1
|
Saatloo MV, Delisi D, Eskandari N, Krieg C, Gentile S. Kv11.1-dependent senescence activates a lethal immune response via tumor necrosis factor alpha. Neoplasia 2025; 63:101148. [PMID: 40117717 PMCID: PMC11981764 DOI: 10.1016/j.neo.2025.101148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025]
Abstract
Understanding the complex relationship between cancer and immune surveillance is essential for leveraging the immune system to control tumor growth. In our study, we discovered that activating the Kv11.1 potassium channel in ER+ breast cancer cells induces a senescent phenotype, which in turn triggers a potent immune response against these senescent cells. Specifically, we found that the senescence-associated secretory phenotype (SASP) plays a crucial role in activating CD4+ T-helper 1 (Th1) cells and memory T cell phenotypes. This activation led to the release of tumor necrosis factor-alpha (TNFα), which induced the death of senescent breast cancer cells, independent of their resistance to endocrine therapy. Our findings suggest that Kv11.1 channel-induced cellular senescence in ER+ breast cancer cells is a key mechanism in immune surveillance, driving a lethal immune response through TNFα. These results highlight the potential immunomodulatory role of Kv11.1 activation in ER-positive breast cancer and provide a foundation for future therapeutic investigations.
Collapse
Affiliation(s)
- Maedeh Vakili Saatloo
- Department of Biochemistry and Molecular Biology; Medical University South Carolina, Charleston, SC 29425, USA; Department of Periodontology, Henry M. Goldman School of Dental Medicine, Boston University, MA, USA
| | - Davide Delisi
- Department of Biochemistry and Molecular Biology; Medical University South Carolina, Charleston, SC 29425, USA
| | - Najmeh Eskandari
- Department of Biochemistry and Molecular Biology; Medical University South Carolina, Charleston, SC 29425, USA
| | - Carsten Krieg
- Department of Pathology and Laboratory Medicine, Medical University South Carolina, Charleston, SC 29425, USA
| | - Saverio Gentile
- Department of Biochemistry and Molecular Biology; Medical University South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
2
|
Mun D, Kang JY, Park M, Yoo G, Yun N, Hwang Y, Joung B. Pathogenic KCNH2-G53S variant in the PAS domain influences the electrophysiological phenotype in long QT syndrome type 2. Front Cardiovasc Med 2025; 12:1524909. [PMID: 40271129 PMCID: PMC12014601 DOI: 10.3389/fcvm.2025.1524909] [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: 11/08/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Background Long QT syndrome type 2 (LQT2) is an arrythmia caused by loss-of-function mutations in KCNH2, leading to impaired Kv11.1 channel function. Objective To better understand LQT2, we examined the electrophysiological differences related to the G53S variant, which is located within the PAS domain of KCNH2, using patient-specific human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hiPSC-CMs). Methods We generated hiPSC-CMs from a patient harboring the KCNH2G53S variant and a healthy control using non-integrative Sendai virus-mediated reprogramming. Their electrophysiological properties were assessed using microelectrode arrays (MEA), and Ca2+ dynamics were characterized using Fluo-4 dye. Results The patient harboring KCNH2G53S experienced aborted sudden cardiac death at 22 years of age, was diagnosed with LQT, and had an implantable cardioverter-defibrillator (ICD) implanted. KCNH2G53S hiPSC-CMs expressed less KCNH2 than normal CMs. Transcriptomic analysis of KCNH2G53S hiPSC-CMs revealed 3,857 differentially expressed genes, highlighting significant changes in pathways related to LQT2 development. Action potential duration was significantly longer in KCNH2G53S hiPSC-CMs than in control (545.3 ± 176.3 ms vs. 339.9 ± 44.5 ms; P = 0.019). Corrected field potential duration was significantly longer in KCNH2G53S hiPSC-CMs than in control (318.0 ± 66.3 ms vs. 234.5 ± 21.0 ms; P = 0.015), indicating altered electrophysiology. KCNH2G53S hiPSC-CMs exhibited significantly increased calcium transient amplitude and prolonged calcium wave duration under isoproterenol stimulation, indicating exacerbated abnormal calcium handling. Conclusion Our analysis of hiPSC-CMs carrying a heterozygous KCNH2G53S mutation, which showed abnormal electrophysiology and impaired calcium handling, provides a basis for developing improved management strategies for patients with LQT2.
Collapse
Affiliation(s)
- Dasom Mun
- Division of Cardiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Young Kang
- Division of Cardiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Malgeum Park
- Division of Cardiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gyeongseo Yoo
- Division of Cardiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nuri Yun
- GNTPharma Science and Technology Center for Health, Incheon, Republic of Korea
| | - YouMi Hwang
- Division of Cardiology, Department of Internal Medicine, St. Vincent’s Hospital, The Catholic University College of Medicine, Suwon, Republic of Korea
- Catholic Research Institute for Intractable Cardiovascular Disease (CRID), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Boyoung Joung
- Division of Cardiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
3
|
Kumawat A, Tavazzani E, Lentini G, Trancuccio A, Kukavica D, Oldani A, Denegri M, Priori SG, Camilloni C. Molecular insights into the rescue mechanism of an HERG activator against severe LQT2 mutations. J Biomed Sci 2025; 32:40. [PMID: 40197385 PMCID: PMC11974032 DOI: 10.1186/s12929-025-01134-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Mutations in the HERG potassium channel are a major cause of long QT syndrome type 2 (LQT2), which can lead to sudden cardiac death. The HERG channel plays a critical role in the repolarization of the myocardial action potential, and loss-of-function mutations prolong cardiac repolarization. METHODS In this study, we investigated the efficacy and underlying molecular mechanism of ICA-105574, an HERG activator, in shortening the duration of cardiac repolarization in severe LQT2 variants. We characterized the efficacy of ICA-105574 in vivo, using an animal model to assess its ability to shorten the QT interval and in vitro, in cellular models mimicking severe HERG channel mutations (A561V, G628S, and L779P) to evaluate its impact in enhancing IKr current. Additionally, molecular dynamics simulations were used to investigate the molecular mechanism of ICA-105574 action. RESULTS In vivo, ICA-105574 significantly shortened the QT interval. LQT2 mutations drastically reduced IKr amplitude and suppressed tail currents in cellular models. ICA-105574 restored IKr in A561V and G628S. Finally, in silico data showed that ICA-105574 stabilizes a pattern of interactions similar to gain-of-function SQT1 mutations and can reverse the G628S modifications, through an allosteric network linking the binding site to the selectivity filter and the S5P turret helix, thereby restoring its K+ ion permeability. CONCLUSIONS Our results support the development of HERG activators like ICA-105574 as promising pharmacological molecules against some severe LQT2 mutations and suggest that molecular dynamics simulations can be used to test the ability of molecules to modulate HERG function in silico, paving the way for the rational design of new HERG activators.
Collapse
Affiliation(s)
- Amit Kumawat
- Department of Biosciences, University of Milan, Milan, Italy
- Department of Physics, University of Cagliari, Cagliari, Italy
| | - Elisa Tavazzani
- IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Molecular Cardiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giovanni Lentini
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Alessandro Trancuccio
- IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Molecular Cardiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Deni Kukavica
- IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
- Molecular Cardiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Amanda Oldani
- Centro Grandi Strumenti of the University of Pavia, Pavia, Italy
| | - Marco Denegri
- IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Silvia G Priori
- IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy.
- Molecular Cardiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.
| | - Carlo Camilloni
- Department of Biosciences, University of Milan, Milan, Italy.
| |
Collapse
|
4
|
Haas B, Roth I, Säcker L, Wos-Maganga M, Beltzig L, Kaina B. Apoptotic and senolytic effects of hERG/Eag1 channel blockers in combination with temozolomide in human glioblastoma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03955-w. [PMID: 40126672 DOI: 10.1007/s00210-025-03955-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 03/26/2025]
Abstract
Temozolomide (TMZ) concomitant with radiotherapy is the first-line treatment for glioblastoma. However, treatment resistance is frequently observed in patients. Cellular senescence (CSEN) induced by TMZ has been proposed to be one underlying mechanism resulting in resting cells, causing inflammation and possibly recurrences if senescent cells re-enter the cell cycle after treatment. Inhibition of the K+ channels human ether-à-go-go type 1 (Eag1) and human ether-à-go-go-related gene (hERG) has shown promising effects in several tumor types including glioblastoma through growth inhibition and induction of apoptosis. In the present study, we analyzed the impact of hERG/Eag1 inhibition on apoptosis and CSEN on its own and in combination with TMZ in a panel of human glioblastoma cell lines and primary glioblastoma cells. hERG/Eag1 protein expression was determined by Western blotting and immunocytochemistry. Cytotoxicity of astemizole and terfenadine alone or in combination with TMZ was assessed by MTT assays. Apoptotic yields were determined by Annexin V/propidium iodide staining, and CSEN was quantified by determining SA-β-galactosidase levels through flow cytometry. We observed a similar protein expression of hERG and Eag1 in all glioblastoma cell lines and primary glioblastoma cells. Astemizole and terfenadine were cytotoxic in glioblastoma cells at low micromolar concentrations (5-10 µM range) through induction of apoptosis. In combination with TMZ, both drugs synergistically sensitized glioblastoma cells to TMZ-induced apoptosis. Moreover, astemizole reduced significantly the TMZ-induced CSEN level, indicating its impact on CSEN induction. Here, we show for the first time that blocking hERG/Eag1 channels in glioblastoma cells can relief TMZ-induced CSEN and synergistically ameliorates cytotoxicity through the induction of apoptosis.
Collapse
Affiliation(s)
- Bodo Haas
- Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany.
| | - Inken Roth
- Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
- Faculty of Applied Natural Sciences, TH Köln - University of Applied Sciences, Campus Platz 1, 51379, Leverkusen, Germany
| | - Luisa Säcker
- Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
- Faculty of Applied Natural Sciences, TH Köln - University of Applied Sciences, Campus Platz 1, 51379, Leverkusen, Germany
| | - Maria Wos-Maganga
- Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Lea Beltzig
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| |
Collapse
|
5
|
Bauer CK, Kortüm F, Möllring A, Grinstein L, Denecke J, Alawi M, Bähring R, Harms FL. Loss-of-function variant in KCNH3 is associated with global developmental delay, autistic behavior, insomnia, and nocturnal seizures. Seizure 2025; 129:14-21. [PMID: 40157307 DOI: 10.1016/j.seizure.2025.03.014] [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: 01/10/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
INTRODUCTION The KCNH gene family encodes voltage-gated potassium (Kv) channels of the EAG subtype covering three subfamilies (Kv10-12). EAG channels are involved in the control of cardiac and neuronal excitation, and pathogenic variants in KCNH genes encoding Kv10 (eag) and Kv11 (erg) subfamily members cause a broad clinical spectrum ranging from cardiac arrhythmia to neurodevelopmental syndromes. However, no pathogenic variants have been hitherto reported for KCNH genes encoding Kv12 (elk) subfamily members. METHODS Clinical, genomic, and functional studies were performed, including voltage-clamp experiments using heterologous channel expression in Xenopus oocytes. RESULTS We examined an eight-year-old girl presenting with global developmental delay, intellectual disability, autistic and aggressive behavior, hyperactivity, insomnia, and nocturnal seizures. Focal seizures were successfully treated with sulthiame, which reduced the occurrence of temporo-parietal spike-wave paroxysms. Trio exome sequencing revealed a heterozygous de novo missense variant, NM_012284.3:c.1112C>T; p.(Ala371Val), in KCNH3, which encodes the Kv channel α-subunit Kv12.2. The amino acid substitution associated with the KCNH3 variant identified in the patient is located at a site highly conserved in EAG channels. The analogous variant in KCNH2 causes long-QT-syndrome 2, and has also been associated with epilepsy. Electrophysiological characterization of the KCNH3 p.(Ala371Val) variant demonstrated loss-of-function of the mutant Kv12.2 channels and strongly reduced current amplitudes upon co-expression of wildtype and mutant channel subunits in a dominant-negative manner. CONCLUSION Our results propose KCNH3, which is primarily expressed in the nervous system, as a new disease gene associated with a neurodevelopmental phenotype including seizures.
Collapse
Affiliation(s)
- Christiane K Bauer
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Anna Möllring
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Lev Grinstein
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany; German Center for Child and Adolescent Health (DZKJ), partner site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Jonas Denecke
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Robert Bähring
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Frederike L Harms
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.
| |
Collapse
|
6
|
Patten-Elliott F, Lei CL, Preston SP, Wilkinson RD, Mirams GR. Optimizing experimental designs for model selection of ion channel drug-binding mechanisms. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2025; 383:20240227. [PMID: 40078143 PMCID: PMC11904620 DOI: 10.1098/rsta.2024.0227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 03/14/2025]
Abstract
The rapid delayed rectifier current carried by the human Ether-à-go-go-Related Gene (hERG) channel is susceptible to drug-induced reduction, which can lead to an increased risk of cardiac arrhythmia. Establishing the mechanism by which a specific drug compound binds to hERG can help reduce uncertainty when quantifying pro-arrhythmic risk. In this study, we introduce a methodology for optimizing experimental voltage protocols to produce data that enable different proposed models for the drug-binding mechanism to be distinguished. We demonstrate the performance of this methodology via a synthetic data study. If the underlying model of hERG current is known exactly, then the optimized protocols generated show noticeable improvements in our ability to select the true model when compared with a simple protocol used in previous studies. However, if the model is not known exactly, and we assume a discrepancy between the data-generating hERG model and the hERG model used in fitting the models, then the optimized protocols become less effective in determining the 'true' binding dynamics. While the introduced methodology shows promise, we must be careful to ensure that, if applied to a real data study, we have a well-calibrated model of hERG current gating.This article is part of the theme issue 'Uncertainty quantification for healthcare and biological systems (Part 1)'.
Collapse
Affiliation(s)
- Frankie Patten-Elliott
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Chon Lok Lei
- Faculty of Health Sciences, Institute of Translational Medicine, University of Macau, Macau, People's Republic of China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Simon P Preston
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Richard D Wilkinson
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
7
|
Liu H, Jiang Z, Shen Y, Shao Y, Su Y, Wang D, Brugada R, Hong K. Topological Distribution of KCNH2 Variants and Genotype-Phenotype Relationship in Patients With Long QT Syndrome. Pacing Clin Electrophysiol 2025; 48:351-362. [PMID: 39913104 DOI: 10.1111/pace.15145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/04/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025]
Abstract
AIMS The aim of this study was to investigate the topological distribution of single nucleotide variants (SNVs) in the KCNH2 gene from patients with type 2 long QT syndrome (LQT2) and to explore the genotype-phenotype relationships. METHODS Information on KCNH2 variants in LQT2 patients was retrospectively obtained from the HGMD, ClinVar, and PubMed databases through October 2022. Pathogenicity of SNV was classified according to the American College of Medical Genetics and Genomics (ACMG) guidelines. Unpaired t-tests and Fisher's exacts were used to analyze the SNV distributions across structural and functional domains, and their correlation with clinical phenotypes. RESULTS A total of 2826 variants were obtained; 2152 were SNVs, 1328 of which were nonsynonymous SNVs (nsSNVs) associated with LQT2. Enrichment analysis revealed that 602 pathogenic (P) and likely pathogenic (LP) nsSNVs were significantly enriched at S5, H5, S6, Extra3, and Extra4. In addition, 759 nsSNVs and 289 P/LP nsSNVs within function domain were enriched at the per-arnt-sim (PAS) and selectivity filter (SF) functional domain. Clinical data revealed that patients with nsSNVs enriched at the N-terminal, S5-H5-S6 region and PAS domain were associated with an increased risk of syncope. Moreover, nsSNVs located at the N-terminal, S5-H5-S6 region, and PAS, SF domains were associated with an increased risk of life-threatening cardiac events, including Torsade de Pointes (TdP) and sudden cardiac death (SCD), and were predominantly female. CONCLUSION KCNH2 nsSNVs located at the N-terminal, S5-H5-S6 region, and the PAS and SF functional domains are associated with an increased risk of life-threatening cardiac events in LQT2 patients.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Medical Genetics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhenhong Jiang
- Molecular Medicine of Jiangxi Key Laboratory, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Yang Shen
- Department of Medical Genetics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Molecular Medicine of Jiangxi Key Laboratory, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Ying Shao
- Nanchang University Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuhao Su
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Daowu Wang
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Ramon Brugada
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain
| | - Kui Hong
- Department of Medical Genetics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Molecular Medicine of Jiangxi Key Laboratory, The Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Fiore CM, Quigley K, Vorobyov I, Clancy CE, Harvey RD. Effect of the Membrane Environment on Pharmacologic Inhibition of hERG K + Channel Activity. JACC Clin Electrophysiol 2025:S2405-500X(24)01022-3. [PMID: 39895450 DOI: 10.1016/j.jacep.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND hERG encodes KV11.1 voltage-gated K+ channels, which generate the rapidly activating delayed rectifier K+ current that contributes to repolarization of the cardiac action potential. In addition to being targeted by many class III antiarrhythmic agents, these channels are also inhibited by a multitude of other pharmacological compounds, which can produce acquired long QT syndrome, leading to polymorphic ventricular tachycardia. While most drugs are thought to interact with a hydrophilic binding site in the channel pore, it has been postulated that some compounds act by perturbing the membrane environment or acting at hydrophobic sites accessed through the plasma membrane. OBJECTIVES Because hERG channels reside in cholesterol rich lipid raft domains, we hypothesized that disrupting the membrane environment by depleting cholesterol might alter inhibition of channel activity by certain drugs. METHODS We tested our hypothesis by examining the effect that depleting membrane cholesterol with methyl-β-cyclodextrin has on the ability of several compounds to inhibit hERG channels expressed in HEK293 cells. RESULTS We found that cholesterol depletion significantly increased the sensitivity of the whole cell current to inhibition by ibutilide, while decreasing the currents sensitivity to dofetilide and amiodarone at negative membrane potentials. CONCLUSIONS These results support the idea that the lipid environment of the plasma membrane plays a role in the ability of certain drugs to inhibit hERG channel activity. Differences in membrane cholesterol content may affect the ability of some hERG channel blockers to produce arrhythmogenic behavior.
Collapse
Affiliation(s)
- Chase M Fiore
- Department of Pharmacology, University of Nevada, Reno, Reno, Nevada, USA
| | - Kate Quigley
- Department of Pharmacology, University of Nevada, Reno, Reno, Nevada, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California- Davis, Davis, California, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California- Davis, Davis, California, USA
| | - Robert D Harvey
- Department of Pharmacology, University of Nevada, Reno, Reno, Nevada, USA.
| |
Collapse
|
9
|
Yu MS, Lee J, Lee Y, Cho D, Oh KS, Jang J, Nong NT, Lee HM, Na D. hERGBoost: A gradient boosting model for quantitative IC 50 prediction of hERG channel blockers. Comput Biol Med 2025; 184:109416. [PMID: 39550914 DOI: 10.1016/j.compbiomed.2024.109416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024]
Abstract
The human ether-a-go-go-related gene (hERG) potassium channel is pivotal in drug discovery due to its susceptibility to blockage by drug candidate molecules, which can cause severe cardiotoxic effects. Consequently, identifying and excluding potential hERG channel blockers at the earliest stages of drug development is crucial. Most traditional machine learning models predict a molecule's cardiotoxicity or non-cardiotoxicity typically at 10 μM, which doesn't account for compounds with low IC50 values that are non-toxic at therapeutic levels due to their high effectiveness at lower concentrations. To address the need for more precise, quantitative predictions, we developed hERGBoost, a cutting-edge machine learning model employing a gradient-boosting algorithm. This model demonstrates superior accuracy in predicting the IC50 of drug candidates. Trained on a specially curated dataset for this study, hERGBoost not only exhibited excellent performance in external validation, achieving an R2 score of 0.394 and a low root mean square error of 0.616 but also significantly outstripped previous models in both qualitative and quantitative assessments. Representing a notable leap forward in the prediction of hERG channel blockers, the hERGBoost model and its datasets are freely available to the drug discovery community on our web server at. http://ssbio.cau.ac.kr/software/hergboost This resource promises to be invaluable in advancing safer pharmaceutical development.
Collapse
Affiliation(s)
- Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jingyu Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Yunhyeok Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Daeahn Cho
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kwang-Seok Oh
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea; Department of Medicinal and Pharmaceutical Chemistry, University of Science and Technology, Daejeon, 34129, Republic of Korea
| | - Jidon Jang
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Nuong Thi Nong
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Vistoli G, Talarico C, Vittorio S, Lunghini F, Mazzolari A, Beccari A, Pedretti A. Approaching Pharmacological Space: Events and Components. Methods Mol Biol 2025; 2834:151-169. [PMID: 39312164 DOI: 10.1007/978-1-0716-4003-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The pharmacological space comprises all the dynamic events that determine the bioactivity (and/or the metabolism and toxicity) of a given ligand. The pharmacological space accounts for the structural flexibility and property variability of the two interacting molecules as well as for the mutual adaptability characterizing their molecular recognition process. The dynamic behavior of all these events can be described by a set of possible states (e.g., conformations, binding modes, isomeric forms) that the simulated systems can assume. For each monitored state, a set of state-dependent ligand- and structure-based descriptors can be calculated. Instead of considering only the most probable state (as routinely done), the pharmacological space proposes to consider all the monitored states. For each state-dependent descriptor, the corresponding space can be evaluated by calculating various dynamic parameters such as mean and range values.The reviewed examples emphasize that the pharmacological space can find fruitful applications in structure-based virtual screening as well as in toxicity prediction. In detail, in all reported examples, the inclusion of the pharmacological space parameters enhances the resulting performances. Beneficial effects are obtained by combining both different binding modes to account for ligand mobility and different target structures to account for protein flexibility/adaptability.The proposed computational workflow that combines docking simulations and rescoring analyses to enrich the arsenal of docking-based descriptors revealed a general applicability regardless of the considered target and utilized docking engine. Finally, the EFO approach that generates consensus models by linearly combining various descriptors yielded highly performing models in all discussed virtual screening campaigns.
Collapse
Affiliation(s)
- Giulio Vistoli
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milan, Italy.
| | | | - Serena Vittorio
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milan, Italy
| | | | - Angelica Mazzolari
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milan, Italy
| | | | - Alessandro Pedretti
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milan, Italy
| |
Collapse
|
11
|
Wilders R. Alleviating the Effects of Short QT Syndrome Type 3 by Allele-Specific Suppression of the KCNJ2 Mutant Allele. Int J Mol Sci 2024; 25:13351. [PMID: 39769116 PMCID: PMC11676537 DOI: 10.3390/ijms252413351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Short QT syndrome type 3 (SQTS3 or SQT3), which is associated with life-threatening cardiac arrhythmias, is caused by heterozygous gain-of-function mutations in the KCNJ2 gene. This gene encodes the pore-forming α-subunit of the ion channel that carries the cardiac inward rectifier potassium current (IK1). These gain-of-function mutations either increase the amplitude of IK1 or attenuate its rectification. The aim of the present in silico study is to test to which extent allele-specific suppression of the KCNJ2 mutant allele can alleviate the effects of SQT3, as recently demonstrated in in vitro studies on specific heterozygous mutations associated with long QT syndrome type 1 and 2 and short QT syndrome type 1. To this end, simulations were carried out with the two most recent comprehensive models of a single human ventricular cardiomyocyte. These simulations showed that suppression of the mutant allele can, at least partially, counteract the effects of the mutation on IK1 and restore the action potential duration for each of the four SQT3 mutations that are known by now. We conclude that allele-specific suppression of the KCNJ2 mutant allele is a promising technique in the treatment of SQT3 that should be evaluated in in vitro and in vivo studies.
Collapse
Affiliation(s)
- Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
12
|
Marjanović JS, Matić JD, Milanović Ž, Divac VM, Kosanić MM, Petković MR, Kostić MD. Molecular modeling studies, in vitro antioxidant and antimicrobial assay and BSA affinity of novel benzyl-amine derived scaffolds as CYP51B inhibitors. Mol Divers 2024. [DOI: 10.1007/s11030-024-11074-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025]
|
13
|
Zhang Y, Ding Y, Zeng Z, Zhu R, Zheng P, Fan S, Cao Q, Chen H, Ren W, Wu M, Wang L, Du J. Intra-channel bi-epitopic crosslinking unleashes ultrapotent antibodies targeting Na V1.7 for pain alleviation. Cell Rep Med 2024; 5:101800. [PMID: 39461335 PMCID: PMC11604545 DOI: 10.1016/j.xcrm.2024.101800] [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: 03/19/2024] [Revised: 09/01/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Crucial for cell activities, ion channels are key drug discovery targets. Although small-molecule and peptide modulators dominate ion channel drug discovery, antibodies are emerging as an alternative modality. However, challenges persist in generating potent antibodies, especially for channels with limited extracellular epitopes. We herein present a bi-epitopic crosslinking strategy to overcome these challenges, focusing on NaV1.7, a potential analgesic target. Aiming to crosslink two non-overlapping epitopes on voltage-sensing domains II and IV, we construct bispecific antibodies and ligand-antibody conjugates. Enhanced affinity and potency are observed in comparison to the monospecific controls. Among them, a ligand-antibody conjugate (1080-PEG7-ACDTB) displays a two-orders-of-magnitude improvement in potency (IC50 of 0.06 ± 0.01 nM) and over 1,000-fold selectivity for NaV1.7. Additionally, this conjugate demonstrates robust analgesic effects in mouse pain models. Our study introduces an approach to developing effective antibodies against NaV1.7, thereby initiating a promising direction for the advancement of pain therapeutics.
Collapse
Affiliation(s)
- Yaning Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China; Peking University-Tsinghua University-National Institute Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanchao Ding
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Ziyan Zeng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Rui Zhu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Peiyuan Zheng
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Shilong Fan
- The Technology Center for Protein Sciences, Tsinghua University, Beijing 100084, China
| | - Qingjuan Cao
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Hang Chen
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Weishuai Ren
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Mengling Wu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Luyao Wang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Juanjuan Du
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
14
|
Qian X, Yao M, Xu J, Dong N, Chen S. From cancer therapy to cardiac safety: the role of proteostasis in drug-induced cardiotoxicity. Front Pharmacol 2024; 15:1472387. [PMID: 39611175 PMCID: PMC11602306 DOI: 10.3389/fphar.2024.1472387] [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: 07/29/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024] Open
Abstract
Drug-induced cardiotoxicity (DICT) poses a significant challenge in the prognosis of cancer patients, particularly with the use of antineoplastic agents like anthracyclines and targeted therapies such as trastuzumab. This review delves into the intricate interplay between drugs and proteins within cardiac cells, focusing on the role of proteostasis as a therapeutic target for mitigating cardiotoxicity. We explore the in vivo modeling of proteostasis, highlighting the complex intracellular environment and the emerging techniques for monitoring proteostasis. Additionally, we discuss how cardiotoxic drugs disrupt protein homeostasis through direct chemical denaturation, endoplasmic reticulum stress, unfolded protein response, chaperone dysfunction, impairment of the proteasome system, and dysregulation of autophagy. Finally, we provide insights into the applications of cardioprotective drugs targeting proteostasis to prevent cardiotoxicity and the adoption of structural proteomics to evaluate potential cardiotoxicity. By gaining a deeper understanding of the role of proteostasis underlying DICT, we can pave the way for the development of targeted therapeutic strategies to safeguard cardiac function while maximizing the therapeutic potential of antineoplastic drugs.
Collapse
Affiliation(s)
- Xingyu Qian
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengdong Yao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingyu Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
15
|
Miyashita Y, Moriya T, Kato T, Kawasaki M, Yasuda S, Adachi N, Suzuki K, Ogasawara S, Saito T, Senda T, Murata T. Improved higher resolution cryo-EM structures reveal the binding modes of hERG channel inhibitors. Structure 2024; 32:1926-1935.e3. [PMID: 39321803 DOI: 10.1016/j.str.2024.08.021] [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: 07/05/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
During drug discovery, it is crucial to exclude compounds with toxic effects. The human ether-à-go-go-related gene (hERG) channel is essential for maintaining cardiac repolarization and is a critical target in drug safety evaluation due to its role in drug-induced arrhythmias. Inhibition of the hERG channel can lead to severe cardiac issues, including Torsades de Pointes tachycardia. Understanding hERG inhibition mechanisms is essential to avoid these toxicities. Several structural studies have elucidated the interactions between inhibitors and hERG. However, orientation and resolution issues have so far limited detailed insights. Here, we used digitonin to analyze the apo state of hERG, which resolved orientation issues and improved the resolution. We determined the structure of hERG bound to astemizole, showing a clear map in the pore pathway. Using this strategy, we also analyzed the binding modes of E-4031 and pimozide. These insights into inhibitor interactions with hERG may aid safer drug design and enhance cardiac safety.
Collapse
Affiliation(s)
- Yasuomi Miyashita
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8670, Japan; Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Toshio Moriya
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan
| | - Takafumi Kato
- Department of Biochemistry, University of Oxford, South Parks Rd, Oxford OX13QC, UK
| | - Masato Kawasaki
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan
| | - Satoshi Yasuda
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Naruhiko Adachi
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Kano Suzuki
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Satoshi Ogasawara
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Tetsuichiro Saito
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo, Chiba 260-8670, Japan
| | - Toshiya Senda
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), 1-1 Oho, Tsukuba 305-0801, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan; Membrane Protein Research Center, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan.
| |
Collapse
|
16
|
Liao J, Yang Z, Yang J, Lin H, Chen B, Fu H, Lin X, Lu B, Gao F. Investigating the cardiotoxicity of N-n-butyl haloperidol iodide: Inhibition mechanisms on hERG channels. Toxicology 2024; 508:153916. [PMID: 39128488 DOI: 10.1016/j.tox.2024.153916] [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: 07/07/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The human Ether-à-go-go-Related Gene (hERG) encodes a protein responsible for forming the alpha subunit of the IKr channel, which plays a crucial role in cardiac repolarization. The proper functioning of hERG channels is paramount in maintaining a normal cardiac rhythm. Inhibition of these channels can result in the prolongation of the QT interval and potentially life-threatening arrhythmias. Cardiotoxicity is a primary concern in the field of drug development. N-n-Butyl haloperidol iodide (F2), a derivative of haloperidol, has been investigated for its therapeutic potential. However, the impact of this compound on cardiac toxicity, specifically on hERG channels, remains uncertain. This study employs computational and experimental methodologies to examine the inhibitory mechanisms of F2 on hERG channels. Molecular docking and molecular dynamics simulations commonly used techniques in computational biology to predict protein-ligand complexes' binding interactions and stability. In the context of the F2-hERG complex, these methods can provide valuable insights into the potential binding modes and strength of interaction between F2 and the hERG protein. On the other hand, electrophysiological assays are experimental techniques used to characterize the extent and nature of hERG channel inhibition caused by various compounds. By measuring the electrical activity of the hERG channel in response to different stimuli, these assays can provide important information about the functional effects of ligand binding to the channel. The study's key findings indicate that F2 interacts with the hERG channel by forming hydrogen bonding, π-cation interactions, and hydrophobic forces. This interaction leads to the inhibition of hERG currents in a concentration-dependent manner, with an IC50 of 3.75 μM. The results presented in this study demonstrate the potential cardiotoxicity of F2 and underscore the significance of considering hERG channel interactions during its clinical development. This study aims to provide comprehensive insights into the interaction between F2 and hERG, which will may guid us in the safe use of F2 and in the development of new derivatives with high efficiency while low toxicity.
Collapse
Affiliation(s)
- Jilin Liao
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China; Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zhenyu Yang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jinhua Yang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Hailing Lin
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Bingxuan Chen
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Hongbo Fu
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xiaojie Lin
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Binger Lu
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China; Department of Pharmacy, the First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China.
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China.
| |
Collapse
|
17
|
Ngo K, Yang PC, Yarov-Yarovoy V, Clancy CE, Vorobyov I. Harnessing AlphaFold to reveal hERG channel conformational state secrets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.577468. [PMID: 38352360 PMCID: PMC10862728 DOI: 10.1101/2024.01.27.577468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
To design safe, selective, and effective new therapies, there must be a deep understanding of the structure and function of the drug target. One of the most difficult problems to solve has been resolution of discrete conformational states of transmembrane ion channel proteins. An example is KV11.1 (hERG), comprising the primary cardiac repolarizing current, I Kr. hERG is a notorious drug anti-target against which all promising drugs are screened to determine potential for arrhythmia. Drug interactions with the hERG inactivated state are linked to elevated arrhythmia risk, and drugs may become trapped during channel closure. However, the structural details of multiple conformational states have remained elusive. Here, we guided AlphaFold2 to predict plausible hERG inactivated and closed conformations, obtaining results consistent with multiple available experimental data. Drug docking simulations demonstrated hERG state-specific drug interactions in good agreement with experimental results, revealing that most drugs bind more effectively in the inactivated state and are trapped in the closed state. Molecular dynamics simulations demonstrated ion conduction for an open but not AlphaFold2 predicted inactivated state that aligned with earlier studies. Finally, we identified key molecular determinants of state transitions by analyzing interaction networks across closed, open, and inactivated states in agreement with earlier mutagenesis studies. Here, we demonstrate a readily generalizable application of AlphaFold2 as an effective and robust method to predict discrete protein conformations, reconcile seemingly disparate data and identify novel linkages from structure to function.
Collapse
Affiliation(s)
- Khoa Ngo
- Center for Precision Medicine and Data Science, University of California, Davis, California
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - Pei-Chi Yang
- Center for Precision Medicine and Data Science, University of California, Davis, California
- Department of Physiology and Membrane Biology, University of California, Davis, California
| | - Vladimir Yarov-Yarovoy
- Center for Precision Medicine and Data Science, University of California, Davis, California
- Department of Physiology and Membrane Biology, University of California, Davis, California
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California
| | - Colleen E. Clancy
- Center for Precision Medicine and Data Science, University of California, Davis, California
- Department of Physiology and Membrane Biology, University of California, Davis, California
- Department of Pharmacology, University of California, Davis, California
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, California
- Department of Pharmacology, University of California, Davis, California
| |
Collapse
|
18
|
Schwarz JR, Freitag S, Pechmann Y, Hermans-Borgmeyer I, Wagner W, Hornig S, Kneussel M. Purkinje cell hyperexcitability and depressive-like behavior in mice lacking erg3 (ether-à-go-go-related gene) K + channel subunits. SCIENCE ADVANCES 2024; 10:eadn6836. [PMID: 39365861 PMCID: PMC11451553 DOI: 10.1126/sciadv.adn6836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 08/30/2024] [Indexed: 10/06/2024]
Abstract
Potassium channels stabilize the resting potential and neuronal excitability. Among them, erg (ether-à-go-go-related gene) K+ channels represent a subfamily of voltage-gated channels, consisting of erg1, erg2, and erg3 subunits; however, their subunit-specific neuronal functions in vivo are barely understood. To find erg3- and erg1-mediated functions, we generated global Kcnh7 (erg3) and conditional Kcnh2 (erg1) knockout mice. We found that erg3 channels stabilize the resting potential and dampen spontaneous activity in cerebellar Purkinje cells (PCs) and hippocampal CA1 neurons, whereas erg1 channels have suprathreshold functions. Lack of erg3 subunits induced hyperexcitability with increased action potential firing in PCs, but not in CA1 neurons. Notably, erg3 depletion caused depressive-like behavior with reduced locomotor activity, strongly decreased digging behavior, and shorter latencies to fall off a rotating wheel, while learning and memory remained unchanged. Our data show that erg K+ channels containing erg3 subunits mediate a neuronal subthreshold K+ current that plays important roles in the regulation of locomotor behavior in vivo.
Collapse
Affiliation(s)
- Jürgen R. Schwarz
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Sandra Freitag
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Yvonne Pechmann
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Core Facility Transgenic Animals, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Wolfgang Wagner
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Sönke Hornig
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
19
|
Wang XC, Zhou Y, Chen HX, Hou HT, He GW, Yang Q. ER stress modulates Kv1.5 channels via PERK branch in HL-1 atrial myocytes: Relevance to atrial arrhythmogenesis and the effect of tetramethylpyrazine. Heliyon 2024; 10:e37767. [PMID: 39318794 PMCID: PMC11420496 DOI: 10.1016/j.heliyon.2024.e37767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is implicated in cardiac arrhythmia whereas the associated mechanisms remain inadequately understood. Kv1.5 channels are essential for atrial repolarization. Whether ER stress affects Kv1.5 channels is unknown. This study aimed to elucidate the response of Kv1.5 channels to ER stress by clarifying the unfolded protein response (UPR) branch responsible for the channel modulation. In addition, the effect of tetramethylpyrazine (TMP) on Kv1.5 channels was studied. Patch clamp and western-blot results revealed that exposure of HL-1 atrial myocytes to ER stress inducer tunicamycin upregulates Kv1.5 expression, increases Kv1.5 channel current (I Kur ) (14.91 ± 1.11 vs. 6.11 ± 1.31 pA/pF, P < 0.001), and shortened action potential duration (APD) (APD90: 82.79 ± 5.25 vs.121.11 ± 6.72 ms, P < 0.01), which could be reverted by ER stress inhibitors. Blockade of the PERK branch while not IRE1 and ATF6 branches of UPR downregulated Kv1.5 expression, accompanied by a decreased I Kur (9.03 ± 0.99 pA/pF) and a prolonged APD90 (113.69 ± 4.41 ms) (P < 0.01). PERK-mediated increases of Kv1.5 expression and I Kur were also observed in HL-1 cells incubated with thapsigargin. TMP suppressed the enhancement of I Kur (10.52 ± 0.97 vs. 17.52 ± 2.25 pA/pF, P < 0.05), prevented the shortening of APD (APD90: 110.16 ± 5.36 vs. 84.84 ± 4.58 ms, P < 0.05), and inhibited the upregulation of Kv1.5 triggered by ER stress. Our study suggests that ER stress induces upregulation and activation of Kv1.5 channels in atrial myocytes through the PERK branch of UPR. TMP prevents Kv1.5 upregulation/activation and the resultant APD shortening by inhibiting ER stress. These results may shed light on the mechanisms of atrial arrhythmogenesis and the antiarrhythmic effect of the traditional Chinese herb TMP.
Collapse
Affiliation(s)
- Xiang-Chong Wang
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
- Department of Pharmacology, Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Hebei International Cooperation Center for Ion channel Function and Innovative Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China
- School of Medicine, Nankai University, Tianjin, 300457, China
| | - Yang Zhou
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Huan-Xin Chen
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Hai-Tao Hou
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Guo-Wei He
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Qin Yang
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| |
Collapse
|
20
|
Hansen J, Xiong Y, Siddiq MM, Dhanan P, Hu B, Shewale B, Yadaw AS, Jayaraman G, Tolentino RE, Chen Y, Martinez P, Beaumont KG, Sebra R, Vidovic D, Schürer SC, Goldfarb J, Gallo JM, Birtwistle MR, Sobie EA, Azeloglu EU, Berger SI, Chan A, Schaniel C, Dubois NC, Iyengar R. Multiscale mapping of transcriptomic signatures for cardiotoxic drugs. Nat Commun 2024; 15:7968. [PMID: 39261481 PMCID: PMC11390749 DOI: 10.1038/s41467-024-52145-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
Drug-induced gene expression profiles can identify potential mechanisms of toxicity. We focus on obtaining signatures for cardiotoxicity of FDA-approved tyrosine kinase inhibitors (TKIs) in human induced-pluripotent-stem-cell-derived cardiomyocytes, using bulk transcriptomic profiles. We use singular value decomposition to identify drug-selective patterns across cell lines obtained from multiple healthy human subjects. Cellular pathways affected by cardiotoxic TKIs include energy metabolism, contractile, and extracellular matrix dynamics. Projecting these pathways to published single cell expression profiles indicates that TKI responses can be evoked in both cardiomyocytes and fibroblasts. Integration of transcriptomic outlier analysis with whole genomic sequencing of our six cell lines enables us to correctly reidentify a genomic variant causally linked to anthracycline-induced cardiotoxicity and predict genomic variants potentially associated with TKI-induced cardiotoxicity. We conclude that mRNA expression profiles when integrated with publicly available genomic, pathway, and single cell transcriptomic datasets, provide multiscale signatures for cardiotoxicity that could be used for drug development and patient stratification.
Collapse
Affiliation(s)
- Jens Hansen
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Yuguang Xiong
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mustafa M Siddiq
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Priyanka Dhanan
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Hu
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bhavana Shewale
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arjun S Yadaw
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gomathi Jayaraman
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rosa E Tolentino
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yibang Chen
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pedro Martinez
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristin G Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dusica Vidovic
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL, 33146, USA
| | - Stephan C Schürer
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL, 33146, USA
| | - Joseph Goldfarb
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - James M Gallo
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- School of Pharmacy and Pharmaceutical Sciences, University of Buffalo SUNY System, Buffalo, NY, 14260, USA
| | - Marc R Birtwistle
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29634, USA
| | - Eric A Sobie
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Evren U Azeloglu
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New, York, NY, 10029, USA
| | - Seth I Berger
- Center for Genetic Medicine Research, Children's National Research Institute, Washington, DC, 20012, USA
| | - Angel Chan
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cardiology Division, Department of Medicine, Memorial Sloan Kettering Cancer Center New York, New York, NY, 10065, USA
| | - Christoph Schaniel
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicole C Dubois
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Ravi Iyengar
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
21
|
Strenja I, Dadić-Hero E, Perković M, Šoša I. Fentanyl and Sudden Death-A Postmortem Perspective for Diagnosing and Predicting Risk. Diagnostics (Basel) 2024; 14:1995. [PMID: 39272779 PMCID: PMC11394624 DOI: 10.3390/diagnostics14171995] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Sudden, unexpected deaths are extremely difficult for families, especially when the victim is a child. Most sudden deaths occur due to cardiovascular issues, and a smaller number (approximately one-quarter) are attributed to other causes, such as epilepsy. The medicinal and non-medicinal use of the synthetic opioid fentanyl, which can cause breathing problems, is frequently involved in these deaths. It is also being found more often in autopsies of sudden death cases, and the number of overdose deaths from illicit drugs containing fentanyl is increasing. There are cases in which it is mixed with other drugs. A gene known as the KCNH2 gene or human ether-a-go-go-related gene (hERG), involved in the heart's electrical activity, can be related to abnormal heart rhythms. This gene, along with others, may play a role in sudden deaths related to fentanyl use. In response, we have examined the scientific literature on genetic variations in the KCNH2 gene that can cause sudden death, the impact of fentanyl on this process, and the potential benefits of genetic testing for the victims to offer genetic counseling for their family members.
Collapse
Affiliation(s)
- Ines Strenja
- Department of Neurology, University Hospital Centre Rijeka, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Elizabeta Dadić-Hero
- Department of Psychiatry, University Hospital Centre Rijeka, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Manuela Perković
- Department of Pathology and Cytology, Pula General Hospital, 52000 Pula, Croatia
| | - Ivan Šoša
- Department of Anatomy, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
22
|
Benn KW, Yuan PH, Chong HK, Stylii SS, Luwor RB, French CR. hERG channel agonist NS1643 strongly inhibits invasive astrocytoma cell line SMA-560. PLoS One 2024; 19:e0309438. [PMID: 39240809 PMCID: PMC11379238 DOI: 10.1371/journal.pone.0309438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/12/2024] [Indexed: 09/08/2024] Open
Abstract
Gliomas are highly malignant brain tumours that remain refractory to treatment. Treatment is typically surgical intervention followed by concomitant temozolomide and radiotherapy; however patient prognosis remains poor. Voltage gated ion channels have emerged as novel targets in cancer therapy and inhibition of a potassium selective subtype (hERG, Kv11.1) has demonstrated antitumour activity. Unfortunately blockade of hERG has been limited by cardiotoxicity, however hERG channel agonists have produced similar chemotherapeutic benefit without significant side effects. In this study, electrophysiological recordings suggest the presence of hERG channels in the anaplastic astrocytoma cell line SMA-560, and treatment with the hERG channel agonist NS1643, resulted in a significant reduction in the proliferation of SMA-560 cells. In addition, NS1643 treatment also resulted in a reduction of the secretion of matrix metalloproteinase-9 and SMA-560 cell migration. When combined with temozolomide, an additive impact was observed, suggesting that NS1643 may be a suitable adjuvant to temozolomide and limit the invasiveness of glioma.
Collapse
Affiliation(s)
- Kieran W Benn
- Neural Dynamics Laboratory, Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick H Yuan
- Neural Dynamics Laboratory, Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Harvey K Chong
- Neural Dynamics Laboratory, Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Stanley S Stylii
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Neurosurgery, Royal Melbourne Hospital, The University of Melbourne, Victoria, Australia
| | - Rodney B Luwor
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christopher R French
- Neural Dynamics Laboratory, Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Zhuang W, Mun SY, Park M, Jeong J, Kim HR, Park H, Han ET, Han JH, Chun W, Li H, Park WS. Second-generation antipsychotic quetiapine blocks voltage-dependent potassium channels in coronary arterial smooth muscle cells. J Appl Toxicol 2024; 44:1446-1453. [PMID: 38797990 DOI: 10.1002/jat.4648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Voltage-dependent K+ (Kv) channels play an important role in restoring the membrane potential to its resting state, thereby maintaining vascular tone. In this study, native smooth muscle cells from rabbit coronary arteries were used to investigate the inhibitory effect of quetiapine, an atypical antipsychotic agent, on Kv channels. Quetiapine showed a concentration-dependent inhibition of Kv channels, with an IC50 of 47.98 ± 9.46 μM. Although quetiapine (50 μM) did not alter the steady-state activation curve, it caused a negative shift in the steady-state inactivation curve. The application of 1 and 2 Hz train steps in the presence of quetiapine significantly increased the inhibition of Kv current. Moreover, the recovery time constants from inactivation were prolonged in the presence of quetiapine, suggesting that its inhibitory action on Kv channels is use (state)-dependent. The inhibitory effects of quetiapine were not significantly affected by pretreatment with Kv1.5, Kv2.1, and Kv7 subtype inhibitors. Based on these findings, we conclude that quetiapine inhibits Kv channels in both a concentration- and use (state)-dependent manner. Given the physiological significance of Kv channels, caution is advised in the use of quetiapine as an antipsychotic due to its potential side effects on cardiovascular Kv channels.
Collapse
MESH Headings
- Quetiapine Fumarate/pharmacology
- Animals
- Rabbits
- Antipsychotic Agents/pharmacology
- Antipsychotic Agents/toxicity
- Potassium Channels, Voltage-Gated/drug effects
- Potassium Channels, Voltage-Gated/antagonists & inhibitors
- Potassium Channels, Voltage-Gated/metabolism
- Coronary Vessels/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Potassium Channel Blockers/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Male
- Dose-Response Relationship, Drug
- Membrane Potentials/drug effects
- Cells, Cultured
Collapse
Affiliation(s)
- Wenwen Zhuang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Seo-Yeong Mun
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Junsu Jeong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Hye Ryung Kim
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Hongliang Li
- Institute of Translational Medicine, Medical College, Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment for Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| |
Collapse
|
24
|
Lu X, Wang T, Hou B, Han N, Li H, Wang X, Xin J, He Y, Zhang D, Jia Z, Wei C. Shensong yangxin, a multi-functional traditional Chinese medicine for arrhythmia: A review of components, pharmacological mechanisms, and clinical applications. Heliyon 2024; 10:e35560. [PMID: 39224243 PMCID: PMC11367280 DOI: 10.1016/j.heliyon.2024.e35560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
As a common cardiovascular disease (CVD), Arrhythmia refers to any abnormality in the origin, frequency, rhythm, conduction velocity, timing, pathway, sequence, or other aspect of cardiac impulses, and it is one of the common cardiovascular diseases in clinical practice. At present, various ion channel blockers are used for treatment of arrhythmia that include Na+ ion channel blockers, K+ ion channel blockers and Ca2+ ion channel blockers. While these drugs offer benefits, they have led to a gradual increase in drug-related adverse reactions across various systems. As a result, the quest for safe and effective antiarrhythmic drugs is pressing. Recent years have seen some advancements in the treatment of ventricular arrhythmias using traditional Chinese medicine(TCM). The theory of Luobing in TCM has proposed a new drug intervention strategy of "fast and slow treatment, integrated regulation" leading to a shift in mindset from "antiarrhythmic" to "rhythm-regulating". Guided by this theory, the development of Shen Song Yang Xin Capsules (SSYX) has involved various Chinese medicinal ingredients that comprehensively regulate the myocardial electrophysiological mechanism, exerting antiarrhythmic effects on multiple ion channels and non-ion channels. Similarly, in clinical studies, evidence-based research has confirmed that SSYX combined with conventional antiarrhythmic drugs can more effectively reduce the occurrence of arrhythmias. Therefore, this article provides a comprehensive review of the composition and mechanisms of action, pharmacological components, network pharmacology analysis, and clinical applications of SSYX guided by the theory of Luobing, aiming to offer valuable insights for improved clinical management of arrhythmias and related research.
Collapse
Affiliation(s)
- Xuan Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Graduate School of Hebei Medical University, 050017, China
| | - Tongxing Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
| | - Bin Hou
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
| | - Ningxin Han
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Graduate School of Hebei Medical University, 050017, China
| | - Hongrong Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
| | - Xiaoqi Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, Hebei, China
| | - Jingjing Xin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Graduate School of Hebei Medical University, 050017, China
| | - Yanling He
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, Hebei, China
| | - Dan Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, Hebei, China
| | - Zhenhua Jia
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Shijiazhuang Compound Traditional Chinese Medicine Technology Innovation Center, Shijiazhuang, 050035, China
| | - Cong Wei
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
- Hebei Provincial Key Laboratory of Luobing, Shijiazhuang, 050035, China
| |
Collapse
|
25
|
Lau CHY, Flood E, Hunter MJ, Williams-Noonan BJ, Corbett KM, Ng CA, Bouwer JC, Stewart AG, Perozo E, Allen TW, Vandenberg JI. Potassium dependent structural changes in the selectivity filter of HERG potassium channels. Nat Commun 2024; 15:7470. [PMID: 39209832 PMCID: PMC11362469 DOI: 10.1038/s41467-024-51208-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
The fine tuning of biological electrical signaling is mediated by variations in the rates of opening and closing of gates that control ion flux through different ion channels. Human ether-a-go-go related gene (HERG) potassium channels have uniquely rapid inactivation kinetics which are critical to the role they play in regulating cardiac electrical activity. Here, we exploit the K+ sensitivity of HERG inactivation to determine structures of both a conductive and non-conductive selectivity filter structure of HERG. The conductive state has a canonical cylindrical shaped selectivity filter. The non-conductive state is characterized by flipping of the selectivity filter valine backbone carbonyls to point away from the central axis. The side chain of S620 on the pore helix plays a central role in this process, by coordinating distinct sets of interactions in the conductive, non-conductive, and transition states. Our model represents a distinct mechanism by which ion channels fine tune their activity and could explain the uniquely rapid inactivation kinetics of HERG.
Collapse
Affiliation(s)
- Carus H Y Lau
- Mark Cowley Lidwill Research Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Emelie Flood
- School of Science, RMIT University, Melbourne, VIC, Australia
| | - Mark J Hunter
- Mark Cowley Lidwill Research Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia
| | | | - Karen M Corbett
- School of Science, RMIT University, Melbourne, VIC, Australia
| | - Chai-Ann Ng
- Mark Cowley Lidwill Research Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - James C Bouwer
- Molecular Horizons and School of Chemistry and Molecular Bioscience, and ARC Centre for Cryoelectron Microscopy of Membrane Proteins, University of Wollongong, Wollongong, NSW, Australia
| | - Alastair G Stewart
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia
- Computational and Structural Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Eduardo Perozo
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Toby W Allen
- School of Science, RMIT University, Melbourne, VIC, Australia.
| | - Jamie I Vandenberg
- Mark Cowley Lidwill Research Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
26
|
Chang G, Aroge FA, Venkateshappa R, Claydon TW, Sun B. Development of an Absolute Quantification Method for hERG Using PRM with Single Isotopologue in-Sample Calibration. ACS OMEGA 2024; 9:33972-33982. [PMID: 39130540 PMCID: PMC11308013 DOI: 10.1021/acsomega.4c04541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 08/13/2024]
Abstract
The human ether-à-go-go-related gene (KCNH2)-encoded protein hERG constitutes the α subunit of the Kv11.1 channel and contributes to the I kr current, which plays an important role in the cardiac action potential. Genetically and xenobiotically triggered malfunctions of hERG can cause arrhythmia. The expression of hERG in various study systems was assessed mainly as the fold change relative to the corresponding control. Here, we developed a simple and sensitive quantitation method using targeted mass spectrometry, i.e., the parallel reaction monitoring approach, to measure the absolute quantity of hERG in copy number. Such measurements do not require controls, and the obtained values can be compared with similar results for any other protein. To effectively avoid matrix effects, we used the heavy-match-light (HML) in-sample calibration approach that requires only a single isotopologue to achieve copy-number quantitation. No significant difference was observed in the results obtained by HML and by the classic standard addition in-sample calibration approach. Using four proteotypic peptides, we quantified the average number of copies of hERG in the HEK293T heterologous expression system as 3.6 ± 0.5 × 106 copies/cell, i.e., 1 million copies/cell for the fully assembled Kv11.1 channel.
Collapse
Affiliation(s)
- Ge Chang
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Fabusuyi A. Aroge
- School
of Mechatronic Systems Engineering, Simon
Fraser University, Surrey, British Columbia V3T0A3, Canada
| | - Ravichandra Venkateshappa
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Tom W. Claydon
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Bingyun Sun
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| |
Collapse
|
27
|
Chen Z, Li N, Zhang P, Li Y, Li X. CardioDPi: An explainable deep-learning model for identifying cardiotoxic chemicals targeting hERG, Cav1.2, and Nav1.5 channels. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134724. [PMID: 38805819 DOI: 10.1016/j.jhazmat.2024.134724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
The cardiotoxic effects of various pollutants have been a growing concern in environmental and material science. These effects encompass arrhythmias, myocardial injury, cardiac insufficiency, and pericardial inflammation. Compounds such as organic solvents and air pollutants disrupt the potassium, sodium, and calcium ion channels cardiac cell membranes, leading to the dysregulation of cardiac function. However, current cardiotoxicity models have disadvantages of incomplete data, ion channels, interpretability issues, and inability of toxic structure visualization. Herein, an interpretable deep-learning model known as CardioDPi was developed, which is capable of discriminating cardiotoxicity induced by the human Ether-à-go-go-related gene (hERG) channel, sodium channel (Na_v1.5), and calcium channel (Ca_v1.5) blockade. External validation yielded promising area under the ROC curve (AUC) values of 0.89, 0.89, and 0.94 for the hERG, Na_v1.5, and Ca_v1.5 channels, respectively. The CardioDPi can be freely accessed on the web server CardioDPipredictor (http://cardiodpi.sapredictor.cn/). Furthermore, the structural characteristics of cardiotoxic compounds were analyzed and structural alerts (SAs) can be extracted using the user-friendly CardioDPi-SAdetector web service (http://cardiosa.sapredictor.cn/). CardioDPi is a valuable tool for identifying cardiotoxic chemicals that are environmental and health risks. Moreover, the SA system provides essential insights for mode-of-action studies concerning cardiotoxic compounds.
Collapse
Affiliation(s)
- Zhaoyang Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Na Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Pei Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Yan Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China.
| |
Collapse
|
28
|
Stevens-Sostre WA, Flores-Aldama L, Bustos D, Li J, Morais-Cabral JH, Delemotte L, Robertson GA. An intracellular hydrophobic nexus critical for hERG1 channel slow deactivation. Biophys J 2024; 123:2024-2037. [PMID: 38219015 PMCID: PMC11309987 DOI: 10.1016/j.bpj.2024.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/17/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Slow deactivation is a critical property of voltage-gated K+ channels encoded by the human Ether-à-go-go-Related Gene 1 (hERG). hERG1 channel deactivation is modulated by interactions between intracellular N-terminal Per-Arnt-Sim (PAS) and C-terminal cyclic nucleotide-binding homology (CNBh) domains. The PAS domain is multipartite, comprising a globular domain (gPAS; residues 26-135) and an N-terminal PAS-cap that is further subdivided into an initial unstructured "tip" (residues 1-12) and an amphipathic α-helical region (residues 13-25). Although the PAS-cap tip has long been considered the effector of slow deactivation, how its position near the gating machinery is controlled has not been elucidated. Here, we show that a triad of hydrophobic interactions among the gPAS, PAS-cap α helix, and the CNBh domains is required to support slow deactivation in hERG1. The primary sequence of this "hydrophobic nexus" is highly conserved among mammalian ERG channels but shows key differences to fast-deactivating Ether-à-go-go 1 (EAG1) channels. Combining sequence analysis, structure-directed mutagenesis, electrophysiology, and molecular dynamics simulations, we demonstrate that polar serine substitutions uncover an intermediate deactivation mode that is also mimicked by deletion of the PAS-cap α helix. Molecular dynamics simulation analyses of the serine-substituted channels show an increase in distance among the residues of the hydrophobic nexus, a rotation of the intracellular gating ring, and a retraction of the PAS-cap tip from its receptor site near the voltage sensor domain and channel gate. These findings provide compelling evidence that the hydrophobic nexus coordinates the respective components of the intracellular gating ring and positions the PAS-cap tip to control hERG1 deactivation gating.
Collapse
Affiliation(s)
- Whitney A Stevens-Sostre
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lisandra Flores-Aldama
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados Del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica Del Maule, Talca, Chile; Laboratorio de Bioinformática y Química Computacional (LBQC), Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica Del Maule, Talca, Chile
| | - Jin Li
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - João H Morais-Cabral
- Instituto de Investigação e Inovação Em Saude da Universidade Do Porto (i3S); Instituto de Biologia Molecular e Celular, Universidade Do Porto, Porto, Portugal
| | - Lucie Delemotte
- KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Gail A Robertson
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
29
|
Wang YJ, Di XJ, Zhang PP, Chen X, Williams MP, Han DY, Nashmi R, Henderson BJ, Moss FJ, Mu TW. Hsp47 promotes biogenesis of multi-subunit neuroreceptors in the endoplasmic reticulum. eLife 2024; 13:e84798. [PMID: 38963323 PMCID: PMC11257679 DOI: 10.7554/elife.84798] [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: 11/18/2022] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Protein homeostasis (proteostasis) deficiency is an important contributing factor to neurological and metabolic diseases. However, how the proteostasis network orchestrates the folding and assembly of multi-subunit membrane proteins is poorly understood. Previous proteomics studies identified Hsp47 (Gene: SERPINH1), a heat shock protein in the endoplasmic reticulum lumen, as the most enriched interacting chaperone for gamma-aminobutyric acid type A (GABAA) receptors. Here, we show that Hsp47 enhances the functional surface expression of GABAA receptors in rat neurons and human HEK293T cells. Furthermore, molecular mechanism study demonstrates that Hsp47 acts after BiP (Gene: HSPA5) and preferentially binds the folded conformation of GABAA receptors without inducing the unfolded protein response in HEK293T cells. Therefore, Hsp47 promotes the subunit-subunit interaction, the receptor assembly process, and the anterograde trafficking of GABAA receptors. Overexpressing Hsp47 is sufficient to correct the surface expression and function of epilepsy-associated GABAA receptor variants in HEK293T cells. Hsp47 also promotes the surface trafficking of other Cys-loop receptors, including nicotinic acetylcholine receptors and serotonin type 3 receptors in HEK293T cells. Therefore, in addition to its known function as a collagen chaperone, this work establishes that Hsp47 plays a critical and general role in the maturation of multi-subunit Cys-loop neuroreceptors.
Collapse
Affiliation(s)
- Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Xiao-Jing Di
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Pei-Pei Zhang
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Xi Chen
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Marnie P Williams
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Dong-Yun Han
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Raad Nashmi
- Department of Biology, University of VictoriaVictoriaCanada
| | - Brandon J Henderson
- Department of Biomedical Sciences, Marshall UniversityHuntingtonUnited States
| | - Fraser J Moss
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| |
Collapse
|
30
|
Peixoto-Neves D, Jaggar JH. Physiological functions and pathological involvement of ion channel trafficking in the vasculature. J Physiol 2024; 602:3275-3296. [PMID: 37818949 PMCID: PMC11006830 DOI: 10.1113/jp285007] [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: 08/09/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
A variety of ion channels regulate membrane potential and calcium influx in arterial smooth muscle and endothelial cells to modify vascular functions, including contractility. The current (I) generated by a population of ion channels is equally dependent upon their number (N), open probability (Po) and single channel current (i), such that I = N.PO.i. A conventional view had been that ion channels traffic to the plasma membrane in a passive manner, resulting in a static surface population. It was also considered that channels assemble with auxiliary subunits prior to anterograde trafficking of the multimeric complex to the plasma membrane. Recent studies have demonstrated that physiological stimuli can regulate the surface abundance (N) of several different ion channels in arterial smooth muscle and endothelial cells to control arterial contractility. Physiological stimuli can also regulate the number of auxiliary subunits present in the plasma membrane to modify the biophysical properties, regulatory mechanisms and physiological functions of some ion channels. Furthermore, ion channel trafficking becomes dysfunctional in the vasculature during hypertension, which negatively impacts the regulation of contractility. The temporal kinetics of ion channel and auxiliary subunit trafficking can also vary depending on the signalling mechanisms and proteins involved. This review will summarize recent work that has uncovered the mechanisms, functions and pathological modifications of ion channel trafficking in arterial smooth muscle and endothelial cells.
Collapse
Affiliation(s)
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis TN 38139
| |
Collapse
|
31
|
Richter JM, Gunaga P, Yadav N, Bora RO, Bhide R, Rajugowda N, Govindrajulu K, Godesi S, Akuthota N, Rao P, Sivaraman A, Panda M, Kaspady M, Gupta A, Mathur A, Levesque PC, Gulia J, Dokania M, Ramarao M, Kole P, Chacko S, Lentz KA, Sivaprasad Lvj S, Thatipamula RP, Sridhar S, Kamble S, Govindrajan A, Soleman SI, Gordon DA, Wexler RR, Priestley ES. Discovery of BMS-986308: A Renal Outer Medullary Potassium Channel Inhibitor for the Treatment of Heart Failure. J Med Chem 2024; 67:9731-9744. [PMID: 38807539 DOI: 10.1021/acs.jmedchem.4c00893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Recent literature reports highlight the importance of the renal outer medullary potassium (ROMK) channel in renal sodium and potassium homeostasis and emphasize the potential impact that ROMK inhibitors could have as a novel mechanism diuretic in heart failure patients. A series of piperazine-based ROMK inhibitors were designed and optimized to achieve excellent ROMK potency, hERG selectivity, and ADME properties, which led to the identification of compound 28 (BMS-986308). BMS-986308 demonstrated efficacy in the volume-loaded rat diuresis model as well as promising in vitro and in vivo profiles and was therefore advanced to clinical development.
Collapse
Affiliation(s)
- Jeremy M Richter
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashantha Gunaga
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Navnath Yadav
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajesh Onkardas Bora
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajeev Bhide
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Nagendra Rajugowda
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Kavitha Govindrajulu
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sreenivasulu Godesi
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Nagarjuna Akuthota
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Prasanna Rao
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Aneesh Sivaraman
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoranjan Panda
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Mahammed Kaspady
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Anuradha Gupta
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Arvind Mathur
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Paul C Levesque
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Jyoti Gulia
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoj Dokania
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manjunath Ramarao
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashant Kole
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Silvi Chacko
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Kimberley A Lentz
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Sankara Sivaprasad Lvj
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | | | - Srikanth Sridhar
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Shyam Kamble
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Arun Govindrajan
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sharif I Soleman
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - David A Gordon
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Ruth R Wexler
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - E Scott Priestley
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| |
Collapse
|
32
|
Ihsan MF, Kawashima D, Li S, Ogasawara S, Murata T, Takei M. Non-invasive hERG channel screening based on electrical impedance tomography and extracellular voltage activation (EIT-EVA). LAB ON A CHIP 2024; 24:3183-3190. [PMID: 38828904 DOI: 10.1039/d4lc00230j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
hERG channel screening has been achieved based on electrical impedance tomography and extracellular voltage activation (EIT-EVA) to improve the non-invasive aspect of drug discovery. EIT-EVA screens hERG channels by considering the change in extracellular ion concentration which modifies the extracellular resistance in cell suspension. The rate of ion passing in cell suspension is calculated from the extracellular resistance Rex, which is obtained from the EIT measurement at a frequency of 500 kHz. In the experiment, non-invasive screening is applied by a novel integrated EIT-EVA printed circuit board (PCB) sensor to human embryonic kidney (HEK) 293 cells transfected with the human ether-a-go-go-related gene (hERG) ion channel, while the E-4031 antiarrhythmic drug is used for hERG channel inhibition. The extracellular resistance Rex of the HEK 293 cells suspension is measured by EIT as the hERG channels are activated by EVA over time. The Rex is reconstructed into extracellular conductivity distribution change Δσ to reflect the extracellular K+ ion concentration change Δc resulting from the activated hERG channel. Δc is increased rapidly during the hERG channel non-inhibition state while Δc is increased slower with increasing drug concentration cd. In order to evaluate the EIT-EVA system, the inhibitory ratio index (IR) was calculated based on the rate of Δc over time. Half-maximal inhibitory concentration (IC50) of 2.7 nM is obtained from the cd and IR dose-response relationship. The IR from EIT-EVA is compared with the results from the patch-clamp method, which gives R2 of 0.85. In conclusion, EIT-EVA is successfully applied to non-invasive hERG channel screening.
Collapse
Affiliation(s)
- Muhammad Fathul Ihsan
- Department of Mechanical Engineering, Graduate School of Science and Engineering, Division of Fundamental Engineering, Chiba University, Chiba 263-8522, Japan
| | - Daisuke Kawashima
- Graduate School of Engineering, Chiba University, Chiba 263-8522, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba 263-8522, Japan.
| | - Songshi Li
- Graduate School of Engineering, Chiba University, Chiba 263-8522, Japan
| | - Satoshi Ogasawara
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
- Molecular Chirality Research Center, Chiba University, Chiba 263-8522, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
- Molecular Chirality Research Center, Chiba University, Chiba 263-8522, Japan
| | - Masahiro Takei
- Graduate School of Engineering, Chiba University, Chiba 263-8522, Japan
| |
Collapse
|
33
|
Ng WM, Wu SN, Huang BM, So EC. Investigating the influence of XAV-939, a tankyrase inhibitor, on the density and gating of erg-mediated K + currents in mouse MA-10 Leydig tumor cells. Eur J Pharmacol 2024; 971:176518. [PMID: 38556119 DOI: 10.1016/j.ejphar.2024.176518] [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: 07/23/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
XAV-939(XAV) is a chemical compound that inhibits the activity of tankyrase. However, the precise way in which XAV alters membrane ionic currents is not well understood. In this study,our goal was to examine the impact of XAV on the ionic currents in mouse MA-10 Leydig cells, specifically focusing on the magnitude, gating properties,and voltage-dependent hysteresis of erg-mediated K+currents(IK(erg)). In our whole-cell current recordings we observed that the addition of XAV inhibited the density of IK(erg) in a concentration-dependent manner with an IC50 of 3.1 μM. Furthermore we found that continued exposure to XAV, further addition of neither liraglutide nor insulin-like growth factor-1 counteracted XAV-mediated inhibition of IK(erg). Additionally the presence of XAV suppressed the mean current versus voltage relationship of IK(erg) across the entire voltage-clamp step analyzed. This compound shifted the steady-state activation curve of IK(erg) to a less negative potential by approximately 12 mV. The presence of XAV increased the time constant of deactivating IK(erg) in MA-10 cells. The voltage-dependent clockwise hysteresis of IK(erg) responding to prolonged upright isosceles-triangular ramp voltage became diminished by adding XAV; moreover subsequent addition of NS3623 effectively reversed XAV-induced decrease of hysteretic area of IK(erg). XAV also inhibited the proliferation of this cell line and the IC50 value of XAV-induced inhibition of cell proliferation was 2.8M. Overall the suppression of IK(erg) by XAV may serve as a significant ionic mechanism that contribute to the functional properties of MA-10 cells. However, it is important to note that this effect cannot be attributed solely to the inhibition of tankyrase.
Collapse
Affiliation(s)
- Woei-Ming Ng
- Department of Urology, An Nan Hospital, China Medical University, Tainan, 70965, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan; School of Medicine, National Sun-Yat Sen University College of Medicine, Kaohsiung, Taiwan; Department of Medical Education and Research, An Nan Hospital, China Medical University, Tainan, Taiwan.
| | - Bu-Miin Huang
- Department of Cell Biology and Anatomy, National Cheng Kung University College of Medicine, Tainan, 70101, Taiwan.
| | - Edmund Cheung So
- Department of Anesthesia, An Nan Hospital, China Medical University, Tainan, 70965, Taiwan.
| |
Collapse
|
34
|
Tikhomirov R, Oakley RH, Anderson C, Xiang Y, Al-Othman S, Smith M, Yaar S, Torre E, Li J, Wilson LR, Goulding DR, Donaldson I, Harno E, Soattin L, Shiels HA, Morris GM, Zhang H, Boyett MR, Cidlowski JA, Mesirca P, Mangoni ME, D’Souza A. Cardiac GR Mediates the Diurnal Rhythm in Ventricular Arrhythmia Susceptibility. Circ Res 2024; 134:1306-1326. [PMID: 38533639 PMCID: PMC11081863 DOI: 10.1161/circresaha.123.323464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/15/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Ventricular arrhythmias (VAs) demonstrate a prominent day-night rhythm, commonly presenting in the morning. Transcriptional rhythms in cardiac ion channels accompany this phenomenon, but their role in the morning vulnerability to VAs and the underlying mechanisms are not understood. We investigated the recruitment of transcription factors that underpins transcriptional rhythms in ion channels and assessed whether this mechanism was pertinent to the heart's intrinsic diurnal susceptibility to VA. METHODS AND RESULTS Assay for transposase-accessible chromatin with sequencing performed in mouse ventricular myocyte nuclei at the beginning of the animals' inactive (ZT0) and active (ZT12) periods revealed differentially accessible chromatin sites annotating to rhythmically transcribed ion channels and distinct transcription factor binding motifs in these regions. Notably, motif enrichment for the glucocorticoid receptor (GR; transcriptional effector of corticosteroid signaling) in open chromatin profiles at ZT12 was observed, in line with the well-recognized ZT12 peak in circulating corticosteroids. Molecular, electrophysiological, and in silico biophysically-detailed modeling approaches demonstrated GR-mediated transcriptional control of ion channels (including Scn5a underlying the cardiac Na+ current, Kcnh2 underlying the rapid delayed rectifier K+ current, and Gja1 responsible for electrical coupling) and their contribution to the day-night rhythm in the vulnerability to VA. Strikingly, both pharmacological block of GR and cardiomyocyte-specific genetic knockout of GR blunted or abolished ion channel expression rhythms and abolished the ZT12 susceptibility to pacing-induced VA in isolated hearts. CONCLUSIONS Our study registers a day-night rhythm in chromatin accessibility that accompanies diurnal cycles in ventricular myocytes. Our approaches directly implicate the cardiac GR in the myocyte excitability rhythm and mechanistically link the ZT12 surge in glucocorticoids to intrinsic VA propensity at this time.
Collapse
Affiliation(s)
- Roman Tikhomirov
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
- Myocardial Function Section, National Heart and Lung Institute, Imperial College London, United Kingdom (R.T., M.S., A.D.)
| | - Robert H. Oakley
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (R.H.O., J.L., L.R.W., D.R.G., J.A.C.)
| | - Cali Anderson
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
| | - Yirong Xiang
- Department of Physics and Astronomy (Y.X., H.Z.), The University of Manchester, United Kingdom
| | - Sami Al-Othman
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
| | - Matthew Smith
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
- Myocardial Function Section, National Heart and Lung Institute, Imperial College London, United Kingdom (R.T., M.S., A.D.)
| | - Sana Yaar
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
| | - Eleonora Torre
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), F-34094 Montpellier France (E.T., P.M., M.E.M.)
| | - Jianying Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (R.H.O., J.L., L.R.W., D.R.G., J.A.C.)
| | - Leslie R. Wilson
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (R.H.O., J.L., L.R.W., D.R.G., J.A.C.)
| | - David R. Goulding
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (R.H.O., J.L., L.R.W., D.R.G., J.A.C.)
| | - Ian Donaldson
- Bioinformatics Core Facility (I.D.), The University of Manchester, United Kingdom
| | - Erika Harno
- Division of Diabetes, Endocrinology and Gastroenterology (E.H.), The University of Manchester, United Kingdom
| | - Luca Soattin
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
| | - Holly A. Shiels
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
| | - Gwilym M. Morris
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
- Department of Cardiology, John Hunter Hospital, Newcastle, NSW, Australia (G.M.M.)
| | - Henggui Zhang
- Department of Physics and Astronomy (Y.X., H.Z.), The University of Manchester, United Kingdom
| | - Mark R. Boyett
- Faculty of Life Sciences, University of Bradford, United Kingdom (M.R.B.)
| | - John A. Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (R.H.O., J.L., L.R.W., D.R.G., J.A.C.)
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), F-34094 Montpellier France (E.T., P.M., M.E.M.)
| | - Matteo E. Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), F-34094 Montpellier France (E.T., P.M., M.E.M.)
| | - Alicia D’Souza
- Division of Cardiovascular Sciences (R.T., C.A., S.A.O., M.S., S.Y., L.S., H.A.S., G.M.M., A.D.), The University of Manchester, United Kingdom
- Myocardial Function Section, National Heart and Lung Institute, Imperial College London, United Kingdom (R.T., M.S., A.D.)
| |
Collapse
|
35
|
Camara MD, Zhou Y, Dara A, Tékété MM, Nóbrega de Sousa T, Sissoko S, Dembélé L, Ouologuem N, Hamidou Togo A, Alhousseini ML, Fofana B, Sagara I, Djimde AA, Gil PJ, Lauschke VM. Population-specific variations in KCNH2 predispose patients to delayed ventricular repolarization upon dihydroartemisinin-piperaquine therapy. Antimicrob Agents Chemother 2024; 68:e0139023. [PMID: 38546223 PMCID: PMC11064487 DOI: 10.1128/aac.01390-23] [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: 11/02/2023] [Accepted: 03/05/2024] [Indexed: 05/03/2024] Open
Abstract
Dihydroartemisinin-piperaquine is efficacious for the treatment of uncomplicated malaria and its use is increasing globally. Despite the positive results in fighting malaria, inhibition of the Kv11.1 channel (hERG; encoded by the KCNH2 gene) by piperaquine has raised concerns about cardiac safety. Whether genetic factors could modulate the risk of piperaquine-mediated QT prolongations remained unclear. Here, we first profiled the genetic landscape of KCNH2 variability using data from 141,614 individuals. Overall, we found 1,007 exonic variants distributed over the entire gene body, 555 of which were missense. By optimizing the gene-specific parametrization of 16 partly orthogonal computational algorithms, we developed a KCNH2-specific ensemble classifier that identified a total of 116 putatively deleterious missense variations. To evaluate the clinical relevance of KCNH2 variability, we then sequenced 293 Malian patients with uncomplicated malaria and identified 13 variations within the voltage sensing and pore domains of Kv11.1 that directly interact with channel blockers. Cross-referencing of genetic and electrocardiographic data before and after piperaquine exposure revealed that carriers of two common variants, rs1805121 and rs41314375, experienced significantly higher QT prolongations (ΔQTc of 41.8 ms and 61 ms, respectively, vs 14.4 ms in controls) with more than 50% of carriers having increases in QTc >30 ms. Furthermore, we identified three carriers of rare population-specific variations who experienced clinically relevant delayed ventricular repolarization. Combined, our results map population-scale genetic variability of KCNH2 and identify genetic biomarkers for piperaquine-induced QT prolongation that could help to flag at-risk patients and optimize efficacy and adherence to antimalarial therapy.
Collapse
Affiliation(s)
- Mahamadou D. Camara
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Antoine Dara
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Mamadou M. Tékété
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Taís Nóbrega de Sousa
- Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Molecular Biology and Malaria Immunology Research Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Sékou Sissoko
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Laurent Dembélé
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Nouhoun Ouologuem
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Amadou Hamidou Togo
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Mohamed L. Alhousseini
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Bakary Fofana
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Issaka Sagara
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Abdoulaye A. Djimde
- Department of Epidemiology of Parasitic Diseases, Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies, Bamako, Mali
| | - Pedro J. Gil
- Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, Nova University of Lisbon, Lisbon, Portugal
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| |
Collapse
|
36
|
Osorio LA, Lozano M, Soto P, Moreno-Hidalgo V, Arévalo-Gil A, Ramírez-Balaguera A, Hevia D, Cifuentes J, Hidalgo Y, Alcayaga-Miranda F, Pasten C, Morales D, Varela D, Urquidi C, Iturriaga A, Rivera-Palma A, Larrea-Gómez R, Irarrázabal CE. Levels of Small Extracellular Vesicles Containing hERG-1 and Hsp47 as Potential Biomarkers for Cardiovascular Diseases. Int J Mol Sci 2024; 25:4913. [PMID: 38732154 PMCID: PMC11084293 DOI: 10.3390/ijms25094913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/03/2024] [Accepted: 04/07/2024] [Indexed: 05/13/2024] Open
Abstract
The diagnosis of cardiovascular disease (CVD) is still limited. Therefore, this study demonstrates the presence of human ether-a-go-go-related gene 1 (hERG1) and heat shock protein 47 (Hsp47) on the surface of small extracellular vesicles (sEVs) in human peripheral blood and their association with CVD. In this research, 20 individuals with heart failure and 26 participants subjected to cardiac stress tests were enrolled. The associations between hERG1 and/or Hsp47 in sEVs and CVD were established using Western blot, flow cytometry, electron microscopy, ELISA, and nanoparticle tracking analysis. The results show that hERG1 and Hsp47 were present in sEV membranes, extravesicularly exposing the sequences 430AFLLKETEEGPPATE445 for hERG1 and 169ALQSINEWAAQTT- DGKLPEVTKDVERTD196 for Hsp47. In addition, upon exposure to hypoxia, rat primary cardiomyocytes released sEVs into the media, and human cardiomyocytes in culture also released sEVs containing hERG1 (EV-hERG1) and/or Hsp47 (EV-Hsp47). Moreover, the levels of sEVs increased in the blood when cardiac ischemia was induced during the stress test, as well as the concentrations of EV-hERG1 and EV-Hsp47. Additionally, the plasma levels of EV-hERG1 and EV-Hsp47 decreased in patients with decompensated heart failure (DHF). Our data provide the first evidence that hERG1 and Hsp47 are present in the membranes of sEVs derived from the human cardiomyocyte cell line, and also in those isolated from human peripheral blood. Total sEVs, EV-hERG1, and EV-Hsp47 may be explored as biomarkers for heart diseases such as heart failure and cardiac ischemia.
Collapse
Affiliation(s)
- Luis A. Osorio
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Mauricio Lozano
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Paola Soto
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Viviana Moreno-Hidalgo
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Angely Arévalo-Gil
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Angie Ramírez-Balaguera
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Daniel Hevia
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Jorge Cifuentes
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
| | - Yessia Hidalgo
- Laboratory of Nano-Regenerative Medicine, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile
| | - Consuelo Pasten
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile
| | - Danna Morales
- Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Diego Varela
- Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Cinthya Urquidi
- Department of Epidemiology and Health Studies, Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Andrés Iturriaga
- Departamento de Matemática y Ciencia de la Computación, Facultad de Ciencia, Universidad de Santiago de Chile, Santiago 9170020, Chile
| | | | | | - Carlos E. Irarrázabal
- Laboratory of Molecular and Integrative Physiology, Physiology Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago 7620001, Chile; (L.A.O.); (C.P.)
- Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile
| |
Collapse
|
37
|
Wei S, Xiao J, Ju F, Liu J, Hu Z. A review on the pharmacology, pharmacokinetics and toxicity of sophocarpine. Front Pharmacol 2024; 15:1353234. [PMID: 38746009 PMCID: PMC11092382 DOI: 10.3389/fphar.2024.1353234] [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: 12/10/2023] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
Sophocarpine is a natural compound that belongs to the quinolizidine alkaloid family, and has a long history of use and widespread distribution in traditional Chinese herbal medicines such as Sophora alopecuroides L., Sophora flavescens Ait., and Sophora subprostrata. This article aims to summarize the pharmacology, pharmacokinetics, and toxicity of sophocarpine, evaluate its potential pharmacological effects in various diseases, and propose the necessity for further research and evaluation to promote its clinical application. A large number of studies have shown that it has anti-inflammatory, analgesic, antiviral, antiparasitic, anticancer, endocrine regulatory, and organ-protective effects as it modulates various signaling pathways, such as the NF-κB, MAPK, PI3K/AKT, and AMPK pathways. The distribution of sophocarpine in the body conforms to a two-compartment model, and sophocarpine can be detected in various tissues with a relatively short half-life. Although the pharmacological effects of sophocarpine have been confirmed, toxicity and safety assessments and reports on molecular mechanisms of its pharmacological actions have been limited. Given its significant pharmacological effects and potential clinical value, further research and evaluation are needed to promote the clinical application of sophocarpine.
Collapse
Affiliation(s)
- Shichao Wei
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junshen Xiao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Ju
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoyang Hu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
38
|
Cheng YJ, Wu Y, Wei HQ, Liao YJ, Qu LP, Pan YH, Liu LJ, Bi WT. A novel mutation in hERG gene associated with azithromycin-induced acquired long QT syndrome. Mol Biol Rep 2024; 51:520. [PMID: 38625436 DOI: 10.1007/s11033-024-09421-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Mutations in human ether-à-go-go-related gene (hERG) potassium channels are closely associated with long QT syndrome (LQTS). Previous studies have demonstrated that macrolide antibiotics increase the risk of cardiovascular diseases. To date, the mechanisms underlying acquired LQTS remain elusive. METHODS A novel hERG mutation I1025N was identified in an azithromycin-treated patient with acquired long QT syndrome via Sanger sequencing. The mutant I1025N plasmid was transfected into HEK-293 cells, which were subsequently incubated with azithromycin. The effect of azithromycin and mutant I1025N on the hERG channel was evaluated via western blot, immunofluorescence, and electrophysiology techniques. RESULTS The protein expression of the mature hERG protein was down-regulated, whereas that of the immature hERG protein was up-regulated in mutant I1025N HEK-293 cells. Azithromycin administration resulted in a negative effect on the maturation of the hERG protein. Additionally, the I1025N mutation exerted an inhibitory effect on hERG channel current. Moreover, azithromycin inhibited hERG channel current in a concentration-dependent manner. The I1025N mutation and azithromycin synergistically decreased hERG channel expression and hERG current. However, the I1025N mutation and azithromycin did not alter channel gating dynamics. CONCLUSIONS These findings suggest that hERG gene mutations might be involved in the genetic susceptibility mechanism underlying acquired LQTS induced by azithromycin.
Collapse
Affiliation(s)
- Yun-Jiu Cheng
- Department of Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yang Wu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Hui-Qiang Wei
- Department of Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Jian Liao
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Li-Ping Qu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Yue-Han Pan
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China
| | - Li-Juan Liu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Sun Yat-sen University, Guangzhou, China.
| | - Wen-Tao Bi
- Department of Cardiovascular Medicine, People's Hospital of Macheng City, Macheng, China.
| |
Collapse
|
39
|
Lei CL, Whittaker DG, Mirams GR. The impact of uncertainty in hERG binding mechanism on in silico predictions of drug-induced proarrhythmic risk. Br J Pharmacol 2024; 181:987-1004. [PMID: 37740435 DOI: 10.1111/bph.16250] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND AND PURPOSE Drug-induced reduction of the rapid delayed rectifier potassium current carried by the human Ether-à-go-go-Related Gene (hERG) channel is associated with increased risk of arrhythmias. Recent updates to drug safety regulatory guidelines attempt to capture each drug's hERG binding mechanism by combining in vitro assays with in silico simulations. In this study, we investigate the impact on in silico proarrhythmic risk predictions due to uncertainty in the hERG binding mechanism and physiological hERG current model. EXPERIMENTAL APPROACH Possible pharmacological binding models were designed for the hERG channel to account for known and postulated small molecule binding mechanisms. After selecting a subset of plausible binding models for each compound through calibration to available voltage-clamp electrophysiology data, we assessed their effects, and the effects of different physiological models, on proarrhythmic risk predictions. KEY RESULTS For some compounds, multiple binding mechanisms can explain the same data produced under the safety testing guidelines, which results in different inferred binding rates. This can result in substantial uncertainty in the predicted torsade risk, which often spans more than one risk category. By comparison, we found that the effect of a different hERG physiological current model on risk classification was subtle. CONCLUSION AND IMPLICATIONS The approach developed in this study assesses the impact of uncertainty in hERG binding mechanisms on predictions of drug-induced proarrhythmic risk. For some compounds, these results imply the need for additional binding data to decrease uncertainty in safety-critical applications.
Collapse
Affiliation(s)
- Chon Lok Lei
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Dominic G Whittaker
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
40
|
Zhang N, Song GY, Yu QH, Fan XM, Zhang WS, Hu YJ, Chao TZ, Wu YY, Duan SY, Wang F, Du RP, Xu P. Evaluation of the lncRNA-miRNA-mRNA ceRNA network in lungs of miR-147 -/- mice. Front Pharmacol 2024; 15:1335374. [PMID: 38510653 PMCID: PMC10953689 DOI: 10.3389/fphar.2024.1335374] [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: 11/08/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Background: Previous studies have documented important roles for microRNA-147 (miR-147) in inflammation, radiation-induced injury, cancer, and a range of other diseases. Murine lungs exhibit high levels of miRNA, mRNA, and lncRNA expression. However, very little research to date has focused on the lncRNA-miRNA-mRNA competing endogenous RNA (ceRNA) networks associated with miR-147, and the regulation of lncRNAs and miRNAs in this setting remains poorly understood. Methods: After establishing a miR-147-/- model mouse, samples of lung tissue were harvested for RNA-sequencing, and differentially expressed lncRNAs, miRNAs, and mRNAs were identified. The miRNA targets of these lncRNAs and the identified miRNAs were first overlapped to facilitate the prediction of target mRNAs, with analyses then examining the overlap between these targets and mRNAs that were differentially expressed. Then, these target mRNAs were subjected to pathway enrichment analyses. These results were ultimately used to establish a miR-147-related ceRNA network. Results: Relative to wild-type mice, the lungs of miR-147-/- mice exhibited 91, 43, and 71 significantly upregulated lncRNAs, miRNAs, and mRNAs, respectively, together with 114, 31, and 156 that were significantly downregulated. The lncRNA-miRNA-mRNA network established based on these results led to the identification of Kcnh6 as a differentially expressed hub gene candidate and enabled the identification of a range of regulatory relationships. KEGG pathway enrichment showed that the mRNA targets of differentially expressed lncRNAs and miRNAs in the mice were associated with tumor-related signaling, endometrial cancer, bladder cancer, and ErbB signaling. Conclusion: These results suggest that the identified ceRNA network in miR-147-/- mice shapes tumor-associated signaling activity, with miR-147 potentially regulating various lncRNAs and miRNAs through Kcnh6, ultimately influencing tumorigenesis. Future studies of the lncRNA, miRNA, and mRNA regulatory targets shown to be associated with miR-147 in the present study may ultimately lead to the identification of novel clinically relevant targets through which miR-147 shapes the pathogenesis of cancer and other diseases.
Collapse
Affiliation(s)
- Nan Zhang
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Gui-Yuan Song
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China
| | - Qing-Hua Yu
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Xin-Ming Fan
- Department of Radiotherapy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Wen-Shuo Zhang
- Department of Radiotherapy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Yong-Jian Hu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Tian-Zhu Chao
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Yao-Yao Wu
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Shu-Yan Duan
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Fei Wang
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Rui-Peng Du
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Ping Xu
- Laboratory of Radiation-Induced Diseases and Molecule-Targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| |
Collapse
|
41
|
Lei M, Salvage SC, Jackson AP, Huang CLH. Cardiac arrhythmogenesis: roles of ion channels and their functional modification. Front Physiol 2024; 15:1342761. [PMID: 38505707 PMCID: PMC10949183 DOI: 10.3389/fphys.2024.1342761] [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: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024] Open
Abstract
Cardiac arrhythmias cause significant morbidity and mortality and pose a major public health problem. They arise from disruptions in the normally orderly propagation of cardiac electrophysiological activation and recovery through successive cardiomyocytes in the heart. They reflect abnormalities in automaticity, initiation, conduction, or recovery in cardiomyocyte excitation. The latter properties are dependent on surface membrane electrophysiological mechanisms underlying the cardiac action potential. Their disruption results from spatial or temporal instabilities and heterogeneities in the generation and propagation of cellular excitation. These arise from abnormal function in their underlying surface membrane, ion channels, and transporters, as well as the interactions between them. The latter, in turn, form common regulatory targets for the hierarchical network of diverse signaling mechanisms reviewed here. In addition to direct molecular-level pharmacological or physiological actions on these surface membrane biomolecules, accessory, adhesion, signal transduction, and cytoskeletal anchoring proteins modify both their properties and localization. At the cellular level of excitation-contraction coupling processes, Ca2+ homeostatic and phosphorylation processes affect channel activity and membrane excitability directly or through intermediate signaling. Systems-level autonomic cellular signaling exerts both acute channel and longer-term actions on channel expression. Further upstream intermediaries from metabolic changes modulate the channels both themselves and through modifying Ca2+ homeostasis. Finally, longer-term organ-level inflammatory and structural changes, such as fibrotic and hypertrophic remodeling, similarly can influence all these physiological processes with potential pro-arrhythmic consequences. These normal physiological processes may target either individual or groups of ionic channel species and alter with particular pathological conditions. They are also potentially alterable by direct pharmacological action, or effects on longer-term targets modifying protein or cofactor structure, expression, or localization. Their participating specific biomolecules, often clarified in experimental genetically modified models, thus constitute potential therapeutic targets. The insights clarified by the physiological and pharmacological framework outlined here provide a basis for a recent modernized drug classification. Together, they offer a translational framework for current drug understanding. This would facilitate future mechanistically directed therapeutic advances, for which a number of examples are considered here. The latter are potentially useful for treating cardiac, in particular arrhythmic, disease.
Collapse
Affiliation(s)
- Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samantha C. Salvage
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Antony P. Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
42
|
Arcangeli A, Iorio J, Duranti C. Targeting the hERG1 and β1 integrin complex for cancer treatment. Expert Opin Ther Targets 2024; 28:145-157. [PMID: 38372580 DOI: 10.1080/14728222.2024.2318449] [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: 10/02/2023] [Accepted: 02/09/2024] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Despite great advances, novel therapeutic targets and strategies are still needed, in particular for some carcinomas in the metastatic stage (breast cancer, colorectal cancer, pancreatic ductal adenocarcinoma and the clear cell renal carcinoma). Ion channels may be considered good cancer biomarkers and targets for antineoplastic therapy. These concepts are particularly relevant considering the hERG1 potassium channel as a novel target for antineoplastic therapy. AREAS COVERED A great deal of evidence demonstrates that hERG1 is aberrantly expressed in human cancers, in particular in aggressive carcinomas. A relevant cornerstone was the discovery that, in cancer cells, the channel is present in a very peculiar conformation, strictly bound to the β1 subunit of integrin receptors. The hERG1/β1 integrin complex does not occur in the heart. Starting from this evidence, we developed a novel single chain bispecific antibody (scDb-hERG1-β1), which specifically targets the hERG1/β1 integrin complex and exerts antineoplastic effects in preclinical experiments. EXPERT OPINION Since hERG1 blockade cannot be pursued for antineoplastic therapy due to the severe cardiac toxic effects (ventricular arrhythmias) that many hERG1 blockers exert, different strategies must be identified to specifically target hERG1 in cancer. The targeting of the hERG1/β1 integrin complex through the bispecific antibody scDb-hERG1-β1 can overcome such hindrances.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Firenze, Italy
- CSDC (Center for the Study of complex dynamics), University of Florence, Sesto Fiorentino (FI), Italy
- MCK Therapeutics srl, Pistoia (PT), Italy
| | - Jessica Iorio
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Firenze, Italy
| | - Claudia Duranti
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, Firenze, Italy
- MCK Therapeutics srl, Pistoia (PT), Italy
| |
Collapse
|
43
|
TeBay C, Vandenberg JI. The real-world incidence of severe QT prolongation in patients taking antipsychotic drugs. Heart Rhythm 2024; 21:329-330. [PMID: 38231169 DOI: 10.1016/j.hrthm.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 01/18/2024]
Affiliation(s)
- Clifford TeBay
- Mark Cowley Lidwill Research Program in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jamie I Vandenberg
- Mark Cowley Lidwill Research Program in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
44
|
Tiakouang EN, Ewonkem MB, Moto JO, Adjieufack AI, Deussom PM, Mbock MA, Ngeufa EH, Toze AFA, Wansi DJ. Synthesis, antimicrobial properties and in silico evaluation of coumarin derivatives mediated by 1,4-dibromobutane. J Biomol Struct Dyn 2024:1-14. [PMID: 38411010 DOI: 10.1080/07391102.2024.2321507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/15/2024] [Indexed: 02/28/2024]
Abstract
In this study, monobrominated coumarins (5-6) and bis-coumarins (7-9) were synthesized from 3-carboxylic coumarin and 7-hydroxy-4-methyl coumarin using 1,4-dibromobutane as a binding agent, according to the synthesis procedures described in the literature. Amongst these coumarins, three are new compounds: monobrominated coumarin 5 and bis-coumarins 7 and 9. The structures of the synthesized coumarins were confirmed by FTIR, NMR and HRMS-ESI. In vitro antimicrobial evaluation of these coumarins against strains of twelve bacteria and four fungi revealed their bactericidal and fungicidal properties, with increased antibacterial activity for monocoumarins and improved antifungal activity for bis-coumarins. It was also found that the antibacterial activity was enhanced by the etheric bond, Br atom and alkyl chain and reduced by the ester bonds at position 3 of the pyrone ring or an additional coumarin unit, while the antifungal activity was reinforced by ester bonds and deactivated by the Br atom. For the first time, the in silico investigations of such coumarins were carried out and it was observed that they are less toxic, suitable for oral administration with good permeability through cell membrane, are able to circulate freely in the bloodstream and cross Blood-Brain-Barriers. Moreover, their molecular docking in DNA indicated stable coumarin-DNA complexes with good scores. The results of molecular dynamics simulations performed for 200 ns revealed the rigidity and stability of bis-coumarins (7-9) in the DNA binding pocket and predict that they are potent binders.
Collapse
Affiliation(s)
- Eunice N Tiakouang
- Department of Chemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Monique B Ewonkem
- Department of Chemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Jean O Moto
- Department of Chemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Abel I Adjieufack
- Physical and Theoretical Chemistry Laboratory, University of Yaoundé 1, Yaoundé, Cameroon
| | - Pascaline M Deussom
- Department of Chemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Michel A Mbock
- Department of Biochemistry, Faculty of Science, Laboratory of Biochemistry, University of Douala, Douala, Cameroon
| | - Emmanuel H Ngeufa
- Department of Chemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Alfred F A Toze
- Department of Chemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Duplex J Wansi
- Department of Chemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| |
Collapse
|
45
|
Seal S, Spjuth O, Hosseini-Gerami L, García-Ortegón M, Singh S, Bender A, Carpenter AE. Insights into Drug Cardiotoxicity from Biological and Chemical Data: The First Public Classifiers for FDA Drug-Induced Cardiotoxicity Rank. J Chem Inf Model 2024; 64:1172-1186. [PMID: 38300851 PMCID: PMC10900289 DOI: 10.1021/acs.jcim.3c01834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
Drug-induced cardiotoxicity (DICT) is a major concern in drug development, accounting for 10-14% of postmarket withdrawals. In this study, we explored the capabilities of chemical and biological data to predict cardiotoxicity, using the recently released DICTrank data set from the United States FDA. We found that such data, including protein targets, especially those related to ion channels (e.g., hERG), physicochemical properties (e.g., electrotopological state), and peak concentration in plasma offer strong predictive ability for DICT. Compounds annotated with mechanisms of action such as cyclooxygenase inhibition could distinguish between most-concern and no-concern DICT. Cell Painting features for ER stress discerned most-concern cardiotoxic from nontoxic compounds. Models based on physicochemical properties provided substantial predictive accuracy (AUCPR = 0.93). With the availability of omics data in the future, using biological data promises enhanced predictability and deeper mechanistic insights, paving the way for safer drug development. All models from this study are available at https://broad.io/DICTrank_Predictor.
Collapse
Affiliation(s)
- Srijit Seal
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Ola Spjuth
- Department
of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box
591, SE-75124 Uppsala, Sweden
| | - Layla Hosseini-Gerami
- Ignota
Labs, The Bradfield Centre, Cambridge Science Park, County Hall, Westminster Bridge Road, Cambridge CB4 0GA, U.K.
| | - Miguel García-Ortegón
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Shantanu Singh
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andreas Bender
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Anne E. Carpenter
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
46
|
Liu Z, Wang F, Yuan H, Tian F, Yang C, Hu F, Liu Y, Tang M, Ping M, Kang C, Luo T, Yang G, Hu M, Gao Z, Li P. An LQT2-related mutation in the voltage-sensing domain is involved in switching the gating polarity of hERG. BMC Biol 2024; 22:29. [PMID: 38317233 PMCID: PMC11380439 DOI: 10.1186/s12915-024-01833-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 01/23/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Cyclic Nucleotide-Binding Domain (CNBD)-family channels display distinct voltage-sensing properties despite sharing sequence and structural similarity. For example, the human Ether-a-go-go Related Gene (hERG) channel and the Hyperpolarization-activated Cyclic Nucleotide-gated (HCN) channel share high amino acid sequence similarity and identical domain structures. hERG conducts outward current and is activated by positive membrane potentials (depolarization), whereas HCN conducts inward current and is activated by negative membrane potentials (hyperpolarization). The structural basis for the "opposite" voltage-sensing properties of hERG and HCN remains unknown. RESULTS We found the voltage-sensing domain (VSD) involves in modulating the gating polarity of hERG. We identified that a long-QT syndrome type 2-related mutation within the VSD, K525N, mediated an inwardly rectifying non-deactivating current, perturbing the channel closure, but sparing the open state and inactivated state. K525N rescued the current of a non-functional mutation in the pore helix region (F627Y) of hERG. K525N&F627Y switched hERG into a hyperpolarization-activated channel. The reactivated inward current induced by hyperpolarization mediated by K525N&F627Y can be inhibited by E-4031 and dofetilide quite well. Moreover, we report an extracellular interaction between the S1 helix and the S5-P region is crucial for modulating the gating polarity. The alanine substitution of several residues in this region (F431A, C566A, I607A, and Y611A) impaired the inward current of K525N&F627Y. CONCLUSIONS Our data provide evidence that a potential cooperation mechanism in the extracellular vestibule of the VSD and the PD would determine the gating polarity in hERG.
Collapse
Affiliation(s)
- Zhipei Liu
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Feng Wang
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
| | - Hui Yuan
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
| | - Fuyun Tian
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chuanyan Yang
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Fei Hu
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yiyao Liu
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
| | - Meiqin Tang
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Meixuan Ping
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunlan Kang
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ting Luo
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Guimei Yang
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
| | - Mei Hu
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China
- Pharmacology Laboratory, Zhongshan Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Zhongshan, 528401, China
| | - Zhaobing Gao
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China.
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China.
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Ping Li
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China.
- Zhongshan Institute for Drug Discovery, Zhongshan, 528400, China.
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
47
|
Wiley AM, Yang J, Madhani R, Nath A, Totah RA. Investigating the association between CYP2J2 inhibitors and QT prolongation: a literature review. Drug Metab Rev 2024; 56:145-163. [PMID: 38478383 DOI: 10.1080/03602532.2024.2329928] [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: 12/22/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
Drug withdrawal post-marketing due to cardiotoxicity is a major concern for drug developers, regulatory agencies, and patients. One common mechanism of cardiotoxicity is through inhibition of cardiac ion channels, leading to prolongation of the QT interval and sometimes fatal arrythmias. Recently, oxylipin signaling compounds have been shown to bind to and alter ion channel function, and disruption in their cardiac levels may contribute to QT prolongation. Cytochrome P450 2J2 (CYP2J2) is the predominant CYP isoform expressed in cardiomyocytes, where it oxidizes arachidonic acid to cardioprotective epoxyeicosatrienoic acids (EETs). In addition to roles in vasodilation and angiogenesis, EETs bind to and activate various ion channels. CYP2J2 inhibition can lower EET levels and decrease their ability to preserve cardiac rhythm. In this review, we investigated the ability of known CYP inhibitors to cause QT prolongation using Certara's Drug Interaction Database. We discovered that among the multiple CYP isozymes, CYP2J2 inhibitors were more likely to also be QT-prolonging drugs (by approximately 2-fold). We explored potential binding interactions between these inhibitors and CYP2J2 using molecular docking and identified four amino acid residues (Phe61, Ala223, Asn231, and Leu402) predicted to interact with QT-prolonging drugs. The four residues are located near the opening of egress channel 2, highlighting the potential importance of this channel in CYP2J2 binding and inhibition. These findings suggest that if a drug inhibits CYP2J2 and interacts with one of these four residues, then it may have a higher risk of QT prolongation and more preclinical studies are warranted to assess cardiovascular safety.
Collapse
Affiliation(s)
- Alexandra M Wiley
- Department of Medicinal Chemistry, University of WA School of Pharmacy, Seattle, WA, USA
| | - Jade Yang
- Department of Medicinal Chemistry, University of WA School of Pharmacy, Seattle, WA, USA
| | - Rivcka Madhani
- Department of Medicinal Chemistry, University of WA School of Pharmacy, Seattle, WA, USA
| | - Abhinav Nath
- Department of Medicinal Chemistry, University of WA School of Pharmacy, Seattle, WA, USA
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of WA School of Pharmacy, Seattle, WA, USA
| |
Collapse
|
48
|
Park NK, Park SJ, Park YG, Moon SH, Woo J, Kim HJ, Kim SJ, Choi SW. Translation reinitiation in c.453delC frameshift mutation of KCNH2 producing functional hERG K+ channels with mild dominant negative effect in the heterozygote patient-derived iPSC cardiomyocytes. Hum Mol Genet 2024; 33:110-121. [PMID: 37769355 DOI: 10.1093/hmg/ddad165] [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: 03/16/2023] [Revised: 08/16/2023] [Accepted: 09/25/2023] [Indexed: 09/30/2023] Open
Abstract
The c.453delC (p.Thr152Profs*14) frameshift mutation in KCNH2 is associated with an elevated risk of Long QT syndrome (LQTS) and fatal arrhythmia. Nevertheless, the loss-of-function mechanism underlying this mutation remains unexplored and necessitates an understanding of electrophysiology. To gain insight into the mechanism of the LQT phenotype, we conducted whole-cell patch-clamp and immunoblot assays, utilizing both a heterologous expression system and patient-derived induced pluripotent stem cell-cardiomyocytes (iPSC-CMs) with 453delC-KCNH2. We also explored the site of translational reinitiation by employing LC/MS mass spectrometry. Contrary to the previous assumption of early termination of translation, the findings of this study indicate that the 453delC-KCNH2 leads to an N-terminally truncated hERG channel, a potential from a non-canonical start codon, with diminished expression and reduced current (IhERG). The co-expression with wildtype KCNH2 produced heteromeric hERG channel with mild dominant-negative effect. Additionally, the heterozygote patient-derived iPSC-CMs exhibited prolonged action potential duration and reduced IhERG, which was ameliorated with the use of a hERG activator, PD-118057. The results of our study offer novel insights into the mechanisms involved in congenital LQTS associated with the 453delC mutation of KCNH2. The mutant results in the formation of less functional N-terminal-truncated channels with reduced amount of membrane expression. A hERG activator is capable of correcting abnormalities in both the heterologous expression system and patient-derived iPSC-CMs.
Collapse
Affiliation(s)
- Na Kyeong Park
- Department of Physiology, Seoul National University College of Medicine, 103, Daehak-ro, Seoul 03080, Korea
| | - Soon-Jung Park
- Stem Cell Research Institute, T&R Biofab Co. Ltd, 237, Sangidaehak-ro, Siheung 15073, Korea
| | - Yun-Gwi Park
- Department of Animal Science and Technology, Chung-Ang University, 4726, Seodong-daero, Anseong 17546, Korea
| | - Sung-Hwan Moon
- Department of Animal Science and Technology, Chung-Ang University, 4726, Seodong-daero, Anseong 17546, Korea
| | - JooHan Woo
- Department of Physiology, Dongguk University College of Medicine, 123, Dongdae-ro, Gyeongju 38066, Korea
| | - Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, 123, Dongdae-ro, Gyeongju 38066, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, 103, Daehak-ro, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, 103, Daehak-ro, Seoul 03080, Korea
| | - Seong Woo Choi
- Department of Physiology, Dongguk University College of Medicine, 123, Dongdae-ro, Gyeongju 38066, Korea
| |
Collapse
|
49
|
Blomquist VG, Niu J, Choudhury P, Al Saneh A, Colecraft HM, Ahern CA. Transfer RNA-mediated restoration of potassium current and electrical correction in premature termination long-QT syndrome hERG mutants. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102032. [PMID: 37842167 PMCID: PMC10568093 DOI: 10.1016/j.omtn.2023.102032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023]
Abstract
Disease-causing premature termination codons (PTCs) individually disrupt the functional expression of hundreds of genes and represent a pernicious clinical challenge. In the heart, loss-of-function mutations in the hERG potassium channel account for approximately 30% of long-QT syndrome arrhythmia, a lethal cardiac disorder with limited treatment options. Premature termination of ribosomal translation produces a truncated and, for potassium channels, a potentially dominant-negative protein that impairs the functional assembly of the wild-type homotetrameric hERG channel complex. We used high-throughput flow cytometry and patch-clamp electrophysiology to assess the trafficking and voltage-dependent activity of hERG channels carrying patient PTC variants that have been corrected by anticodon engineered tRNA. Adenoviral-mediated expression of mutant hERG channels in cultured adult guinea pig cardiomyocytes prolonged action potential durations, and this deleterious effect was corrected upon adenoviral delivery of a human ArgUGA tRNA to restore full-length hERG protein. The results demonstrate mutation-specific, context-agnostic PTC correction and elevate the therapeutic potential of this approach for rare genetic diseases caused by stop codons.
Collapse
Affiliation(s)
- Viggo G. Blomquist
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032, USA
| | - Jacqueline Niu
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032, USA
| | - Papiya Choudhury
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032, USA
| | - Ahmad Al Saneh
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Henry M. Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032, USA
| | - Christopher A. Ahern
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
50
|
Arcos-Hernández C, Nishigaki T. Ion currents through the voltage sensor domain of distinct families of proteins. J Biol Phys 2023; 49:393-413. [PMID: 37851173 PMCID: PMC10651576 DOI: 10.1007/s10867-023-09645-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/11/2023] [Indexed: 10/19/2023] Open
Abstract
The membrane potential of a cell (Vm) regulates several physiological processes. The voltage sensor domain (VSD) is a region that confers voltage sensitivity to different types of transmembrane proteins such as the following: voltage-gated ion channels, the voltage-sensing phosphatase (Ci-VSP), and the sperm-specific Na+/H+ exchanger (sNHE). VSDs contain four transmembrane segments (S1-S4) and several positively charged amino acids in S4, which are essential for the voltage sensitivity of the protein. Generally, in response to changes of the Vm, the positive residues of S4 displace along the plasma membrane without generating ionic currents through this domain. However, some native (e.g., Hv1 channel) and mutants of VSDs produce ionic currents. These gating pore currents are usually observed in VSDs that lack one or more of the conserved positively charged amino acids in S4. The gating pore currents can also be induced by the isolation of a VSD from the rest of the protein domains. In this review, we summarize gating pore currents from all families of proteins with VSDs with classification into three cases: (1) pathological, (2) physiological, and (3) artificial currents. We reinforce the model in which the position of S4 that lacks the positively charged amino acid determines the voltage dependency of the gating pore current of all VSDs independent of protein families.
Collapse
Affiliation(s)
- César Arcos-Hernández
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, 62210, Mexico.
| | - Takuya Nishigaki
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, 62210, Mexico
| |
Collapse
|