) where NK cells were incubated for 24 hours in two protocols: treated with 200 µM NTX, or non-treated; TRPM3 channel function was assessed with patch-clamp protocol. Results This investigation confirmed impaired TRPM3 ion channel function in NK cells from post COVID-19 condition and ME/CFS patients. Importantly, PregS-induced TRPM3 currents were significantly restored in NTX-treated NK cells from post COVID-19 condition compared with HC. Furthermore, the sensitivity of NK cells to ononetin was not significantly different between post COVID-19 condition and HC after treatment with NTX. Discussion Our findings provide further evidence identifying similarities of TRPM3 ion channel dysfunction between ME/CFS and post COVID-19 condition patients. This study also reports, for the first time, TRPM3 ion channel activity was restored in NK cells isolated from post COVID-19 condition patients after in vitro treatment with NTX. The TRPM3 restoration consequently may re-establish TRPM3-dependent calcium (Ca2+) influx. This investigation proposes NTX as a potential therapeutic intervention and TRPM3 as a treatment biomarker for post COVID-19 condition.
Collapse
Affiliation(s)
- Etianne Martini Sasso
- 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, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Katsuhiko Muraki
- Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, 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, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Peter Smith
- Consortium Health International for Myalgic Encephalomyelitis, National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- Clinical Medicine, Griffith University, Gold Coast, QLD, Australia
| | - Andrew Jeremijenko
- 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, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Paul Griffin
- Department of Medicine and Infectious Diseases, Mater Hospital and Mater Medical Research Institute, Brisbane, 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, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
23
|
Chubanov V, Köttgen M, Touyz RM, Gudermann T. TRPM channels in health and disease. Nat Rev Nephrol 2024; 20:175-187. [PMID: 37853091 DOI: 10.1038/s41581-023-00777-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Different cell channels and transporters tightly regulate cytoplasmic levels and the intraorganelle distribution of cations. Perturbations in these processes lead to human diseases that are frequently associated with kidney impairment. The family of melastatin-related transient receptor potential (TRPM) channels, which has eight members in mammals (TRPM1-TRPM8), includes ion channels that are highly permeable to divalent cations, such as Ca2+, Mg2+ and Zn2+ (TRPM1, TRPM3, TRPM6 and TRPM7), non-selective cation channels (TRPM2 and TRPM8) and monovalent cation-selective channels (TRPM4 and TRPM5). Three family members contain an enzymatic protein moiety: TRPM6 and TRPM7 are fused to α-kinase domains, whereas TRPM2 is linked to an ADP-ribose-binding NUDT9 homology domain. TRPM channels also function as crucial cellular sensors involved in many physiological processes, including mineral homeostasis, blood pressure, cardiac rhythm and immunity, as well as photoreception, taste reception and thermoreception. TRPM channels are abundantly expressed in the kidney. Mutations in TRPM genes cause several inherited human diseases, and preclinical studies in animal models of human disease have highlighted TRPM channels as promising new therapeutic targets. Here, we provide an overview of this rapidly evolving research area and delineate the emerging role of TRPM channels in kidney pathophysiology.
Collapse
Affiliation(s)
- Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| | - Michael Köttgen
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | - Rhian M Touyz
- Research Institute of McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| |
Collapse
|
24
|
Rohacs T. Phosphoinositide Regulation of TRP Channels: A Functional Overview in the Structural Era. Annu Rev Physiol 2024; 86:329-355. [PMID: 37871124 DOI: 10.1146/annurev-physiol-042022-013956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Transient receptor potential (TRP) ion channels have diverse activation mechanisms including physical stimuli, such as high or low temperatures, and a variety of intracellular signaling molecules. Regulation by phosphoinositides and their derivatives is their only known common regulatory feature. For most TRP channels, phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] serves as a cofactor required for activity. Such dependence on PI(4,5)P2 has been demonstrated for members of the TRPM subfamily and for the epithelial TRPV5 and TRPV6 channels. Intracellular TRPML channels show specific activation by PI(3,5)P2. Structural studies uncovered the PI(4,5)P2 and PI(3,5)P2 binding sites for these channels and shed light on the mechanism of channel opening. PI(4,5)P2 regulation of TRPV1-4 as well as some TRPC channels is more complex, involving both positive and negative effects. This review discusses the functional roles of phosphoinositides in TRP channel regulation and molecular insights gained from recent cryo-electron microscopy structures.
Collapse
Affiliation(s)
- Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey;
| |
Collapse
|
25
|
Ujisawa T, Lei J, Kashio M, Tominaga M. Thermal gradient ring for analysis of temperature-dependent behaviors involving TRP channels in mice. J Physiol Sci 2024; 74:9. [PMID: 38331738 PMCID: PMC10851596 DOI: 10.1186/s12576-024-00903-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024]
Abstract
There are a lot of temperature-sensitive proteins including transient receptor potential (TRP) channels. Some TRP channels are temperature receptors having specific activation temperatures in vitro that are within the physiological temperature range. Mice deficient in specific TRP channels show abnormal thermal behaviors, but the role of TRP channels in these behaviors is not fully understood. The Thermal Gradient Ring is a new apparatus that allows mice to freely move around the ring floor and not stay in a corner. The system can analyze various factors (e.g., 'Spent time', 'Travel distance', 'Moving speed', 'Acceleration') associated with temperature-dependent behaviors of TRP-deficient mice. For example, the Ring system clearly discriminated differences in temperature-dependent phenotypes between mice with diabetic peripheral neuropathy and TRPV1-/- mice, and demonstrated the importance of TRPV3 in temperature detection in skin. Studies using the Thermal Gradient Ring system can increase understanding of the molecular basis of thermal behaviors in mice and in turn help develop strategies to affect responses to different temperature conditions in humans.
Collapse
Affiliation(s)
- Tomoyo Ujisawa
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
| | - Jing Lei
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
| | - Makiko Kashio
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.
- Course of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan.
| |
Collapse
|
26
|
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
|
27
|
Wu F, Bu S, Wang H. Role of TRP Channels in Metabolism-Related Diseases. Int J Mol Sci 2024; 25:692. [PMID: 38255767 PMCID: PMC10815096 DOI: 10.3390/ijms25020692] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
Metabolic syndrome (MetS), with its high prevalence and significant impact on cardiovascular disease, poses a substantial threat to human health. The early identification of pathological abnormalities related to MetS and prevention of the risk of associated diseases is of paramount importance. Transient Receptor Potential (TRP) channels, a type of nonselective cation channel, are expressed in a variety of tissues and have been implicated in the onset and progression of numerous metabolism-related diseases. This study aims to review and discuss the expression and function of TRP channels in metabolism-related tissues and blood vessels, and to elucidate the interactions and mechanisms between TRP channels and metabolism-related diseases. A comprehensive literature search was conducted using keywords such as TRP channels, metabolic syndrome, pancreas, liver, oxidative stress, diabetes, hypertension, and atherosclerosis across various academic databases including PubMed, Google Scholar, Elsevier, Web of Science, and CNKI. Our review of the current research suggests that TRP channels may be involved in the development of metabolism-related diseases by regulating insulin secretion and release, lipid metabolism, vascular functional activity, oxidative stress, and inflammatory response. TRP channels, as nonselective cation channels, play pivotal roles in sensing various intra- and extracellular stimuli and regulating ion homeostasis by osmosis. They present potential new targets for the diagnosis or treatment of metabolism-related diseases.
Collapse
Affiliation(s)
| | | | - Hongmei Wang
- School of Medicine, Southeast University, Nanjing 210009, China; (F.W.); (S.B.)
| |
Collapse
|
28
|
Humer C, Radiskovic T, Horváti K, Lindinger S, Groschner K, Romanin C, Höglinger C. Bidirectional Allosteric Coupling between PIP 2 Binding and the Pore of the Oncochannel TRPV6. Int J Mol Sci 2024; 25:618. [PMID: 38203789 PMCID: PMC10779433 DOI: 10.3390/ijms25010618] [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: 12/08/2023] [Revised: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
The epithelial ion channel TRPV6 plays a pivotal role in calcium homeostasis. Channel function is intricately regulated at different stages, involving the lipid phosphatidylinositol-4,5-bisphosphate (PIP2). Given that dysregulation of TRPV6 is associated with various diseases, including different types of cancer, there is a compelling need for its pharmacological targeting. Structural studies provide insights on how TRPV6 is affected by different inhibitors, with some binding to sites else occupied by lipids. These include the small molecule cis-22a, which, however, also binds to and thereby blocks the pore. By combining calcium imaging, electrophysiology and optogenetics, we identified residues within the pore and the lipid binding site that are relevant for regulation by cis-22a and PIP2 in a bidirectional manner. Yet, mutation of the cytosolic pore exit reduced inhibition by cis-22a but preserved sensitivity to PIP2 depletion. Our data underscore allosteric communication between the lipid binding site and the pore and vice versa for most sites along the pore.
Collapse
Affiliation(s)
- Christina Humer
- Institute of Biophysics, Johannes Kepler University Linz, 4020 Linz, Austria; (C.H.); (T.R.); (C.R.)
| | - Tamara Radiskovic
- Institute of Biophysics, Johannes Kepler University Linz, 4020 Linz, Austria; (C.H.); (T.R.); (C.R.)
| | - Kata Horváti
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary;
| | - Sonja Lindinger
- Institute of Biophysics, Johannes Kepler University Linz, 4020 Linz, Austria; (C.H.); (T.R.); (C.R.)
| | - Klaus Groschner
- Gottfried Schatz Research Center, Division of Biophysics, Medical University of Graz, 8010 Graz, Austria;
| | - Christoph Romanin
- Institute of Biophysics, Johannes Kepler University Linz, 4020 Linz, Austria; (C.H.); (T.R.); (C.R.)
| | - Carmen Höglinger
- Institute of Biophysics, Johannes Kepler University Linz, 4020 Linz, Austria; (C.H.); (T.R.); (C.R.)
| |
Collapse
|
29
|
Vlachova V, Barvik I, Zimova L. Human Transient Receptor Potential Ankyrin 1 Channel: Structure, Function, and Physiology. Subcell Biochem 2024; 104:207-244. [PMID: 38963489 DOI: 10.1007/978-3-031-58843-3_10] [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: 07/05/2024]
Abstract
The transient receptor potential ion channel TRPA1 is a Ca2+-permeable nonselective cation channel widely expressed in sensory neurons, but also in many nonneuronal tissues typically possessing barrier functions, such as the skin, joint synoviocytes, cornea, and the respiratory and intestinal tracts. Here, the primary role of TRPA1 is to detect potential danger stimuli that may threaten the tissue homeostasis and the health of the organism. The ability to directly recognize signals of different modalities, including chemical irritants, extreme temperatures, or osmotic changes resides in the characteristic properties of the ion channel protein complex. Recent advances in cryo-electron microscopy have provided an important framework for understanding the molecular basis of TRPA1 function and have suggested novel directions in the search for its pharmacological regulation. This chapter summarizes the current knowledge of human TRPA1 from a structural and functional perspective and discusses the complex allosteric mechanisms of activation and modulation that play important roles under physiological or pathophysiological conditions. In this context, major challenges for future research on TRPA1 are outlined.
Collapse
Affiliation(s)
- Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Ivan Barvik
- Division of Biomolecular Physics, Institute of Physics, Faculty of Mathematics and Physics, Charles University, Prague, Czech Republic.
| | - Lucie Zimova
- Department of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
30
|
Sundaramurthi JC, Bagley AM, Blau H, Carmody L, Crandall A, Danis D, Gargano MA, Gustafson AG, Raney EM, Shingle M, Davids JR, Robinson PN. De novo TRPM3 missense variant associated with neurodevelopmental delay and manifestations of cerebral palsy. Cold Spring Harb Mol Case Stud 2023; 9:a006293. [PMID: 37684057 PMCID: PMC10815282 DOI: 10.1101/mcs.a006293] [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: 05/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
We identified a de novo heterozygous transient receptor potential cation channel subfamily M (melastatin) member 3 (TRPM3) missense variant, p.(Asn1126Asp), in a patient with developmental delay and manifestations of cerebral palsy (CP) using phenotype-driven prioritization analysis of whole-genome sequencing data with Exomiser. The variant is localized in the functionally important ion transport domain of the TRPM3 protein and predicted to impact the protein structure. Our report adds TRPM3 to the list of Mendelian disease-associated genes that can be associated with CP and provides further evidence for the pathogenicity of the variant p.(Asn1126Asp).
Collapse
Affiliation(s)
| | - Anita M Bagley
- Shriners Children's Northern California, Sacramento, California 95817, USA
| | - Hannah Blau
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Leigh Carmody
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Amy Crandall
- Shriners Children's, Portland, Oregon 97239, USA
| | - Daniel Danis
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Michael A Gargano
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | | | | | - Mallory Shingle
- Shriners Children's Northern California, Sacramento, California 95817, USA
| | - Jon R Davids
- Shriners Children's Northern California, Sacramento, California 95817, USA
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, California 95817, USA
| | - Peter N Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA;
- Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut 06032, USA
| |
Collapse
|
31
|
Wu W, Zheng J, Wang R, Wang Y. Ion channels regulate energy homeostasis and the progression of metabolic disorders: Novel mechanisms and pharmacology of their modulators. Biochem Pharmacol 2023; 218:115863. [PMID: 37863328 DOI: 10.1016/j.bcp.2023.115863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
The progression of metabolic diseases, featured by dysregulated metabolic signaling pathways, is orchestrated by numerous signaling networks. Among the regulators, ion channels transport ions across the membranes and trigger downstream signaling transduction. They critically regulate energy homeostasis and pathogenesis of metabolic diseases and are potential therapeutic targets for treating metabolic disorders. Ion channel blockers have been used to treat diabetes for decades by stimulating insulin secretion, yet with hypoglycemia and other adverse effects. It calls for deeper understanding of the largely elusive regulatory mechanisms, which facilitates the identification of new therapeutic targets and safe drugs against ion channels. In the article, we critically assess the two principal regulatory mechanisms, protein-channel interaction and post-translational modification on the activities of ion channels to modulate energy homeostasis and metabolic disorders through multiple novel mechanisms. Moreover, we discuss the multidisciplinary methods that provide the tools for elucidation of the regulatory mechanisms mediating metabolic disorders by ion channels. In terms of translational perspective, the mechanistic analysis of recently validated ion channels that regulate insulin resistance, body weight control, and adverse effects of current ion channel antagonists are discussed in details. Their small molecule modulators serve as promising new drug candidates to combat metabolic disorders.
Collapse
Affiliation(s)
- Wenyi Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Jianan Zheng
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, China
| | - Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, China.
| |
Collapse
|
32
|
Falzone ME, MacKinnon R. The mechanism of Gα q regulation of PLCβ3 -catalyzed PIP2 hydrolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555394. [PMID: 37693483 PMCID: PMC10491199 DOI: 10.1101/2023.08.29.555394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
PLCβ enzymes cleave PIP2 producing IP3 and DAG. PIP2 modulates the function of many ion channels, while IP3 and DAG regulate intracellular Ca 2+ levels and protein phosphorylation by protein kinase C, respectively. PLCβ enzymes are under the control of GPCR signaling through direct interactions with G proteins Gβγ and Gα q and have been shown to be coincidence detectors for dual stimulation of Gα q and G α i coupled receptors. PLCβs are aqueous-soluble cytoplasmic enzymes, but partition onto the membrane surface to access their lipid substrate, complicating their functional and structural characterization. Using newly developed methods, we recently showed that Gβγ activates PLCβ3 by recruiting it to the membrane. Using these same methods, here we show that Gα q increases the catalytic rate constant, k cat , of PLCβ3 . Since stimulation of PLCβ3 by Gα q depends on an autoinhibitory element (the X-Y linker), we propose that Gα q produces partial relief of the X-Y linker autoinhibition through an allosteric mechanism. We also determined membrane-bound structures of the PLCβ3-Gα q , and PLCβ3-Gβγ(2)-Gα q complexes, which show that these G proteins can bind simultaneously and independently of each other to regulate PLCβ3 activity. The structures rationalize a finding in the enzyme assay, that co-stimulation by both G proteins follows a product rule of each independent stimulus. We conclude that baseline activity of PLCβ3 is strongly suppressed, but the effect of G proteins, especially acting together, provides a robust stimulus upon G protein stimulation. Significance Statement For certain cellular signaling processes, the background activity of signaling enzymes must be minimal and stimulus-dependent activation robust. Nowhere is this truer than in signaling by PLCβ3 , whose activity regulates intracellular Ca 2+ , phosphorylation by Protein Kinase C, and the activity of numerous ion channels and membrane receptors. In this study we show how PLCβ3 enzymes are regulated by two kinds of G proteins, Gβγ and Gα q . Enzyme activity studies and structures on membranes show how these G proteins act by separate, independent mechanisms, leading to a product rule of co-stimulation when they act together. The findings explain how cells achieve robust stimulation of PLCβ3 in the setting of very low background activity, properties essential to cell health and survival.
Collapse
|
33
|
Szollosi A, Almássy J. Functional characterization of the transient receptor potential melastatin 2 (TRPM2) cation channel from Nematostella vectensis reconstituted into lipid bilayer. Sci Rep 2023; 13:11471. [PMID: 37454209 PMCID: PMC10349829 DOI: 10.1038/s41598-023-38640-6] [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: 05/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2) cation channel activity is required for insulin secretion, immune cell activation and body heat control. Channel activation upon oxidative stress is involved in the pathology of stroke and neurodegenerative disorders. Cytosolic Ca2+, ADP-ribose (ADPR) and phosphatidylinositol-4,5-bisphosphate (PIP2) are the obligate activators of the channel. Several TRPM2 cryo-EM structures have been resolved to date, yet functionality of the purified protein has not been tested. Here we reconstituted overexpressed and purified TRPM2 from Nematostella vectensis (nvTRPM2) into lipid bilayers and found that the protein is fully functional. Consistent with the observations in native membranes, nvTRPM2 in lipid bilayers is co-activated by cytosolic Ca2+ and either ADPR or ADPR-2'-phosphate (ADPRP). The physiological metabolite ADPRP has a higher apparent affinity than ADPR. In lipid bilayers nvTRPM2 displays a large linear unitary conductance, its open probability (Po) shows little voltage dependence and is stable over several minutes. Po is high without addition of exogenous PIP2, but is largely blunted by treatment with poly-L-Lysine, a polycation that masks PIP2 headgroups. These results indicate that PIP2 or some other activating phosphoinositol lipid co-purifies with nvTRPM2, suggesting a high PIP2 binding affinity of nvTRPM2 under physiological conditions.
Collapse
Affiliation(s)
- Andras Szollosi
- Department of Biochemistry, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary.
- ELKH-SE Ion Channel Research Group, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary.
- HCEMM-SE Molecular Channelopathies Research Group, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary.
| | - János Almássy
- Department of Physiology, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary
| |
Collapse
|
34
|
Zhang M, Ma Y, Ye X, Zhang N, Pan L, Wang B. TRP (transient receptor potential) ion channel family: structures, biological functions and therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:261. [PMID: 37402746 DOI: 10.1038/s41392-023-01464-x] [Citation(s) in RCA: 167] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/26/2023] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Transient receptor potential (TRP) channels are sensors for a variety of cellular and environmental signals. Mammals express a total of 28 different TRP channel proteins, which can be divided into seven subfamilies based on amino acid sequence homology: TRPA (Ankyrin), TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipin), TRPN (NO-mechano-potential, NOMP), TRPP (Polycystin), TRPV (Vanilloid). They are a class of ion channels found in numerous tissues and cell types and are permeable to a wide range of cations such as Ca2+, Mg2+, Na+, K+, and others. TRP channels are responsible for various sensory responses including heat, cold, pain, stress, vision and taste and can be activated by a number of stimuli. Their predominantly location on the cell surface, their interaction with numerous physiological signaling pathways, and the unique crystal structure of TRP channels make TRPs attractive drug targets and implicate them in the treatment of a wide range of diseases. Here, we review the history of TRP channel discovery, summarize the structures and functions of the TRP ion channel family, and highlight the current understanding of the role of TRP channels in the pathogenesis of human disease. Most importantly, we describe TRP channel-related drug discovery, therapeutic interventions for diseases and the limitations of targeting TRP channels in potential clinical applications.
Collapse
Affiliation(s)
- Miao Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yueming Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lei Pan
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
35
|
Nadezhdin KD, Correia L, Narangoda C, Patel DS, Neuberger A, Gudermann T, Kurnikova MG, Chubanov V, Sobolevsky AI. Structural mechanisms of TRPM7 activation and inhibition. Nat Commun 2023; 14:2639. [PMID: 37156763 PMCID: PMC10167348 DOI: 10.1038/s41467-023-38362-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/27/2023] [Indexed: 05/10/2023] Open
Abstract
The transient receptor potential channel TRPM7 is a master regulator of the organismal balance of divalent cations that plays an essential role in embryonic development, immune responses, cell mobility, proliferation, and differentiation. TRPM7 is implicated in neuronal and cardiovascular disorders, tumor progression and has emerged as a new drug target. Here we use cryo-EM, functional analysis, and molecular dynamics simulations to uncover two distinct structural mechanisms of TRPM7 activation by a gain-of-function mutation and by the agonist naltriben, which show different conformational dynamics and domain involvement. We identify a binding site for highly potent and selective inhibitors and show that they act by stabilizing the TRPM7 closed state. The discovered structural mechanisms provide foundations for understanding the molecular basis of TRPM7 channelopathies and drug development.
Collapse
Affiliation(s)
- Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Leonor Correia
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Chamali Narangoda
- Chemistry Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Dhilon S Patel
- Chemistry Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center, German Center for Lung Research (DZL), Munich, Germany
| | - Maria G Kurnikova
- Chemistry Department, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
36
|
Kahler JP, Aloi VD, Miedes Aliaga J, Kerselaers S, Voets T, Vriens J, Verhelst SHL, Barniol-Xicota M. Clotrimazole-Based Modulators of the TRPM3 Ion Channel Reveal Narrow Structure-Activity Relationship. ACS Chem Biol 2023; 18:456-464. [PMID: 36762958 DOI: 10.1021/acschembio.2c00672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
TRPM3 is an ion channel that is highly expressed in nociceptive neurons and plays a key role in pain perception. In the presence of the endogenous TRPM3 ligand, pregnenolone sulfate (PS), the antifungal compound clotrimazole (Clt) augments Ca2+ signaling and opens a non-canonical pore, permeable to Na+, which aggravates TRPM3-induced pain. To date, little is known about structural features that govern the Clt modulatory effect of TRPM3. Here, we synthesized and evaluated several Clt analogues in order to gain insights into their structure-activity relationship. Our results reveal a tight SAR with the three phenyl rings on the trityl moiety being essential for the activity, as well as the presence of fluorine or chlorine substituents on the trityl group. Imidazole as a heterocycle is also necessary for activity. Interestingly, we identified a pentafluoro-trityl analogue (29a) that is able to act as a TRPM3 agonist in the absence of PS. The compounds we report in this work will be useful tools for the further study of TRPM3 modulation and its effect on pain perception.
Collapse
Affiliation(s)
- Jan Pascal Kahler
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven, Herestraat 49, Box 901b, 3000 Leuven, Belgium
| | - Vincenzo Davide Aloi
- Laboratory of Ion Channel Research, VIB Center for Brain and Disease Research, Herestraat 49, Box 802, 3000 Leuven, Belgium.,Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, Herestraat 49, Box 802, 3000 Leuven, Belgium.,Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Julia Miedes Aliaga
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven, Herestraat 49, Box 901b, 3000 Leuven, Belgium
| | - Sara Kerselaers
- Laboratory of Ion Channel Research, VIB Center for Brain and Disease Research, Herestraat 49, Box 802, 3000 Leuven, Belgium.,Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, Herestraat 49, Box 802, 3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB Center for Brain and Disease Research, Herestraat 49, Box 802, 3000 Leuven, Belgium.,Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, Herestraat 49, Box 802, 3000 Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Steven H L Verhelst
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven, Herestraat 49, Box 901b, 3000 Leuven, Belgium.,Leibniz Institute for Analytical Sciences, ISAS, e.V., Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| | - Marta Barniol-Xicota
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven, Herestraat 49, Box 901b, 3000 Leuven, Belgium
| |
Collapse
|
37
|
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
|