1
|
Zheng Z, Song Y, Tan X. Deciphering hERG Mutation in Long QT Syndrome Type 2 Using Antisense Oligonucleotide-Mediated Techniques: Lessons from Cystic Fibrosis. Heart Rhythm 2023:S1547-5271(23)02180-X. [PMID: 37121422 DOI: 10.1016/j.hrthm.2023.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023]
Abstract
Long QT syndrome type 2 (LQT2) is a genetic disorder caused by mutations in the KCNH2 gene, also known as the human ether-a-go-go-related gene (hERG). Over 30% of hERG mutations result in a premature termination codon (PTC) that triggers a process called nonsense-mediated mRNA decay (NMD), where the mRNA transcript is degraded. NMD is a quality control mechanism that removes faulty mRNA to prevent the translation of truncated proteins. Recent advances in antisense oligonucleotide (ASO) technology in the field of cystic fibrosis (CF) have yielded significant progress, including the ASO-mediated comprehensive characterization of key NMD factors and exon-skipping therapy. These advances have contributed to our understanding of the role of PTC-containing mutations in disease phenotypes and have also led to the development of potentially useful therapeutic strategies. Historically, studies of CF have provided valuable insights for the research on LQT2, particularly concerning increasing the expression of hERG. In this article, we outline the current state of knowledge regarding ASO, NMD, and hERG and discuss the introduction of ASO technology in the CF to elucidate the pathogenic mechanisms through targeting NMD. We also discuss the potential clinical therapeutic benefits and limitations of ASO for the management of LQT2. By drawing on lessons learned from CF research, we explore the potential translational values of these advances into LQT2 studies.
Collapse
Affiliation(s)
- Zequn Zheng
- Department of Cardiology, Shantou University Medical College, Shantou, China; Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, China; Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, China.
| | - Yongfei Song
- Ningbo Institute for Medicine &Biomedical Engineering Combined Innovation, Ningbo, China
| | - Xuerui Tan
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, China; Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
2
|
van Ham WB, Cornelissen CM, van Veen TAB. Uremic toxins in chronic kidney disease highlight a fundamental gap in understanding their detrimental effects on cardiac electrophysiology and arrhythmogenesis. Acta Physiol (Oxf) 2022; 236:e13888. [PMID: 36148604 PMCID: PMC9787632 DOI: 10.1111/apha.13888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/09/2022] [Accepted: 09/18/2022] [Indexed: 01/29/2023]
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) have an estimated 700-800 and 523 million cases worldwide, respectively, with CVD being the leading cause of death in CKD patients. The pathophysiological interplay between the heart and kidneys is defined as the cardiorenal syndrome (CRS), in which worsening of kidney function is represented by increased plasma concentrations of uremic toxins (UTs), culminating in dialysis patients. As there is a high incidence of CVD in CKD patients, accompanied by arrhythmias and sudden cardiac death, knowledge on electrophysiological remodeling would be instrumental for understanding the CRS. While the interplay between both organs is clearly of importance in CRS, the involvement of UTs in pro-arrhythmic remodeling is only poorly investigated, especially regarding the mechanistic background. Currently, the clinical approach against potential arrhythmic events is mainly restricted to symptom treatment, stressing the need for fundamental research on UT in relation to electrophysiology. This review addresses the existing knowledge of UTs and cardiac electrophysiology, and the experimental research gap between fundamental research and clinical research of the CRS. Clinically, mainly absorbents like ibuprofen and AST-120 are studied, which show limited safe and efficient usability. Experimental research shows disturbances in cardiac electrical activation and conduction after inducing CKD or exposure to UTs, but are scarcely present or focus solely on already well-investigated UTs. Based on UTs data derived from CKD patient cohort studies, a clinically relevant overview of physiological and pathological UTs concentrations is created. Using this, future experimental research is stimulated to involve electrophysiologically translatable animals, such as rabbits, or in vitro engineered heart tissues.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Carlijn M. Cornelissen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Toon A. B. van Veen
- Department of Medical Physiology, Division Heart & LungsUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
3
|
Kekenes-Huskey PM, Burgess DE, Sun B, Bartos DC, Rozmus ER, Anderson CL, January CT, Eckhardt LL, Delisle BP. Mutation-Specific Differences in Kv7.1 ( KCNQ1) and Kv11.1 ( KCNH2) Channel Dysfunction and Long QT Syndrome Phenotypes. Int J Mol Sci 2022; 23:7389. [PMID: 35806392 PMCID: PMC9266926 DOI: 10.3390/ijms23137389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
The electrocardiogram (ECG) empowered clinician scientists to measure the electrical activity of the heart noninvasively to identify arrhythmias and heart disease. Shortly after the standardization of the 12-lead ECG for the diagnosis of heart disease, several families with autosomal recessive (Jervell and Lange-Nielsen Syndrome) and dominant (Romano-Ward Syndrome) forms of long QT syndrome (LQTS) were identified. An abnormally long heart rate-corrected QT-interval was established as a biomarker for the risk of sudden cardiac death. Since then, the International LQTS Registry was established; a phenotypic scoring system to identify LQTS patients was developed; the major genes that associate with typical forms of LQTS were identified; and guidelines for the successful management of patients advanced. In this review, we discuss the molecular and cellular mechanisms for LQTS associated with missense variants in KCNQ1 (LQT1) and KCNH2 (LQT2). We move beyond the "benign" to a "pathogenic" binary classification scheme for different KCNQ1 and KCNH2 missense variants and discuss gene- and mutation-specific differences in K+ channel dysfunction, which can predispose people to distinct clinical phenotypes (e.g., concealed, pleiotropic, severe, etc.). We conclude by discussing the emerging computational structural modeling strategies that will distinguish between dysfunctional subtypes of KCNQ1 and KCNH2 variants, with the goal of realizing a layered precision medicine approach focused on individuals.
Collapse
Affiliation(s)
- Peter M. Kekenes-Huskey
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Don E. Burgess
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (D.E.B.); (E.R.R.)
| | - Bin Sun
- Department of Pharmacology, Harbin Medical University, Harbin 150081, China;
| | | | - Ezekiel R. Rozmus
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (D.E.B.); (E.R.R.)
| | - Corey L. Anderson
- Cellular and Molecular Arrythmias Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (C.L.A.); (C.T.J.); (L.L.E.)
| | - Craig T. January
- Cellular and Molecular Arrythmias Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (C.L.A.); (C.T.J.); (L.L.E.)
| | - Lee L. Eckhardt
- Cellular and Molecular Arrythmias Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (C.L.A.); (C.T.J.); (L.L.E.)
| | - Brian P. Delisle
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (D.E.B.); (E.R.R.)
| |
Collapse
|
4
|
Saponara S, Fusi F, Iovinelli D, Ahmed A, Trezza A, Spiga O, Sgaragli G, Valoti M. Flavonoids and hERG channels: Friends or foes? Eur J Pharmacol 2021; 899:174030. [PMID: 33727059 DOI: 10.1016/j.ejphar.2021.174030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/28/2021] [Accepted: 03/11/2021] [Indexed: 01/24/2023]
Abstract
The cardiac action potential is regulated by several ion channels. Drugs capable to block these channels, in particular the human ether-à-go-go-related gene (hERG) channel, also known as KV11.1 channel, may lead to a potentially lethal ventricular tachyarrhythmia called "Torsades de Pointes". Thus, evaluation of the hERG channel off-target activity of novel chemical entities is nowadays required to safeguard patients as well as to avoid attrition in drug development. Flavonoids, a large class of natural compounds abundantly present in food, beverages, herbal medicines, and dietary food supplements, generally escape this assessment, though consumed in consistent amounts. Continuously growing evidence indicates that these compounds may interact with the hERG channel and block it. The present review, by examining numerous studies, summarizes the state-of-the-art in this field, describing the most significant examples of direct and indirect inhibition of the hERG channel current operated by flavonoids. A description of the molecular interactions between a few of these natural molecules and the Rattus norvegicus channel protein, achieved by an in silico approach, is also presented.
Collapse
Affiliation(s)
- Simona Saponara
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Fabio Fusi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy.
| | - Daniele Iovinelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Amer Ahmed
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Alfonso Trezza
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Ottavia Spiga
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Giampietro Sgaragli
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy; Accademia Italiana della Vite e del Vino, via Logge degli Uffizi Corti 1, 50122, Florence, Italy
| | - Massimo Valoti
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, via A. Moro 2, 53100, Siena, Italy
| |
Collapse
|
5
|
Long QT Syndrome Type 2: Emerging Strategies for Correcting Class 2 KCNH2 ( hERG) Mutations and Identifying New Patients. Biomolecules 2020. [PMID: 32759882 DOI: 10.3390/biom10081144s] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Significant advances in our understanding of the molecular mechanisms that cause congenital long QT syndrome (LQTS) have been made. A wide variety of experimental approaches, including heterologous expression of mutant ion channel proteins and the use of inducible pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from LQTS patients offer insights into etiology and new therapeutic strategies. This review briefly discusses the major molecular mechanisms underlying LQTS type 2 (LQT2), which is caused by loss-of-function (LOF) mutations in the KCNH2 gene (also known as the human ether-à-go-go-related gene or hERG). Almost half of suspected LQT2-causing mutations are missense mutations, and functional studies suggest that about 90% of these mutations disrupt the intracellular transport, or trafficking, of the KCNH2-encoded Kv11.1 channel protein to the cell surface membrane. In this review, we discuss emerging strategies that improve the trafficking and functional expression of trafficking-deficient LQT2 Kv11.1 channel proteins to the cell surface membrane and how new insights into the structure of the Kv11.1 channel protein will lead to computational approaches that identify which KCNH2 missense variants confer a high-risk for LQT2.
Collapse
|
6
|
Ono M, Burgess DE, Schroder EA, Elayi CS, Anderson CL, January CT, Sun B, Immadisetty K, Kekenes-Huskey PM, Delisle BP. Long QT Syndrome Type 2: Emerging Strategies for Correcting Class 2 KCNH2 ( hERG) Mutations and Identifying New Patients. Biomolecules 2020; 10:E1144. [PMID: 32759882 PMCID: PMC7464307 DOI: 10.3390/biom10081144] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
Significant advances in our understanding of the molecular mechanisms that cause congenital long QT syndrome (LQTS) have been made. A wide variety of experimental approaches, including heterologous expression of mutant ion channel proteins and the use of inducible pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from LQTS patients offer insights into etiology and new therapeutic strategies. This review briefly discusses the major molecular mechanisms underlying LQTS type 2 (LQT2), which is caused by loss-of-function (LOF) mutations in the KCNH2 gene (also known as the human ether-à-go-go-related gene or hERG). Almost half of suspected LQT2-causing mutations are missense mutations, and functional studies suggest that about 90% of these mutations disrupt the intracellular transport, or trafficking, of the KCNH2-encoded Kv11.1 channel protein to the cell surface membrane. In this review, we discuss emerging strategies that improve the trafficking and functional expression of trafficking-deficient LQT2 Kv11.1 channel proteins to the cell surface membrane and how new insights into the structure of the Kv11.1 channel protein will lead to computational approaches that identify which KCNH2 missense variants confer a high-risk for LQT2.
Collapse
Affiliation(s)
- Makoto Ono
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| | - Don E. Burgess
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| | - Elizabeth A. Schroder
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| | | | - Corey L. Anderson
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI 53706, USA; (C.L.A.); (C.T.J.)
| | - Craig T. January
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI 53706, USA; (C.L.A.); (C.T.J.)
| | - Bin Sun
- Department of Cellular & Molecular Physiology, Loyola University Chicago, Chicago, IL 60153, USA; (B.S.); (K.I.); (P.M.K.-H.)
| | - Kalyan Immadisetty
- Department of Cellular & Molecular Physiology, Loyola University Chicago, Chicago, IL 60153, USA; (B.S.); (K.I.); (P.M.K.-H.)
| | - Peter M. Kekenes-Huskey
- Department of Cellular & Molecular Physiology, Loyola University Chicago, Chicago, IL 60153, USA; (B.S.); (K.I.); (P.M.K.-H.)
| | - Brian P. Delisle
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| |
Collapse
|
7
|
Tay YL, Amanah A, Adenan MI, Wahab HA, Tan ML. Mitragynine, an euphoric compound inhibits hERG1a/1b channel current and upregulates the complexation of hERG1a-Hsp90 in HEK293-hERG1a/1b cells. Sci Rep 2019; 9:19757. [PMID: 31874991 PMCID: PMC6930223 DOI: 10.1038/s41598-019-56106-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 11/06/2019] [Indexed: 11/24/2022] Open
Abstract
Mitragyna speciosa Korth (M. speciosa) has been widely used as a recreational product, however, there are growing concerns on the abuse potentials and toxicity of the plant. Several poisoning and fatal cases involving kratom and mitragynine have been reported but the underlying causes remain unclear. The human ether-a-go-go-related gene 1 (hERG1) encodes the pore-forming subunit underlying cardiac rapidly delayed rectifier potassium current (IKr). Pharmacological blockade of the IKr can cause acquired long QT syndrome, leading to lethal cardiac arrhythmias. This study aims to elucidate the mechanisms of mitragynine-induced inhibition on hERG1a/1b current. Electrophysiology experiments were carried out using Port-a-Patch system. Quantitative RT-PCR, Western blot analysis, immunofluorescence and co-immunoprecipitation methods were used to determine the effects of mitragynine on hERG1a/1b expression and hERG1-cytosolic chaperones interaction. Mitragynine was found to inhibit the IKr current with an IC50 value of 332.70 nM. It causes a significant reduction of the fully-glycosylated (fg) hERG1a protein expression but upregulates both core-glycosylated (cg) expression and hERG1a-Hsp90 complexes, suggesting possible impaired hERG1a trafficking. In conclusion, mitragynine inhibits hERG1a/1b current through direct channel blockade at lower concentration, but at higher concentration, it upregulates the complexation of hERG1a-Hsp90 which may be inhibitory towards channel trafficking.
Collapse
Affiliation(s)
- Yea Lu Tay
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, NIBM, Ministry of Energy, Science, Technology, Environment and Climate Change (MESTECC), Pulau Pinang, 11700, Malaysia
| | - Azimah Amanah
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, NIBM, Ministry of Energy, Science, Technology, Environment and Climate Change (MESTECC), Pulau Pinang, 11700, Malaysia
| | - Mohd Ilham Adenan
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Selangor Darul Ehsan, 42300, Malaysia
| | - Habibah Abdul Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, 11700, Malaysia
| | - Mei Lan Tan
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, NIBM, Ministry of Energy, Science, Technology, Environment and Climate Change (MESTECC), Pulau Pinang, 11700, Malaysia. .,School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, 11700, Malaysia. .,Advanced Medical and Dental Institute, Universiti Sains Malaysia, SAINS@BERTAM, Kepala Batas, Pulau Pinang, 13200, Malaysia.
| |
Collapse
|
8
|
Mauerhöfer M, Bauer CK. Effects of Temperature on Heteromeric Kv11.1a/1b and Kv11.3 Channels. Biophys J 2017; 111:504-523. [PMID: 27508435 DOI: 10.1016/j.bpj.2016.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 01/17/2023] Open
Abstract
Kv11.1 channels are crucial in cardiac physiology, and there is increasing evidence of physiological roles of different Kv11 channels outside the heart. The HERG (human Kv11.1a) channel has previously been shown to carry substantially more current at elevated temperatures, and we have now comparably investigated the temperature dependence of neuronal Kv11.3 channels and the more ubiquitous heteromeric Kv11.1a/1b channels. Transiently expressed rat Kv11 channels were studied at 21°C, 30°C, and 35°C. At near-physiological temperature, the maximal sustained outward current density was almost three times the mean value obtained at room temperature for Kv11.1a/1b, and increased by ∼150% for Kv11.3. For both channels, reduced inactivation contributed to the current increase at higher temperature. Elevated temperature moved Kv11.1a/1b isochronal activation curves to more negative potentials, but shifted the potential of half-maximal Kv11.3 channel activation to more depolarized values and reduced its voltage sensitivity. Thus, increased temperature stabilized the open state over the closed state of Kv11.1a/1b channels and exerted the opposite effect on Kv11.3 channel activation. Both Kv11 channels exhibited an overall high temperature sensitivity of most gating parameters, with remarkably high Q10 factors of ∼5 for the rate of Kv11.1a/1b activation. The Q10 factors for Kv11.3 gating were more uniform, but still higher for activation than for inactivation kinetics. The results demonstrate that characteristic differences between Kv11.1a/1b and Kv11.3 determined at room temperature do not necessarily apply to physiological conditions. The data provided here can aid in the design of models that will enhance our understanding of the role of Kv11 currents in excitable cells.
Collapse
Affiliation(s)
- Maike Mauerhöfer
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christiane K Bauer
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
9
|
Evidence for splice transcript variants of TMEM165, a gene involved in CDG. Biochim Biophys Acta Gen Subj 2017; 1861:737-748. [DOI: 10.1016/j.bbagen.2017.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
|
10
|
Bronte E, Bronte G, Novo G, Bronte F, Bavetta MG, Lo Re G, Brancatelli G, Bazan V, Natoli C, Novo S, Russo A. What links BRAF to the heart function? New insights from the cardiotoxicity of BRAF inhibitors in cancer treatment. Oncotarget 2016; 6:35589-601. [PMID: 26431495 PMCID: PMC4742127 DOI: 10.18632/oncotarget.5853] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/22/2015] [Indexed: 12/14/2022] Open
Abstract
The RAS-related signalling cascade has a fundamental role in cell. It activates differentiation and survival. It is particularly important one of its molecules, B-RAF. B-RAF has been a central point for research, especially in melanoma. Indeed, it lacked effective therapeutic weapons since the early years of its study. Molecules targeting B-RAF have been developed. Nowadays, two classes of molecules are approved by FDA. Multi-target molecules, such as Sorafenib and Regorafenib, and selective molecules, such as Vemurafenib and Dabrafenib. Many other molecules are still under investigation. Most of them are studied in phase 1 trials. Clinical studies correlate B-RAF inhibitors and QT prolongation. Though this cardiovascular side effect is not common using these drugs, it must be noticed early and recognize its signals. Indeed, Oncologists and Cardiologists should work in cooperation to prevent lethal events, such as fatal arrhythmias or sudden cardiac death. These events could originate from an uncontrolled QT prolongation.
Collapse
Affiliation(s)
- Enrico Bronte
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Bronte
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppina Novo
- Department of Internal Medicine and Cardiovascular Disease, University of Palermo, Palermo, Italy
| | - Fabrizio Bronte
- DiBiMIS, Section of Gastroenterology, University of Palermo, Palermo, Italy
| | | | - Giuseppe Lo Re
- Department of Radiology, University of Palermo, Palermo, Italy
| | | | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy
| | - Salvatore Novo
- Department of Internal Medicine and Cardiovascular Disease, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
11
|
Abstract
Approximately 80 genes in the human genome code for pore-forming subunits of potassium (K(+)) channels. Rare variants (mutations) in K(+) channel-encoding genes may cause heritable arrhythmia syndromes. Not all rare variants in K(+) channel-encoding genes are necessarily disease-causing mutations. Common variants in K(+) channel-encoding genes are increasingly recognized as modifiers of phenotype in heritable arrhythmia syndromes and in the general population. Although difficult, distinguishing pathogenic variants from benign variants is of utmost importance to avoid false designations of genetic variants as disease-causing mutations.
Collapse
Affiliation(s)
- Ahmad S Amin
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands; King Abdulaziz University, Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders, PO Box 80200, Jeddah 21589, Kingdom of Saudi Arabia.
| |
Collapse
|
12
|
Smith JL, Anderson CL, Burgess DE, Elayi CS, January CT, Delisle BP. Molecular pathogenesis of long QT syndrome type 2. J Arrhythm 2016; 32:373-380. [PMID: 27761161 PMCID: PMC5063260 DOI: 10.1016/j.joa.2015.11.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/21/2015] [Accepted: 11/17/2015] [Indexed: 12/26/2022] Open
Abstract
The molecular mechanisms underlying congenital long QT syndrome (LQTS) are now beginning to be understood. New insights into the etiology and therapeutic strategies are emerging from heterologous expression studies of LQTS-linked mutant proteins, as well as inducible pluripotent stem cell derived cardiomyocytes (iPSC-CMs) from LQTS patients. This review focuses on the major molecular mechanism that underlies LQTS type 2 (LQT2). LQT2 is caused by loss of function (LOF) mutations in KCNH2 (also known as the human Ether-à-go-go-Related Gene or hERG). Most LQT2-linked mutations are missense mutations and functional studies suggest that ~90% of them disrupt the intracellular transport (trafficking) of KCNH2-encoded Kv11.1 proteins to the cell membrane. Trafficking deficient LQT2 mutations disrupt Kv11.1 protein folding and misfolded Kv11.1 proteins are retained in the endoplasmic reticulum (ER) until they are degraded in the ER associated degradation pathway (ERAD). This review focuses on the quality control mechanisms in the ER that contribute to the folding and ERAD of Kv11.1 proteins; the mechanism for ER export of Kv11.1 proteins in the secretory pathway; different subclasses of trafficking deficient LQT2 mutations; and strategies being developed to mitigate or correct trafficking deficient LQT2-related phenotypes.
Collapse
Affiliation(s)
- Jennifer L Smith
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Corey L Anderson
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Don E Burgess
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Claude S Elayi
- Department of Cardiology, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Craig T January
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Brian P Delisle
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
13
|
Dubois VFS, de Witte WEA, Visser SAG, Danhof M, Della Pasqua O. Assessment of Interspecies Differences in Drug-Induced QTc Interval Prolongation in Cynomolgus Monkeys, Dogs and Humans. Pharm Res 2015; 33:40-51. [PMID: 26553352 PMCID: PMC4689776 DOI: 10.1007/s11095-015-1760-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 07/16/2015] [Indexed: 11/30/2022]
Abstract
Background and Purpose The selection of the most suitable animal species and subsequent translation of the concentration-effect relationship to humans are critical steps for accurate assessment of the pro-arrhythmic risk of candidate molecules. The objective of this investigation was to assess quantitatively the differences in the QTc prolonging effects of moxifloxacin between cynomolgus monkeys, dogs and humans. The impact of interspecies differences is also illustrated for a new candidate molecule. Experimental Approach Pharmacokinetic data and ECG recordings from pre-clinical protocols in monkeys and dogs and from a phase I trial in healthy subjects were identified for the purpose of this analysis. A previously established Bayesian model describing the combined effect of heart rate, circadian variation and drug effect on the QT interval was used to describe the pharmacokinetic-pharmacodynamic relationships. The probability of a ≥10 ms increase in QT was derived as measure of the pro-arrhythmic effect. Key Results For moxifloxacin, the concentrations associated with a 50% probability of QT prolongation ≥10 ms (Cp50) varied from 20.3 to 6.4 and 2.6 μM in dogs, monkeys and humans, respectively. For NCE05, these values were 0.4 μM vs 2.0 μM for monkeys and humans, respectively. Conclusions and Implications Our findings reveal significant interspecies differences in the QT-prolonging effect of moxifloxacin. In addition to the dissimilarity in pharmacokinetics across species, it is likely that differences in pharmacodynamics also play an important role. It appears that, regardless of the animal model used, a translation function is needed to predict concentration-effect relationships in humans.
Collapse
Affiliation(s)
- V F S Dubois
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | - W E A de Witte
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | - S A G Visser
- Global DMPK, AstraZeneca R&D, Sodertalje, Sweden
| | - M Danhof
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands
| | - O Della Pasqua
- Leiden Academic Centre for Drug Research, Division of Pharmacology, Leiden University, Leiden, The Netherlands. .,Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Stockley Park, Uxbridge, UK. .,Clinical Pharmacology & Therapeutics, University College London, London, UK.
| | | | | |
Collapse
|
14
|
Pedersen PJ, Thomsen KB, Olander ER, Hauser F, Tejada MDLA, Poulsen KL, Grubb S, Buhl R, Calloe K, Klaerke DA. Molecular Cloning and Functional Expression of the Equine K+ Channel KV11.1 (Ether à Go-Go-Related/KCNH2 Gene) and the Regulatory Subunit KCNE2 from Equine Myocardium. PLoS One 2015; 10:e0138320. [PMID: 26376488 PMCID: PMC4574097 DOI: 10.1371/journal.pone.0138320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 08/28/2015] [Indexed: 11/18/2022] Open
Abstract
The KCNH2 and KCNE2 genes encode the cardiac voltage-gated K+ channel KV11.1 and its auxiliary β subunit KCNE2. KV11.1 is critical for repolarization of the cardiac action potential. In humans, mutations or drug therapy affecting the KV11.1 channel are associated with prolongation of the QT intervals on the ECG and increased risk of ventricular tachyarrhythmia and sudden cardiac death—conditions known as congenital or acquired Long QT syndrome (LQTS), respectively. In horses, sudden, unexplained deaths are a well-known problem. We sequenced the cDNA of the KCNH2 and KCNE2 genes using RACE and conventional PCR on mRNA purified from equine myocardial tissue. Equine KV11.1 and KCNE2 cDNA had a high homology to human genes (93 and 88%, respectively). Equine and human KV11.1 and KV11.1/KCNE2 were expressed in Xenopus laevis oocytes and investigated by two-electrode voltage-clamp. Equine KV11.1 currents were larger compared to human KV11.1, and the voltage dependence of activation was shifted to more negative values with V1/2 = -14.2±1.1 mV and -17.3±0.7, respectively. The onset of inactivation was slower for equine KV11.1 compared to the human homolog. These differences in kinetics may account for the larger amplitude of the equine current. Furthermore, the equine KV11.1 channel was susceptible to pharmacological block with terfenadine. The physiological importance of KV11.1 was investigated in equine right ventricular wedge preparations. Terfenadine prolonged action potential duration and the effect was most pronounced at slow pacing. In conclusion, these findings indicate that horses could be disposed to both congenital and acquired LQTS.
Collapse
Affiliation(s)
- Philip Juul Pedersen
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Kirsten Brolin Thomsen
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Emma Rie Olander
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Frank Hauser
- Center for Functional and Comparative Insect Genomics, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Maria de los Angeles Tejada
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Kristian Lundgaard Poulsen
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Soren Grubb
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Rikke Buhl
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Kirstine Calloe
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
- * E-mail:
| | - Dan Arne Klaerke
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
15
|
Abriel H, Rougier JS, Jalife J. Ion channel macromolecular complexes in cardiomyocytes: roles in sudden cardiac death. Circ Res 2015; 116:1971-88. [PMID: 26044251 DOI: 10.1161/circresaha.116.305017] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The movement of ions across specific channels embedded on the membrane of individual cardiomyocytes is crucial for the generation and propagation of the cardiac electric impulse. Emerging evidence over the past 20 years strongly suggests that the normal electric function of the heart is the result of dynamic interactions of membrane ion channels working in an orchestrated fashion as part of complex molecular networks. Such networks work together with exquisite temporal precision to generate each action potential and contraction. Macromolecular complexes play crucial roles in transcription, translation, oligomerization, trafficking, membrane retention, glycosylation, post-translational modification, turnover, function, and degradation of all cardiac ion channels known to date. In addition, the accurate timing of each cardiac beat and contraction demands, a comparable precision on the assembly and organizations of sodium, calcium, and potassium channel complexes within specific subcellular microdomains, where physical proximity allows for prompt and efficient interaction. This review article, part of the Compendium on Sudden Cardiac Death, discusses the major issues related to the role of ion channel macromolecular assemblies in normal cardiac electric function and the mechanisms of arrhythmias leading to sudden cardiac death. It provides an idea of how these issues are being addressed in the laboratory and in the clinic, which important questions remain unanswered, and what future research will be needed to improve knowledge and advance therapy.
Collapse
Affiliation(s)
- Hugues Abriel
- From the Department of Clinical Research, University of Bern, Bern, Switzerland (H.A., J.-S.R.); Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor (J.J.); and Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (J.J.)
| | - Jean-Sébastien Rougier
- From the Department of Clinical Research, University of Bern, Bern, Switzerland (H.A., J.-S.R.); Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor (J.J.); and Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (J.J.)
| | - José Jalife
- From the Department of Clinical Research, University of Bern, Bern, Switzerland (H.A., J.-S.R.); Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor (J.J.); and Area of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (J.J.).
| |
Collapse
|
16
|
Li P, Chen X, Zhang Q, Zheng Y, Jiang H, Yang H, Gao Z. The human ether-a-go-go-related gene activator NS1643 enhances epilepsy-associated KCNQ channels. J Pharmacol Exp Ther 2014; 351:596-604. [PMID: 25232191 DOI: 10.1124/jpet.114.217703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human ether-a-go-go-related gene (hERG) and KCNQ channels are two classes of voltage-gated potassium channels. Specific mutations have been identified that are causal for type II long QT (LQT2) syndrome, neonatal epilepsy, and benign familial neonatal convulsions. Increasing evidence from clinical studies suggests that LQT2 and epilepsy coexist in some patients. Therefore, an integral approach to investigating and treating the two diseases is likely more effective. In the current study, we found that NS1643 [1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea], a previously reported hERG activator, is also an activator of KCNQ channels. It potentiates the neuronal KCNQ2, KCNQ4, and KCNQ2/Q3 channels, but not the cardiac KCNQ1. The effects of NS1643 on the KCNQ2 channel include left shifting of voltage for reaching 50% of the maximum conductance and slowing of deactivation. Analysis of the dose-response curve of NS1643 revealed an EC50 value of 2.44 ± 0.25 μM. A hydrophobic phenylalanine (F137) located at the middle region of the voltage-sensing domain was identified as critical for NS1643 activity on KCNQ2. When testing NS1643 effects in rescuing LQT2 hERG mutants and the KCNQ2 BFNC mutants, we found it is particularly efficacious in some cases. Considering the substantial relationship between LQT2 and epilepsy, these findings reveal that NS1643 is a useful compound to elucidate the causal connection of LQT2 and epilepsy. More generally, this may provide a strategy in the development of therapeutics for LQT2 and epilepsy.
Collapse
Affiliation(s)
- Ping Li
- CAS Key Laboratory of Receptor Research (P.L., X.C., Y.Z., Z.G.), and State Key Laboratory of Drug Research (Q.Z., H.J., H.Y.), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xueqin Chen
- CAS Key Laboratory of Receptor Research (P.L., X.C., Y.Z., Z.G.), and State Key Laboratory of Drug Research (Q.Z., H.J., H.Y.), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qiansen Zhang
- CAS Key Laboratory of Receptor Research (P.L., X.C., Y.Z., Z.G.), and State Key Laboratory of Drug Research (Q.Z., H.J., H.Y.), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yueming Zheng
- CAS Key Laboratory of Receptor Research (P.L., X.C., Y.Z., Z.G.), and State Key Laboratory of Drug Research (Q.Z., H.J., H.Y.), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hualiang Jiang
- CAS Key Laboratory of Receptor Research (P.L., X.C., Y.Z., Z.G.), and State Key Laboratory of Drug Research (Q.Z., H.J., H.Y.), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Huaiyu Yang
- CAS Key Laboratory of Receptor Research (P.L., X.C., Y.Z., Z.G.), and State Key Laboratory of Drug Research (Q.Z., H.J., H.Y.), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhaobing Gao
- CAS Key Laboratory of Receptor Research (P.L., X.C., Y.Z., Z.G.), and State Key Laboratory of Drug Research (Q.Z., H.J., H.Y.), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
17
|
Schmitt N, Grunnet M, Olesen SP. Cardiac potassium channel subtypes: new roles in repolarization and arrhythmia. Physiol Rev 2014; 94:609-53. [PMID: 24692356 DOI: 10.1152/physrev.00022.2013] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
About 10 distinct potassium channels in the heart are involved in shaping the action potential. Some of the K+ channels are primarily responsible for early repolarization, whereas others drive late repolarization and still others are open throughout the cardiac cycle. Three main K+ channels drive the late repolarization of the ventricle with some redundancy, and in atria this repolarization reserve is supplemented by the fairly atrial-specific KV1.5, Kir3, KCa, and K2P channels. The role of the latter two subtypes in atria is currently being clarified, and several findings indicate that they could constitute targets for new pharmacological treatment of atrial fibrillation. The interplay between the different K+ channel subtypes in both atria and ventricle is dynamic, and a significant up- and downregulation occurs in disease states such as atrial fibrillation or heart failure. The underlying posttranscriptional and posttranslational remodeling of the individual K+ channels changes their activity and significance relative to each other, and they must be viewed together to understand their role in keeping a stable heart rhythm, also under menacing conditions like attacks of reentry arrhythmia.
Collapse
|
18
|
Erg potassium currents of neonatal mouse Purkinje cells exhibit fast gating kinetics and are inhibited by mGluR1 activation. J Neurosci 2013; 33:16729-40. [PMID: 24133274 DOI: 10.1523/jneurosci.5523-12.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
We investigated the subthreshold properties of an erg (ether-à-go-go-related gene) K(+) current in Purkinje cells of neonatal mice. Action potentials recorded from Purkinje cells in cerebellar slices exhibited a decreased threshold potential and increased frequency of spontaneous and repetitive activity following application of the specific erg channel blocker E-4031. Accommodation was absent before and after drug application. The erg current of these Purkinje cells activated at membrane potentials near -60 mV and exhibited fast gating kinetics. The functional importance of fast gating subthreshold erg channels in Purkinje cells was corroborated by comparing the results of action potential clamp experiments with erg1a, erg1b, erg2, and erg3 currents heterologously expressed in HEK cells. Computer simulations based on a NEURON model of Purkinje cells only reproduced the effects of the native erg current when an erg channel conductance like that of erg3 was included. Experiments with subunit-sensitive toxins (BeKm-1, APETx1) indicated that erg channels in Purkinje cells are presumably mediated by heteromeric erg1/erg3 or modified erg1 channels. Following mGluR1 activation, the native erg current was reduced by ∼70%, brought about by reduction of the maximal erg current and a shift of the activation curve to more positive potentials. The Purkinje cell erg current contributed to the sustained current component of the biphasic mGluR1 response. Activation of mGluR1 by the agonist 3,4-dihydroxyphenylglycol increased Purkinje cell excitability, similar to that induced by E-4031. The results indicated that erg currents can be modulated and may contribute to the mGluR1-induced plasticity changes in Purkinje cells.
Collapse
|
19
|
Levit A, Nowak S, Peters M, Wiener A, Meyerhof W, Behrens M, Niv MY. The bitter pill: clinical drugs that activate the human bitter taste receptor TAS2R14. FASEB J 2013; 28:1181-97. [PMID: 24285091 DOI: 10.1096/fj.13-242594] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bitter taste receptors (TAS2Rs) mediate aversive response to toxic food, which is often bitter. These G-protein-coupled receptors are also expressed in extraoral tissues, and emerge as novel targets for therapeutic indications such as asthma and infection. Our goal was to identify ligands of the broadly tuned TAS2R14 among clinical drugs. Molecular properties of known human bitter taste receptor TAS2R14 agonists were incorporated into pharmacophore- and shape-based models and used to computationally predict additional ligands. Predictions were tested by calcium imaging of TAS2R14-transfected HEK293 cells. In vitro testing of the virtual screening predictions resulted in 30-80% success rates, and 15 clinical drugs were found to activate the TAS2R14. hERG potassium channel, which is predominantly expressed in the heart, emerged as a common off-target of bitter drugs. Despite immense chemical diversity of known TAS2R14 ligands, novel ligands and previously unknown polypharmacology of drugs were unraveled by in vitro screening of computational predictions. This enables rational repurposing of traditional and standard drugs for bitter taste signaling modulation for therapeutic indications.
Collapse
Affiliation(s)
- Anat Levit
- 2The Institute of Biochemistry, Food Science, and Nutrition, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, P.O. Box 12, Rehovot 76100, Israel.
| | | | | | | | | | | | | |
Collapse
|