1
|
Chen S, Luo Y, Yu P, Yue X, Yang W. [Advances in the development of transient receptor potential melastatin 2 channel inhibitors]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2025; 54:120-130. [PMID: 39909465 PMCID: PMC11956857 DOI: 10.3724/zdxbyxb-2024-0586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025]
Abstract
Studies on specific transient receptor potential melastatin 2 (TRPM2) channel inhibitors can deepen our understanding of the pathological mechanism of related diseases, and allow discovery of novel, effective targets and drugs for therapy. The development of TRPM2 channel inhibitors can be broadly classified into four categories with distinct characteristics: reutilization and structural modification of homologous ion channel modulators to produce a diverse array of TRPM2 channel inhibitors with strong inhibitory effects; TRPM2 channel inhibitors based on channel gating mechanism with high specificity; inhibitors identified through high-throughput screening with novel chemical structures; inhibitors developed from natural antioxidants with higher safety. In recent years, the application of computer-aided drug design has significantly accelerated the development of TRPM2 channel inhibitors. Several promising compounds such as ZA18, A1 and D9 have been discovered, and it is expected that more potent and selective TRPM2 channel inhibitor scaffolds will be discovered in the future. This article reviews the advances on the studies of TRPM2 channel inhibitors, aiming to provide insights for further research and clinical application of TRPM2 channel inhibitors.
Collapse
Affiliation(s)
- Shiyao Chen
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Yanping Luo
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University School of Medicine , Hangzhou 310058, China
| | - Xiaomin Yue
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Wei Yang
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
2
|
Liu W, Deng W, Hu L, Zou H. Advances in TRPV6 inhibitors for tumors by targeted therapies: Macromolecular proteins, synthetic small molecule compounds, and natural compounds. Eur J Med Chem 2024; 270:116379. [PMID: 38588625 DOI: 10.1016/j.ejmech.2024.116379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/30/2024] [Accepted: 03/31/2024] [Indexed: 04/10/2024]
Abstract
TRPV6, a Ca2+-selective member of the transient receptor potential vanilloid (TRPV) family, plays a key role in extracellular calcium transport, calcium ion reuptake, and maintenance of a local low calcium environment. An increasing number of studies have shown that TRPV6 is involved in the regulation of various diseases. Notably, overexpression of TRPV6 is closely related to the occurrence of various cancers. Research confirmed that knocking down TRPV6 could effectively reduce the proliferation and invasiveness of tumors by mainly mediating the calcium signaling pathway. Hence, TRPV6 has become a promising new drug target for numerous tumor treatments. However, the development of TRPV6 inhibitors is still in the early stage, and the existing TRPV6 inhibitors have poor selectivity and off-target effects. In this review, we focus on summarizing and describing the structure characters, and mechanisms of existing TRPV6 inhibitors to provide new ideas and directions for the development of novel TRPV6 inhibitors.
Collapse
Affiliation(s)
- Weikang Liu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Wenwen Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Liqing Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China.
| | - Hui Zou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China.
| |
Collapse
|
3
|
Pumroy RA, De Jesús-Pérez JJ, Protopopova AD, Rocereta JA, Fluck EC, Fricke T, Lee BH, Rohacs T, Leffler A, Moiseenkova-Bell V. Molecular details of ruthenium red pore block in TRPV channels. EMBO Rep 2024; 25:506-523. [PMID: 38225355 PMCID: PMC10897480 DOI: 10.1038/s44319-023-00050-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Transient receptor potential vanilloid (TRPV) channels play a critical role in calcium homeostasis, pain sensation, immunological response, and cancer progression. TRPV channels are blocked by ruthenium red (RR), a universal pore blocker for a wide array of cation channels. Here we use cryo-electron microscopy to reveal the molecular details of RR block in TRPV2 and TRPV5, members of the two TRPV subfamilies. In TRPV2 activated by 2-aminoethoxydiphenyl borate, RR is tightly coordinated in the open selectivity filter, blocking ion flow and preventing channel inactivation. In TRPV5 activated by phosphatidylinositol 4,5-bisphosphate, RR blocks the selectivity filter and closes the lower gate through an interaction with polar residues in the pore vestibule. Together, our results provide a detailed understanding of TRPV subfamily pore block, the dynamic nature of the selectivity filter and allosteric communication between the selectivity filter and lower gate.
Collapse
Affiliation(s)
- Ruth A Pumroy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - José J De Jesús-Pérez
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anna D Protopopova
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julia A Rocereta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Edwin C Fluck
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tabea Fricke
- Institute for Neurophysiology, Hannover Medical School, 30625, Hannover, Germany
| | - Bo-Hyun Lee
- Department of Physiology and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University Medical School, Jinju, Korea
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Andreas Leffler
- Institute for Neurophysiology, Hannover Medical School, 30625, Hannover, Germany
| | - Vera Moiseenkova-Bell
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
De Jesús-Pérez JJ, Gabrielle M, Raheem S, Fluck EC, Rohacs T, Moiseenkova-Bell VY. Structural mechanism of TRPV5 inhibition by econazole. Structure 2024; 32:148-156.e5. [PMID: 38141613 PMCID: PMC10872542 DOI: 10.1016/j.str.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/01/2023] [Accepted: 11/28/2023] [Indexed: 12/25/2023]
Abstract
The calcium-selective TRPV5 channel activated by phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is involved in calcium homeostasis. Recently, cryoelectron microscopy (cryo-EM) provided molecular details of TRPV5 modulation by exogenous and endogenous molecules. However, the details of TRPV5 inhibition by the antifungal agent econazole (ECN) remain elusive due to the low resolution of the currently available structure. In this study, we employ cryo-EM to comprehensively examine how the ECN inhibits TRPV5. By combining our structural findings with site-directed mutagenesis, calcium measurements, electrophysiology, and molecular dynamics simulations, we determined that residues F472 and L475 on the S4 helix, along with residue W495 on the S5 helix, collectively constitute the ECN-binding site. Additionally, the structure of TRPV5 in the presence of ECN and PI(4,5)P2, which does not show the bound activator, reveals a potential inhibition mechanism in which ECN competes with PI(4,5)P2, preventing the latter from binding, and ultimately pore closure.
Collapse
Affiliation(s)
- José J De Jesús-Pérez
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Gabrielle
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Sumiyya Raheem
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Edwin C Fluck
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Vera Y Moiseenkova-Bell
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Melancon K, Pliushcheuskaya P, Meiler J, Künze G. Targeting ion channels with ultra-large library screening for hit discovery. Front Mol Neurosci 2024; 16:1336004. [PMID: 38249296 PMCID: PMC10796734 DOI: 10.3389/fnmol.2023.1336004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024] Open
Abstract
Ion channels play a crucial role in a variety of physiological and pathological processes, making them attractive targets for drug development in diseases such as diabetes, epilepsy, hypertension, cancer, and chronic pain. Despite the importance of ion channels in drug discovery, the vastness of chemical space and the complexity of ion channels pose significant challenges for identifying drug candidates. The use of in silico methods in drug discovery has dramatically reduced the time and cost of drug development and has the potential to revolutionize the field of medicine. Recent advances in computer hardware and software have enabled the screening of ultra-large compound libraries. Integration of different methods at various scales and dimensions is becoming an inevitable trend in drug development. In this review, we provide an overview of current state-of-the-art computational chemistry methodologies for ultra-large compound library screening and their application to ion channel drug discovery research. We discuss the advantages and limitations of various in silico techniques, including virtual screening, molecular mechanics/dynamics simulations, and machine learning-based approaches. We also highlight several successful applications of computational chemistry methodologies in ion channel drug discovery and provide insights into future directions and challenges in this field.
Collapse
Affiliation(s)
- Kortney Melancon
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
| | | | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
- Medical Faculty, Institute for Drug Discovery, Leipzig University, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University, Leipzig, Germany
| | - Georg Künze
- Medical Faculty, Institute for Drug Discovery, Leipzig University, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University, Leipzig, Germany
- Interdisciplinary Center for Bioinformatics, Leipzig University, Leipzig, Germany
| |
Collapse
|
6
|
Neuberger A, Sobolevsky AI. Molecular pharmacology of the onco-TRP channel TRPV6. Channels (Austin) 2023; 17:2266669. [PMID: 37838981 PMCID: PMC10578198 DOI: 10.1080/19336950.2023.2266669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023] Open
Abstract
TRPV6, a representative of the vanilloid subfamily of TRP channels, serves as the principal calcium uptake channel in the gut. Dysregulation of TRPV6 results in disturbed calcium homeostasis leading to a variety of human diseases, including many forms of cancer. Inhibitors of this oncochannel are therefore particularly needed. In this review, we provide an overview of recent advances in structural pharmacology that uncovered the molecular mechanisms of TRPV6 inhibition by a variety of small molecules, including synthetic and natural, plant-derived compounds as well as some prospective and clinically approved drugs.
Collapse
Affiliation(s)
- Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | | |
Collapse
|
7
|
Okada Y, Numata T, Sabirov RZ, Kashio M, Merzlyak PG, Sato-Numata K. Cell death induction and protection by activation of ubiquitously expressed anion/cation channels. Part 3: the roles and properties of TRPM2 and TRPM7. Front Cell Dev Biol 2023; 11:1246955. [PMID: 37842082 PMCID: PMC10576435 DOI: 10.3389/fcell.2023.1246955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell volume regulation (CVR) is a prerequisite for animal cells to survive and fulfill their functions. CVR dysfunction is essentially involved in the induction of cell death. In fact, sustained normotonic cell swelling and shrinkage are associated with necrosis and apoptosis, and thus called the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. Since a number of ubiquitously expressed ion channels are involved in the CVR processes, these volume-regulatory ion channels are also implicated in the NVI and AVD events. In Part 1 and Part 2 of this series of review articles, we described the roles of swelling-activated anion channels called VSOR or VRAC and acid-activated anion channels called ASOR or PAC in CVR and cell death processes. Here, Part 3 focuses on therein roles of Ca2+-permeable non-selective TRPM2 and TRPM7 cation channels activated by stress. First, we summarize their phenotypic properties and molecular structure. Second, we describe their roles in CVR. Since cell death induction is tightly coupled to dysfunction of CVR, third, we focus on their participation in the induction of or protection against cell death under oxidative, acidotoxic, excitotoxic, and ischemic conditions. In this regard, we pay attention to the sensitivity of TRPM2 and TRPM7 to a variety of stress as well as to their capability to physicall and functionally interact with other volume-related channels and membrane enzymes. Also, we summarize a large number of reports hitherto published in which TRPM2 and TRPM7 channels are shown to be involved in cell death associated with a variety of diseases or disorders, in some cases as double-edged swords. Lastly, we attempt to describe how TRPM2 and TRPM7 are organized in the ionic mechanisms leading to cell death induction and protection.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan
| | - Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| | - Ravshan Z. Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Makiko Kashio
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
| | - Peter G. Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| |
Collapse
|
8
|
Neuberger A, Trofimov YA, Yelshanskaya MV, Nadezhdin KD, Krylov NA, Efremov RG, Sobolevsky AI. Structural mechanism of human oncochannel TRPV6 inhibition by the natural phytoestrogen genistein. Nat Commun 2023; 14:2659. [PMID: 37160865 PMCID: PMC10169861 DOI: 10.1038/s41467-023-38352-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 04/27/2023] [Indexed: 05/11/2023] Open
Abstract
Calcium-selective oncochannel TRPV6 is the major driver of cell proliferation in human cancers. While significant effort has been invested in the development of synthetic TRPV6 inhibitors, natural channel blockers have been largely neglected. Here we report the structure of human TRPV6 in complex with the plant-derived phytoestrogen genistein, extracted from Styphnolobium japonicum, that was shown to inhibit cell invasion and metastasis in cancer clinical trials. Despite the pharmacological value, the molecular mechanism of TRPV6 inhibition by genistein has remained enigmatic. We use cryo-EM combined with electrophysiology, calcium imaging, mutagenesis, and molecular dynamics simulations to show that genistein binds in the intracellular half of the TRPV6 pore and acts as an ion channel blocker and gating modifier. Genistein binding to the open channel causes pore closure and a two-fold symmetrical conformational rearrangement in the S4-S5 and S6-TRP helix regions. The unprecedented mechanism of TRPV6 inhibition by genistein uncovers new possibilities in structure-based drug design.
Collapse
Affiliation(s)
- Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Yury A Trofimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Maria V Yelshanskaya
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Nikolay A Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
9
|
Fujii T, Shimizu T, Kaji Y, Katoh M, Sakai H. Activation of mouse Otop3 proton channels by Zn2+. Biochem Biophys Res Commun 2023; 658:55-61. [PMID: 37023615 DOI: 10.1016/j.bbrc.2023.03.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Otopetrins (Otop1-Otop3) belong to a newly identified family of proton (H+) channels activated by extracellular acidification. Here, we found that Zn2+ activates the mouse Otop3 (mOtop3) proton channels by using electrophysiological patch-clamp techniques. In mOtop3-expressing human embryonic kidney HEK293T cells, a biphasic inward mOtop3 H+ current comprising a fast transient current followed by a sustained current was observed upon extracellular acidification at pH 5.0. No significant activation of the mOtop3 channel was observed at pH 6.5 and 7.4, but interestingly, Zn2+ dose-dependently induced a sustained activation of mOtop3 under these pH conditions. Increasing the Zn2+ concentration had no effect on the reversal potential of the channel currents, suggesting that Zn2+ does not permeate through the mOtop3. The activation of the mOtop3 channel was specific to Zn2+ among divalent metal cations. Our findings reveal a novel modulatory mechanism of mOtop3 proton channels by Zn2+.
Collapse
|
10
|
Rohacs T, Fluck EC, De Jesús-Pérez JJ, Moiseenkova-Bell VY. What structures did, and did not, reveal about the function of the epithelial Ca 2+ channels TRPV5 and TRPV6. Cell Calcium 2022; 106:102620. [PMID: 35834842 PMCID: PMC11500022 DOI: 10.1016/j.ceca.2022.102620] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 12/15/2022]
Abstract
Transient Receptor Potential Vanilloid 5 and 6 (TRPV5 and TRPV6) are Ca2+ selective epithelial ion channels. They are the products of a relatively recent gene duplication in mammals, and have high sequence homology to each other. Their functional properties are also much more similar to each other than to other members of the TRPV subfamily. They are both constitutively active, and this activity depends on the endogenous cofactor phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Both channels undergo Ca2+-induced inactivation, which is mediated by direct binding of the ubiquitous Ca2+ binding protein calmodulin (CaM) to the channels, and by a decrease in PI(4,5)P2 levels by Ca2+ -induced activation of phospholipase C (PLC). Recent cryo electron microscopy (cryo-EM) and X-ray crystallography structures provided detailed structural information for both TRPV5 and TRPV6. This review will discuss this structural information in the context of the function of these channels focusing on the mechanism of CaM inhibition, activation by PI(4,5)P2 and binding of pharmacological modulators.
Collapse
Affiliation(s)
- Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, USA.
| | - Edwin C Fluck
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - José J De Jesús-Pérez
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Vera Y Moiseenkova-Bell
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
11
|
Hird C, Franklin CE, Cramp RL. The role of environmental calcium in the extreme acid tolerance of northern banjo frog (Limnodynastes terraereginae) larvae. J Exp Biol 2022; 225:275908. [PMID: 35702935 DOI: 10.1242/jeb.244376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/10/2022] [Indexed: 11/20/2022]
Abstract
Many aquatically respiring animals acutely exposed to low pH waters suffer inhibition of ion uptake, and loss of branchial (gill) epithelial integrity, culminating in a fatal loss of body Na+. Environmental calcium levels ([Ca2+]e) are pivotal in maintaining branchial junction integrity, with supplemental Ca2+ reversing the negative effects of low pH in some animals. Tolerance of some naturally acidic environments by aquatic animals is further complicated by low [Ca2+]e, yet many of these environments are surprisingly biodiverse. How animals overcome the damaging actions of low pH and low environmental Ca2+ remains unknown. We examined the effects of [Ca2+]e on the response to low pH in larvae of the highly acid tolerant frog Limnodynastes terraereginae. Acute exposure to low pH water in the presence of low (5 µmol L-1) [Ca2+]e increased net Na+ efflux. Provision of additional [Ca2+]e reduced net Na+ efflux, but the effect was saturable. Acclimation to both low and high (250 µmol L-1) [Ca2+]e improved the resistance of larvae to Na+ efflux at low pH. Exposure to the Ca2+ channel inhibitor ruthenium red resulted in an abrupt loss of tolerance in low pH acclimated larvae. Acclimation to acidic water increased branchial gene expression of the intracellular Ca2+ transport protein calbindin, consistent with a role for increased transcellular Ca2+ trafficking in the tolerance of acidic water. This study supports a role for [Ca2+]e in promoting branchial integrity and highlights a potential mechanism via the maintenance of transcellular Ca2+ uptake in the acid tolerance of L. terraereginae larvae.
Collapse
Affiliation(s)
- Coen Hird
- School of Biological Sciences, The University of Queensland, Brisbane 4072, Australia
| | - Craig E Franklin
- School of Biological Sciences, The University of Queensland, Brisbane 4072, Australia
| | - Rebecca L Cramp
- School of Biological Sciences, The University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
12
|
Yelshanskaya MV, Sobolevsky AI. Ligand-Binding Sites in Vanilloid-Subtype TRP Channels. Front Pharmacol 2022; 13:900623. [PMID: 35652046 PMCID: PMC9149226 DOI: 10.3389/fphar.2022.900623] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/06/2022] [Indexed: 02/02/2023] Open
Abstract
Vanilloid-subfamily TRP channels TRPV1-6 play important roles in various physiological processes and are implicated in numerous human diseases. Advances in structural biology, particularly the "resolution revolution" in cryo-EM, have led to breakthroughs in molecular characterization of TRPV channels. Structures with continuously improving resolution uncover atomic details of TRPV channel interactions with small molecules and protein-binding partners. Here, we provide a classification of structurally characterized binding sites in TRPV channels and discuss the progress that has been made by structural biology combined with mutagenesis, functional recordings, and molecular dynamics simulations toward understanding of the molecular mechanisms of ligand action. Given the similarity in structural architecture of TRP channels, 16 unique sites identified in TRPV channels may be shared between TRP channel subfamilies, although the chemical identity of a particular ligand will likely depend on the local amino-acid composition. The characterized binding sites and molecular mechanisms of ligand action create a diversity of druggable targets to aid in the design of new molecules for tuning TRP channel function in disease conditions.
Collapse
Affiliation(s)
| | - Alexander I. Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, United States
| |
Collapse
|
13
|
Lees JA, Dias JM, Han S. Applications of Cryo-EM in small molecule and biologics drug design. Biochem Soc Trans 2021; 49:2627-2638. [PMID: 34812853 PMCID: PMC8786282 DOI: 10.1042/bst20210444] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 02/03/2023]
Abstract
Electron cryo-microscopy (cryo-EM) is a powerful technique for the structural characterization of biological macromolecules, enabling high-resolution analysis of targets once inaccessible to structural interrogation. In recent years, pharmaceutical companies have begun to utilize cryo-EM for structure-based drug design. Structural analysis of integral membrane proteins, which comprise a large proportion of druggable targets and pose particular challenges for X-ray crystallography, by cryo-EM has enabled insights into important drug target families such as G protein-coupled receptors (GPCRs), ion channels, and solute carrier (SLCs) proteins. Structural characterization of biologics, such as vaccines, viral vectors, and gene therapy agents, has also become significantly more tractable. As a result, cryo-EM has begun to make major impacts in bringing critical therapeutics to market. In this review, we discuss recent instructive examples of impacts from cryo-EM in therapeutics design, focusing largely on its implementation at Pfizer. We also discuss the opportunities afforded by emerging technological advances in cryo-EM, and the prospects for future development of the technique.
Collapse
Affiliation(s)
- Joshua A. Lees
- Discovery Sciences, Medicine Design, Pfizer Worldwide Research and Development, Groton, CT 06340, U.S.A
| | - Joao M. Dias
- Discovery Sciences, Medicine Design, Pfizer Worldwide Research and Development, Groton, CT 06340, U.S.A
| | - Seungil Han
- Discovery Sciences, Medicine Design, Pfizer Worldwide Research and Development, Groton, CT 06340, U.S.A
| |
Collapse
|
14
|
Neuberger A, Nadezhdin KD, Sobolevsky AI. Structural mechanisms of TRPV6 inhibition by ruthenium red and econazole. Nat Commun 2021; 12:6284. [PMID: 34725357 PMCID: PMC8560856 DOI: 10.1038/s41467-021-26608-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/14/2021] [Indexed: 11/26/2022] Open
Abstract
TRPV6 is a calcium-selective ion channel implicated in epithelial Ca2+ uptake. TRPV6 inhibitors are needed for the treatment of a broad range of diseases associated with disturbed calcium homeostasis, including cancers. Here we combine cryo-EM, calcium imaging, and mutagenesis to explore molecular bases of human TRPV6 inhibition by the antifungal drug econazole and the universal ion channel blocker ruthenium red (RR). Econazole binds to an allosteric site at the channel's periphery, where it replaces a lipid. In contrast, RR inhibits TRPV6 by binding in the middle of the ion channel's selectivity filter and plugging its pore like a bottle cork. Despite different binding site locations, both inhibitors induce similar conformational changes in the channel resulting in closure of the gate formed by S6 helices bundle crossing. The uncovered molecular mechanisms of TRPV6 inhibition can guide the design of a new generation of clinically useful inhibitors.
Collapse
Affiliation(s)
- Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
15
|
Zhang H, Zhao S, Yu J, Yang W, Liu Z, Zhang L. Medicinal chemistry perspective of TRPM2 channel inhibitors: where we are and where we might be heading? Drug Discov Today 2020; 25:2326-2334. [PMID: 33065292 DOI: 10.1016/j.drudis.2020.09.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/26/2020] [Accepted: 09/30/2020] [Indexed: 01/06/2023]
Abstract
Transient receptor potential melastatin 2 (TRPM2) is a Ca2+- permeable nonselective cation channel that is involved in diverse biological functions as a cellular sensor for oxidative stress and temperature. It has been considered a promising therapeutic target for the treatment of ischemia/reperfusion (IR) injury, inflammation, cancer, and neurodegenerative diseases. Development of highly potent and selective TRPM2 inhibitors and validation of their use in relevant disease models will advance drug discovery. In this review, we describe the molecular structures and gating mechanism of the TRPM2 channel, and offer a comprehensive review of advances in the discovery of TRPM2 inhibitors. Furthermore, we analyze the properties of reported TRPM2 inhibitors with an emphasis on how specific inhibitors targeting this channel could be better developed.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Siqi Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jie Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wei Yang
- Department of Biophysics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
16
|
López-Romero AE, Hernández-Araiza I, Torres-Quiroz F, Tovar-Y-Romo LB, Islas LD, Rosenbaum T. TRP ion channels: Proteins with conformational flexibility. Channels (Austin) 2020; 13:207-226. [PMID: 31184289 PMCID: PMC6602575 DOI: 10.1080/19336950.2019.1626793] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Ion channels display conformational changes in response to binding of their agonists and antagonists. The study of the relationships between the structure and the function of these proteins has witnessed considerable advances in the last two decades using a combination of techniques, which include electrophysiology, optical approaches (i.e. patch clamp fluorometry, incorporation of non-canonic amino acids, etc.), molecular biology (mutations in different regions of ion channels to determine their role in function) and those that have permitted the resolution of their structures in detail (X-ray crystallography and cryo-electron microscopy). The possibility of making correlations among structural components and functional traits in ion channels has allowed for more refined conclusions on how these proteins work at the molecular level. With the cloning and description of the family of Transient Receptor Potential (TRP) channels, our understanding of several sensory-related processes has also greatly moved forward. The response of these proteins to several agonists, their regulation by signaling pathways as well as by protein-protein and lipid-protein interactions and, in some cases, their biophysical characteristics have been studied thoroughly and, recently, with the resolution of their structures, the field has experienced a new boom. This review article focuses on the conformational changes in the pores, concentrating on some members of the TRP family of ion channels (TRPV and TRPA subfamilies) that result in changes in their single-channel conductances, a phenomenon that may lead to fine-tuning the electrical response to a given agonist in a cell.
Collapse
Affiliation(s)
- Ana Elena López-Romero
- a Departamento de Neurociencia Cognitiva, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico , Mexico
| | - Ileana Hernández-Araiza
- a Departamento de Neurociencia Cognitiva, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico , Mexico
| | - Francisco Torres-Quiroz
- b Departamento de Bioquímica y Biología Estructural, División Investigación Básica , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Luis B Tovar-Y-Romo
- c Departamento de Neuropatología Molecular, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - León D Islas
- d Departamento de Fisiología, Facultad de Medicina , Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Tamara Rosenbaum
- a Departamento de Neurociencia Cognitiva, División Neurociencias , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico , Mexico
| |
Collapse
|
17
|
Abstract
Two decades ago a class of ion channels, hitherto unsuspected, was discovered. In mammals these Transient Receptor Potential channels (TRPs) have not only expanded in number (to 26 functional channels) but also expanded the view of our interface with the physical and chemical environment. Some are heat and cold sensors while others monitor endogenous and/or exogenous chemical signals. Some TRP channels monitor osmotic potential, and others measure cell movement, stretching, and fluid flow. Many TRP channels are major players in nociception and integration of pain signals. One member of the vanilloid sub-family of channels is TRPV6. This channel is highly selective for divalent cations, particularly calcium, and plays a part in general whole-body calcium homeostasis, capturing calcium in the gut from the diet. TRPV6 can be greatly elevated in a number of cancers deriving from epithelia and considerable study has been made of its role in the cancer phenotype where calcium control is dysfunctional. This review compiles and updates recent published work on TRPV6 as a promising drug target in a number of cancers including those afflicting breast, ovarian, prostate and pancreatic tissues.
Collapse
Affiliation(s)
- John M. Stewart
- Soricimed Biopharma Inc. 18 Botsford Street, Moncton, NB, Canada, E1C 4W7
| |
Collapse
|
18
|
Hughes TE, Del Rosario JS, Kapoor A, Yazici AT, Yudin Y, Fluck EC, Filizola M, Rohacs T, Moiseenkova-Bell VY. Structure-based characterization of novel TRPV5 inhibitors. eLife 2019; 8:49572. [PMID: 31647410 PMCID: PMC6834369 DOI: 10.7554/elife.49572] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/23/2019] [Indexed: 12/31/2022] Open
Abstract
Transient receptor potential vanilloid 5 (TRPV5) is a highly calcium selective ion channel that acts as the rate-limiting step of calcium reabsorption in the kidney. The lack of potent, specific modulators of TRPV5 has limited the ability to probe the contribution of TRPV5 in disease phenotypes such as hypercalcemia and nephrolithiasis. Here, we performed structure-based virtual screening (SBVS) at a previously identified TRPV5 inhibitor binding site coupled with electrophysiology screening and identified three novel inhibitors of TRPV5, one of which exhibits high affinity, and specificity for TRPV5 over other TRP channels, including its close homologue TRPV6. Cryo-electron microscopy of TRPV5 in the presence of the specific inhibitor and its parent compound revealed novel binding sites for this channel. Structural and functional analysis have allowed us to suggest a mechanism of action for the selective inhibition of TRPV5 and lay the groundwork for rational design of new classes of TRPV5 modulators.
Collapse
Affiliation(s)
- Taylor Et Hughes
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - John Smith Del Rosario
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, United States
| | - Abhijeet Kapoor
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Aysenur Torun Yazici
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, United States
| | - Yevgen Yudin
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, United States
| | - Edwin C Fluck
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, United States
| | - Vera Y Moiseenkova-Bell
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
19
|
Cheng HH, Liang WZ, Kuo CC, Hao LJ, Chou CT, Jan CR. The exploration of effect of terfenadine on Ca 2+ signaling in renal tubular cells. J Recept Signal Transduct Res 2019; 39:73-79. [PMID: 31184240 DOI: 10.1080/10799893.2019.1620777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Terfenadine, an antihistamine used for the treatment of allergic conditions, affected Ca2+-related physiological responses in various models. However, the effect of terfenadine on cytosolic free Ca2+ levels ([Ca2+]i) and its related physiology in renal tubular cells is unknown. This study examined whether terfenadine altered Ca2+ signaling and caused cytotoxicity in Madin-Darby canine kidney (MDCK) renal tubular cells. The Ca2+-sensitive fluorescent dye fura-2 was used to measure [Ca2+]i. Cell viability was measured by the fluorescent reagent 4-[3-[4-lodophenyl]-2-4(4-nitrophenyl)-2H-5-tetrazolio-1,3-benzene disulfonate] water soluble tetrazolium-1 (WST-1) assay. Terfenadine at concentrations of 100-1000 μM induced [Ca2+]i rises concentration dependently. The response was reduced by approximately 35% by removing extracellular Ca2+. In Ca2+-free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor 2,5-di-tert-butylhydroquinone (BHQ) partly inhibited terfenadine-evoked [Ca2+]i rises. Conversely, treatment with terfenadine abolished BHQ-evoked [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 inhibited 95% of terfenadine-induced Ca2+ release. Terfenadine-induced Ca2+ entry was supported by Mn2+-caused quenching of fura-2 fluorescence. Terfenadine-induced Ca2+ entry was partly inhibited by an activator of protein kinase C (PKC), phorbol 12-myristate 13 acetate (PMA) and by three modulators of store-operated Ca2+ channels (nifedipine, econazole, and SKF96365). Terfenadine at 200-300 μM decreased cell viability, which was not reversed by pretreatment with the Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM). Together, in MDCK cells, terfenadine induced [Ca2+]i rises by evoking PLC-dependent Ca2+ release from the endoplasmic reticulum and Ca2+ entry via PKC-sensitive store-operated Ca2+ entry. Furthermore, terfenadine caused cell death that was not triggered by preceding [Ca2+]i rises.
Collapse
Affiliation(s)
- He-Hsiung Cheng
- a Department of Medicine , Chang Bing Show Chwan Memorial Hospital , Changhua , Taiwan
| | - Wei-Zhe Liang
- b Department of Medical Education and Research , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan.,c Department of Pharmacy , Tajen University , Pingtung , Taiwan
| | - Chun-Chi Kuo
- d Department of Nursing , Tzu Hui Institute of Technology , Pingtung , Taiwan
| | - Lyh-Jyh Hao
- e Department of Metabolism , Kaohsiung Veterans General Hospital Tainan Branch , Tainan , Taiwan
| | - Chiang-Ting Chou
- f Department of Nursing, Division of Basic Medical Sciences , Chang Gung University of Science and Technology , Chia-Yi , Taiwan
| | - Chung-Ren Jan
- b Department of Medical Education and Research , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan
| |
Collapse
|
20
|
Haustrate A, Hantute-Ghesquier A, Prevarskaya N, Lehen’kyi V. RETRACTED: TRPV6 calcium channel regulation, downstream pathways, and therapeutic targeting in cancer. Cell Calcium 2019; 80:117-124. [DOI: 10.1016/j.ceca.2019.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 11/30/2022]
|
21
|
Hughes TET, Lodowski DT, Huynh KW, Yazici A, Del Rosario J, Kapoor A, Basak S, Samanta A, Han X, Chakrapani S, Zhou ZH, Filizola M, Rohacs T, Han S, Moiseenkova-Bell VY. Structural basis of TRPV5 channel inhibition by econazole revealed by cryo-EM. Nat Struct Mol Biol 2018; 25:53-60. [PMID: 29323279 PMCID: PMC5951624 DOI: 10.1038/s41594-017-0009-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/13/2017] [Indexed: 12/18/2022]
Abstract
The transient receptor potential vanilloid 5 (TRPV5) channel is a member of the transient receptor potential (TRP) channel family, which is highly selective for Ca2+, that is present primarily at the apical membrane of distal tubule epithelial cells in the kidney and plays a key role in Ca2+ reabsorption. Here we present the structure of the full-length rabbit TRPV5 channel as determined using cryo-EM in complex with its inhibitor econazole. This structure reveals that econazole resides in a hydrophobic pocket analogous to that occupied by phosphatidylinositides and vanilloids in TRPV1, thus suggesting conserved mechanisms for ligand recognition and lipid binding among TRPV channels. The econazole-bound TRPV5 structure adopts a closed conformation with a distinct lower gate that occludes Ca2+ permeation through the channel. Structural comparisons between TRPV5 and other TRPV channels, complemented with molecular dynamics (MD) simulations of the econazole-bound TRPV5 structure, allowed us to gain mechanistic insight into TRPV5 channel inhibition by small molecules.
Collapse
Affiliation(s)
- Taylor E T Hughes
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David T Lodowski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kevin W Huynh
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aysenur Yazici
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - John Del Rosario
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Abhijeet Kapoor
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sandip Basak
- Department of Physiology and Biophysics School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Amrita Samanta
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Physiology and Biophysics School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Xu Han
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sudha Chakrapani
- Department of Physiology and Biophysics School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Z Hong Zhou
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Seungil Han
- Pfizer Research and Development, Groton, CT, USA
| | - Vera Y Moiseenkova-Bell
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Physiology and Biophysics School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
A Gate Hinge Controls the Epithelial Calcium Channel TRPV5. Sci Rep 2017; 7:45489. [PMID: 28374795 PMCID: PMC5379628 DOI: 10.1038/srep45489] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/27/2017] [Indexed: 12/03/2022] Open
Abstract
TRPV5 is unique within the large TRP channel family for displaying a high Ca2+ selectivity together with Ca2+-dependent inactivation. Our study aims to uncover novel insights into channel gating through in-depth structure-function analysis. We identify an exceptional tryptophan (W583) at the terminus of the intracellular pore that is unique for TRPV5 (and TRPV6). A combination of site-directed mutagenesis, biochemical and electrophysiological analysis, together with homology modeling, demonstrates that W583 is part of the gate for Ca2+ permeation. The W583 mutants show increased cell death due to profoundly enhanced Ca2+ influx, resulting from altered channel function. A glycine residue above W583 might act as flexible linker to rearrange the tryptophan gate. Furthermore, we hypothesize functional crosstalk between the pore region and carboxy terminus, involved in Ca2+-calmodulin-mediated inactivation. This study proposes a unique channel gating mechanism and delivers detailed molecular insight into the Ca2+ permeation pathway that can be extrapolated to other Ca2+-selective channels.
Collapse
|
23
|
Mohammed SG, Arjona FJ, Latta F, Bindels RJM, Roepman R, Hoenderop JGJ. Fluid shear stress increases transepithelial transport of Ca
2+
in ciliated distal convoluted and connecting tubule cells. FASEB J 2017; 31:1796-1806. [DOI: 10.1096/fj.201600687rrr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 01/03/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Sami G. Mohammed
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Francisco J. Arjona
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Femke Latta
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - René J. M. Bindels
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Ronald Roepman
- Department of Human GeneticsRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Joost G. J. Hoenderop
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
24
|
Lepannetier S, Zanou N, Yerna X, Emeriau N, Dufour I, Masquelier J, Muccioli G, Tajeddine N, Gailly P. Sphingosine-1-phosphate-activated TRPC1 channel controls chemotaxis of glioblastoma cells. Cell Calcium 2016; 60:373-383. [PMID: 27638096 DOI: 10.1016/j.ceca.2016.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 01/21/2023]
Abstract
TRP channels are involved in the control of a broad range of cellular functions such as cell proliferation and motility. We investigated the gating mechanism of TRPC1 channel and its role in U251 glioblastoma cells migration in response to chemotaxis by platelet-derived growth factor (PDGF). PDGF induced an influx of Ca2+ that was partially inhibited after pretreatment of the cells with SKI-II, a specific inhibitor of sphingosine kinase producing sphingosine-1-P (S1P). S1P by itself also induced an entry of Ca2+. Interestingly, PDGF- and S1P-induced entries of Ca2+ were lost in siRNA-TRPC1 treated cells. PDGF-induced chemotaxis of U251 cells was dramatically inhibited in cells treated with SKI-II. This effect was almost completely rescued by addition of synthetic S1P. Chemotaxis was also completely lost in siRNA-TRPC1 treated cells and interestingly, the rescue of migration of cells treated with SKI-II by S1P was dependent on the expression of TRPC1. Immunocytochemistry revealed that, in response to PDGF, TRPC1 translocated from inside of the cell to the front of migration (lamellipodes). This effect seemed PI3K dependent as it was inhibited by cell pre-treatment with LY294002, a PI3-kinase inhibitor. Our results thus identify S1P as a potential activator of TRPC1, a channel involved in cell orientation during chemotaxis by PDGF.
Collapse
Affiliation(s)
- Sophie Lepannetier
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Nadège Zanou
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Xavier Yerna
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Noémie Emeriau
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Inès Dufour
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Julien Masquelier
- Université catholique de Louvain, Louvain Drug Research Institute, av. Mounier 72, box B1.72.01, 1200 Brussels, Belgium
| | - Giulio Muccioli
- Université catholique de Louvain, Louvain Drug Research Institute, av. Mounier 72, box B1.72.01, 1200 Brussels, Belgium
| | - Nicolas Tajeddine
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Philippe Gailly
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium.
| |
Collapse
|
25
|
Leunissen EHP, Blanchard MG, Sheedfar F, Lavrijsen M, van der Wijst J, Bindels RJM, Hoenderop JGJ. Urinary β-galactosidase stimulates Ca2+ transport by stabilizing TRPV5 at the plasma membrane. Glycobiology 2016; 26:472-81. [PMID: 26747426 DOI: 10.1093/glycob/cwv172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 12/28/2015] [Indexed: 01/26/2023] Open
Abstract
Transcellular Ca(2+)transport in the late distal convoluted tubule and connecting tubule (DCT2/CNT) of the kidney is a finely controlled process mediated by the transient receptor potential vanilloid type 5 (TRPV5) channel. A complex-type-N-glycan bound at the extracellular residue Asn358 of TRPV5 through post-translational glycosylation has been postulated to regulate the activity of TRPV5 channels. Using in vitro Ca(2+)transport assays, immunoblot analysis, immunohistochemistry, patch clamp electrophysiology and total internal reflection fluorescence microscopy, it is demonstrated that the glycosidase β-galactosidase (β-gal), an enzyme that hydrolyzes galactose, stimulates TRPV5 channel activity. However, the activity of the non-glycosylated TRPV(N358Q)mutant was not altered in the presence of β-gal, showing that the stimulation is dependent on the presence of the TRPV5N-glycan. In addition, β-gal was found to stimulate transcellular Ca(2+)transport in isolated mouse primary DCT2/CNT cells. β-gal expression was detected in the apical membrane of the proximal tubules, and the protein was found in mouse urine. In summary, β-gal is present in the pro-urine from where it is thought to stimulate TRPV5 activity.
Collapse
Affiliation(s)
- Elizabeth H P Leunissen
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Maxime G Blanchard
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Fareeba Sheedfar
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Marla Lavrijsen
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Jenny van der Wijst
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - René J M Bindels
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6500 HB, The Netherlands
| |
Collapse
|
26
|
The involvement of mitochondrial apoptotic pathway in eugenol-induced cell death in human glioblastoma cells. Toxicol Lett 2015; 232:122-32. [DOI: 10.1016/j.toxlet.2014.10.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/30/2014] [Accepted: 10/15/2014] [Indexed: 12/27/2022]
|
27
|
Ortega P, Custódio MR, Zanotto FP. Characterization of cadmium plasma membrane transport in gills of a mangrove crab Ucides cordatus. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 157:21-29. [PMID: 25456216 DOI: 10.1016/j.aquatox.2014.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/16/2014] [Accepted: 09/19/2014] [Indexed: 06/04/2023]
Abstract
Membrane pathway for intracellular cadmium (Cd(2+)) accumulation is not fully elucidated in many organisms and has not been studied in crab gill cells. To characterize membrane Cd(2+) transport of anterior and posterior gill cells of Ucides cordatus, a hypo-hyper-regulating crab, a change in intracellular Cd(2+) concentration under various experimental conditions was examined by using FluoZin, a fluorescent probe. The membrane Cd(2+) transport was estimated by the augmentation of FluoZin fluorescence induced by extracellular application of CdCl2 and different inhibitors. Addition of extracellular calcium (Ca(2+)) to the cells affected little the fluorescence of FluoZin, confirming that Cd(2+) was the main ion increasing intracellular fluorescence. Ca(2+) channels blockers (nimodipine and verapamil) decreased Cd(2+) influx as well as vanadate, a Ca(2+)-ATPase blocker. Chelating intracellular Ca(2+) (BAPTA) decreased Cd(2+) influx in gill cells, while increasing intracellular Ca(2+) (caffeine) augmented Cd influx. Cd(2+) and ATP added at different temporal conditions were not effective at increasing intracellular Cd(2+) accumulation. Ouabain (Na(+)/K(+)-ATPase inhibitor) increased Cd(2+) influx probably through a change in intracellular Na and/or a change in cell membrane potential. Routes of Cd(2+) influx, a non-essential metal, through the gill cell plasma membrane of crabs are suggested.
Collapse
Affiliation(s)
- P Ortega
- Instituto de Biociências, Departamento de Fisiologia, Universidade de São Paulo, Rua do Matão, Travessa 14, #101, São Paulo 05508-900, SP, Brazil
| | - M R Custódio
- Instituto de Biociências, Departamento de Fisiologia, Universidade de São Paulo, Rua do Matão, Travessa 14, #101, São Paulo 05508-900, SP, Brazil
| | - F P Zanotto
- Instituto de Biociências, Departamento de Fisiologia, Universidade de São Paulo, Rua do Matão, Travessa 14, #101, São Paulo 05508-900, SP, Brazil; Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Três de Maio 100, São Paulo 04044-020, Brazil.
| |
Collapse
|
28
|
Liu W, Liu MR, Zhang H. Novel calcium ion channels TRPV5 and TRPV6 and gastrointestinal tumors. Shijie Huaren Xiaohua Zazhi 2014; 22:1966-1971. [DOI: 10.11569/wcjd.v22.i14.1966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential cation channel, subfamily V, member 5 (TRPV5) and TRPV6 are the subfamily members of the transient receptor potential (TRP), representing new highly selective Ca2+ membrane transport channels, which are mainly responsible for active transport of Ca2+ across the cell membrane and participate in regulation of many physiological activities in the body. This paper discusses the structures and electrophysiological properties of TRPV5 and TRPV6, their related factors and their relationship with gastrointestinal tumors, highlighting the role of TRPV5 and TRPV6 in the formation of gastrointestinal tumors.
Collapse
|
29
|
Abstract
TRPV5 is one of the two channels in the TRPV family that exhibit high selectivity to Ca(2+) ions. TRPV5 mediates Ca(2+) influx into cells as the first step to transport Ca(2+) across epithelia. The specialized distribution in the distal tubule of the kidney positions TRPV5 as a key player in Ca(2+) reabsorption. The responsiveness in expression and/or activity of TRPV5 to hormones such as 1,25-dihydroxyvitamin D3, parathyroid hormone, estrogen, and testosterone makes TRPV5 suitable for its role in the fine-tuning of Ca(2+) reabsorption. This role is further optimized by the modulation of TRPV5 trafficking and activity via its binding partners; co-expressed proteins; tubular factors such as calbindin-D28k, calmodulin, klotho, uromodulin, and plasmin; extracellular and intracellular factors such as proton, Mg(2+), Ca(2+), and phosphatidylinositol-4,5-bisphosphate; and fluid flow. These regulations allow TRPV5 to adjust its overall activity in response to the body's demand for Ca(2+) and to prevent kidney stone formation. A point mutation in mouse Trpv5 gene leads to hypercalciuria similar to Trpv5 knockout mice, suggesting a possible role of TRPV5 in hypercalciuric disorders in humans. In addition, the single nucleotide polymorphisms in Trpv5 gene prevalently present in African descents may contribute to the efficient renal Ca(2+) reabsorption among African descendants. TRPV5 represents a potential therapeutic target for disorders with altered Ca(2+) homeostasis.
Collapse
Affiliation(s)
- Tao Na
- Cell Collection and Research Center, Institute for Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
| | | |
Collapse
|
30
|
Abstract
Drug-like compounds that exert biological activity towards TRP channels are either being used as cell biological tools or further developed into pharmacological lead structures aiming at therapeutic use in diseased states. Although drug-likeliness is not easy to predict, common rules include a relatively low molecular weight, physicochemical constraints, and the absence of known reactive or otherwise toxic groups. Small molecules that exert a biological activity to block, activate, or modulate TRP channels are intensely sought. Such tool compounds may be useful to assign native currents to a certain TRP channel and to validate the channel as a candidate target for future pharmacological intervention. Depending on the TRP channel isotype, these activities have reached different levels, with only few TRP channels modulators already being clinically tested in humans, whereas other compounds only underwent a preliminary validation. For some TRP channels, reliable low molecular weight inhibitors are not yet available. Hence, further efforts need to be undertaken in order to explore the physiological impact and possible therapeutic potential of TRP channel targeting with drug-like compounds.
Collapse
Affiliation(s)
- Michael Schaefer
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Härtelstraße 16-18, 04107, Leipzig, Germany,
| |
Collapse
|
31
|
Mechanisms of calcium absorption by anterior and posterior segments of the intestinal tract of juvenile lake sturgeon. Comp Biochem Physiol A Mol Integr Physiol 2013; 166:293-301. [DOI: 10.1016/j.cbpa.2013.06.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/27/2013] [Accepted: 06/27/2013] [Indexed: 11/23/2022]
|
32
|
Derler I, Fritsch R, Schindl R, Romanin C. CRAC inhibitors: identification and potential. Expert Opin Drug Discov 2013; 3:787-800. [PMID: 23496221 DOI: 10.1517/17460441.3.7.787] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Ca(2+) release-activated Ca(2+) (CRAC) channels, a subfamily of store-operated channels, play an essential role in various diseases such as immune disorders and allergic responses. OBJECTIVE The successful treatment of these diseases requires the identification of specific inhibitors. So far, a variety of chemical compounds blocking CRAC have been identified; however, they have all turned out to be less specific. Recently two proteins, STIM1 and ORAI1, have been identified as the essential components that fully reconstitute CRAC currents with a similar biophysical fingerprint. METHOD These two proteins and their activation process represent direct targets for the application of specific CRAC inhibitors. RESULTS/CONCLUSION For drug development, fluorescence microscopy adaptable for high-throughput screening will provide a powerful assay to mechanistically identify potential CRAC inhibitors that act on various stages within the STIM1/ORAI1 activation pathway visualized by fluorescent-tagged proteins.
Collapse
Affiliation(s)
- Isabella Derler
- University of Linz, Institute of Biophysics, A-4040 Linz, Austria +43 732 2468 9272 ; +43 732 2468 9280 ; ;
| | | | | | | |
Collapse
|
33
|
Pecze L, Winter Z, Jósvay K, Ötvös F, Kolozsi C, Vizler C, Budai D, Letoha T, Dombi G, Szakonyi G, Oláh Z. Divalent heavy metal cations block the TRPV1 Ca(2+) channel. Biol Trace Elem Res 2013; 151:451-61. [PMID: 23264033 PMCID: PMC3566393 DOI: 10.1007/s12011-012-9570-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 12/03/2012] [Indexed: 01/17/2023]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a non-selective cation channel involved in pain sensation and in a wide range of non-pain-related physiological and pathological conditions. The aim of the present study was to explore the effects of selected heavy metal cations on the function of TRPV1. The cations ranked in the following sequence of pore-blocking activity: Co(2+) [half-maximal inhibitory concentration (IC(50)) = 13 μM] > Cd(2+) (I (50) = 38 μM) > Ni(2+) (IC(50) = 62 μM) > Cu(2+) (IC(50) = 200 μM). Zn(2+) proved to be a weak (IC(50) = 27 μM) and only partial inhibitor of the channel function, whereas Mg(2+), Mn(2+) and La(3+) did not exhibit any substantial effect. Co(2+), the most potent channel blocker, was able not only to compete with Ca(2+) but also to pass with it through the open channel of TRPV1. In response to heat activation or vanilloid treatment, Co(2+) accumulation was verified in TRPV1-transfected cell lines and in the TRPV1+ dorsal root ganglion neurons. The inhibitory effect was also demonstrated in vivo. Co(2+) applied together with vanilloid agonists attenuated the nocifensive eye wipe response in mice. Different rat TRPV1 pore point mutants (Y627W, N628W, D646N and E651W) were created that can validate the binding site of previously used channel blockers in agonist-evoked (45)Ca(2+) influx assays in cells expressing TRPV1. The IC(50) of Co(2+) on these point mutants were determined to be reasonably comparable to those on the wild type, which suggests that divalent cations passing through the TRPV1 channel use the same negatively charged amino acids as Ca(2+).
Collapse
Affiliation(s)
- László Pecze
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Zoltán Winter
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Katalin Jósvay
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Ferenc Ötvös
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
- Greenformatix Nonprofit Ltd., Szeged, Hungary
| | - Csongor Kolozsi
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Csaba Vizler
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | | | - Tamás Letoha
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Pharmacoidea Ltd., Szeged, Hungary
| | - György Dombi
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Gerda Szakonyi
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Zoltán Oláh
- Institute of Pharmaceutical Analysis, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Acheuron Hungary Ltd., Szeged, Hungary
| |
Collapse
|
34
|
Guo Z, Grimm C, Becker L, Ricci AJ, Heller S. A novel ion channel formed by interaction of TRPML3 with TRPV5. PLoS One 2013; 8:e58174. [PMID: 23469151 PMCID: PMC3585263 DOI: 10.1371/journal.pone.0058174] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/31/2013] [Indexed: 01/15/2023] Open
Abstract
TRPML3 and TRPV5 are members of the mucolipin (TRPML) and TRPV subfamilies of transient receptor potential (TRP) cation channels. Based on sequence similarities of the pore forming regions and on structure-function evidence, we hypothesized that the pore forming domains of TRPML and TRPV5/TRPV6 channels have similarities that indicate possible functional interactions between these TRP channel subfamilies. Here we show that TRPML3 and TRPV5 associate to form a novel heteromeric ion channel. This novel conductance is detectable under conditions that do not activate either TRPML3 or TRPV5. It has pharmacological similarity with TRPML3 and requires functional TRPML3 as well as functional TRPV5. Single channel analyses revealed that TRPML3 and TRPV5 heteromers have different features than the respective homomers, and furthermore, that they occur in potentially distinct stoichiometric configurations. Based on overlapping expression of TRPML3 and TRPV5 in the kidney and the inner ear, we propose that TRPML3 and TRPV5 heteromers could have a biological function in these organs.
Collapse
Affiliation(s)
- Zhaohua Guo
- Departments of Otolaryngology – HNS and Molecular & Cellular Physiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Christian Grimm
- Department of Pharmacy – Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig-Maximilians-Universität, München, Germany
| | - Lars Becker
- Departments of Otolaryngology – HNS and Molecular & Cellular Physiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Anthony J. Ricci
- Departments of Otolaryngology – HNS and Molecular & Cellular Physiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Stefan Heller
- Departments of Otolaryngology – HNS and Molecular & Cellular Physiology, Stanford University School of Medicine, Palo Alto, California, United States of America
- * E-mail:
| |
Collapse
|
35
|
Ding J, Zhang JR, Wang Y, Li CL, Lu D, Guan SM, Chen J. Effects of a non-selective TRPC channel blocker, SKF-96365, on melittin-induced spontaneous persistent nociception and inflammatory pain hypersensitivity. Neurosci Bull 2012; 28:173-81. [PMID: 22466128 DOI: 10.1007/s12264-012-1213-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
OBJECTIVE Melittin is the main peptide in bee venom and causes both persistent spontaneous nociception and pain hypersensitivity. Our recent studies indicated that both transient receptor potential (TRP) vanilloid receptor 1 (TRPV1) and canonical TRPs (TRPCs) are involved in mediating the melittin-induced activation of different subpopulations of primary nociceptive cells. Here, we further determined whether TRPC channels are involved in melittin-induced inflammatory nociceptive responses in behavioral assays. METHODS The anti-nociceptive and anti-hyperalgesic effects of localized peripheral administration of three doses of the non-selective TRPC antagonist, SKF-96365 (1-{β-[3-(4-methoxyphenyl)propoxy]-4-methoxyphenyl}-1H-imidazole hydrochloride), were evaluated in melittin tests. Pain-related behaviors were rated by counting the number of paw flinches, and measuring paw withdrawal thermal latency (s) and paw withdrawl mechanical threshold (g), over a 1-h time-course. RESULTS Localized peripheral SKF-96365 given before melittin prevented, and given after melittin significantly suppressed, the melittin-evoked persistent spontaneous nociception. Pre-blockade and post-suppression of activation of primary nociceptive activity resulted in decreased hypersensitivity to both thermal and mechanical stimuli applied to the primary injury site of the ipsilateral hindpaw, despite dose-effect differences between thermal and mechanical hyperalgesia. However, local administration of SKF-96365 into the contralateral hindpaw had no significant effect on any pain-associated behaviors. In addition, SKF-96365 had no effect on baseline threshold for either thermal or mechanical sensitivity under normal conditions. CONCLUSION Besides TRPV1, SKF-96365-sensitive TRPC channels might also be involved in the pathophysiological processing of melittin-induced inflammatory pain and hypersensitivity. Therapeutically, SKF-96365 is equally effective in preventing primary thermal and mechanical hyperalgesia as well as persistent spontaneous nociception. However, this drug is likely to be more effective in the relief of thermal hyperalgesia than mechanical hyperalgesia when applied 5 min after establishment of primary afferent activation.
Collapse
Affiliation(s)
- Jing Ding
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Derler I, Schindl R, Fritsch R, Heftberger P, Riedl MC, Begg M, House D, Romanin C. The action of selective CRAC channel blockers is affected by the Orai pore geometry. Cell Calcium 2012; 53:139-51. [PMID: 23218667 PMCID: PMC3580291 DOI: 10.1016/j.ceca.2012.11.005] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/02/2012] [Accepted: 11/03/2012] [Indexed: 11/10/2022]
Abstract
As the molecular composition of calcium-release activated calcium (CRAC) channels has been unknown for two decades, elucidation of selective inhibitors has been considerably hampered. By the identification of the two key components of CRAC channels, STIM1 and Orai1 have emerged as promising targets for CRAC blockers. The aim of this study was to thoroughly characterize the effects of two selective CRAC channel blockers on currents derived from STIM1/Orai heterologoulsy expressed in HEK293 cells. The novel compounds GSK-7975A and GSK-5503A were tested for effects on STIM1 mediated Orai1 or Orai3 currents by whole-cell patch-clamp recordings and for the effects on STIM1 oligomerisation or STIM1/Orai coupling by FRET microscopy. To investigate their site of action, inhibitory effects of these molecules were explored using Orai pore mutants. The GSK blockers inhibited Orai1 and Orai3 currents with an IC50 of approximately 4 μM and exhibited a substantially slower rate of onset than the typical pore blocker La3+, together with almost no current recovery upon wash-out over 4 min. For the less Ca2+-selective Orai1 E106D pore mutant, ICRAC inhibition was significantly reduced. FRET experiments indicated that neither STIM1–STIM1 oligomerization nor STIM1–Orai1 coupling was affected by these compounds. These CRAC channel blockers are acting downstream of STIM1 oligomerization and STIM1/Orai1 interaction, potentially via an allosteric effect on the selectivity filter of Orai. The elucidation of these CRAC current blockers represents a significant step toward the identification of CRAC channel-selective drug compounds.
Collapse
Affiliation(s)
- Isabella Derler
- Institute of Biophysics, University of Linz, 4040 Linz, Austria
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Kovacs G, Montalbetti N, Simonin A, Danko T, Balazs B, Zsembery A, Hediger MA. Inhibition of the human epithelial calcium channel TRPV6 by 2-aminoethoxydiphenyl borate (2-APB). Cell Calcium 2012; 52:468-80. [PMID: 23040501 DOI: 10.1016/j.ceca.2012.08.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 08/25/2012] [Accepted: 08/27/2012] [Indexed: 12/30/2022]
Abstract
TRPV6, a highly calcium-selective member of the transient receptor potential (TRP) channel superfamily, is a major pathway for calcium absorption in the fetal and adult body. It is expressed abundantly in the duodenum, the placenta and exocrine tissues. TRVP6 was postulated to contribute to store-operated calcium channel (SOC) activity in certain cell types such as exocrine cells. In this study, we tested 2-APB, a widely used SOC inhibitor on human TRPV6 (hTRPV6) activity using fluorescence imaging, patch clamp and radioactive tracer techniques in transiently and stably transfected HEK293 cells. We found that the basal calcium and cadmium influx was higher in HEK293 cells transfected with hTRPV6 than in non-transfected cells. 2-APB inhibited hTRPV6 activity in both transient and stably transfected cells. This effect was slightly sensitive toward extracellular calcium. The extracellular sodium concentration did not affect the inhibition of hTRPV6 by 2-APB. However, N-methyl-d-glucamine significantly diminished the inhibitory effect of 2-APB presumably through direct interaction with this compound. Furthermore, 2-APB inhibited the activity of TRPV6 orthologs but not human TRPV5. 2-APB may serve as a parental compound for the development of therapeutic strategies specifically targeting the hTRPV6 calcium channel.
Collapse
Affiliation(s)
- Gergely Kovacs
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
38
|
Cheng HH, Lu YC, Lu T, Cheng JS, Mar GY, Fang YC, Chai KL, Jan CR. Effect of methoxychlor on Ca(2+) movement and viability in MDCK renal tubular cells. Basic Clin Pharmacol Toxicol 2012; 111:224-31. [PMID: 22458924 DOI: 10.1111/j.1742-7843.2012.00887.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 03/22/2012] [Indexed: 12/13/2022]
Abstract
The effect of the insecticide methoxychlor on the physiology of renal tubular cells is unknown. This study aimed to explore the effect of methoxychlor on cytosolic Ca(2+) concentrations ([Ca(2+) ](i) ) in MDCK renal tubular cells using the Ca(2+) -sensitive fluorescent dye fura-2. Methoxychlor at 5-20 μM increased [Ca(2+) ](i) in a concentration-dependent manner. The signal was reduced by 80% by removing extracellular Ca(2+) . Methoxychlor-induced Ca(2+) entry was not affected by nifedipine and SK&F96365 but was inhibited by econazole and protein kinase C modulators. In Ca(2+) -free medium, treatment with the endoplasmic reticulum Ca(2+) pump inhibitor thapsigargin or 2,5-di-tert-butylhydroquinone (BHQ) partly inhibited methoxychlor-induced [Ca(2+) ](i) rise. Incubation with methoxychlor also inhibited thapsigargin- or BHQ-induced [Ca(2+) ](i) rise. Inhibition of phospholipase C with U73122 nearly abolished methoxychlor-induced [Ca(2+) ](i) rise. At 5-15 μM, methoxychlor slightly increased cell viability, whereas at 20 μM, it decreased viability. The cytotoxic effect of methoxychlor was not reversed by chelating cytosolic Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N,N-tetraacetic acid/AM (BAPTA/AM). Annexin V-FITC data suggest that 10 μM methoxychlor inhibited apoptosis, while 20 μM methoxychlor enhanced apoptosis. Methoxychlor (10 and 20 μM) increased the production of reactive oxygen species. Together, in renal tubular cells, methoxychlor induced [Ca(2+) ](i) rise by inducing phospholipase C-dependent Ca(2+) release from multiple stores and Ca(2+) entry via protein kinase C- and econazole-sensitive channels. Methoxychlor slightly enhanced or inhibited cell viability in a concentration-dependent, Ca(2+) -independent manner. Methoxychlor induced cell death that may involve apoptosis via mitochondrial pathways.
Collapse
Affiliation(s)
- He-Hsiung Cheng
- Department of Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua County, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Cehak A, Wilkens MR, Guschlbauer M, Mrochen N, Schröder B, Feige K, Breves G. In vitro studies on intestinal calcium and phosphate transport in horses. Comp Biochem Physiol A Mol Integr Physiol 2012; 161:259-64. [DOI: 10.1016/j.cbpa.2011.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 11/05/2011] [Accepted: 11/06/2011] [Indexed: 12/14/2022]
|
40
|
Lewis R, Asplin KE, Bruce G, Dart C, Mobasheri A, Barrett-Jolley R. The role of the membrane potential in chondrocyte volume regulation. J Cell Physiol 2011; 226:2979-86. [PMID: 21328349 PMCID: PMC3229839 DOI: 10.1002/jcp.22646] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 01/05/2011] [Indexed: 02/06/2023]
Abstract
Many cell types have significant negative resting membrane potentials (RMPs) resulting from the activity of potassium-selective and chloride-selective ion channels. In excitable cells, such as neurones, rapid changes in membrane permeability underlie the generation of action potentials. Chondrocytes have less negative RMPs and the role of the RMP is not clear. Here we examine the basis of the chondrocyte RMP and possible physiological benefits. We demonstrate that maintenance of the chondrocyte RMP involves gadolinium-sensitive cation channels. Pharmacological inhibition of these channels causes the RMP to become more negative (100 µM gadolinium: ΔV(m) = -30 ± 4 mV). Analysis of the gadolinium-sensitive conductance reveals a high permeability to calcium ions (PCa/PNa ≈80) with little selectivity between monovalent ions; similar to that reported elsewhere for TRPV5. Detection of TRPV5 by PCR and immunohistochemistry and the sensitivity of the RMP to the TRPV5 inhibitor econazole (ΔV(m) = -18 ± 3 mV) suggests that the RMP may be, in part, controlled by TRPV5. We investigated the physiological advantage of the relatively positive RMP using a mathematical model in which membrane stretch activates potassium channels allowing potassium efflux to oppose osmotic water uptake. At very negative RMP potassium efflux is negligible, but at more positive RMP it is sufficient to limit volume increase. In support of our model, cells clamped at -80 mV and challenged with a reduced osmotic potential swelled approximately twice as much as cells at +10 mV. The positive RMP may be a protective adaptation that allows chondrocytes to respond to the dramatic osmotic changes, with minimal changes in cell volume.
Collapse
Affiliation(s)
- Rebecca Lewis
- Department of Musculoskeletal Biology, Institute of Aging and Chronic Disease, Faculty of Health and Life Sciences, University of LiverpoolLiverpool, UK
| | - Katie E Asplin
- Musculoskeletal Research Group, Division of Veterinary Medicine, Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of NottinghamLoughborough, UK
| | - Gareth Bruce
- Institute of Membrane and Systems Biology, University of LeedsLeeds, UK
| | - Caroline Dart
- Institute of Integrative Biology, Faculty of Health and Life Sciences, University of LiverpoolLiverpool, UK
| | - Ali Mobasheri
- Musculoskeletal Research Group, Division of Veterinary Medicine, Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of NottinghamLoughborough, UK
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Institute of Aging and Chronic Disease, Faculty of Health and Life Sciences, University of LiverpoolLiverpool, UK
| |
Collapse
|
41
|
Yan P, Li T, Bo M, Die L, Xing L. Inhibition of bone resorption by econazole in rat osteoclast-like cells through suppressing TRPV5. Arch Pharm Res 2011; 34:1007-13. [PMID: 21725822 DOI: 10.1007/s12272-011-0618-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/27/2010] [Accepted: 11/14/2010] [Indexed: 11/30/2022]
Abstract
Osteoclasts are primary bone resorption cells and intervention in osteoclast activation is considered an effective therapeutic approach to treatment of bone diseases involving osteoclasts. TRPV5 was detected in osteoclasts and it has been thought to take part in the transportation of the degraded calcium in the resorption lacuna, which is essential for bone resorption. The aim of the present study was to examine the effects of a modulator of calcium dynamics, econazole, on the expression of TRPV5 and bone resorption activity in rat osteoclast-like cells (OLCs). OLCs were obtained by co-culturing rat bone marrow cells with osteoblasts and then culturing with different concentrations of econazole (0.01, 0.1, 1.0, 10.0 μmol/L). Cell counting and staining protocols were used to determine whether econazole influenced the survival of OLCs. Expression of TRPV5 in response to econazole treatment was assessed by western blotting. Bone resorption activity of OLCs was determined by measuring the resorption area of dentin slices with a microscope and a digital image analysis system. Additionally, Ca(2+) inside OLCs was tested. We found that econazole inhibited expression of TRPV5 in a dose dependent manner while it had no influence on the survival of OLCs and it therefore inhibited bone resorption activity in rat OLCs. Ca(2+) inside OLCs increased, suggesting a limited compensatory mechanism to make up for inhibition of TRPV5 effects.
Collapse
Affiliation(s)
- Peng Yan
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, China
| | | | | | | | | |
Collapse
|
42
|
Hofmeister MV, Füchtbauer EM, Fenton RA, Praetorius J. The TRPV5 promoter as a tool for generation of transgenic mouse models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:277-86. [PMID: 21290301 DOI: 10.1007/978-94-007-0265-3_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transient receptor potential vanilloid 5 (TRPV5) is a Ca(2+) channel, which is expressed in renal late distal convoluted tubules (DCT2s) and connecting tubules (CNTs). These tubules play a major role in hormone controlled renal Ca(2+) reabsorption, and thereby in body Ca(2+) homeostasis, as well as urinary excretion of other electrolytes, including Na(+) and K(+). DCT2 and CNT are difficult to distinguish from the surrounding structures and thereby to study by direct functional methods. We developed a transgenic mouse model expressing enhanced green fluorescent protein (EGFP) driven by the TRPV5 promoter to identify these specific tubules. Expression of EGFP in the DCT2 and CNT allows the isolation of pure DCT2 and CNT populations for proteomic and physiological analyses. The TRPV5 promoter is also useful for generating conditional knockout mouse models in a cell-specific manner. TRPV5 promoter driven Cre recombinase expression will be useful for inducing DCT2 and CNT specific gene silencing of various channels, pumps, carriers, and receptors. In this chapter, we describe the strategy for developing transgenic mouse lines involving the TRPV5 promoter, provide a description of extensive validation of these mouse lines, and discuss possible uses and limitations.
Collapse
Affiliation(s)
- Marlene Vind Hofmeister
- Department of Anatomy, Water and Salt Research Center, Aarhus University, DK-8000 Aarhus, Denmark.
| | | | | | | |
Collapse
|
43
|
Kennedy BG, Torabi AJ, Kurzawa R, Echtenkamp SF, Mangini NJ. Expression of transient receptor potential vanilloid channels TRPV5 and TRPV6 in retinal pigment epithelium. Mol Vis 2010; 16:665-75. [PMID: 20405023 PMCID: PMC2855730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 04/07/2010] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Hydration and ionic composition of the subretinal space (SRS) is modulated by the retinal pigment epithelium (RPE). In particular calcium concentration (Ca(2+)) in the SRS varies with light exposure, and although this change is regulated by RPE transport activity, the specific transport proteins involved have yet to be defined. Two members of the transient receptor potential vanilloid family, TRPV5 and TRPV6, are calcium selective ion channels and are known to be expressed in calcium-transporting epithelial tissues. The present work characterizes of TRPV5 and TRPV6 in RPE. METHODS Reverse transcriptase PCR was used to examine the presence of TRPV5 and TRPV6 mRNA in cultured human RPE. Protein expression was assessed by western blotting using TRPV5- and TRPV6-specific antibodies. Immunocytochemistry was employed to examine subcellular localization of TRPV5 and TRPV6 in frozen, formaldehyde-fixed sections of native RPE-choroid tissue and in cultured human RPE monolayers. Finally, TRPV5/TRPV6 activity was assessed in cultured RPE, using Ca(2+) indicator dyes to follow [Ca(2+)](i) as a function of changes in [Ca(2+)](o) with and without addition of the TRPV5/TRPV6 inhibitor ruthenium red. RESULTS Direct sequencing of PCR DNAs documented the presence of TRPV5 and TRPV6 transcripts in human RPE. Immunocytochemistry showed that TRPV5 and TRPV6 are expressed in native RPE-choroid tissue with strong immunoreactivity for both channels on the apical as well as the basal plasma membranes. Immunostaining for both channels was also positive in monolayers of cultured RPE cells. In cultured cells subcellular localization was variable with immunoreactivity present in the cytoplasmic domain as well as on the plasma membrane. Plasma membrane staining was increased with phagocytosis. The reported molecular weight of the core protein for both TRPV5 and TRPV6 is about 75 kDa, with the expected size of the glycosylated proteins in the range of 85-100 kDa. Western blot analysis of TRPV6 in RPE detected a distinct band at approximately 85 kDa, with another strong band at approximately 60 kDa. A similar pattern was seen for TRPV5, with strong bands at 82 kDa and 71 kDa. In live-cell imaging experiments, [Ca(2+)](i) was lower in the presence of the TRPV5/TRPV6 inhibitor ruthenium red. CONCLUSIONS RPE expresses the epithelial calcium channels TRPV5 and TRPV6, the most calcium-selective channels of the TRP superfamily. Present findings suggest that these channels could function in RPE to mediate calcium influx from SRS and thus regulate changes in SRS calcium composition that accompany light/dark transitions.
Collapse
Affiliation(s)
- Brian G. Kennedy
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine-Northwest, Gary, IN
| | | | - Rafal Kurzawa
- Department of Anatomy and Cell Biology, Indiana University School of Medicine-Northwest, Gary, IN
| | - Stephen F. Echtenkamp
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine-Northwest, Gary, IN
| | - Nancy J. Mangini
- Department of Anatomy and Cell Biology, Indiana University School of Medicine-Northwest, Gary, IN
| |
Collapse
|
44
|
Malkia A, Pertusa M, Fernández-Ballester G, Ferrer-Montiel A, Viana F. Differential role of the menthol-binding residue Y745 in the antagonism of thermally gated TRPM8 channels. Mol Pain 2009; 5:62. [PMID: 19886999 PMCID: PMC2778643 DOI: 10.1186/1744-8069-5-62] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 11/03/2009] [Indexed: 11/10/2022] Open
Abstract
Background TRPM8 is a non-selective cation channel that belongs to the melastatin subfamily of the transient receptor potential (TRP) ion channels. TRPM8 is activated by voltage, cold and cooling compounds such as menthol. Despite its essential role for cold temperature sensing in mammals, the pharmacology of TRPM8 is still in its infancy. Recently, tyrosine 745 (Y745) was identified as a critical residue for menthol sensitivity of the channel. In this report, we study the effect of mutating this residue on the action of several known TRPM8 antagonists: BCTC, capsazepine, SKF96365, and clotrimazole as well as two new inhibitor candidates, econazole and imidazole. Results We show that Y745 at the menthol binding site is critical for inhibition mediated by SKF96365 of cold- and voltage-activated TRPM8 currents. In contrast, the inhibition by other antagonists was unaffected by the mutation (BCTC) or only partially reduced (capsazepine, clotrimazole, econazole), suggesting that additional binding sites exist on the TRPM8 channel from where the inhibitors exert their negative modulation. Indeed, a molecular docking model implies that menthol and SKF96365 interact readily with Y745, while BCTC is unable to bind to this residue. Conclusion In summary, we identify structural elements on the TRPM8 channel that are critical for the action of channel antagonists, providing valuable information for the future design of new, specific modulator compounds.
Collapse
Affiliation(s)
- Annika Malkia
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain.
| | | | | | | | | |
Collapse
|
45
|
Romanovsky AA, Almeida MC, Garami A, Steiner AA, Norman MH, Morrison SF, Nakamura K, Burmeister JJ, Nucci TB. The transient receptor potential vanilloid-1 channel in thermoregulation: a thermosensor it is not. Pharmacol Rev 2009; 61:228-61. [PMID: 19749171 PMCID: PMC2763780 DOI: 10.1124/pr.109.001263] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The development of antagonists of the transient receptor potential vanilloid-1 (TRPV1) channel as pain therapeutics has revealed that these compounds cause hyperthermia in humans. This undesirable on-target side effect has triggered a surge of interest in the role of TRPV1 in thermoregulation and revived the hypothesis that TRPV1 channels serve as thermosensors. We review literature data on the distribution of TRPV1 channels in the body and on thermoregulatory responses to TRPV1 agonists and antagonists. We propose that two principal populations of TRPV1-expressing cells have connections with efferent thermoeffector pathways: 1) first-order sensory (polymodal), glutamatergic dorsal-root (and possibly nodose) ganglia neurons that innervate the abdominal viscera and 2) higher-order sensory, glutamatergic neurons presumably located in the median preoptic hypothalamic nucleus. We further hypothesize that all thermoregulatory responses to TRPV1 agonists and antagonists and thermoregulatory manifestations of TRPV1 desensitization stem from primary actions on these two neuronal populations. Agonists act primarily centrally on population 2; antagonists act primarily peripherally on population 1. We analyze what roles TRPV1 might play in thermoregulation and conclude that this channel does not serve as a thermosensor, at least not under physiological conditions. In the hypothalamus, TRPV1 channels are inactive at common brain temperatures. In the abdomen, TRPV1 channels are tonically activated, but not by temperature. However, tonic activation of visceral TRPV1 by nonthermal factors suppresses autonomic cold-defense effectors and, consequently, body temperature. Blockade of this activation by TRPV1 antagonists disinhibits thermoeffectors and causes hyperthermia. Strategies for creating hyperthermia-free TRPV1 antagonists are outlined. The potential physiological and pathological significance of TRPV1-mediated thermoregulatory effects is discussed.
Collapse
Affiliation(s)
- Andrej A Romanovsky
- Systemic Inflammation Laboratory, St. Joseph's Hospital and Medical Center, Phoenix, Arizona 85013, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Irnaten M, Blanchard-Gutton N, Praetorius J, Harvey BJ. Rapid effects of 17beta-estradiol on TRPV5 epithelial Ca2+ channels in rat renal cells. Steroids 2009; 74:642-9. [PMID: 19463684 DOI: 10.1016/j.steroids.2009.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 01/27/2009] [Accepted: 02/09/2009] [Indexed: 10/21/2022]
Abstract
The renal distal tubules and collecting ducts play a key role in the control of electrolyte and fluid homeostasis. The discovery of highly calcium selective channels, Transient Receptor Potential Vanilloid 5 (TRPV5) of the TRP superfamily, has clarified the nature of the calcium entry channels. It has been proposed that this channel mediates the critical Ca(2+) entry step in transcellular Ca(2+) re-absorption in the kidney. The regulation of transmembrane Ca(2+) flux through TRPV5 is of particular importance for whole body calcium homeostasis.In this study, we provide evidence that the TRPV5 channel is present in rat cortical collecting duct (RCCD(2)) cells at mRNA and protein levels. We demonstrate that 17beta-estradiol (E(2)) is involved in the regulation of Ca(2+) influx in these cells via the epithelial Ca(2+) channels TRPV5. By combining whole-cell patch-clamp and Ca(2+)-imaging techniques, we have characterized the electrophysiological properties of the TRPV5 channel and showed that treatment with 20-50nM E(2) rapidly (<5min) induced a transient increase in inward whole-cell currents and intracellular Ca(2+) via TRPV5 channels. This rise was significantly prevented when cells were pre-treated with ruthenium red and completely abolished in cells treated with siRNA specifically targeting TRPV5.These data demonstrate for the first time, a novel rapid modulation of endogenously expressed TRPV5 channels by E(2) in kidney cells. Furthermore, the results suggest calcitropic effects of E(2). The results are discussed in relation to present concepts of non-genomic actions of E(2) in Ca(2+) homeostasis.
Collapse
Affiliation(s)
- Mustapha Irnaten
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Beaumont Hospital, PO Box 9063, Dublin 9, Ireland.
| | | | | | | |
Collapse
|
47
|
Vriens J, Appendino G, Nilius B. Pharmacology of vanilloid transient receptor potential cation channels. Mol Pharmacol 2009; 75:1262-79. [PMID: 19297520 DOI: 10.1124/mol.109.055624] [Citation(s) in RCA: 311] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Depending on their primary structure, the 28 mammalian transient receptor potential (TRP) cation channels identified so far can be sorted into 6 subfamilies: TRPC ("Canonical"), TRPV ("Vanilloid"), TRPM ("Melastatin"), TRPP ("Polycystin"), TRPML ("Mucolipin"), and TRPA ("Ankyrin"). The TRPV subfamily (vanilloid receptors) comprises channels critically involved in nociception and thermosensing (TRPV1, TRPV2, TRPV3, and TRPV4), whereas TRPV5 and TRPV6 are involved in renal Ca(2+) absorption/reabsorption. Apart from TRPV1, the pharmacology of these channels is still insufficiently known. Furthermore, only few small-molecule ligands for non-TRPV1 vanilloid receptors have been identified, and little is known of their endogenous ligands, resulting in a substantial "orphan" state for these channels. In this review, we summarize the pharmacological properties of members of the TRPV subfamily, highlighting the critical issues and challenges facing their "deorphanization" and clinical exploitation.
Collapse
Affiliation(s)
- Joris Vriens
- Department of Molecular Cell Biology, Katholieke Universiteit Leuven, Belgium
| | | | | |
Collapse
|
48
|
de Groot T, Lee K, Langeslag M, Xi Q, Jalink K, Bindels RJM, Hoenderop JGJ. Parathyroid hormone activates TRPV5 via PKA-dependent phosphorylation. J Am Soc Nephrol 2009; 20:1693-704. [PMID: 19423690 DOI: 10.1681/asn.2008080873] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Low extracellular calcium (Ca(2+)) promotes release of parathyroid hormone (PTH), which acts on multiple organs to maintain overall Ca(2+) balance. In the distal part of the nephron, PTH stimulates active Ca(2+) reabsorption via the adenylyl cyclase-cAMP-protein kinase A (PKA) pathway, but the molecular target of this pathway is unknown. The transient receptor potential vanilloid 5 (TRPV5) channel constitutes the luminal gate for Ca(2+) entry in the distal convoluted tubule and has several putative PKA phosphorylation sites. Here, we investigated the effect of PTH-induced cAMP signaling on TRPV5 activity. Using fluorescence resonance energy transfer, we studied cAMP and Ca(2+) dynamics during PTH stimulation of HEK293 cells that coexpressed the PTH receptor and TRPV5. PTH increased cAMP levels, followed by a rise in TRPV5-mediated Ca(2+) influx. PTH (1 to 31) and forskolin, which activate the cAMP pathway, mimicked the stimulation of TRPV5 activity. Remarkably, TRPV5 activation was limited to conditions of strong intracellular Ca(2+) buffering. Cell surface biotinylation studies demonstrated that forskolin did not affect TRPV5 expression on the cell surface, suggesting that it alters the single-channel activity of a fixed number of TRPV5 channels. Application of the PKA catalytic subunit, which phosphorylated TRPV5, directly increased TRPV5 channel open probability. Alanine substitution of threonine-709 abolished both in vitro phosphorylation and PTH-mediated stimulation of TRPV5. In summary, PTH activates the cAMP-PKA signaling cascade, which rapidly phosphorylates threonine-709 of TRPV5, increasing the channel's open probability and promoting Ca(2+) reabsorption in the distal nephron.
Collapse
Affiliation(s)
- Theun de Groot
- Department of Physiology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | | | | | | | | | | | | |
Collapse
|
49
|
Semenova SB, Vassilieva IO, Fomina AF, Runov AL, Negulyaev YA. Endogenous expression of TRPV5 and TRPV6 calcium channels in human leukemia K562 cells. Am J Physiol Cell Physiol 2009; 296:C1098-C1104. [PMID: 19295174 DOI: 10.1152/ajpcell.00435.2008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In blood cells, changes in intracellular Ca(2+) concentration ([Ca(2+)](i)) are associated with multiple cellular events, including activation of cellular kinases and phosphatases, degranulation, regulation of cytoskeleton binding proteins, transcriptional control, and modulation of surface receptors. Although there is no doubt as to the significance of Ca(2+) signaling in blood cells, there is sparse knowledge about the molecular identities of the plasmalemmal Ca(2+) permeable channels that control Ca(2+) fluxes across the plasma membrane and mediate changes in [Ca(2+)](i) in blood cells. Using RNA expression analysis, we have shown that human leukemia K562 cells endogenously coexpress transient receptor potential vanilloid channels type 5 (TRPV5) and type 6 (TRPV6) mRNAs. Moreover, we demonstrated that TRPV5 and TRPV6 channel proteins are present in both the total lysates and the crude membrane preparations from leukemia cells. Immunoprecipitation revealed that a physical interaction between TRPV5 and TRPV6 may take place. Single-channel patch-clamp experiments demonstrated the presence of inwardly rectifying monovalent currents that displayed kinetic characteristics of unitary TRPV5 and/or TRPV6 currents and were blocked by extracellular Ca(2+) and ruthenium red. Taken together, our data strongly indicate that human myeloid leukemia cells coexpress functional TRPV5 and TRPV6 calcium channels that may interact with each other and contribute into intracellular Ca(2+) signaling.
Collapse
MESH Headings
- Calcium/pharmacokinetics
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/physiology
- Gene Expression Regulation, Leukemic
- Humans
- Indicators and Reagents/pharmacokinetics
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology
- Membrane Potentials/physiology
- Patch-Clamp Techniques
- RNA, Messenger/metabolism
- Ruthenium Red/pharmacokinetics
- TRPV Cation Channels/genetics
- TRPV Cation Channels/metabolism
Collapse
|
50
|
Rosenquist TH, Bennett GD, Brauer PR, Stewart ML, Chaudoin TR, Finnell RH. Microarray analysis of homocysteine-responsive genes in cardiac neural crest cells in vitro. Dev Dyn 2007; 236:1044-54. [PMID: 17326132 DOI: 10.1002/dvdy.21101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The amino acid homocysteine increases in the serum when there is insufficient folic acid or vitamin B(12), or with certain mutations in enzymes important in methionine metabolism. Elevated homocysteine is related to increased risk for cardiovascular and other diseases in adults and elevated maternal homocysteine increases the risk for certain congenital defects, especially those that result from abnormal development of the neural crest and neural tube. Experiments with the avian embryo model have shown that elevated homocysteine perturbs neural crest/neural tube migration in vitro and in vivo. Whereas there have been numerous studies of homocysteine-induced changes in gene expression in adult cells, there is no previous report of a homocysteine-responsive transcriptome in the embryonic neural crest. We treated neural crest cells in vitro with exogenous homocysteine in a protocol that induces significant changes in neural crest cell migration. We used microarray analysis and expression profiling to identify 65 transcripts of genes of known function that were altered by homocysteine. The largest set of effected genes (19) included those with a role in cell migration and adhesion. Other major groups were genes involved in metabolism (13); DNA/RNA interaction (11); cell proliferation/apoptosis (10); and transporter/receptor (6). Although the genes identified in this experiment were consistent with prior observations of the effect of homocysteine upon neural crest cell function, none had been identified previously as response to homocysteine in adult cells.
Collapse
Affiliation(s)
- T H Rosenquist
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 68198-7878, USA.
| | | | | | | | | | | |
Collapse
|