1
|
Antinociceptive Activity of Vanilloids in Caenorhabditis elegans is Mediated by the Desensitization of the TRPV Channel OCR-2 and Specific Signal Transduction Pathways. Neurochem Res 2023; 48:1900-1911. [PMID: 36737562 DOI: 10.1007/s11064-023-03876-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Vanilloids, including capsaicin and eugenol, are ligands of transient receptor potential channel vanilloid subfamily member 1 (TRPV1). Prolonged treatment with vanilloids triggered the desensitization of TRPV1, leading to analgesic or antinociceptive effects. Caenorhabditis elegans (C. elegans) is a model organism expressing vanilloid receptor orthologs (e.g., OSM-9 and OCR-2) that are associated with behavioral and physiological processes, including sensory transduction. We have shown that capsaicin and eugenol hamper the nocifensive response to noxious heat in C. elegans. The objective of this study was to perform proteomics to identify proteins and pathways responsible for the induced phenotype and to identify capsaicin and eugenol targets using a thermal proteome profiling (TPP) strategy. The results indicated hierarchical differences following Reactome Pathway enrichment analyses between capsaicin- and eugenol-treated nematodes. However, both treated groups were associated mainly with signal transduction pathways, energy generation, biosynthesis and structural processes. Wnt signaling, a specific signal transduction pathway, is involved following treatment with both molecules. Wnt signaling pathway is noticeably associated with pain. The TPP results show that capsaicin and eugenol target OCR-2 but not OSM-9. Further protein-protein interaction (PPI) analyses showed other targets associated with enzymatic catalysis and calcium ion binding activity. The resulting data help to better understand the broad-spectrum pharmacological activity of vanilloids.
Collapse
|
2
|
Caballero J. A new era for the design of TRPV1 antagonists and agonists with the use of structural information and molecular docking of capsaicin-like compounds. J Enzyme Inhib Med Chem 2022; 37:2169-2178. [PMID: 35975286 PMCID: PMC9387342 DOI: 10.1080/14756366.2022.2110089] [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] [Indexed: 11/20/2022] Open
Abstract
The design of TRPV1 antagonists and agonists has reached a new era since TRPV1 structures at near-atomic resolution are available. Today, the ligand-binding forms of several classical antagonists and agonists are known; therefore, the specific role of key TRPV1’s residues in binding of ligands can be elucidated. It is possible to place the well-defined pharmacophore of TRPV1 ligands, conformed by head, neck, and tail groups, in the right pocket regions of TRPV1. It will allow a more thorough use of molecular modelling methods to conduct more effective rational drug design protocols. In this work, important points about the interactions between TRPV1 and capsaicin-like compounds are spelled out, based on the known pharmacophore of the ligands and the already available TRPV1 structures. These points must be addressed to generate reliable poses of novel candidates and should be considered during the design of novel TRPV1 antagonists and agonists.
Collapse
Affiliation(s)
- Julio Caballero
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Talca, Chile
| |
Collapse
|
3
|
Trkulja CL, Jungholm O, Davidson M, Jardemark K, Marcus MM, Hägglund J, Karlsson A, Karlsson R, Bruton J, Ivarsson N, Srinivasa SP, Cavallin A, Svensson P, Jeffries GDM, Christakopoulou MN, Reymer A, Ashok A, Willman G, Papadia D, Johnsson E, Orwar O. Rational antibody design for undruggable targets using kinetically controlled biomolecular probes. SCIENCE ADVANCES 2021; 7:7/16/eabe6397. [PMID: 33863724 PMCID: PMC8051879 DOI: 10.1126/sciadv.abe6397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/26/2021] [Indexed: 05/07/2023]
Abstract
Several important drug targets, e.g., ion channels and G protein-coupled receptors, are extremely difficult to approach with current antibody technologies. To address these targets classes, we explored kinetically controlled proteases as structural dynamics-sensitive druggability probes in native-state and disease-relevant proteins. By using low-Reynolds number flows, such that a single or a few protease incisions are made, we could identify antibody binding sites (epitopes) that were translated into short-sequence antigens for antibody production. We obtained molecular-level information of the epitope-paratope region and could produce high-affinity antibodies with programmed pharmacological function against difficult-to-drug targets. We demonstrate the first stimulus-selective monoclonal antibodies targeting the transient receptor potential vanilloid 1 (TRPV1) channel, a clinically validated pain target widely considered undruggable with antibodies, and apoptosis-inducing antibodies selectively mediating cytotoxicity in KRAS-mutated cells. It is our hope that this platform will widen the scope of antibody therapeutics for the benefit of patients.
Collapse
Affiliation(s)
| | - Oscar Jungholm
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Max Davidson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
| | - Kent Jardemark
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Monica M Marcus
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Jessica Hägglund
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anders Karlsson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Nanoxis Consulting AB, SE-40016 Gothenburg, Sweden
| | - Roger Karlsson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Nanoxis Consulting AB, SE-40016 Gothenburg, Sweden
| | - Joseph Bruton
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Niklas Ivarsson
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | - Alexandra Cavallin
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Peder Svensson
- Integrative Research Laboratories, SE-41346, Gothenburg, Sweden
| | | | | | - Anna Reymer
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden
| | | | | | | | - Emma Johnsson
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden
| | - Owe Orwar
- Oblique Therapeutics AB, SE-41346 Gothenburg, Sweden.
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
4
|
The structural changes of the mutated ankyrin repeat domain of the human TRPV4 channel alter its ATP binding ability. J Mech Behav Biomed Mater 2019; 101:103407. [PMID: 31493693 DOI: 10.1016/j.jmbbm.2019.103407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/23/2019] [Accepted: 08/24/2019] [Indexed: 11/20/2022]
Abstract
The transient receptor potential (TRP) channel TRPV4 is a calcium-permeable cation channel protein which plays a mechanosensory and osmosensory role in several musculoskeletal tissues. Previous studies have shown that some specific mutations in the ankyrin repeat domain (ARD) of TRPV4 can reduce channel activity and further cause the osteoarthropathy related disease. Mutations in this region probably influence the constitutive activity of the channel, which mainly regulated by the binding of a small ligand such as adenosine triphosphate (ATP). These findings suggest that it is crucial to understand the fundamental mechanisms regulated by chemical ligands such as ATP binding with the ankyrin repeat domain (ARD) of TRPV4. However, how these mutations at the molecular level resulting in the related diseases are still unclear. Here we use full atomistic simulations to investigate the mutation induced conformational changes and ATP binding ability differences of TRPV4-ARD. Conformation characteristics of different mutations of TRPV4-ARD are explored. Optimal communication paths are studied to explain how a point mutation away from aim region (Finger 3) can cause a significant alteration on the conformation. We identify two molecular mechanisms through the conformation of Finger 3 and through alter the ATP binding mechanism correspondently to explain these unknowns. Our study provides fundamental insights into the mutation induced structural changes of the TRPV4-ARD and helps to explain how the mutations alter the ATP binding ability of the TRPV4-ARD.
Collapse
|
5
|
Thongnum K, Chanthai S. Inhibitory Reactivity of Capsaicin with α-Amylase and α-Glucosidase Related to Antidiabetes using Molecular Docking and Quantum Calculation Methods. ACTA ACUST UNITED AC 2018. [DOI: 10.13005/ojc/340501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This work aims to investigate the inhibitory activity of capsaicin, which is one of capsaicinoid compounds, on these enzymes using a molecular docking and quantum calculation. Acarbose, a commercial diabetes drug, was also investigated for comparison. The docking results revealed that acarbose yields better inhibition efficiency with binding free energy (ΔGbinding) of about -8.2 to -11.9 kcal/mol, and inhibition constant (Ki) of about 0.0002 to 0.4 µM, whereas capsaicin provided the ΔGbinding of -5.8 to -6.1 kcal/mol and Ki of 23.7 to 45.9 µM. The total binding energy (ΔEbinding) between each inhibitor and amino acids in active site of enzyme obtained from quantum calculation with MP2/6-31G(d,p) level is in agreement with the ΔGbinding, i.e. the ΔEbinding of acarbose was larger negative than that of capsaicin. The amino acids interacting with inhibitor as hydrogen bond mainly contribute to the total binding energy. Nevertheless, it could be concluded that capsaicinoids have high potential to be developed as an alternative drug for diabetes disease.
Collapse
Affiliation(s)
- Kultida Thongnum
- Materials Chemistry Research Center, Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Saksit Chanthai
- Materials Chemistry Research Center, Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
6
|
Geron M, Hazan A, Priel A. Animal Toxins Providing Insights into TRPV1 Activation Mechanism. Toxins (Basel) 2017; 9:toxins9100326. [PMID: 29035314 PMCID: PMC5666373 DOI: 10.3390/toxins9100326] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Beyond providing evolutionary advantages, venoms offer unique research tools, as they were developed to target functionally important proteins and pathways. As a key pain receptor in the nociceptive pathway, transient receptor potential vanilloid 1 (TRPV1) of the TRP superfamily has been shown to be a target for several toxins, as a way of producing pain to deter predators. Importantly, TRPV1 is involved in thermoregulation, inflammation, and acute nociception. As such, toxins provide tools to understand TRPV1 activation and modulation, a critical step in advancing pain research and the development of novel analgesics. Indeed, the phytotoxin capsaicin, which is the spicy chemical in chili peppers, was invaluable in the original cloning and characterization of TRPV1. The unique properties of each subsequently characterized toxin have continued to advance our understanding of functional, structural, and biophysical characteristics of TRPV1. By building on previous reviews, this work aims to provide a comprehensive summary of the advancements made in TRPV1 research in recent years by employing animal toxins, in particular DkTx, RhTx, BmP01, Echis coloratus toxins, APHCs and HCRG21. We examine each toxin’s functional aspects, behavioral effects, and structural features, all of which have contributed to our current knowledge of TRPV1. We additionally discuss the key features of TRPV1’s outer pore domain, which proves to be the target of the currently discussed toxins.
Collapse
Affiliation(s)
- Matan Geron
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Adina Hazan
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Avi Priel
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
7
|
White JPM, Cibelli M, Urban L, Nilius B, McGeown JG, Nagy I. TRPV4: Molecular Conductor of a Diverse Orchestra. Physiol Rev 2017; 96:911-73. [PMID: 27252279 DOI: 10.1152/physrev.00016.2015] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential vanilloid type 4 (TRPV4) is a calcium-permeable nonselective cation channel, originally described in 2000 by research teams led by Schultz (Nat Cell Biol 2: 695-702, 2000) and Liedtke (Cell 103: 525-535, 2000). TRPV4 is now recognized as being a polymodal ionotropic receptor that is activated by a disparate array of stimuli, ranging from hypotonicity to heat and acidic pH. Importantly, this ion channel is constitutively expressed and capable of spontaneous activity in the absence of agonist stimulation, which suggests that it serves important physiological functions, as does its widespread dissemination throughout the body and its capacity to interact with other proteins. Not surprisingly, therefore, it has emerged more recently that TRPV4 fulfills a great number of important physiological roles and that various disease states are attributable to the absence, or abnormal functioning, of this ion channel. Here, we review the known characteristics of this ion channel's structure, localization and function, including its activators, and examine its functional importance in health and disease.
Collapse
Affiliation(s)
- John P M White
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Mario Cibelli
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Laszlo Urban
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Bernd Nilius
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - J Graham McGeown
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Istvan Nagy
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
8
|
Goldmann D, Zdrazil B, Digles D, Ecker GF. Empowering pharmacoinformatics by linked life science data. J Comput Aided Mol Des 2017; 31:319-328. [PMID: 27830428 PMCID: PMC5385323 DOI: 10.1007/s10822-016-9990-4] [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: 07/29/2016] [Accepted: 10/24/2016] [Indexed: 11/11/2022]
Abstract
With the public availability of large data sources such as ChEMBLdb and the Open PHACTS Discovery Platform, retrieval of data sets for certain protein targets of interest with consistent assay conditions is no longer a time consuming process. Especially the use of workflow engines such as KNIME or Pipeline Pilot allows complex queries and enables to simultaneously search for several targets. Data can then directly be used as input to various ligand- and structure-based studies. In this contribution, using in-house projects on P-gp inhibition, transporter selectivity, and TRPV1 modulation we outline how the incorporation of linked life science data in the daily execution of projects allowed to expand our approaches from conventional Hansch analysis to complex, integrated multilayer models.
Collapse
Affiliation(s)
- Daria Goldmann
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Barbara Zdrazil
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Daniela Digles
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, 1090, Vienna, Austria.
| |
Collapse
|
9
|
Understand spiciness: mechanism of TRPV1 channel activation by capsaicin. Protein Cell 2017; 8:169-177. [PMID: 28044278 PMCID: PMC5326624 DOI: 10.1007/s13238-016-0353-7] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/22/2016] [Indexed: 11/06/2022] Open
Abstract
Capsaicin in chili peppers bestows the sensation of spiciness. Since the discovery of its receptor, transient receptor potential vanilloid 1 (TRPV1) ion channel, how capsaicin activates this channel has been under extensive investigation using a variety of experimental techniques including mutagenesis, patch-clamp recording, crystallography, cryo-electron microscopy, computational docking and molecular dynamic simulation. A framework of how capsaicin binds and activates TRPV1 has started to merge: capsaicin binds to a pocket formed by the channel’s transmembrane segments, where it takes a “tail-up, head-down” configuration. Binding is mediated by both hydrogen bonds and van der Waals interactions. Upon binding, capsaicin stabilizes the open state of TRPV1 by “pull-and-contact” with the S4-S5 linker. Understanding the ligand-host interaction will greatly facilitate pharmaceutical efforts to develop novel analgesics targeting TRPV1.
Collapse
|
10
|
Peters M, Katz B, Lev S, Zaguri R, Gutorov R, Minke B. Depletion of Membrane Cholesterol Suppresses Drosophila Transient Receptor Potential-Like (TRPL) Channel Activity. CURRENT TOPICS IN MEMBRANES 2017; 80:233-254. [DOI: 10.1016/bs.ctm.2017.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
11
|
Monastyrnaya M, Peigneur S, Zelepuga E, Sintsova O, Gladkikh I, Leychenko E, Isaeva M, Tytgat J, Kozlovskaya E. Kunitz-Type Peptide HCRG21 from the Sea Anemone Heteractis crispa Is a Full Antagonist of the TRPV1 Receptor. Mar Drugs 2016; 14:E229. [PMID: 27983679 PMCID: PMC5192466 DOI: 10.3390/md14120229] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 12/18/2022] Open
Abstract
Sea anemone venoms comprise multifarious peptides modulating biological targets such as ion channels or receptors. The sequence of a new Kunitz-type peptide, HCRG21, belonging to the Heteractis crispa RG (HCRG) peptide subfamily was deduced on the basis of the gene sequence obtained from the Heteractis crispa cDNA. HCRG21 shares high structural homology with Kunitz-type peptides APHC1-APHC3 from H. crispa, and clusters with the peptides from so named "analgesic cluster" of the HCGS peptide subfamily but forms a separate branch on the NJ-phylogenetic tree. Three unique point substitutions at the N-terminus of the molecule, Arg1, Gly2, and Ser5, distinguish HCRG21 from other peptides of this cluster. The trypsin inhibitory activity of recombinant HCRG21 (rHCRG21) was comparable with the activity of peptides from the same cluster. Inhibition constants for trypsin and α-chymotrypsin were 1.0 × 10-7 and 7.0 × 10-7 M, respectively. Electrophysiological experiments revealed that rHCRG21 inhibits 95% of the capsaicin-induced current through transient receptor potential family member vanilloid 1 (TRPV1) and has a half-maximal inhibitory concentration of 6.9 ± 0.4 μM. Moreover, rHCRG21 is the first full peptide TRPV1 inhibitor, although displaying lower affinity for its receptor in comparison with other known ligands. Macromolecular docking and full atom Molecular Dynamics (MD) simulations of the rHCRG21-TRPV1 complex allow hypothesizing the existence of two feasible, intra- and extracellular, molecular mechanisms of blocking. These data provide valuable insights in the structural and functional relationships and pharmacological potential of bifunctional Kunitz-type peptides.
Collapse
Affiliation(s)
- Margarita Monastyrnaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok 690022, Russia.
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, Herestraat 49, P.O. Box 922, Leuven B-3000, Belgium.
| | - Elena Zelepuga
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok 690022, Russia.
| | - Oksana Sintsova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok 690022, Russia.
| | - Irina Gladkikh
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok 690022, Russia.
| | - Elena Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok 690022, Russia.
| | - Marina Isaeva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok 690022, Russia.
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, Herestraat 49, P.O. Box 922, Leuven B-3000, Belgium.
| | - Emma Kozlovskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok 690022, Russia.
| |
Collapse
|
12
|
Gregorio-Teruel L, Valente P, Liu B, Fernández-Ballester G, Qin F, Ferrer-Montiel A. The Integrity of the TRP Domain Is Pivotal for Correct TRPV1 Channel Gating. Biophys J 2016; 109:529-41. [PMID: 26244735 DOI: 10.1016/j.bpj.2015.06.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 01/19/2023] Open
Abstract
Transient receptor potential vanilloid subtype I (TRPV1) is a thermosensory ion channel that is also gated by chemical substances such as vanilloids. Adjacent to the channel gate, this polymodal thermoTRP channel displays a TRP domain, referred to as AD1, that plays a role in subunit association and channel gating. Previous studies have shown that swapping the AD1 in TRPV1 with the cognate from the TRPV2 channel (AD2) reduces protein expression and produces a nonfunctional chimeric channel (TRPV1-AD2). Here, we used a stepwise, sequential, cumulative site-directed mutagenesis approach, based on rebuilding the AD1 domain in the TRPV1-AD2 chimera, to unveil the minimum number of amino acids needed to restore protein expression and polymodal channel activity. Unexpectedly, we found that virtually full restitution of the AD1 sequence is required to reinstate channel expression and responses to capsaicin, temperature, and voltage. This strategy identified E692, R701, and T704 in the TRP domain as important for TRPV1 activity. Even conservative mutagenesis at these sites (E692D/R701K/T704S) impaired channel expression and abolished TRPV1 activity. However, the sole mutation of these positions in the TRPV1-AD2 chimera (D692E/K701R/S704T) was not sufficient to rescue channel gating, implying that other residues in the TRP domain are necessary to endow activity to TRPV1-AD2. A biophysical analysis of a functional chimera suggested that mutations in the TRP domain raised the energetics of channel gating by altering the coupling of stimuli sensing and pore opening. These findings indicate that inter- and/or intrasubunit interactions in the TRP domain are essential for correct TRPV1 gating.
Collapse
Affiliation(s)
- Lucia Gregorio-Teruel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante, Spain
| | - Pierluigi Valente
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genova, Italy
| | - Beiying Liu
- Department of Physiology and Biophysical Sciences, State University of New York, Buffalo, New York
| | | | - Feng Qin
- Department of Physiology and Biophysical Sciences, State University of New York, Buffalo, New York
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante, Spain.
| |
Collapse
|
13
|
Hyaluronan modulates TRPV1 channel opening, reducing peripheral nociceptor activity and pain. Nat Commun 2015; 6:8095. [PMID: 26311398 PMCID: PMC4560824 DOI: 10.1038/ncomms9095] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/17/2015] [Indexed: 01/31/2023] Open
Abstract
Hyaluronan (HA) is present in the extracellular matrix of all body tissues, including synovial fluid in joints, in which it behaves as a filter that buffers transmission of mechanical forces to nociceptor nerve endings thereby reducing pain. Using recombinant systems, mouse-cultured dorsal root ganglia (DRG) neurons and in vivo experiments, we found that HA also modulates polymodal transient receptor potential vanilloid subtype 1 (TRPV1) channels. HA diminishes heat, pH and capsaicin (CAP) responses, thus reducing the opening probability of the channel by stabilizing its closed state. Accordingly, in DRG neurons, HA decreases TRPV1-mediated impulse firing and channel sensitization by bradykinin. Moreover, subcutaneous HA injection in mice reduces heat and capsaicin nocifensive responses, whereas the intra-articular injection of HA in rats decreases capsaicin joint nociceptor fibres discharge. Collectively, these results indicate that extracellular HA reduces the excitability of the ubiquitous TRPV1 channel, thereby lowering impulse activity in the peripheral nociceptor endings underlying pain. Hyaluronan is a major component of the extracellular matrix, and is used to treat joint pain in osteoarthritis. In this study, Caires et al. show hyaluronan achieves its analgesic effects by targeting TRPV1 and stabilising the channel in its closed state.
Collapse
|
14
|
Hilton JK, Rath P, Helsell CVM, Beckstein O, Van Horn WD. Understanding Thermosensitive Transient Receptor Potential Channels as Versatile Polymodal Cellular Sensors. Biochemistry 2015; 54:2401-13. [DOI: 10.1021/acs.biochem.5b00071] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Jacob K. Hilton
- Center
for Personalized Diagnostics, Magnetic Resonance Research Center,
and Department of Chemistry and Biochemistry, Arizona State University, 551 East University Drive, PSG-106, Tempe, Arizona 85287, United States
| | - Parthasarathi Rath
- Center
for Personalized Diagnostics, Magnetic Resonance Research Center,
and Department of Chemistry and Biochemistry, Arizona State University, 551 East University Drive, PSG-106, Tempe, Arizona 85287, United States
| | - Cole V. M. Helsell
- Center
for Personalized Diagnostics, Magnetic Resonance Research Center,
and Department of Chemistry and Biochemistry, Arizona State University, 551 East University Drive, PSG-106, Tempe, Arizona 85287, United States
| | - Oliver Beckstein
- Center
for Biological Physics and Department of Physics, Arizona State University, 550 East Tyler Mall, Tempe, Arizona 85287, United States
| | - Wade D. Van Horn
- Center
for Personalized Diagnostics, Magnetic Resonance Research Center,
and Department of Chemistry and Biochemistry, Arizona State University, 551 East University Drive, PSG-106, Tempe, Arizona 85287, United States
| |
Collapse
|
15
|
Wang J, Li Y, Yang Y, Du J, Zhang S, Yang L. In silico research to assist the investigation of carboxamide derivatives as potent TRPV1 antagonists. MOLECULAR BIOSYSTEMS 2015; 11:2885-99. [DOI: 10.1039/c5mb00356c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The transient receptor potential vanilloid type 1 (TRPV1), a non-selective cation channel, is known for its essential role in the pathogenesis of various pain conditions such as nerve damage induced hyperalgesia, diabetic neuropathy and cancer pain.
Collapse
Affiliation(s)
- Jinghui Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE)
- Faculty of Chemical
- Environmental and Biological Science and Technology
- Dalian University of Technology
- Dalian
| | - Yan Li
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE)
- Faculty of Chemical
- Environmental and Biological Science and Technology
- Dalian University of Technology
- Dalian
| | - Yinfeng Yang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE)
- Faculty of Chemical
- Environmental and Biological Science and Technology
- Dalian University of Technology
- Dalian
| | - Jian Du
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE)
- Faculty of Chemical
- Environmental and Biological Science and Technology
- Dalian University of Technology
- Dalian
| | - Shuwei Zhang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE)
- Faculty of Chemical
- Environmental and Biological Science and Technology
- Dalian University of Technology
- Dalian
| | - Ling Yang
- Laboratory of Pharmaceutical Resource Discovery
- Dalian Institute of Chemical Physics
- Graduate School of the Chinese Academy of Sciences
- Dalian
- China
| |
Collapse
|
16
|
Gregorio-Teruel L, Valente P, González-Ros JM, Fernández-Ballester G, Ferrer-Montiel A. Mutation of I696 and W697 in the TRP box of vanilloid receptor subtype I modulates allosteric channel activation. ACTA ACUST UNITED AC 2014; 143:361-75. [PMID: 24567510 PMCID: PMC3933934 DOI: 10.1085/jgp.201311070] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Residues I696 and W697 are crucial to coupling between the TRPV1 ligand- and voltage-sensing domains and the channel pore. The transient receptor potential vanilloid receptor subtype I (TRPV1) channel acts as a polymodal sensory receptor gated by chemical and physical stimuli. Like other TRP channels, TRPV1 contains in its C terminus a short, conserved domain called the TRP box, which is necessary for channel gating. Substitution of two TRP box residues—I696 and W697—with Ala markedly affects TRPV1’s response to all activating stimuli, which indicates that these two residues play a crucial role in channel gating. We systematically replaced I696 and W697 with 18 native l-amino acids (excluding cysteine) and evaluated the effect on voltage- and capsaicin-dependent gating. Mutation of I696 decreased channel activation by either voltage or capsaicin; furthermore, gating was only observed with substitution of hydrophobic amino acids. Substitution of W697 with any of the 18 amino acids abolished gating in response to depolarization alone, shifting the threshold to unreachable voltages, but not capsaicin-mediated gating. Moreover, vanilloid-activated responses of W697X mutants showed voltage-dependent gating along with a strong voltage-independent component. Analysis of the data using an allosteric model of activation indicates that mutation of I696 and W697 primarily affects the allosteric coupling constants of the ligand and voltage sensors to the channel pore. Together, our findings substantiate the notion that inter- and/or intrasubunit interactions at the level of the TRP box are critical for efficient coupling of stimulus sensing and gate opening. Perturbation of these interactions markedly reduces the efficacy and potency of the activating stimuli. Furthermore, our results identify these interactions as potential sites for pharmacological intervention.
Collapse
Affiliation(s)
- Lucia Gregorio-Teruel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, 03202 Elche, Spain
| | | | | | | | | |
Collapse
|
17
|
Trkulja CL, Jansson ET, Jardemark K, Orwar O. Probing structure and function of ion channels using limited proteolysis and microfluidics. J Am Chem Soc 2014; 136:14875-82. [PMID: 25254316 DOI: 10.1021/ja507285w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Even though gain, loss, or modulation of ion channel function is implicated in many diseases, both rare and common, the development of new pharmaceuticals targeting this class has been disappointing, where it has been a major problem to obtain correlated structural and functional information. Here, we present a microfluidic method in which the ion channel TRPV1, contained in proteoliposomes or in excised patches, was exposed to limited trypsin proteolysis. Cleaved-off peptides were identified by MS, and electrophysiological properties were recorded by patch clamp. Thus, the structure-function relationship was evaluated by correlating changes in function with removal of structural elements. Using this approach, we pinpointed regions of TRPV1 that affect channel properties upon their removal, causing changes in current amplitude, single-channel conductance, and EC50 value toward its agonist, capsaicin. We have provided a fast "shotgun" method for chemical truncation of a membrane protein, which allows for functional assessments of various peptide regions.
Collapse
Affiliation(s)
- Carolina L Trkulja
- Department of Chemical and Biological Engineering, Chalmers University of Technology , SE-412 96 Göteborg, Sweden
| | | | | | | |
Collapse
|
18
|
Lindy AS, Parekh PK, Zhu R, Kanju P, Chintapalli SV, Tsvilovskyy V, Patterson RL, Anishkin A, van Rossum DB, Liedtke WB. TRPV channel-mediated calcium transients in nociceptor neurons are dispensable for avoidance behaviour. Nat Commun 2014; 5:4734. [PMID: 25178952 PMCID: PMC4164786 DOI: 10.1038/ncomms5734] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/17/2014] [Indexed: 12/21/2022] Open
Abstract
Animals need to sense and react to potentially dangerous environments. TRP ion channels participate in nociception, presumably via Ca2+ influx, in most animal species. However, the relationship between ion permeation and animals’ nocifensive behaviour is unknown. Here we use an invertebrate animal model with relevance for mammalian pain. We analyse the putative selectivity filter of OSM-9, a TRPV channel, in osmotic avoidance behaviour of Caenorhabditis elegans. Using mutagenized OSM-9 expressed in the head nociceptor neuron, ASH, we study nocifensive behaviour and Ca2+ influx. Within the selectivity filter, M601-F609, Y604G strongly reduces avoidance behaviour and eliminates Ca2+ transients. Y604F also abolishes Ca2+ transients in ASH, while sustaining avoidance behaviour, yet it disrupts behavioral plasticity. Homology modelling of the OSM-9 pore suggests that Y604 may assume a scaffolding role. Thus, aromatic residues in the OSM-9 selectivity filter are critical for pain behaviour and ion permeation. These findings have relevance for understanding evolutionary roots of mammalian nociception. TRPs are calcium-permeable channels involved in the sensing of damaging stimuli but the relationship between calcium influx and pain behaviour has been elusive. Here the authors find that the TRP channel OSM-9 functions as an ion channel in vivo in C. elegans, and establish residues that are critical for worm pain-like behaviour.
Collapse
Affiliation(s)
- Amanda S Lindy
- 1] Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, USA [2] Department of Neurology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Puja K Parekh
- Department of Neurology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Richard Zhu
- Department of Neurology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Patrick Kanju
- Department of Neurology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Sree V Chintapalli
- 1] Department of Membrane Biology and Physiology, University of California, Davis, California 95616, USA [2] Department of Biochemistry and Molecular Medicine, University of California, Davis, California 95616, USA
| | - Volodymyr Tsvilovskyy
- Department of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Randen L Patterson
- 1] Department of Membrane Biology and Physiology, University of California, Davis, California 95616, USA [2] Department of Biochemistry and Molecular Medicine, University of California, Davis, California 95616, USA
| | - Andriy Anishkin
- 1] Center for Computational Proteomics, The Pennsylvania State University, University Park, Pennsylvania 16801, USA [2] Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16801, USA
| | - Damian B van Rossum
- 1] Center for Computational Proteomics, The Pennsylvania State University, University Park, Pennsylvania 16801, USA [2] Department of Biology, The Pennsylvania State University, University Park, Pennsylvania 16801, USA
| | - Wolfgang B Liedtke
- 1] Department of Neurology, Duke University Medical Center, Durham, North Carolina 27710, USA [2] Duke University Clinics for Pain and Palliative Care, 932 Morreene Road, Durham, North Carolina 27705, USA [3] Departments of Anesthesiology and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA [4] Department of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
19
|
Steinberg X, Lespay-Rebolledo C, Brauchi S. A structural view of ligand-dependent activation in thermoTRP channels. Front Physiol 2014; 5:171. [PMID: 24847275 PMCID: PMC4017155 DOI: 10.3389/fphys.2014.00171] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/11/2014] [Indexed: 11/26/2022] Open
Abstract
Transient Receptor Potential (TRP) proteins are a large family of ion channels, grouped into seven sub-families. Although great advances have been made regarding the activation and modulation of TRP channel activity, detailed molecular mechanisms governing TRP channel gating are still needed. Sensitive to electric, chemical, mechanical, and thermal cues, TRP channels are tightly associated with the detection and integration of sensory input, emerging as a model to study the polymodal activation of ion channel proteins. Among TRP channels, the temperature-activated kind constitute a subgroup by itself, formed by Vanilloid receptors 1–4, Melastatin receptors 2, 4, 5, and 8, TRPC5, and TRPA1. Some of the so-called “thermoTRP” channels participate in the detection of noxious stimuli making them an interesting pharmacological target for the treatment of pain. However, the poor specificity of the compounds available in the market represents an important obstacle to overcome. Understanding the molecular mechanics underlying ligand-dependent modulation of TRP channels may help with the rational design of novel synthetic analgesics. The present review focuses on the structural basis of ligand-dependent activation of TRPV1 and TRPM8 channels. Special attention is drawn to the dissection of ligand-binding sites within TRPV1, PIP2-dependent modulation of TRP channels, and the structure of natural and synthetic ligands.
Collapse
Affiliation(s)
- Ximena Steinberg
- Faculty of Medicine, Institute of Physiology, Universidad Austral de Chile Campus Isla Teja, Valdivia, Chile ; Faculty of Sciences, Graduate School, Universidad Austral de Chile Campus Isla Teja, Valdivia, Chile
| | - Carolyne Lespay-Rebolledo
- Faculty of Chemical and Pharmaceutical Sciences, Graduate School, Universidad de Chile Santiago, Chile
| | - Sebastian Brauchi
- Faculty of Medicine, Institute of Physiology, Universidad Austral de Chile Campus Isla Teja, Valdivia, Chile
| |
Collapse
|
20
|
Cohen MR, Moiseenkova-Bell VY. Structure of thermally activated TRP channels. CURRENT TOPICS IN MEMBRANES 2014; 74:181-211. [PMID: 25366237 DOI: 10.1016/b978-0-12-800181-3.00007-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Temperature sensation is important for adaptation and survival of organisms. While temperature has the potential to affect all biological macromolecules, organisms have evolved specific thermosensitive molecular detectors that are able to transduce temperature changes into physiologically relevant signals. Among these thermosensors are ion channels from the transient receptor potential (TRP) family. Prime candidates include TRPV1-4, TRPA1, and TRPM8 (the so-called "thermoTRP" channels), which are expressed in sensory neurons and gated at specific temperatures. Electrophysiological and thermodynamic approaches have been employed to determine the nature by which thermoTRPs detect temperature and couple temperature changes to channel gating. To further understand how thermoTRPs sense temperature, high-resolution structures of full-length thermoTRPs channels will be required. Here, we will discuss current progress in unraveling the structures of thermoTRP channels.
Collapse
Affiliation(s)
- Matthew R Cohen
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Vera Y Moiseenkova-Bell
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
21
|
Gohlke BO, Preissner R, Preissner S. SuperPain--a resource on pain-relieving compounds targeting ion channels. Nucleic Acids Res 2013; 42:D1107-12. [PMID: 24271391 PMCID: PMC3964982 DOI: 10.1093/nar/gkt1176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pain is more than an unpleasant sensory experience associated with actual or potential tissue damage: it is the most common reason for physician consultation and often dramatically affects quality of life. The management of pain is often difficult and new targets are required for more effective and specific treatment. SuperPain (http://bioinformatics.charite.de/superpain/) is freely available database for pain-stimulating and pain-relieving compounds, which bind or potentially bind to ion channels that are involved in the transmission of pain signals to the central nervous system, such as TRPV1, TRPM8, TRPA1, TREK1, TRESK, hERG, ASIC, P2X and voltage-gated sodium channels. The database consists of ∼8700 ligands, which are characterized by experimentally measured binding affinities. Additionally, 100 000 putative ligands are included. Moreover, the database provides 3D structures of receptors and predicted ligand-binding poses. These binding poses and a structural classification scheme provide hints for the design of new analgesic compounds. A user-friendly graphical interface allows similarity searching, visualization of ligands docked into the receptor, etc.
Collapse
Affiliation(s)
- Björn O Gohlke
- German Cancer Consortium (DKTK), Lindenberger Weg 80, 13125 Berlin, Germany, Charité - Universitätsmedizin Berlin, Structural Bioinformatics Group, Lindenberger Weg 80, 13125 Berlin, Germany and Dental, Oral and Maxillary Medicine, Charité - Universitätsmedizin Berlin, CC3, Assmannshauser Straße 4-6, 14197 Berlin, Germany
| | | | | |
Collapse
|
22
|
Klausen TK, Janssens A, Prenen J, Owsianik G, Hoffmann EK, Pedersen SF, Nilius B. Single point mutations of aromatic residues in transmembrane helices 5 and -6 differentially affect TRPV4 activation by 4α-PDD and hypotonicity: implications for the role of the pore region in regulating TRPV4 activity. Cell Calcium 2013; 55:38-47. [PMID: 24342753 DOI: 10.1016/j.ceca.2013.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 11/08/2013] [Indexed: 12/20/2022]
Abstract
The importance of the TRPV4 channel for human physiology has been highlighted in recent years with the identification of an increasing number of hereditary diseases associated with mutations of this channel. However, the functional understanding of TRPV4 associated pathologies remains a puzzle due to incomplete understanding of the polymodal regulation of TRPV4 channels and lack of insight into the structure-function relationship of the channel. In this work, we identified a series of highly conserved aromatic residues in transmembrane (TM) helices 5-6 with profound importance for TRPV4 activity. Substituting F617, Y621 or F624 in TM5 with leucine reduced channel sensitivity to the agonist 4α-PDD and heat, yet two of these mutants - F617L and Y621L - showed increased activation in response to cell swelling. In TM6, a Y702L mutation significantly reduced sensitivity to all of the above stimuli. In conclusion, we have identified residues in TM5-6 which differentially affect heat and agonist activation, and we have demonstrated distinct activation pathways for 4α-PDD and osmolarity.
Collapse
Affiliation(s)
- Thomas Kjær Klausen
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, Bus 802, Leuven, Belgium; Department of Biology, The August Krogh Building, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark.
| | - Annelies Janssens
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, Bus 802, Leuven, Belgium
| | - Jean Prenen
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, Bus 802, Leuven, Belgium
| | - Grzegorz Owsianik
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, Bus 802, Leuven, Belgium
| | - Else Kay Hoffmann
- Department of Biology, The August Krogh Building, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark
| | - Stine Falsig Pedersen
- Department of Biology, The August Krogh Building, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark
| | - Bernd Nilius
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, Bus 802, Leuven, Belgium
| |
Collapse
|
23
|
Abstract
Transient receptor potential (TRP) channels are cellular sensors for a wide spectrum of physical and chemical stimuli. They are involved in the formation of sight, hearing, touch, smell, taste, temperature, and pain sensation. TRP channels also play fundamental roles in cell signaling and allow the host cell to respond to benign or harmful environmental changes. As TRP channel activation is controlled by very diverse processes and, in many cases, exhibits complex polymodal properties, understanding how each TRP channel responds to its unique forms of activation energy is both crucial and challenging. The past two decades witnessed significant advances in understanding the molecular mechanisms that underlie TRP channels activation. This review focuses on our current understanding of the molecular determinants for TRP channel activation.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, USA.
| |
Collapse
|
24
|
De-la-Rosa V, Rangel-Yescas GE, Ladrón-de-Guevara E, Rosenbaum T, Islas LD. Coarse architecture of the transient receptor potential vanilloid 1 (TRPV1) ion channel determined by fluorescence resonance energy transfer. J Biol Chem 2013; 288:29506-17. [PMID: 23965996 DOI: 10.1074/jbc.m113.479618] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The transient receptor potential vanilloid 1 ion channel is responsible for the perception of high temperatures and low extracellular pH, and it is also involved in the response to some pungent compounds. Importantly, it is also associated with the perception of pain and noxious stimuli. Here, we attempt to discern the molecular organization and location of the N and C termini of the transient receptor potential vanilloid 1 ion channel by measuring FRET between genetically attached enhanced yellow and cyan fluorescent protein to the N or C terminus of the channel protein, expressed in transfected HEK 293 cells or Xenopus laevis oocytes. The static measurements of the domain organization were mapped into an available cryo-electron microscopy density of the channel with good agreement. These measurements also provide novel insights into the organization of terminal domains and their proximity to the plasma membrane.
Collapse
|
25
|
Winter Z, Buhala A, Ötvös F, Jósvay K, Vizler C, Dombi G, Szakonyi G, Oláh Z. Functionally important amino acid residues in the transient receptor potential vanilloid 1 (TRPV1) ion channel--an overview of the current mutational data. Mol Pain 2013; 9:30. [PMID: 23800232 PMCID: PMC3707783 DOI: 10.1186/1744-8069-9-30] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/17/2013] [Indexed: 12/30/2022] Open
Abstract
This review aims to create an overview of the currently available results of site-directed mutagenesis studies on transient receptor potential vanilloid type 1 (TRPV1) receptor. Systematization of the vast number of data on the functionally important amino acid mutations of TRPV1 may provide a clearer picture of this field, and may promote a better understanding of the relationship between the structure and function of TRPV1. The review summarizes information on 112 unique mutated sites along the TRPV1, exchanged to multiple different residues in many cases. These mutations influence the effect or binding of different agonists, antagonists, and channel blockers, alter the responsiveness to heat, acid, and voltage dependence, affect the channel pore characteristics, and influence the regulation of the receptor function by phosphorylation, glycosylation, calmodulin, PIP2, ATP, and lipid binding. The main goal of this paper is to publish the above mentioned data in a form that facilitates in silico molecular modelling of the receptor by promoting easier establishment of boundary conditions. The better understanding of the structure-function relationship of TRPV1 may promote discovery of new, promising, more effective and safe drugs for treatment of neurogenic inflammation and pain-related diseases and may offer new opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Zoltán Winter
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Zelepuga EA, Tabakmakher VM, Chausova VE, monastyrnaia MM, Isaeva MP, Kozlovskaia ÉP. [Interaction of sea amemone Heteractis crispa Kunitz type polypeptides with pain vanilloid receptor TRPV1: in silico investigation]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2012; 38:185-98. [PMID: 22792722 DOI: 10.1134/s106816201202015x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Using methods of molecular biology we defined the structures of the 31 sea anemone Heteractis crispa genes encoding polypeptides which are structurally homologous to the Kunitz proteinase inhibitor family. Identified amino acid sequences have point residue substitutions, high degree of homology with sequences of known H. crispa Kunitz family members, and represent a combinatorial library of polypeptides. We generated their three-dimensional structures by homologous modeling methods. Analysis of their molecular electrostatic potential enabled us to divide given polypeptides into three clusters. One of them includes polypeptides APHC1, APHC2 and APHC3, which were earlier shown to possess a unique property of inhibiting of the pain vanilloid receptor TRPV1 in vitro and providing the analgesic effects in vivo in addition to their trypsin inhibitory activity. Molecular docking made possible establishing the spatial structure of the complexes, the nature of the polypeptides binding with TRPV1, as well as functionally important structural elements involved in the complex formation. Structural models have enabled us to propose a hypothesis contributing to understanding the APHC1-3 impact mechanism for the pain signals transduction by TRPV1: apparently, there is an increase of the receptor relaxation time resulted in binding of its two chains with the polypeptide molecule, which disrupt the functioning of the TRPV1 and leads to partial inhibition of signal transduction in electrophysiological experiments.
Collapse
|
27
|
Goswami C. TRPV1-tubulin complex: involvement of membrane tubulin in the regulation of chemotherapy-induced peripheral neuropathy. J Neurochem 2012; 123:1-13. [PMID: 22845740 DOI: 10.1111/j.1471-4159.2012.07892.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/16/2012] [Accepted: 07/19/2012] [Indexed: 12/18/2022]
Abstract
Existence of microtubule cytoskeleton at the membrane and submembranous regions, referred as 'membrane tubulin' has remained controversial for a long time. Since we reported physical and functional interaction of Transient Receptor Potential Vanilloid Sub Type 1 (TRPV1) with microtubules and linked the importance of TRPV1-tubulin complex in the context of chemotherapy-induced peripheral neuropathy, a few more reports have characterized this interaction in in vitro and in in vivo condition. However, the cross-talk between TRPs with microtubule cytoskeleton, and the complex feedback regulations are not well understood. Sequence analysis suggests that other than TRPV1, few TRPs can potentially interact with microtubules. The microtubule interaction with TRPs has evolutionary origin and has a functional significance. Biochemical evidence, Fluorescence Resonance Energy Transfer analysis along with correlation spectroscopy and fluorescence anisotropy measurements have confirmed that TRPV1 interacts with microtubules in live cell and this interaction has regulatory roles. Apart from the transport of TRPs and maintaining the cellular structure, microtubules regulate signaling and functionality of TRPs at the single channel level. Thus, TRPV1-tubulin interaction sets a stage where concept and parameters of 'membrane tubulin' can be tested in more details. In this review, I critically analyze the advancements made in biochemical, pharmacological, behavioral as well as cell-biological observations and summarize the limitations that need to be overcome in the future.
Collapse
Affiliation(s)
- Chandan Goswami
- National Institute of Science Education and Research, Bhubaneswar, Orissa, India.
| |
Collapse
|
28
|
Aguado-Llera D, Bacarizo J, Gregorio-Teruel L, Taberner FJ, Cámara-Artigas A, Neira JL. Biophysical characterization of the isolated C-terminal region of the transient receptor potential vanilloid 1. FEBS Lett 2012; 586:1154-9. [DOI: 10.1016/j.febslet.2012.03.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 03/14/2012] [Accepted: 03/14/2012] [Indexed: 11/28/2022]
|
29
|
Deering-Rice CE, Johansen ME, Roberts JK, Thomas KC, Romero EG, Lee J, Yost GS, Veranth JM, Reilly CA. Transient receptor potential vanilloid-1 (TRPV1) is a mediator of lung toxicity for coal fly ash particulate material. Mol Pharmacol 2012; 81:411-9. [PMID: 22155782 PMCID: PMC3286291 DOI: 10.1124/mol.111.076067] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 12/08/2011] [Indexed: 12/29/2022] Open
Abstract
Environmental particulate matter (PM) pollutants adversely affect human health, but the molecular basis is poorly understood. The ion channel transient receptor potential vanilloid-1 (TRPV1) has been implicated as a sensor for environmental PM and a mediator of adverse events in the respiratory tract. The objectives of this study were to determine whether TRPV1 can distinguish chemically and physically unique PM that represents important sources of air pollution; to elucidate the molecular basis of TRPV1 activation by PM; and to ascertain the contributions of TRPV1 to human lung cell and mouse lung tissue responses exposed to an insoluble PM agonist, coal fly ash (CFA1). The major findings of this study are that TRPV1 is activated by some, but not all of the prototype PM materials evaluated, with rank-ordered responses of CFA1 > diesel exhaust PM > crystalline silica; TRP melastatin-8 is also robustly activated by CFA1, whereas other TRP channels expressed by airway sensory neurons and lung epithelial cells that may also be activated by CFA1, including TRPs ankyrin 1 (A1), canonical 4α (C4α), M2, V2, V3, and V4, were either slightly (TRPA1) or not activated by CFA1; activation of TRPV1 by CFA1 occurs via cell surface interactions between the solid components of CFA1 and specific amino acid residues of TRPV1 that are localized in the putative pore-loop region; and activation of TRPV1 by CFA1 is not exclusive in mouse lungs but represents a pathway by which CFA1 affects the expression of selected genes in lung epithelial cells and airway tissue.
Collapse
|
30
|
Wang L, Cvetkov TL, Chance MR, Moiseenkova-Bell VY. Identification of in vivo disulfide conformation of TRPA1 ion channel. J Biol Chem 2011; 287:6169-76. [PMID: 22207754 DOI: 10.1074/jbc.m111.329748] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
TRPA1 (transient receptor potential ankyrin 1) is an ion channel expressed in the termini of sensory neurons and is activated in response to a broad array of noxious exogenous and endogenous thiol-reactive compounds, making it a crucial player in chemical nociception. A number of conserved cysteine residues on the N-terminal domain of the channel have been identified as critical for sensing these electrophilic pungent chemicals, and our recent EM structure with modeled domains predicts that these cysteines form a ligand-binding pocket, allowing for the possibility of disulfide bonding between the cysteine residues. Here, we present a comprehensive mass spectrometry investigation of the in vivo disulfide bonding conformation and in vitro reactivity of 30 of the 31 cysteine residues in the TRPA1 ion channel. Four disulfide bonds were detected in the in vivo TRPA1 structure: Cys-666-Cys-622, Cys-666-Cys-463, Cys-622-Cys-609, and Cys-666-Cys-193. All of the cysteines detected were reactive to N-methylmaleimide (NMM) in vitro, with varying degrees of labeling efficiency. Comparison of the ratio of the labeling efficiency at 300 μM versus 2 mM NMM identified a number of cysteine residues that were outliers from the mean labeling ratio, suggesting that protein conformation changes rendered these cysteines either more or less protected from labeling at the higher NMM concentrations. These results indicate that the activation mechanism of TRPA1 may involve N-terminal conformation changes and disulfide bonding between critical cysteine residues.
Collapse
Affiliation(s)
- Liwen Wang
- Department of Pharmacology, School of Medicine, Case Western Reserve University,Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
31
|
New strategies to develop novel pain therapies: addressing thermoreceptors from different points of view. Pharmaceuticals (Basel) 2011; 5:16-48. [PMID: 24288041 PMCID: PMC3763626 DOI: 10.3390/ph5010016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 12/16/2011] [Accepted: 12/21/2011] [Indexed: 01/23/2023] Open
Abstract
One approach to develop successful pain therapies is the modulation of dysfunctional ion channels that contribute to the detection of thermal, mechanical and chemical painful stimuli. These ion channels, known as thermoTRPs, promote the sensitization and activation of primary sensory neurons known as nociceptors. Pharmacological blockade and genetic deletion of thermoTRP have validated these channels as therapeutic targets for pain intervention. Several thermoTRP modulators have progressed towards clinical development, although most failed because of the appearance of unpredicted side effects. Thus, there is yet a need to develop novel channel modulators with improved therapeutic index. Here, we review the current state-of-the art and illustrate new pharmacological paradigms based on TRPV1 that include: (i) the identification of activity-dependent modulators of this thermoTRP channel; (ii) the design of allosteric modulators that interfere with protein-protein interaction involved in the functional coupling of stimulus sensing and gate opening; and (iii) the development of compounds that abrogate the inflammation-mediated increase of receptor expression in the neuronal surface. These new sites of action represent novel strategies to modulate pathologically active TRPV1, while minimizing an effect on the TRPV1 subpopulation involved in physiological and protective roles, thus increasing their potential therapeutic use.
Collapse
|
32
|
Fernández-Ballester G, Fernández-Carvajal A, González-Ros JM, Ferrer-Montiel A. Ionic channels as targets for drug design: a review on computational methods. Pharmaceutics 2011; 3:932-53. [PMID: 24309315 PMCID: PMC3857065 DOI: 10.3390/pharmaceutics3040932] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 10/26/2011] [Accepted: 11/30/2011] [Indexed: 01/21/2023] Open
Abstract
Ion channels are involved in a broad range of physiological and pathological processes. The implications of ion channels in a variety of diseases, including diabetes, epilepsy, hypertension, cancer and even chronic pain, have signaled them as pivotal drug targets. Thus far, drugs targeting ion channels were developed without detailed knowledge of the molecular interactions between the lead compounds and the target channels. In recent years, however, the emergence of high-resolution structures for a plethora of ion channels paves the way for computer-assisted drug design. Currently, available functional and structural data provide an attractive platform to generate models that combine substrate-based and protein-based approaches. In silico approaches include homology modeling, quantitative structure-activity relationships, virtual ligand screening, similarity and pharmacophore searching, data mining, and data analysis tools. These strategies have been frequently used in the discovery and optimization of novel molecules with enhanced affinity and specificity for the selected therapeutic targets. In this review we summarize recent applications of in silico methods that are being used for the development of ion channel drugs.
Collapse
|
33
|
Cvetkov TL, Huynh KW, Cohen MR, Moiseenkova-Bell VY. Molecular architecture and subunit organization of TRPA1 ion channel revealed by electron microscopy. J Biol Chem 2011; 286:38168-38176. [PMID: 21908607 DOI: 10.1074/jbc.m111.288993] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is a non-selective ion channel, which is expressed in nociceptor sensory neurons and transduces chemical, inflammatory, and neuropathic pain signals. Numerous non-reactive compounds and electrophilic compounds, such as endogenous inflammatory mediators and exogenous pungent chemicals, can activate TRPA1. Here we report a 16-Å resolution structure of purified, functional, amphipol-stabilized TRPA1 analyzed by single-particle EM. Molecular models of the N and C termini of the channel were generated using the I-TASSER protein structure prediction server and docked into the EM density to provide insight into the TRPA1 subunit organization. This structural analysis suggests a location for critical N-terminal cysteine residues involved in electrophilic activation at the interface between neighboring subunits. Our results indicate that covalent modifications within this pocket may alter interactions between subunits and promote conformational changes that lead to channel activation.
Collapse
Affiliation(s)
- Teresa L Cvetkov
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Kevin W Huynh
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Matthew R Cohen
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Vera Y Moiseenkova-Bell
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
34
|
Yang W, Manna PT, Zou J, Luo J, Beech DJ, Sivaprasadarao A, Jiang LH. Zinc inactivates melastatin transient receptor potential 2 channels via the outer pore. J Biol Chem 2011; 286:23789-98. [PMID: 21602277 PMCID: PMC3129160 DOI: 10.1074/jbc.m111.247478] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 05/19/2011] [Indexed: 10/18/2022] Open
Abstract
Zinc ion (Zn(2+)) is an endogenous allosteric modulator that regulates the activity of a wide variety of ion channels in a reversible and concentration-dependent fashion. Here we used patch clamp recording to study the effects of Zn(2+) on the melastatin transient receptor potential 2 (TRPM2) channel. Zn(2+) inhibited the human (h) TRPM2 channel currents, and the steady-state inhibition was largely not reversed upon washout and concentration-independent in the range of 30-1000 μM, suggesting that Zn(2+) induces channel inactivation. Zn(2+) inactivated the channels fully when they conducted inward currents, but only by half when they passed outward currents, indicating profound influence of the permeant ion on Zn(2+) inactivation. Alanine substitution scanning mutagenesis of 20 Zn(2+)-interacting candidate residues in the outer pore region of the hTRPM2 channel showed that mutation of Lys(952) in the extracellular end of the fifth transmembrane segment and Asp(1002) in the large turret strongly attenuated or abolished Zn(2+) inactivation, and mutation of several other residues dramatically changed the inactivation kinetics. The mouse (m) TRPM2 channels were also inactivated by Zn(2+), but the kinetics were remarkably slower. Reciprocal mutation of His(995) in the hTRPM2 channel and the equivalent Gln(992) in the mTRPM2 channel completely swapped the kinetics, but no such opposing effects resulted from exchanging another pair of species-specific residues, Arg(961)/Ser(958). We conclude from these results that Zn(2+) inactivates the TRPM2 channels and that residues in the outer pore are critical determinants of the inactivation.
Collapse
Affiliation(s)
- Wei Yang
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
- the Department of Neurobiology, Zhejiang University School of Medicine, Zhejiang 310058, China
| | - Paul T. Manna
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Jie Zou
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Jianhong Luo
- the Department of Neurobiology, Zhejiang University School of Medicine, Zhejiang 310058, China
| | - David J. Beech
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Asipu Sivaprasadarao
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| | - Lin-Hua Jiang
- From the Institute of Membrane and Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom and
| |
Collapse
|
35
|
Lin Z, Reilly CA, Antemano R, Hughen RW, Marett L, Concepcion GP, Haygood MG, Olivera BM, Light A, Schmidt EW. Nobilamides A-H, long-acting transient receptor potential vanilloid-1 (TRPV1) antagonists from mollusk-associated bacteria. J Med Chem 2011; 54:3746-55. [PMID: 21524089 PMCID: PMC3133741 DOI: 10.1021/jm101621u] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
New compounds nobilamides A-H and related known compounds A-3302-A and A-3302-B were isolated based upon their suppression of capsaicin-induced calcium uptake in a mouse dorsal root ganglion primary cell culture assay. Two of these compounds, nobilamide B and A-3302-A, were shown to be long-acting antagonists of mouse and human TRPV1 channels, abolishing activity for >1 h after removal of drug presumably via a covalent attachment. Other derivatives also inhibited the TRPV1 channel, albeit with low potency, affording a structure-activity profile to support the proposed mechanism of action. While the activities were modest, we propose a new mechanism of action and a new site of binding for these inhibitors that may spur development of related analogues for treatment of pain.
Collapse
Affiliation(s)
- Zhenjian Lin
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | - Christopher A. Reilly
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, USA
| | - Rowena Antemano
- Marine Science Institute, University of the Philippines, Diliman, Quezon City 1101, Philippines
| | - Ronald W. Hughen
- Department of Anesthesiology, University of Utah, Salt Lake City, Utah 84112, USA
| | - Lenny Marett
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | - Gisela P. Concepcion
- Marine Science Institute, University of the Philippines, Diliman, Quezon City 1101, Philippines
| | - Margo G. Haygood
- Department of Environmental and Biomolecular Systems, OGI School of Science & Engineering, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | | | - Alan Light
- Department of Anesthesiology, University of Utah, Salt Lake City, Utah 84112, USA
| | - Eric W. Schmidt
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
- Department of Biology, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
36
|
Goswami C, Kuhn J, Dina OA, Fernández-Ballester G, Levine JD, Ferrer-Montiel A, Hucho T. Estrogen destabilizes microtubules through an ion-conductivity-independent TRPV1 pathway. J Neurochem 2011; 117:995-1008. [PMID: 21480900 DOI: 10.1111/j.1471-4159.2011.07270.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recently, we described estrogen and agonists of the G-protein coupled estrogen receptor GPR30 to induce protein kinase C (PKC)ε-dependent pain sensitization. PKCε phosphorylates the ion channel transient receptor potential, vanilloid subclass I (TRPV1) close to a novel microtubule-TRPV1 binding site. We now modeled the binding of tubulin to the TRPV1 C-terminus. The model suggests PKCε phosphorylation of TRPV1-S800 to abolish the tubulin-TRPV1 interaction. Indeed, in vitro PKCε phosphorylation of TRPV1 hindered tubulin-binding to TRPV1. In vivo, treatment of sensory neurons and F-11 cells with estrogen and the GPR30 agonist, G-1, resulted in microtubule destabilization and retraction of microtubules from filopodial structures. We found estrogen and G-1 to regulate the stability of the microtubular network via PKC phosphorylation of the PKCε-phosphorylation site TRPV1-S800. Microtubule disassembly was not, however, dependent on TRPV1 ion conductivity. TRPV1 knock-down in rats inverted the effect of the microtubule-modulating drugs, Taxol and Nocodazole, on estrogen-induced and PKCε-dependent mechanical pain sensitization. Thus, we suggest the C-terminus of TRPV1 to be a signaling intermediate downstream of estrogen and PKCε, regulating microtubule-stability and microtubule-dependent pain sensitization.
Collapse
Affiliation(s)
- Chandan Goswami
- Department for Molecular Human Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Picazo-Juárez G, Romero-Suárez S, Nieto-Posadas A, Llorente I, Jara-Oseguera A, Briggs M, McIntosh TJ, Simon SA, Ladrón-de-Guevara E, Islas LD, Rosenbaum T. Identification of a binding motif in the S5 helix that confers cholesterol sensitivity to the TRPV1 ion channel. J Biol Chem 2011; 286:24966-76. [PMID: 21555515 DOI: 10.1074/jbc.m111.237537] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The TRPV1 ion channel serves as an integrator of noxious stimuli with its activation linked to pain and neurogenic inflammation. Cholesterol, a major component of cell membranes, modifies the function of several types of ion channels. Here, using measurements of capsaicin-activated currents in excised patches from TRPV1-expressing HEK cells, we show that enrichment with cholesterol, but not its diastereoisomer epicholesterol, markedly decreased wild-type rat TRPV1 currents. Substitutions in the S5 helix, rTRPV1-R579D, and rTRPV1-F582Q, decreased this cholesterol response and rTRPV1-L585I was insensitive to cholesterol addition. Two human TRPV1 variants, with different amino acids at position 585, had different responses to cholesterol with hTRPV1-Ile(585) being insensitive to this molecule. However, hTRPV1-I585L was inhibited by cholesterol addition similar to rTRPV1 with the same S5 sequence. In the absence of capsaicin, cholesterol enrichment also inhibited TRPV1 currents induced by elevated temperature and voltage. These data suggest that there is a cholesterol-binding site in TRPV1 and that the functions of TRPV1 depend on the genetic variant and membrane cholesterol content.
Collapse
Affiliation(s)
- Giovanni Picazo-Juárez
- Departamento de Neurodesarrollo y Fisiología, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, D.F. 04510, México
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Valente P, Fernández-Carvajal A, Camprubí-Robles M, Gomis A, Quirce S, Viana F, Fernández-Ballester G, González-Ros JM, Belmonte C, Planells-Cases R, Ferrer-Montiel A. Membrane-tethered peptides patterned after the TRP domain (TRPducins) selectively inhibit TRPV1 channel activity. FASEB J 2011; 25:1628-40. [PMID: 21307333 DOI: 10.1096/fj.10-174433] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The transient receptor potential vanilloid 1 (TRPV1) channel is a thermosensory receptor implicated in diverse physiological and pathological processes. The TRP domain, a highly conserved region in the C terminus adjacent to the internal channel gate, is critical for subunit tetramerization and channel gating. Here, we show that cell-penetrating, membrane-anchored peptides patterned after this protein domain are moderate and selective TRPV1 antagonists both in vitro and in vivo, blocking receptor activity in intact rat primary sensory neurons and their peripheral axons with mean decline time of 30 min. The most potent lipopeptide, TRP-p5, blocked all modes of TRPV1 gating with micromolar efficacy (IC(50)<10 μM), without significantly affecting other thermoTRP channels. In contrast, its retrosequence or the corresponding sequences of other TRPV channels did not alter TRPV1 channel activity (IC(50)>100 μM). TRP-p5 did not affect the capsaicin sensitivity of the vanilloid receptor. Our data suggest that TRP-p5 interferes with protein-protein interactions at the level of the TRP domain that are essential for the "conformational" change that leads to gate opening. Therefore, these palmitoylated peptides, which we termed TRPducins, are noncompetitive, voltage-independent, sequence-specific TRPV1 blockers. Our findings indicate that TRPducin-like peptides may embody a novel molecular strategy that can be exploited to generate a selective pharmacological arsenal for the TRP superfamily of ion channels.
Collapse
Affiliation(s)
- Pierluigi Valente
- Instituto de Biología Molecular y Celular, Universidad Miguel Herna´ndez, Elche, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Boukalova S, Marsakova L, Teisinger J, Vlachova V. Conserved residues within the putative S4-S5 region serve distinct functions among thermosensitive vanilloid transient receptor potential (TRPV) channels. J Biol Chem 2010; 285:41455-62. [PMID: 21044960 DOI: 10.1074/jbc.m110.145466] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The vanilloid transient receptor potential channel TRPV1 is a tetrameric six-transmembrane segment (S1-S6) channel that can be synergistically activated by various proalgesic agents such as capsaicin, protons, heat, or highly depolarizing voltages, and also by 2-aminoethoxydiphenyl borate (2-APB), a common activator of the related thermally gated vanilloid TRP channels TRPV1, TRPV2, and TRPV3. In these channels, the conserved charged residues in the intracellular S4-S5 region have been proposed to constitute part of a voltage sensor that acts in concert with other stimuli to regulate channel activation. The molecular basis of this gating event is poorly understood. We mutated charged residues all along the S4 and the S4-S5 linker of TRPV1 and identified four potential voltage-sensing residues (Arg(557), Glu(570), Asp(576), and Arg(579)) that, when specifically mutated, altered the functionality of the channel with respect to voltage, capsaicin, heat, 2-APB, and/or their interactions in different ways. The nonfunctional charge-reversing mutations R557E and R579E were partially rescued by the charge-swapping mutations R557E/E570R and D576R/R579E, indicating that electrostatic interactions contribute to allosteric coupling between the voltage-, temperature- and capsaicin-dependent activation mechanisms. The mutant K571E was normal in all aspects of TRPV1 activation except for 2-APB, revealing the specific role of Lys(571) in chemical sensitivity. Surprisingly, substitutions at homologous residues in TRPV2 or TRPV3 had no effect on temperature- and 2-APB-induced activity. Thus, the charged residues in S4 and the S4-S5 linker contribute to voltage sensing in TRPV1 and, despite their highly conserved nature, regulate the temperature and chemical gating in the various TRPV channels in different ways.
Collapse
Affiliation(s)
- Stepana Boukalova
- Department of Cellular Neurophysiology, Institute of Physiology, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic
| | | | | | | |
Collapse
|
40
|
Schumacher MA, Eilers H. TRPV1 splice variants: structure and function. Front Biosci (Landmark Ed) 2010; 15:872-82. [PMID: 20515731 DOI: 10.2741/3651] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The capsaicin receptor (TRPV1) is a non-selective cation channel predominantly expressed in specialized sensory neurons that detect painful stimuli. Although its many functional roles continue to be revealed, it has been confirmed to play a critical role in the perception of peripheral inflammatory hyperalgesia and pain. TRPV1 not only is sensitized and/or activated under a wide range of conditions including inflammation and nerve injury but also undergoes changes in expressed levels in response to these same pathologic conditions. Just as our understanding of the structural requirements of TRPV1 activation has grown, there is evidence that TRPV1 forms heteromeric channel complexes. This review is focused on the structural and functional consequence of TRPV1 splice variants: VR.5'sv, TRPV1b/beta and TRPV1var. Through their co-expression and formation of heteromeric complexes with TRPV1, they have been shown to modulate TRPV1 activation. Moreover, TRPV1 splice variant subunits may also contribute unique properties of activation such as the detection of hypertonic conditions.
Collapse
Affiliation(s)
- Mark A Schumacher
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143-0427, USA.
| | | |
Collapse
|
41
|
|
42
|
Thermosensitive TRP channel pore turret is part of the temperature activation pathway. Proc Natl Acad Sci U S A 2010; 107:7083-8. [PMID: 20351268 DOI: 10.1073/pnas.1000357107] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Temperature sensing is crucial for homeotherms, including human beings, to maintain a stable body core temperature and respond to the ambient environment. A group of exquisitely temperature-sensitive transient receptor potential channels, termed thermoTRPs, serve as cellular temperature sensors. How thermoTRPs convert thermal energy (heat) into protein conformational changes leading to channel opening remains unknown. Here we demonstrate that the pathway for temperature-dependent activation is distinct from those for ligand- and voltage-dependent activation and involves the pore turret. We found that mutant channels with an artificial pore turret sequence lose temperature sensitivity but maintain normal ligand responses. Using site-directed fluorescence recordings we observed that temperature change induces a significant rearrangement of TRPV1 pore turret that is coupled to channel opening. This movement is specifically associated to temperature-dependent activation and is not observed during ligand- and voltage-dependent channel activation. These observations suggest that the turret is part of the temperature-sensing apparatus in thermoTRP channels, and its conformational change may give rise to the large entropy that defines high temperature sensitivity.
Collapse
|
43
|
Shigematsu H, Sokabe T, Danev R, Tominaga M, Nagayama K. A 3.5-nm structure of rat TRPV4 cation channel revealed by Zernike phase-contrast cryoelectron microscopy. J Biol Chem 2009; 285:11210-8. [PMID: 20044482 DOI: 10.1074/jbc.m109.090712] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel responsive to various stimuli including cell swelling, warm temperatures (27-35 degrees C), and chemical compounds such as phorbol ester derivatives. Here we report the three-dimensional structure of full-length rat TRPV4 purified from baculovirus-infected Sf9 cells. Hexahistidine-tagged rat TRPV4 (His-rTRPV4) was solubilized with detergent and purified through affinity chromatography and size-exclusion chromatography. Chemical cross-linking analysis revealed that detergent-solubilized His-rTRPV4 was a tetramer. The 3.5-nm structure of rat TRPV4 was determined by cryoelectron microscopy using single-particle reconstruction from Zernike phase-contrast images. The overall structure comprises two distinct regions; a larger dense component, likely corresponding to the cytoplasmic N- and C-terminal regions, and a smaller component corresponding to the transmembrane region.
Collapse
Affiliation(s)
- Hideki Shigematsu
- Division of Nano-Structure Physiology, Okazaki Institute for Integrative Bioscience, Higashiyama, Myodaiji, Okazaki 444-8787 Japan
| | | | | | | | | |
Collapse
|
44
|
Pedretti A, Marconi C, Bettinelli I, Vistoli G. Comparative modeling of the quaternary structure for the human TRPM8 channel and analysis of its binding features. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:973-82. [DOI: 10.1016/j.bbamem.2009.02.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Revised: 01/24/2009] [Accepted: 02/04/2009] [Indexed: 11/15/2022]
|