1
|
Davis J, Cornwell JD, Campagna N, Guo J, Li W, Yang T, Wang T, Zhang S. Rescue of expression and function of long QT syndrome-causing mutant hERG channels by enhancing channel stability in the plasma membrane. J Biol Chem 2024; 300:107526. [PMID: 38960041 PMCID: PMC11325228 DOI: 10.1016/j.jbc.2024.107526] [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: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024] Open
Abstract
The human ether-a-go-go-related gene (hERG) encodes the Kv11.1 (or hERG) channel that conducts the rapidly activating delayed rectifier potassium current (IKr). Naturally occurring mutations in hERG impair the channel function and cause long QT syndrome type 2. Many missense hERG mutations lead to a lack of channel expression on the cell surface, representing a major mechanism for the loss-of-function of mutant channels. While it is generally thought that a trafficking defect underlies the lack of channel expression on the cell surface, in the present study, we demonstrate that the trafficking defective mutant hERG G601S can reach the plasma membrane but is unstable and quickly degrades, which is akin to WT hERG channels under low K+ conditions. We previously showed that serine (S) residue at 624 in the innermost position of the selectivity filter of hERG is involved in hERG membrane stability such that substitution of serine 624 with threonine (S624T) enhances hERG stability and renders hERG insensitive to low K+ culture. Here, we report that the intragenic addition of S624T substitution to trafficking defective hERG mutants G601S, N470D, and P596R led to a complete rescue of the function of these otherwise loss-of-function mutant channels to a level similar to the WT channel, representing the most effective rescue means for the function of mutant hERG channels. These findings not only provide novel insights into hERG mutation-mediated channel dysfunction but also point to the critical role of S624 in hERG stability on the plasma membrane.
Collapse
Affiliation(s)
- Jordan Davis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - James D Cornwell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Noah Campagna
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Tingzhong Wang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
2
|
Taniguchi S, Berenger F, Doi Y, Mimura A, Yamanishi Y, Okiyoneda T. Ligand-based virtual-screening identified a novel CFTR ligand which improves the defective cell surface expression of misfolded ABC transporters. Front Pharmacol 2024; 15:1370676. [PMID: 38666024 PMCID: PMC11043560 DOI: 10.3389/fphar.2024.1370676] [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: 01/15/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Cystic fibrosis (CF) is a monogenetic disease caused by the mutation of CFTR, a cAMP-regulated Cl- channel expressing at the apical plasma membrane (PM) of epithelia. ∆F508-CFTR, the most common mutant in CF, fails to reach the PM due to its misfolding and premature degradation at the endoplasmic reticulum (ER). Recently, CFTR modulators have been developed to correct CFTR abnormalities, with some being used as therapeutic agents for CF treatment. One notable example is Trikafta, a triple combination of CFTR modulators (TEZ/ELX/IVA), which significantly enhances the functionality of ΔF508-CFTR on the PM. However, there's room for improvement in its therapeutic effectiveness since TEZ/ELX/IVA doesn't fully stabilize ΔF508-CFTR on the PM. To discover new CFTR modulators, we conducted a virtual screening of approximately 4.3 million compounds based on the chemical structures of existing CFTR modulators. This effort led us to identify a novel CFTR ligand named FR3. Unlike clinically available CFTR modulators, FR3 appears to operate through a distinct mechanism of action. FR3 enhances the functional expression of ΔF508-CFTR on the apical PM in airway epithelial cell lines by stabilizing NBD1. Notably, FR3 counteracted the degradation of mature ΔF508-CFTR, which still occurs despite the presence of TEZ/ELX/IVA. Furthermore, FR3 corrected the defective PM expression of a misfolded ABCB1 mutant. Therefore, FR3 may be a potential lead compound for addressing diseases resulting from the misfolding of ABC transporters.
Collapse
Affiliation(s)
- Shogo Taniguchi
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Nishinomiya, Japan
| | - Francois Berenger
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yukako Doi
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Nishinomiya, Japan
| | - Ayana Mimura
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Nishinomiya, Japan
| | - Yoshihiro Yamanishi
- Department of Complex Systems Science, Graduate School of Informatics, Nagoya University, Graduate School of Informatics, Nagoya University, Nagoya, Japan
| | - Tsukasa Okiyoneda
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Nishinomiya, Japan
| |
Collapse
|
3
|
Egly CL, Barny L, Do T, McDonald EF, Plate L, Knollmann BC. The proteostasis interactomes of trafficking-deficient K V 11.1 variants associated with Long QT Syndrome and pharmacological chaperone rescue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.574410. [PMID: 38352392 PMCID: PMC10862811 DOI: 10.1101/2024.01.31.574410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Introduction The voltage gated potassium ion channel K V 11.1 plays a critical role in cardiac repolarization. Genetic variants that render Kv11.1 dysfunctional cause Long QT Syndrome (LQTS), which is associated with fatal arrhythmias. Approximately 90% of LQTS-associated variants cause intracellular protein transport (trafficking) dysfunction, which can be rescued by pharmacological chaperones like E-4031. Protein folding and trafficking decisions are regulated by chaperones, protein quality control factors, and trafficking machinery, comprising the cellular proteostasis network. Here, we test whether trafficking dysfunction is associated with alterations in the proteostasis network of pathogenic Kv11.1 variants, and whether pharmacological chaperones can normalize the proteostasis network of responsive variants. Methods We used affinity-purification coupled with tandem mass tag-based quantitative mass spectrometry to assess protein interaction changes in human embryonic kidney (HEK293) cells expressing wild-type (WT) K V 11.1 or trafficking-deficient channel variants in the presence or absence of E-4031. Resultsa We identified 573 core K V 11.1 protein interactors. Both variants K V 11.1-G601S and K V 11.1-G601S-G965* had significantly increased interactions with proteins responsible for folding, trafficking, and degradation compared to WT. We found that proteasomal degradation is a key component for K V 11.1 degradation and that the K V 11.1-G601S-G965* variant was more responsive to E-4031 treatment. This suggests a role in the C-terminal domain and the ER retention motif of K V 11.1 in regulating trafficking. Conclusion Our report characterizes the proteostasis network of K V 11.1, two trafficking deficient K V 11.1 variants, and variants treated with a pharmacological chaperone. The identified protein interactions could be targeted therapeutically to improve K V 11.1 trafficking and treat Long QT Syndrome.
Collapse
|
4
|
Cai Q, Zha HJ, Yuan SY, Sun X, Lin X, Zheng XY, Qian YX, Xia RF, Luo YS, Shi Z, Su JC, Wan LS. Diterpenoids from Euphorbia fischeriana with Kv1.3 Inhibitory Activity. JOURNAL OF NATURAL PRODUCTS 2023; 86:2379-2390. [PMID: 37796721 DOI: 10.1021/acs.jnatprod.3c00580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Euphorbia diterpenoids possess inhibitory effects of Kv1.3 ion channel, but most of this research has focused on diterpenoids with jatrophane-related or ingenane-related skeletons. In the present study, nine undescribed (1-9) and 16 known (10-25) diterpenoids, based on jatrophane, lathyrane, ingenane, abietane, and atisane skeletons, were identified from the methanol extract of the aerial parts of Euphorbia fischeriana. The structures were established by analysis of the spectroscopic data as well as by single-crystal X-ray diffraction analysis. Among the isolated diterpenoids, macrocyclic jatrophanes and lathyranes exerted Kv1.3 blocking activity. Compound 8 exhibited good selectivity on the inhibition of the Kv 1.3 channel rather than hERG channel, with a selectivity index over 7.0. The selective activity of lathyrane diterpenoids indicates that macrocyclic diterpenoids have the potential to be further investigated as therapeutic agents for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Qin Cai
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Hong-Jing Zha
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Shi-Ying Yuan
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xing Sun
- Hebei Chemical and Pharmaceutical College, Shijiazhuang 050026, People's Republic of China
| | - Xin Lin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xin-Yu Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Ying-Xian Qian
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Ru-Feng Xia
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yue-Shan Luo
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zhimian Shi
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, People's Republic of China
| | - Jun-Cheng Su
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, People's Republic of China
| | - Luo-Sheng Wan
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
5
|
Campagna N, Wall E, Lee K, Guo J, Li W, Yang T, Baranchuk A, El-Diasty M, Zhang S. Differential Effects of Remdesivir and Lumacaftor on Homomeric and Heteromeric hERG Channels. Mol Pharmacol 2023; 104:164-173. [PMID: 37419691 DOI: 10.1124/molpharm.123.000708] [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: 04/19/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 07/09/2023] Open
Abstract
The human ether-a-go-go-related gene (hERG) encodes for the pore-forming subunit of the channel that conducts the rapidly activating delayed K+ current (IKr) in the heart. The hERG channel is important for cardiac repolarization, and reduction of its expression in the plasma membrane due to mutations causes long QT syndrome type 2 (LQT2). As such, promoting hERG membrane expression is a strategy to rescue mutant channel function. In the present study, we applied patch clamp, western blots, immunocytochemistry, and quantitative reverse transcription polymerase chain reaction techniques to investigate the rescue effects of two drugs, remdesivir and lumacaftor, on trafficking-defective mutant hERG channels. As our group has recently reported that the antiviral drug remdesivir increases wild-type (WT) hERG current and surface expression, we studied the effects of remdesivir on trafficking-defective LQT2-causing hERG mutants G601S and R582C expressed in HEK293 cells. We also investigated the effects of lumacaftor, a drug used to treat cystic fibrosis, that promotes CFTR protein trafficking and has been shown to rescue membrane expression of some hERG mutations. Our results show that neither remdesivir nor lumacaftor rescued the current or cell-surface expression of homomeric mutants G601S and R582C. However, remdesivir decreased while lumacaftor increased the current and cell-surface expression of heteromeric channels formed by WT hERG and mutant G601S or R582C hERG. We concluded that drugs can differentially affect homomeric WT and heteromeric WT+G601S (or WT+R582C) hERG channels. These findings extend our understanding of drug-channel interaction and may have clinical implications for patients with hERG mutations. SIGNIFICANCE STATEMENT: Various naturally occurring mutations in a cardiac potassium channel called hERG can impair channel function by decreasing cell-surface channel expression, resulting in cardiac electrical disturbances and even sudden cardiac death. Promotion of cell-surface expression of mutant hERG channels represents a strategy to rescue channel function. This work demonstrates that drugs such as remdesivir and lumacaftor can differently affect homomeric and heteromeric mutant hERG channels, which have biological and clinical implications.
Collapse
Affiliation(s)
- Noah Campagna
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Erika Wall
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Kevin Lee
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Jun Guo
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Wentao Li
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Tonghua Yang
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Adrian Baranchuk
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Mohammad El-Diasty
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| | - Shetuan Zhang
- Department of Biomedical and Molecular Sciences (N.C., E.W., K.L., J.G., W.L., T.Y., S.Z.); Division of Cardiology, Department of Medicine (A.B.); and Division of Cardiac Surgery, Department of Surgery (M.E.-D.), Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
6
|
Alameh M, Oliveira-Mendes BR, Kyndt F, Rivron J, Denjoy I, Lesage F, Schott JJ, De Waard M, Loussouarn G. A need for exhaustive and standardized characterization of ion channels activity. The case of K V11.1. Front Physiol 2023; 14:1132533. [PMID: 36860515 PMCID: PMC9968853 DOI: 10.3389/fphys.2023.1132533] [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: 12/27/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
hERG, the pore-forming subunit of the rapid component of the delayed rectifier K+ current, plays a key role in ventricular repolarization. Mutations in the KCNH2 gene encoding hERG are associated with several cardiac rhythmic disorders, mainly the Long QT syndrome (LQTS) characterized by prolonged ventricular repolarization, leading to ventricular tachyarrhythmias, sometimes progressing to ventricular fibrillation and sudden death. Over the past few years, the emergence of next-generation sequencing has revealed an increasing number of genetic variants including KCNH2 variants. However, the potential pathogenicity of the majority of the variants remains unknown, thus classifying them as variants of uncertain significance or VUS. With diseases such as LQTS being associated with sudden death, identifying patients at risk by determining the variant pathogenicity, is crucial. The purpose of this review is to describe, on the basis of an exhaustive examination of the 1322 missense variants, the nature of the functional assays undertaken so far and their limitations. A detailed analysis of 38 hERG missense variants identified in Long QT French patients and studied in electrophysiology also underlies the incomplete characterization of the biophysical properties for each variant. These analyses lead to two conclusions: first, the function of many hERG variants has never been looked at and, second, the functional studies done so far are excessively heterogeneous regarding the stimulation protocols, cellular models, experimental temperatures, homozygous and/or the heterozygous condition under study, a context that may lead to conflicting conclusions. The state of the literature emphasizes how necessary and important it is to perform an exhaustive functional characterization of hERG variants and to standardize this effort for meaningful comparison among variants. The review ends with suggestions to create a unique homogeneous protocol that could be shared and adopted among scientists and that would facilitate cardiologists and geneticists in patient counseling and management.
Collapse
Affiliation(s)
- Malak Alameh
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France,Labex ICST, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Barbara Ribeiro Oliveira-Mendes
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France,*Correspondence: Barbara Ribeiro Oliveira-Mendes,
| | - Florence Kyndt
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| | - Jordan Rivron
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| | - Isabelle Denjoy
- Service de Cardiologie et CNMR Maladies Cardiaques Héréditaires Rares, Hôpital Bichat, Paris, France
| | - Florian Lesage
- Labex ICST, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Jean-Jacques Schott
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| | - Michel De Waard
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France,Labex ICST, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Gildas Loussouarn
- CNRS, INSERM, l’institut du thorax, Nantes Université, CHU Nantes, Nantes, France
| |
Collapse
|
7
|
MacRae CA, Peterson RT. Zebrafish as a Mainstream Model for In Vivo Systems Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2023; 63:43-64. [PMID: 36151053 DOI: 10.1146/annurev-pharmtox-051421-105617] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pharmacology and toxicology are part of a much broader effort to understand the relationship between chemistry and biology. While biomedicine has necessarily focused on specific cases, typically of direct human relevance, there are real advantages in pursuing more systematic approaches to characterizing how health and disease are influenced by small molecules and other interventions. In this context, the zebrafish is now established as the representative screenable vertebrate and, through ongoing advances in the available scale of genome editing and automated phenotyping, is beginning to address systems-level solutions to some biomedical problems. The addition of broader efforts to integrate information content across preclinical model organisms and the incorporation of rigorous analytics, including closed-loop deep learning, will facilitate efforts to create systems pharmacology and toxicology with the ability to continuously optimize chemical biological interactions around societal needs. In this review, we outline progress toward this goal.
Collapse
Affiliation(s)
- Calum A MacRae
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA;
| | | |
Collapse
|
8
|
Blandin CE, Gravez BJ, Hatem SN, Balse E. Remodeling of Ion Channel Trafficking and Cardiac Arrhythmias. Cells 2021; 10:cells10092417. [PMID: 34572065 PMCID: PMC8468138 DOI: 10.3390/cells10092417] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 01/08/2023] Open
Abstract
Both inherited and acquired cardiac arrhythmias are often associated with the abnormal functional expression of ion channels at the cellular level. The complex machinery that continuously traffics, anchors, organizes, and recycles ion channels at the plasma membrane of a cardiomyocyte appears to be a major source of channel dysfunction during cardiac arrhythmias. This has been well established with the discovery of mutations in the genes encoding several ion channels and ion channel partners during inherited cardiac arrhythmias. Fibrosis, altered myocyte contacts, and post-transcriptional protein changes are common factors that disorganize normal channel trafficking during acquired cardiac arrhythmias. Channel availability, described notably for hERG and KV1.5 channels, could be another potent arrhythmogenic mechanism. From this molecular knowledge on cardiac arrhythmias will emerge novel antiarrhythmic strategies.
Collapse
Affiliation(s)
- Camille E. Blandin
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
| | - Basile J. Gravez
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
| | - Stéphane N. Hatem
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
- ICAN—Institute of Cardiometabolism and Nutrition, Institute of Cardiology, Pitié-Salpêtrière Hospital, Sorbonne University, F-75013 Paris, France
| | - Elise Balse
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
- Correspondence:
| |
Collapse
|
9
|
Long QT Syndrome Type 2: Emerging Strategies for Correcting Class 2 KCNH2 ( hERG) Mutations and Identifying New Patients. Biomolecules 2020. [PMID: 32759882 DOI: 10.3390/biom10081144s] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Significant advances in our understanding of the molecular mechanisms that cause congenital long QT syndrome (LQTS) have been made. A wide variety of experimental approaches, including heterologous expression of mutant ion channel proteins and the use of inducible pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from LQTS patients offer insights into etiology and new therapeutic strategies. This review briefly discusses the major molecular mechanisms underlying LQTS type 2 (LQT2), which is caused by loss-of-function (LOF) mutations in the KCNH2 gene (also known as the human ether-à-go-go-related gene or hERG). Almost half of suspected LQT2-causing mutations are missense mutations, and functional studies suggest that about 90% of these mutations disrupt the intracellular transport, or trafficking, of the KCNH2-encoded Kv11.1 channel protein to the cell surface membrane. In this review, we discuss emerging strategies that improve the trafficking and functional expression of trafficking-deficient LQT2 Kv11.1 channel proteins to the cell surface membrane and how new insights into the structure of the Kv11.1 channel protein will lead to computational approaches that identify which KCNH2 missense variants confer a high-risk for LQT2.
Collapse
|
10
|
Ono M, Burgess DE, Schroder EA, Elayi CS, Anderson CL, January CT, Sun B, Immadisetty K, Kekenes-Huskey PM, Delisle BP. Long QT Syndrome Type 2: Emerging Strategies for Correcting Class 2 KCNH2 ( hERG) Mutations and Identifying New Patients. Biomolecules 2020; 10:E1144. [PMID: 32759882 PMCID: PMC7464307 DOI: 10.3390/biom10081144] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
Significant advances in our understanding of the molecular mechanisms that cause congenital long QT syndrome (LQTS) have been made. A wide variety of experimental approaches, including heterologous expression of mutant ion channel proteins and the use of inducible pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from LQTS patients offer insights into etiology and new therapeutic strategies. This review briefly discusses the major molecular mechanisms underlying LQTS type 2 (LQT2), which is caused by loss-of-function (LOF) mutations in the KCNH2 gene (also known as the human ether-à-go-go-related gene or hERG). Almost half of suspected LQT2-causing mutations are missense mutations, and functional studies suggest that about 90% of these mutations disrupt the intracellular transport, or trafficking, of the KCNH2-encoded Kv11.1 channel protein to the cell surface membrane. In this review, we discuss emerging strategies that improve the trafficking and functional expression of trafficking-deficient LQT2 Kv11.1 channel proteins to the cell surface membrane and how new insights into the structure of the Kv11.1 channel protein will lead to computational approaches that identify which KCNH2 missense variants confer a high-risk for LQT2.
Collapse
Affiliation(s)
- Makoto Ono
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| | - Don E. Burgess
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| | - Elizabeth A. Schroder
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| | | | - Corey L. Anderson
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI 53706, USA; (C.L.A.); (C.T.J.)
| | - Craig T. January
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI 53706, USA; (C.L.A.); (C.T.J.)
| | - Bin Sun
- Department of Cellular & Molecular Physiology, Loyola University Chicago, Chicago, IL 60153, USA; (B.S.); (K.I.); (P.M.K.-H.)
| | - Kalyan Immadisetty
- Department of Cellular & Molecular Physiology, Loyola University Chicago, Chicago, IL 60153, USA; (B.S.); (K.I.); (P.M.K.-H.)
| | - Peter M. Kekenes-Huskey
- Department of Cellular & Molecular Physiology, Loyola University Chicago, Chicago, IL 60153, USA; (B.S.); (K.I.); (P.M.K.-H.)
| | - Brian P. Delisle
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA; (M.O.); (D.E.B.); (E.A.S.)
| |
Collapse
|
11
|
Kozek KA, Glazer AM, Ng CA, Blackwell D, Egly CL, Vanags LR, Blair M, Mitchell D, Matreyek KA, Fowler DM, Knollmann BC, Vandenberg JI, Roden DM, Kroncke BM. High-throughput discovery of trafficking-deficient variants in the cardiac potassium channel K V11.1. Heart Rhythm 2020; 17:2180-2189. [PMID: 32522694 DOI: 10.1016/j.hrthm.2020.05.041] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/27/2020] [Accepted: 05/31/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND KCHN2 encodes the KV11.1 potassium channel responsible for IKr, a major repolarization current during the cardiomyocyte action potential. Variants in KCNH2 that lead to decreased IKr have been associated with long QT syndrome type 2 (LQT2). The mechanism of LQT2 is most often induced loss of KV11.1 trafficking to the cell surface. Accurately discriminating between variants with normal and abnormal trafficking would aid in understanding the deleterious nature of these variants; however, the volume of reported nonsynonymous KCNH2 variants precludes the use of conventional methods for functional study. OBJECTIVE The purpose of this study was to report a high-throughput, multiplexed screening method for KCNH2 genetic variants capable of measuring the cell surface abundance of hundreds of missense variants in the resulting KV11.1 channel. METHODS We developed a method to quantitate KV11.1 variant trafficking on a pilot region of 11 residues in the S5 helix. RESULTS We generated trafficking scores for 220 of 231 missense variants in the pilot region. For 5 of 5 variants, high-throughput trafficking scores validated when tested in single variant flow cytometry and confocal microscopy experiments. We further explored these results with planar patch electrophysiology and found that loss-of-trafficking variants do not produce IKr. Conversely, but expectedly, some variants that traffic normally were still functionally compromised. CONCLUSION We describe a new method for detecting KV11.1 trafficking-deficient variants in a multiplexed assay. This new method accurately generated trafficking data for variants in KV11.1 and is extendable both to all residues in KV11.1 and to other cell surface proteins.
Collapse
Affiliation(s)
- Krystian A Kozek
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Andrew M Glazer
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Chai-Ann Ng
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St Vincent's Clinical School, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Daniel Blackwell
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christian L Egly
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Loren R Vanags
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marcia Blair
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Devyn Mitchell
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kenneth A Matreyek
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, Washington; Department of Bioengineering, University of Washington, Seattle, Washington
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jamie I Vandenberg
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St Vincent's Clinical School, UNSW Sydney, Darlinghurst, New South Wales, Australia
| | - Dan M Roden
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brett M Kroncke
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
12
|
Melgari D, Barbier C, Dilanian G, Rücker-Martin C, Doisne N, Coulombe A, Hatem SN, Balse E. Microtubule polymerization state and clathrin-dependent internalization regulate dynamics of cardiac potassium channel: Microtubule and clathrin control of K V1.5 channel. J Mol Cell Cardiol 2020; 144:127-139. [PMID: 32445844 DOI: 10.1016/j.yjmcc.2020.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/19/2022]
Abstract
Ion channel trafficking powerfully influences cardiac electrical activity as it regulates the number of available channels at the plasma membrane. Studies have largely focused on identifying the molecular determinants of the trafficking of the atria-specific KV1.5 channel, the molecular basis of the ultra-rapid delayed rectifier current IKur. Besides, regulated KV1.5 channel recycling upon changes in homeostatic state and mechanical constraints in native cardiomyocytes has been well documented. Here, using cutting-edge imaging in live myocytes, we investigated the dynamics of this channel in the plasma membrane. We demonstrate that the clathrin pathway is a major regulator of the functional expression of KV1.5 channels in atrial myocytes, with the microtubule network as the prominent organizer of KV1.5 transport within the membrane. Both clathrin blockade and microtubule disruption result in channel clusterization with reduced membrane mobility and internalization, whereas disassembly of the actin cytoskeleton does not. Mobile KV1.5 channels are associated with the microtubule plus-end tracking protein EB1 whereas static KV1.5 clusters are associated with stable acetylated microtubules. In human biopsies from patients in atrial fibrillation associated with atrial remodeling, drastic modifications in the trafficking balance occurs together with alteration in microtubule polymerization state resulting in modest reduced endocytosis and increased recycling. Consequently, hallmark of atrial KV1.5 dynamics within the membrane is clathrin- and microtubule- dependent. During atrial remodeling, predominance of anterograde trafficking activity over retrograde trafficking could result in accumulation ok KV1.5 channels in the plasma membrane.
Collapse
Affiliation(s)
- Dario Melgari
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Camille Barbier
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Gilles Dilanian
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | | | - Nicolas Doisne
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Alain Coulombe
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Stéphane N Hatem
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France; Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Elise Balse
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France.
| |
Collapse
|
13
|
Mesquita FCP, Arantes PC, Kasai-Brunswick TH, Araujo DS, Gubert F, Monnerat G, Silva Dos Santos D, Neiman G, Leitão IC, Barbosa RAQ, Coutinho JL, Vaz IM, Dos Santos MN, Borgonovo T, Cruz FES, Miriuka S, Medei EH, Campos de Carvalho AC, Carvalho AB. R534C mutation in hERG causes a trafficking defect in iPSC-derived cardiomyocytes from patients with type 2 long QT syndrome. Sci Rep 2019; 9:19203. [PMID: 31844156 PMCID: PMC6915575 DOI: 10.1038/s41598-019-55837-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023] Open
Abstract
Patient-specific cardiomyocytes obtained from induced pluripotent stem cells (CM-iPSC) offer unprecedented mechanistic insights in the study of inherited cardiac diseases. The objective of this work was to study a type 2 long QT syndrome (LQTS2)-associated mutation (c.1600C > T in KCNH2, p.R534C in hERG) in CM-iPSC. Peripheral blood mononuclear cells were isolated from two patients with the R534C mutation and iPSCs were generated. In addition, the same mutation was inserted in a control iPSC line by genome editing using CRISPR/Cas9. Cells expressed pluripotency markers and showed spontaneous differentiation into the three embryonic germ layers. Electrophysiology demonstrated that action potential duration (APD) of LQTS2 CM-iPSC was significantly longer than that of the control line, as well as the triangulation of the action potentials (AP), implying a longer duration of phase 3. Treatment with the IKr inhibitor E4031 only caused APD prolongation in the control line. Patch clamp showed a reduction of IKr on LQTS2 CM-iPSC compared to control, but channel activation was not significantly affected. Immunofluorescence for hERG demonstrated perinuclear staining in LQTS2 CM-iPSC. In conclusion, CM-iPSC recapitulated the LQTS2 phenotype and our findings suggest that the R534C mutation in KCNH2 leads to a channel trafficking defect to the plasma membrane.
Collapse
Affiliation(s)
- Fernanda C P Mesquita
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Paulo C Arantes
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Tais H Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Dayana S Araujo
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Fernanda Gubert
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco F, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Gustavo Monnerat
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Danúbia Silva Dos Santos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Gabriel Neiman
- FLENI Foundation, Sede Escobar. Ruta 9, Km 53, Belen de Escobar, BA, B1625, Argentina
| | - Isabela C Leitão
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Raiana A Q Barbosa
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Jorge L Coutinho
- National Institute of Cardiology, Rua das Laranjeiras 374, Rio de Janeiro, RJ, 22240-006, Brazil
| | - Isadora M Vaz
- Pontifical Catholic University of Parana. Rua Imaculada Conceição 1155, Curitiba, PR, 80215-901, Brazil
| | - Marcus N Dos Santos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Tamara Borgonovo
- Pontifical Catholic University of Parana. Rua Imaculada Conceição 1155, Curitiba, PR, 80215-901, Brazil
| | - Fernando E S Cruz
- National Institute of Cardiology, Rua das Laranjeiras 374, Rio de Janeiro, RJ, 22240-006, Brazil
| | - Santiago Miriuka
- FLENI Foundation, Sede Escobar. Ruta 9, Km 53, Belen de Escobar, BA, B1625, Argentina
| | - Emiliano H Medei
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Antonio C Campos de Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Institute of Cardiology, Rua das Laranjeiras 374, Rio de Janeiro, RJ, 22240-006, Brazil.
- National Institute for Science and Technology in Regenerative Medicine. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Adriana B Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Institute for Science and Technology in Regenerative Medicine. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
14
|
Mattivi CL, Ye D, Tester DJ, Clemens DJ, Zhou W, Giudicessi JR, Ackerman MJ. Utilization of the genome aggregation database, in silico tools, and heterologous expression patch-clamp studies to identify and demote previously published type 2 long QT syndrome: Causative variants from pathogenic to likely benign. Heart Rhythm 2019; 17:315-323. [PMID: 31493592 DOI: 10.1016/j.hrthm.2019.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Indexed: 10/26/2022]
Abstract
BACKGROUND Loss-of-function variants in the KCNH2-encoded Kv11.1 potassium channel cause long QT syndrome (LQTS) type 2 (LQT2). Presently, hundreds of KCNH2 missense variants (MVs) have been published as "disease-causative." However, an estimated 10% of rare published LQTS MVs may be "false positives." OBJECTIVE The purpose of this study was to determine which published KCNH2 MVs are likely false positives and warrant demotion to "likely benign" status. METHODS A list of 337 LQT2-associated MVs from 6 large compendia was compiled. MV frequency within the Genome Aggregation Database (gnomAD) (n = 141,352 individuals) was assessed, and MVs were analyzed with 8 in silico tools. Variants with minor allele frequency (MAF) >7*10E-6, the calculated maximum credible frequency of LQT2, and predicted "benign" by all tools were demoted to "likely benign." Ultra-rare variants (n = 8) absent in gnomAD but predicted "benign" by all tools were considered as potential false positives and were characterized functionally using whole-cell patch clamp. RESULTS Overall, 14 of 337 published KCNH2 MVs (4%) were observed at MAF >7*10E-6, whereas 252 of 337 (75%) were absent in gnomAD. Among the latter, 8 variants (I96V, G187S, A203T, P241L, H254Q, G314S, P935S, P963T) were predicted benign by 8 tools and lacked characterization. Patch clamp showed no functional perturbation for these 8 MVs. CONCLUSION This study offers compelling evidence for the demotion of 22 of 337 KCNH2 variants (6.5%) in the literature. Meticulous "pruning" of compendia using exome/genome databases, in silico tools, and in vitro functional studies must be conducted not only for putatively pathogenic LQTS MVs but for the entire field of genetic heart disease.
Collapse
Affiliation(s)
- Connor L Mattivi
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Dan Ye
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota
| | - David J Tester
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota; Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Mayo Clinic, Rochester, Minnesota
| | - Daniel J Clemens
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Wei Zhou
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota
| | - John R Giudicessi
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Mayo Clinic, Rochester, Minnesota
| | - Michael J Ackerman
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota; Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
15
|
Needham PG, Guerriero CJ, Brodsky JL. Chaperoning Endoplasmic Reticulum-Associated Degradation (ERAD) and Protein Conformational Diseases. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a033928. [PMID: 30670468 DOI: 10.1101/cshperspect.a033928] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Misfolded proteins compromise cellular homeostasis. This is especially problematic in the endoplasmic reticulum (ER), which is a high-capacity protein-folding compartment and whose function requires stringent protein quality-control systems. Multiprotein complexes in the ER are able to identify, remove, ubiquitinate, and deliver misfolded proteins to the 26S proteasome for degradation in the cytosol, and these events are collectively termed ER-associated degradation, or ERAD. Several steps in the ERAD pathway are facilitated by molecular chaperone networks, and the importance of ERAD is highlighted by the fact that this pathway is linked to numerous protein conformational diseases. In this review, we discuss the factors that constitute the ERAD machinery and detail how each step in the pathway occurs. We then highlight the underlying pathophysiology of protein conformational diseases associated with ERAD.
Collapse
Affiliation(s)
- Patrick G Needham
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | | | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
16
|
Capera J, Serrano-Novillo C, Navarro-Pérez M, Cassinelli S, Felipe A. The Potassium Channel Odyssey: Mechanisms of Traffic and Membrane Arrangement. Int J Mol Sci 2019; 20:ijms20030734. [PMID: 30744118 PMCID: PMC6386995 DOI: 10.3390/ijms20030734] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/29/2022] Open
Abstract
Ion channels are transmembrane proteins that conduct specific ions across biological membranes. Ion channels are present at the onset of many cellular processes, and their malfunction triggers severe pathologies. Potassium channels (KChs) share a highly conserved signature that is necessary to conduct K⁺ through the pore region. To be functional, KChs require an exquisite regulation of their subcellular location and abundance. A wide repertoire of signatures facilitates the proper targeting of the channel, fine-tuning the balance that determines traffic and location. These signature motifs can be part of the secondary or tertiary structure of the protein and are spread throughout the entire sequence. Furthermore, the association of the pore-forming subunits with different ancillary proteins forms functional complexes. These partners can modulate traffic and activity by adding their own signatures as well as by exposing or masking the existing ones. Post-translational modifications (PTMs) add a further dimension to traffic regulation. Therefore, the fate of a KCh is not fully dependent on a gene sequence but on the balance of many other factors regulating traffic. In this review, we assemble recent evidence contributing to our understanding of the spatial expression of KChs in mammalian cells. We compile specific signatures, PTMs, and associations that govern the destination of a functional channel.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Clara Serrano-Novillo
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Silvia Cassinelli
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| |
Collapse
|
17
|
Hou ZS, Ulloa-Aguirre A, Tao YX. Pharmacoperone drugs: targeting misfolded proteins causing lysosomal storage-, ion channels-, and G protein-coupled receptors-associated conformational disorders. Expert Rev Clin Pharmacol 2018; 11:611-624. [DOI: 10.1080/17512433.2018.1480367] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM) and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
18
|
Hall AR, Anderson CL, Smith JL, Mirshahi T, Elayi CS, January CT, Delisle BP. Visualizing Mutation-Specific Differences in the Trafficking-Deficient Phenotype of Kv11.1 Proteins Linked to Long QT Syndrome Type 2. Front Physiol 2018; 9:584. [PMID: 29875689 PMCID: PMC5974211 DOI: 10.3389/fphys.2018.00584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/02/2018] [Indexed: 02/05/2023] Open
Abstract
KCNH2 encodes the Kv11.1 α-subunit that underlies the rapidly activating delayed-rectifier K+ current in the heart. Loss-of-function KCNH2 mutations cause long QT syndrome type 2 (LQT2), and most LQT2-linked missense mutations inhibit the trafficking of Kv11.1 channel protein to the cell surface membrane. Several trafficking-deficient LQT2 mutations (e.g., G601S) generate Kv11.1 proteins that are sequestered in a microtubule-dependent quality control (QC) compartment in the transitional endoplasmic reticulum (ER). We tested the hypothesis that the QC mechanisms that regulate LQT2-linked Kv11.1 protein trafficking are mutation-specific. Confocal imaging analyses of HEK293 cells stably expressing the trafficking-deficient LQT2 mutation F805C showed that, unlike G601S-Kv11.1 protein, F805C-Kv11.1 protein was concentrated in several transitional ER subcompartments. The microtubule depolymerizing drug nocodazole differentially affected G601S- and F805C-Kv11.1 protein immunostaining. Nocodazole caused G601S-Kv11.1 protein to distribute into peripheral reticular structures, and it increased the diffuse immunostaining of F805C-Kv11.1 protein around the transitional ER subcompartments. Proteasome inhibition also affected the immunostaining of G601S- and F805C-Kv11.1 protein differently. Incubating cells in MG132 minimally impacted G601S-Kv11.1 immunostaining, but it dramatically increased the diffuse immunostaining of F805C-Kv11.1 protein in the transitional ER. Similar results were seen after incubating cells in the proteasome inhibitor lactacystin. Differences in the cellular distribution of G601S-Kv11.1 and F805C-Kv11.1 protein persisted in transfected human inducible pluripotent stem cell derived cardiomyocytes. These are the first data to visually demonstrate mutation-specific differences in the trafficking-deficient LQT2 phenotype, and this study has identified a novel way to categorize trafficking-deficient LQT2 mutations based on differences in intracellular retention.
Collapse
Affiliation(s)
- Allison R. Hall
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Corey L. Anderson
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin–Madison, Madison, WI, United States
| | - Jennifer L. Smith
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Tooraj Mirshahi
- Department of Molecular and Functional Genomics, Genomic Medicine Institute, Geisinger Clinic, Danville, PA, United States
| | - Claude S. Elayi
- Department of Cardiology, Gill Heart Institute, University of Kentucky, Lexington, KY, United States
| | - Craig T. January
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin–Madison, Madison, WI, United States
| | - Brian P. Delisle
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
19
|
Ion Channel Disorders and Sudden Cardiac Death. Int J Mol Sci 2018; 19:ijms19030692. [PMID: 29495624 PMCID: PMC5877553 DOI: 10.3390/ijms19030692] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/19/2022] Open
Abstract
Long QT syndrome, short QT syndrome, Brugada syndrome and catecholaminergic polymorphic ventricular tachycardia are inherited primary electrical disorders that predispose to sudden cardiac death in the absence of structural heart disease. Also known as cardiac channelopathies, primary electrical disorders respond to mutations in genes encoding cardiac ion channels and/or their regulatory proteins, which result in modifications in the cardiac action potential or in the intracellular calcium handling that lead to electrical instability and life-threatening ventricular arrhythmias. These disorders may have low penetrance and expressivity, making clinical diagnosis often challenging. However, because sudden cardiac death might be the first presenting symptom of the disease, early diagnosis becomes essential. Genetic testing might be helpful in this regard, providing a definite diagnosis in some patients. Yet important limitations still exist, with a significant proportion of patients remaining with no causative mutation identifiable after genetic testing. This review aims to provide the latest knowledge on the genetic basis of cardiac channelopathies and discuss the role of the affected proteins in the pathophysiology of each one of these diseases.
Collapse
|
20
|
Bridges RJ, Bradbury NA. Cystic Fibrosis, Cystic Fibrosis Transmembrane Conductance Regulator and Drugs: Insights from Cellular Trafficking. Handb Exp Pharmacol 2018; 245:385-425. [PMID: 29460152 DOI: 10.1007/164_2018_103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The eukaryotic cell is organized into membrane-delineated compartments that are characterized by specific cadres of proteins sustaining biochemically distinct cellular processes. The appropriate subcellular localization of proteins is key to proper organelle function and provides a physiological context for cellular processes. Disruption of normal trafficking pathways for proteins is seen in several genetic diseases, where a protein's absence for a specific subcellular compartment leads to organelle disruption, and in the context of an individual, a disruption of normal physiology. Importantly, several drug therapies can also alter protein trafficking, causing unwanted side effects. Thus, a deeper understanding of trafficking pathways needs to be appreciated as novel therapeutic modalities are proposed. Despite the promising efficacy of novel therapeutic agents, the intracellular bioavailability of these compounds has proved to be a potential barrier, leading to failures in treatments for various diseases and disorders. While endocytosis of drug moieties provides an efficient means of getting material into cells, the subsequent release and endosomal escape of materials into the cytosol where they need to act has been a barrier. An understanding of cellular protein/lipid trafficking pathways has opened up strategies for increasing drug bioavailability. Approaches to enhance endosomal exit have greatly increased the cytosolic bioavailability of drugs and will provide a means of investigating previous drugs that may have been shelved due to their low cytosolic concentration.
Collapse
Affiliation(s)
- Robert J Bridges
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA.
| |
Collapse
|
21
|
Balse E, Boycott HE. Ion Channel Trafficking: Control of Ion Channel Density as a Target for Arrhythmias? Front Physiol 2017; 8:808. [PMID: 29089904 PMCID: PMC5650974 DOI: 10.3389/fphys.2017.00808] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/02/2017] [Indexed: 12/20/2022] Open
Abstract
The shape of the cardiac action potential (AP) is determined by the contributions of numerous ion channels. Any dysfunction in the proper function or expression of these ion channels can result in a change in effective refractory period (ERP) and lead to arrhythmia. The processes underlying the correct targeting of ion channels to the plasma membrane are complex, and have not been fully characterized in cardiac myocytes. Emerging evidence highlights ion channel trafficking as a potential causative factor in certain acquired and inherited arrhythmias, and therapies which target trafficking as opposed to pore block are starting to receive attention. In this review we present the current evidence for the mechanisms which underlie precise control of cardiac ion channel trafficking and targeting.
Collapse
Affiliation(s)
- Elise Balse
- Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition, Faculté de Médecine Pitié-Salpêtrière, Sorbonne Universités, UPMC Univ. Paris VI, Inserm, UMRS 1166, Université Pierre et Marie Curie, Paris, France
| | - Hannah E. Boycott
- Department of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Sale H, Roy S, Warrier J, Thangathirupathy S, Vadari Y, Gopal SK, Krishnamurthy P, Ramarao M. Modulation of K v 11.1 (hERG) channels by 5-(((1H-indazol-5-yl)oxy)methyl)-N-(4-(trifluoromethoxy)phenyl)pyrimidin-2-amine (ITP-2), a novel small molecule activator. Br J Pharmacol 2017; 174:2484-2500. [PMID: 28500657 DOI: 10.1111/bph.13859] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Activators of Kv 11.1 (hERG) channels have potential utility in the treatment of acquired and congenital long QT (LQT) syndrome. Here, we describe a new hERG channel activator, 5-(((1H-indazol-5-yl)oxy)methyl)-N-(4-(trifluoromethoxy)phenyl)pyrimidin-2-amine (ITP-2), with a chemical structure distinct from previously reported compounds. EXPERIMENTAL APPROACH Conventional electrophysiological methods were used to assess the effects of ITP-2 on hERG1a and hERG1a/1b channels expressed heterologously in HEK-293 cells. KEY RESULTS ITP-2 selectively increased test pulse currents (EC50 1.0 μM) and decreased tail currents. ITP-2 activated hERG1a homomeric channels primarily by causing large depolarizing shifts in the midpoint of voltage-dependent inactivation and hyperpolarizing shifts in the voltage-dependence of activation. In addition, ITP-2 slowed rates of inactivation and made recovery from inactivation faster. hERG1a/1b heteromeric channels showed reduced sensitivity to ITP-2 and their inactivation properties were differentially modulated. Effects on midpoint of voltage-dependent inactivation and rates of inactivation were less pronounced for hERG1a/1b channels. Effects on voltage-dependent activation and activation kinetics were not different from hERG1a channels. Interestingly, hERG1b channels were inhibited by ITP-2. Inactivation-impairing mutations abolished activation by ITP-2 and led to inhibition of hERG channels. ITP-2 exerted agonistic effect from extracellular side of the membrane and could activate one of the arrhythmia-associated trafficking-deficient LQT2 mutants. CONCLUSIONS AND IMPLICATIONS ITP-2 may serve as another novel lead molecule for designing robust activators of hERG channels. hERG1a/1b gating kinetics were differentially modulated by ITP-2 leading to altered sensitivity. ITP-2 is capable of activating an LQT2 mutant and may be potentially useful in the development of LQT2 therapeutics.
Collapse
Affiliation(s)
- Harinath Sale
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| | - Samrat Roy
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| | - Jayakumar Warrier
- Medicinal Chemistry, Biocon Bristol Myers-Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| | - Srinivasan Thangathirupathy
- Medicinal Chemistry, Biocon Bristol Myers-Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| | - Yoganand Vadari
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| | - Shruthi K Gopal
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| | - Prasad Krishnamurthy
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| | - Manjunath Ramarao
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research and Development Center, Syngene International Limited, Bangalore, India
| |
Collapse
|
23
|
Kithcart A, MacRae CA. Using Zebrafish for High-Throughput Screening of Novel Cardiovascular Drugs. JACC Basic Transl Sci 2017; 2:1-12. [PMID: 30167552 PMCID: PMC6113531 DOI: 10.1016/j.jacbts.2017.01.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 01/17/2017] [Accepted: 01/17/2017] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases remain a major challenge for modern drug discovery. The diseases are chronic, complex, and the result of sophisticated interactions between genetics and environment involving multiple cell types and a host of systemic factors. The clinical events are often abrupt, and the diseases may be asymptomatic until a highly morbid event. Target selection is often based on limited information, and though highly specific agents are often identified in screening, their final efficacy is often compromised by unanticipated systemic responses, a narrow therapeutic index, or substantial toxicities. Our understanding of complexity of cardiovascular disease has grown dramatically over the past 2 decades, and the range of potential disease mechanisms now includes pathways previously thought only tangentially involved in cardiac or vascular disease. Despite these insights, the majority of active cardiovascular agents derive from a remarkably small number of classes of agents and target a very limited number of pathways. These agents have often been used initially for particular indications and then discovered serendipitously to have efficacy in other cardiac disorders or in a manner unrelated to their original mechanism of action. In this review, the rationale for in vivo screening is described, and the utility of the zebrafish for this approach and for complementary work in functional genomics is discussed. Current limitations of the model in this setting and the need for careful validation in new disease areas are also described. An overview is provided of the complex mechanisms underlying most clinical cardiovascular diseases, and insight is offered into the limits of single downstream pathways as drug targets. The zebrafish is introduced as a model organism, in particular for cardiovascular biology. Potential approaches to overcoming the hurdles to drug discovery in the face of complex biology are discussed, including in vivo screening of zebrafish genetic disease models.
Collapse
Affiliation(s)
- Aaron Kithcart
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Harvard Stem Cell Institute, Boston, Massachusetts
| | - Calum A MacRae
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Harvard Stem Cell Institute, Boston, Massachusetts
| |
Collapse
|
24
|
Bohnen MS, Peng G, Robey SH, Terrenoire C, Iyer V, Sampson KJ, Kass RS. Molecular Pathophysiology of Congenital Long QT Syndrome. Physiol Rev 2017; 97:89-134. [PMID: 27807201 PMCID: PMC5539372 DOI: 10.1152/physrev.00008.2016] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ion channels represent the molecular entities that give rise to the cardiac action potential, the fundamental cellular electrical event in the heart. The concerted function of these channels leads to normal cyclical excitation and resultant contraction of cardiac muscle. Research into cardiac ion channel regulation and mutations that underlie disease pathogenesis has greatly enhanced our knowledge of the causes and clinical management of cardiac arrhythmia. Here we review the molecular determinants, pathogenesis, and pharmacology of congenital Long QT Syndrome. We examine mechanisms of dysfunction associated with three critical cardiac currents that comprise the majority of congenital Long QT Syndrome cases: 1) IKs, the slow delayed rectifier current; 2) IKr, the rapid delayed rectifier current; and 3) INa, the voltage-dependent sodium current. Less common subtypes of congenital Long QT Syndrome affect other cardiac ionic currents that contribute to the dynamic nature of cardiac electrophysiology. Through the study of mutations that cause congenital Long QT Syndrome, the scientific community has advanced understanding of ion channel structure-function relationships, physiology, and pharmacological response to clinically employed and experimental pharmacological agents. Our understanding of congenital Long QT Syndrome continues to evolve rapidly and with great benefits: genotype-driven clinical management of the disease has improved patient care as precision medicine becomes even more a reality.
Collapse
Affiliation(s)
- M S Bohnen
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - G Peng
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - S H Robey
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - C Terrenoire
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - V Iyer
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - K J Sampson
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| | - R S Kass
- Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York Stem Cell Foundation Research Institute, New York, New York
| |
Collapse
|
25
|
Hantouche C, Williamson B, Valinsky WC, Solomon J, Shrier A, Young JC. Bag1 Co-chaperone Promotes TRC8 E3 Ligase-dependent Degradation of Misfolded Human Ether a Go-Go-related Gene (hERG) Potassium Channels. J Biol Chem 2016; 292:2287-2300. [PMID: 27998983 DOI: 10.1074/jbc.m116.752618] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/02/2016] [Indexed: 11/06/2022] Open
Abstract
Cardiac long QT syndrome type 2 is caused by mutations in the human ether a go-go-related gene (hERG) potassium channel, many of which cause misfolding and degradation at the endoplasmic reticulum instead of normal trafficking to the cell surface. The Hsc70/Hsp70 chaperones assist the folding of the hERG cytosolic domains. Here, we demonstrate that the Hsp70 nucleotide exchange factor Bag1 promotes hERG degradation by the ubiquitin-proteasome system at the endoplasmic reticulum to regulate hERG levels and channel activity. Dissociation of hERG complexes containing Hsp70 and the E3 ubiquitin ligase CHIP requires the interaction of Bag1 with Hsp70, but this does not involve the Bag1 ubiquitin-like domain. The interaction with Bag1 then shifts hERG degradation to the membrane-anchored E3 ligase TRC8 and its E2-conjugating enzyme Ube2g2, as determined by siRNA screening. TRC8 interacts through the transmembrane region with hERG and decreases hERG functional expression. TRC8 also mediates degradation of the misfolded hERG-G601S disease mutant, but pharmacological stabilization of the mutant structure prevents degradation. Our results identify TRC8 as a previously unknown Hsp70-independent quality control E3 ligase for hERG.
Collapse
Affiliation(s)
- Christine Hantouche
- From the Departments of Physiology and.,Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Brittany Williamson
- Biochemistry and.,Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - William C Valinsky
- From the Departments of Physiology and.,Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Joshua Solomon
- From the Departments of Physiology and.,Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Alvin Shrier
- From the Departments of Physiology and .,Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Jason C Young
- Biochemistry and .,Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec H3G 0B1, Canada
| |
Collapse
|
26
|
Holzem KM, Gomez JF, Glukhov AV, Madden EJ, Koppel AC, Ewald GA, Trenor B, Efimov IR. Reduced response to IKr blockade and altered hERG1a/1b stoichiometry in human heart failure. J Mol Cell Cardiol 2016; 96:82-92. [PMID: 26093152 PMCID: PMC4683114 DOI: 10.1016/j.yjmcc.2015.06.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/27/2015] [Accepted: 06/01/2015] [Indexed: 01/27/2023]
Abstract
Heart failure (HF) claims 250,000 lives per year in the US, and nearly half of these deaths are sudden and presumably due to ventricular tachyarrhythmias. QT interval and action potential (AP) prolongation are hallmark proarrhythmic changes in the failing myocardium, which potentially result from alterations in repolarizing potassium currents. Thus, we aimed to examine whether decreased expression of the rapid delayed rectifier potassium current, IKr, contributes to repolarization abnormalities in human HF. To map functional IKr expression across the left ventricle (LV), we optically imaged coronary-perfused LV free wall from donor and end-stage failing human hearts. The LV wedge preparation was used to examine transmural AP durations at 80% repolarization (APD80), and treatment with the IKr-blocking drug, E-4031, was utilized to interrogate functional expression. We assessed the percent change in APD80 post-IKr blockade relative to baseline APD80 (∆APD80) and found that ∆APD80s are reduced in failing versus donor hearts in each transmural region, with 0.35-, 0.43-, and 0.41-fold reductions in endo-, mid-, and epicardium, respectively (p=0.008, 0.037, and 0.022). We then assessed hERG1 isoform gene and protein expression levels using qPCR and Western blot. While we did not observe differences in hERG1a or hERG1b gene expression between donor and failing hearts, we found a shift in the hERG1a:hERG1b isoform stoichiometry at the protein level. Computer simulations were then conducted to assess IKr block under E-4031 influence in failing and nonfailing conditions. Our results confirmed the experimental observations and E-4031-induced relative APD80 prolongation was greater in normal conditions than in failing conditions, provided that the cellular model of HF included a significant downregulation of IKr. In human HF, the response to IKr blockade is reduced, suggesting decreased functional IKr expression. This attenuated functional response is associated with altered hERG1a:hERG1b protein stoichiometry in the failing human LV, and failing cardiomyoctye simulations support the experimental findings. Thus, of IKr protein and functional expression may be important determinants of repolarization remodeling in the failing human LV.
Collapse
Affiliation(s)
- Katherine M Holzem
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Juan F Gomez
- Polytechnic University of Valencia, Valencia, Spain
| | - Alexey V Glukhov
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Eli J Madden
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Aaron C Koppel
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Gregory A Ewald
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | | | - Igor R Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA; Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia.
| |
Collapse
|
27
|
Spence S, Deurinck M, Ju H, Traebert M, McLean L, Marlowe J, Emotte C, Tritto E, Tseng M, Shultz M, Friedrichs GS. Histone Deacetylase Inhibitors Prolong Cardiac Repolarization through Transcriptional Mechanisms. Toxicol Sci 2016; 153:39-54. [PMID: 27255383 DOI: 10.1093/toxsci/kfw104] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are an emerging class of anticancer agents that modify gene expression by altering the acetylation status of lysine residues of histone proteins, thereby inducing transcription, cell cycle arrest, differentiation, and cell death or apoptosis of cancer cells. In the clinical setting, treatment with HDAC inhibitors has been associated with delayed cardiac repolarization and in rare instances a lethal ventricular tachyarrhythmia known as torsades de pointes. The mechanism(s) of HDAC inhibitor-induced effects on cardiac repolarization is unknown. We demonstrate that administration of structurally diverse HDAC inhibitors to dogs causes delayed but persistent increases in the heart rate corrected QT interval (QTc), an in vivo measure of cardiac repolarization, at timepoints far removed from the Tmax for parent drug and metabolites. Transcriptional profiling of ventricular myocardium from dogs treated with various HDAC inhibitors demonstrated effects on genes involved in protein trafficking, scaffolding and insertion of various ion channels into the cell membrane as well as genes for specific ion channel subunits involved in cardiac repolarization. Extensive in vitro ion channel profiling of various structural classes of HDAC inhibitors (and their major metabolites) by binding and acute patch clamp assays failed to show any consistent correlations with direct ion channel blockade. Drug-induced rescue of an intracellular trafficking-deficient mutant potassium ion channel, hERG (G601S), and decreased maturation (glycosylation) of wild-type hERG expressed by CHO cells in vitro correlated with prolongation of QTc intervals observed in vivo The results suggest that HDAC inhibitor-induced prolongation of cardiac repolarization may be mediated in part by transcriptional changes of genes required for ion channel trafficking and localization to the sarcolemma. These data have broad implications for the development of these drug classes and suggest that the optimal time to assess potentially transcriptionally mediated physiologic effects will be delayed relative to an epigenetic drug's Tmax/Cmax.
Collapse
Affiliation(s)
- Stan Spence
- *Preclinical Safety, Novartis Institutes of Biomedical Research, 100 Technology Square, Cambridge, MA 02139
| | - Mark Deurinck
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - Haisong Ju
- Preclinical Safety, Novartis Institutes of Biomedical Research, One Health Plaza, East Hanover, NJ 07936-1080
| | - Martin Traebert
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - LeeAnne McLean
- Oncology Global Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ 07936-1080
| | - Jennifer Marlowe
- *Preclinical Safety, Novartis Institutes of Biomedical Research, 100 Technology Square, Cambridge, MA 02139
| | - Corinne Emotte
- Drug Metabolism and Pharmacokinetics, Novartis Pharmaceuticals Corporation, Fabrikstrasse 14-3.02.08, Basel, CH
| | - Elaine Tritto
- Preclinical Safety, Novartis Institutes of Biomedical Research, Klybeckstrasse 141, CH-4057, Basel, CH
| | - Min Tseng
- Development Sciences, Safety Assessment, Investigative Toxicology, Genentech Inc, 550 Grandview Drive, South San Francisco, CA 94080
| | - Michael Shultz
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research Inc, 250 Massachusetts Avenue, Cambridge, MA 02139
| | - Gregory S Friedrichs
- Preclinical Safety, Novartis Institutes of Biomedical Research, One Health Plaza, East Hanover, NJ 07936-1080
| |
Collapse
|
28
|
Cotranslational association of mRNA encoding subunits of heteromeric ion channels. Proc Natl Acad Sci U S A 2016; 113:4859-64. [PMID: 27078096 DOI: 10.1073/pnas.1521577113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Oligomers of homomeric voltage-gated potassium channels associate early in biogenesis as the nascent proteins emerge from the polysome. Less is known about how proteins emerging from different polysomes associate to form hetero-oligomeric channels. Here, we report that alternate mRNA transcripts encoding human ether-à-go-go-related gene (hERG) 1a and 1b subunits, which assemble to produce ion channels mediating cardiac repolarization, are physically associated during translation. We show that shRNA specifically targeting either hERG 1a or 1b transcripts reduced levels of both transcripts, but only when they were coexpressed heterologously. Both transcripts could be copurified with an Ab against the nascent hERG 1a N terminus. This interaction occurred even when translation of 1b was prevented, indicating the transcripts associate independent of their encoded proteins. The association was also demonstrated in cardiomyocytes, where levels of both hERG transcripts were reduced by either 1a or 1b shRNA, but native KCNE1 and ryanodine receptor 2 (RYR2) transcripts were unaffected. Changes in protein levels and membrane currents mirrored changes in transcript levels, indicating the targeted transcripts were undergoing translation. The physical association of transcripts encoding different subunits provides the spatial proximity required for nascent proteins to interact during biogenesis, and may represent a general mechanism facilitating assembly of heteromeric protein complexes involved in a range of biological processes.
Collapse
|
29
|
LI GUOLIANG, SHI RUI, WU JINE, HAN WENQI, ZHANG AIFENG, CHENG GONG, XUE XIAOLIN, SUN CHAOFENG. Association of the hERG mutation with long-QT syndrome type 2, syncope and epilepsy. Mol Med Rep 2016; 13:2467-75. [PMID: 26847485 PMCID: PMC4768985 DOI: 10.3892/mmr.2016.4859] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 08/04/2015] [Indexed: 01/08/2023] Open
Abstract
Mutations in the human ether‑à‑go‑go‑related gene (hERG) are responsible for long‑QT syndrome (LQTS) type 2 (LQT2). In the present study, a heterozygous missense mutation (A561V) linked to LQT2, syncope and epilepsy was identified in the S5/pore region of the hERG protein. The mutation, A561V, was prepared and subcloned into hERG‑pcDNA3.0. Mutant plasmids were co‑transfected into HEK‑293 cells, which stably express wild‑type (WT) hERG, in order to mimic a heterozygous genotype, and the whole‑cell current was recorded using a patch‑clamp technique. Confocal microscopy was performed to evaluate the membrane distribution of the hERG channel protein using a green fluorescent protein tagged to the N‑terminus of hERG. A561V‑hERG decreased the amplitude of the WT‑hERG currents in a concentration‑dependent manner. In addition, A561V‑hERG resulted in alterations to activation, inactivation and recovery from inactivation in the hERG protein channels. Further evaluation of hERG membrane localization indicated that the A561V‑hERG mutant protein was unable to travel to the plasma membrane, which resulted in a trafficking‑deficient WT‑hERG protein. In conclusion, A561V‑hERG exerts a potent dominant‑negative effect on WT‑hERG channels, resulting in decreased hERG currents and impairment of hERG membrane localization. This may partially elucidate the clinical manifestations of LQTS patients who carry the A561V mutation.
Collapse
Affiliation(s)
- GUOLIANG LI
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - RUI SHI
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - JINE WU
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - WENQI HAN
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - AIFENG ZHANG
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710004, P.R. China
| | - GONG CHENG
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - XIAOLIN XUE
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - CHAOFENG SUN
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
30
|
Smith JL, Anderson CL, Burgess DE, Elayi CS, January CT, Delisle BP. Molecular pathogenesis of long QT syndrome type 2. J Arrhythm 2016; 32:373-380. [PMID: 27761161 PMCID: PMC5063260 DOI: 10.1016/j.joa.2015.11.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/21/2015] [Accepted: 11/17/2015] [Indexed: 12/26/2022] Open
Abstract
The molecular mechanisms underlying congenital long QT syndrome (LQTS) are now beginning to be understood. New insights into the etiology and therapeutic strategies are emerging from heterologous expression studies of LQTS-linked mutant proteins, as well as inducible pluripotent stem cell derived cardiomyocytes (iPSC-CMs) from LQTS patients. This review focuses on the major molecular mechanism that underlies LQTS type 2 (LQT2). LQT2 is caused by loss of function (LOF) mutations in KCNH2 (also known as the human Ether-à-go-go-Related Gene or hERG). Most LQT2-linked mutations are missense mutations and functional studies suggest that ~90% of them disrupt the intracellular transport (trafficking) of KCNH2-encoded Kv11.1 proteins to the cell membrane. Trafficking deficient LQT2 mutations disrupt Kv11.1 protein folding and misfolded Kv11.1 proteins are retained in the endoplasmic reticulum (ER) until they are degraded in the ER associated degradation pathway (ERAD). This review focuses on the quality control mechanisms in the ER that contribute to the folding and ERAD of Kv11.1 proteins; the mechanism for ER export of Kv11.1 proteins in the secretory pathway; different subclasses of trafficking deficient LQT2 mutations; and strategies being developed to mitigate or correct trafficking deficient LQT2-related phenotypes.
Collapse
Affiliation(s)
- Jennifer L Smith
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Corey L Anderson
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Don E Burgess
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Claude S Elayi
- Department of Cardiology, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Craig T January
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Brian P Delisle
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
31
|
Dumotier BM. Republished: A straightforward guide to the basic science behind arrhythmogenesis. Postgrad Med J 2015; 91:221-9. [DOI: 10.1136/postgradmedj-2014-305647rep] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Affiliation(s)
- Ying Fu
- From Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.X., T.T.H., R.M.S.); and Department of Medicine, University of California Los Angeles (T.T.H., R.M.S.)
| | - Shaohua Xiao
- From Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.X., T.T.H., R.M.S.); and Department of Medicine, University of California Los Angeles (T.T.H., R.M.S.)
| | - TingTing Hong
- From Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.X., T.T.H., R.M.S.); and Department of Medicine, University of California Los Angeles (T.T.H., R.M.S.)
| | - Robin M Shaw
- From Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (Y.F., S.X., T.T.H., R.M.S.); and Department of Medicine, University of California Los Angeles (T.T.H., R.M.S.).
| |
Collapse
|
33
|
Paul F. Cranefield Award to Matthew Trudeau. J Gen Physiol 2015; 145:3-4. [PMID: 25512597 PMCID: PMC4278183 DOI: 10.1085/jgp.201411331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
34
|
Structural insight into concealed long QT type 1. Heart Rhythm 2014; 11:469-70. [DOI: 10.1016/j.hrthm.2013.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Indexed: 11/20/2022]
|
35
|
Abstract
Ion channels are essential for basic cellular function and for processes including sensory perception and intercellular communication in multicellular organisms. Voltage-gated potassium (Kv) channels facilitate dynamic cellular repolarization during an action potential, opening in response to membrane depolarization to facilitate K+ efflux. In both excitable and nonexcitable cells other, constitutively active, K+ channels provide a relatively constant repolarizing force to control membrane potential, ion homeostasis, and secretory processes. Of the forty known human Kv channel pore-forming α subunits that coassemble in various combinations to form the fundamental tetrameric channel pore and voltage sensor module, KCNQ1 is unique. KCNQ1 stands alone in having the capacity to form either channels that are voltage-dependent and require membrane depolarization for activation, or constitutively active channels. In mammals, KCNQ1 regulates processes including gastric acid secretion, thyroid hormone biosynthesis, salt and glucose homeostasis, and cell volume and in some species is required for rhythmic beating of the heart. In this review, the author discusses the unique functional properties, regulation, cell biology, diverse physiological roles, and involvement in human disease states of this chameleonic K+ channel.
Collapse
|
36
|
Barajas-Martínez H, Hu D, Antzelevitch C. [Genetic and molecular basis for sodium channel-mediated Brugada syndrome]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2013; 83:295-302. [PMID: 24269159 DOI: 10.1016/j.acmx.2013.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 10/18/2013] [Accepted: 10/21/2013] [Indexed: 11/30/2022] Open
Abstract
Brugada syndrome is a genetic disease that is characterized by abnormal electrocardiogram findings and an increased risk of sudden cardiac death. This syndrome is linked to mutations in the SCN5A gene in approximately 20% of Brugada syndrome probands. SCN5A encodes the α subunit of the cardiac sodium channel. Studies conducted over the past decade have identified 11 other Brugada syndrome susceptibility genes besides to SCN5A, pointing to genetic heterogeneity of the syndrome. Transmission of the disease shows an autosomal dominant inheritance pattern. This brief review focuses on a reported case of sodium channel-mediated Brugada syndrome, guiding the reader through the process of identification of the genetic variants responsible for the clinically-diagnosed syndrome, mutagenesis to clone SCN5A with and without the 2 variants identified and transfection of the 2 variants into TSA201 cells to determine the functional consequence of these genetic variants on sodium channel expression and function.
Collapse
Affiliation(s)
- Héctor Barajas-Martínez
- Departamento de Genética Molecular, Masonic Medical Research Laboratory, Utica, Nueva York, Estados Unidos.
| | - Dan Hu
- Departamento de Genética Molecular, Masonic Medical Research Laboratory, Utica, Nueva York, Estados Unidos
| | - Charles Antzelevitch
- Departamento de Genética Molecular, Masonic Medical Research Laboratory, Utica, Nueva York, Estados Unidos
| |
Collapse
|
37
|
Bartos DC, Giudicessi JR, Tester DJ, Ackerman MJ, Ohno S, Horie M, Gollob MH, Burgess DE, Delisle BP. A KCNQ1 mutation contributes to the concealed type 1 long QT phenotype by limiting the Kv7.1 channel conformational changes associated with protein kinase A phosphorylation. Heart Rhythm 2013; 11:459-68. [PMID: 24269949 DOI: 10.1016/j.hrthm.2013.11.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND Type 1 long QT syndrome (LQT1) is caused by loss-of-function mutations in the KCNQ1-encoded Kv7.1 channel that conducts the slowly activating component of the delayed rectifier K(+) current (IKs). Clinically, the diagnosis of LQT1 is complicated by variable phenotypic expressivity, whereby approximately 25% of genotype-positive individuals present with concealed LQT1 (resting corrected QT [QTc] interval ≤460 ms). OBJECTIVE To determine whether a specific molecular mechanism contributes to concealed LQT1. METHODS We identified a multigenerational LQT1 family whereby 79% of the patients genotype-positive for p.Ile235Asn-KCNQ1 (I235N-Kv7.1) have concealed LQT1. We assessed the effect I235N-Kv7.1 has on IKs and the ventricular action potential (AP) by using in vitro analysis and computational simulations. RESULTS Clinical data showed that all 10 patients with I235N-Kv7.1 have normal resting QTc intervals but abnormal QTc interval prolongation during the recovery phase of an electrocardiographic treadmill stress test. Voltage-clamping HEK293 cells coexpressing wild-type Kv7.1 and I235N-Kv7.1 (to mimic the patients' genotypes) showed that I235N-Kv7.1 generated relatively normal functioning Kv7.1 channels but were insensitive to protein kinase A (PKA) activation. Phosphomimetic and quinidine sensitivity studies suggest that I235N-Kv7.1 limits the conformational changes in Kv7.1 channels, which are necessary to upregulate IKs after PKA phosphorylation. Computational ventricular AP simulations predicted that the PKA insensitivity of I235N-Kv7.1 is primarily responsible for prolonging the AP with β-adrenergic stimulation, especially at slower cycle lengths. CONCLUSIONS KCNQ1 mutations that generate relatively normal Kv7.1 channels, but limit the upregulation of IKs by PKA activation, likely contribute to concealed LQT1.
Collapse
Affiliation(s)
- Daniel C Bartos
- Department of Physiology, Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - John R Giudicessi
- Departments of Medicine, Pediatrics, and Molecular Pharmacology and Experimental Therapeutics, Divisions of Cardiovascular Diseases and Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota
| | - David J Tester
- Departments of Medicine, Pediatrics, and Molecular Pharmacology and Experimental Therapeutics, Divisions of Cardiovascular Diseases and Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota
| | - Michael J Ackerman
- Departments of Medicine, Pediatrics, and Molecular Pharmacology and Experimental Therapeutics, Divisions of Cardiovascular Diseases and Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota
| | - Seiko Ohno
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Sciences, Seta-tsukinowa, Otsu, Japan
| | - Minoru Horie
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Sciences, Seta-tsukinowa, Otsu, Japan
| | - Michael H Gollob
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Don E Burgess
- Department of Physiology, Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Brian P Delisle
- Department of Physiology, Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
38
|
Apaja PM, Foo B, Okiyoneda T, Valinsky WC, Barriere H, Atanasiu R, Ficker E, Lukacs GL, Shrier A. Ubiquitination-dependent quality control of hERG K+ channel with acquired and inherited conformational defect at the plasma membrane. Mol Biol Cell 2013; 24:3787-804. [PMID: 24152733 PMCID: PMC3861077 DOI: 10.1091/mbc.e13-07-0417] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Membrane trafficking in concert with the peripheral quality control machinery plays a critical role in preserving plasma membrane (PM) protein homeostasis. Unfortunately, the peripheral quality control may also dispose of partially or transiently unfolded polypeptides and thereby contribute to the loss-of-expression phenotype of conformational diseases. Defective functional PM expression of the human ether-a-go-go-related gene (hERG) K(+) channel leads to the prolongation of the ventricular action potential that causes long QT syndrome 2 (LQT2), with increased propensity for arrhythmia and sudden cardiac arrest. LQT2 syndrome is attributed to channel biosynthetic processing defects due to mutation, drug-induced misfolding, or direct channel blockade. Here we provide evidence that a peripheral quality control mechanism can contribute to development of the LQT2 syndrome. We show that PM hERG structural and metabolic stability is compromised by the reduction of extracellular or intracellular K(+) concentration. Cardiac glycoside-induced intracellular K(+) depletion conformationally impairs the complex-glycosylated channel, which provokes chaperone- and C-terminal Hsp70-interacting protein-dependent polyubiquitination, accelerated internalization, and endosomal sorting complex required for transport-dependent lysosomal degradation. A similar mechanism contributes to the down-regulation of PM hERG harboring LQT2 missense mutations, with incomplete secretion defect. These results suggest that PM quality control plays a determining role in the loss-of-expression phenotype of hERG in certain hereditary and acquired LTQ2 syndromes.
Collapse
Affiliation(s)
- Pirjo M Apaja
- Department of Physiology and Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montréal, QC H3E 1Y6, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ayon RJ, Fernandez RA, Yuan JXJ. Mutant hERG channel traffic jam. Focus on "Pharmacological correction of long QT-linked mutations in KCNH2 (hERG) increases the trafficking of Kv11.1 channels stored in the transitional endoplasmic reticulum". Am J Physiol Cell Physiol 2013; 305:C916-8. [PMID: 23986200 DOI: 10.1152/ajpcell.00256.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Ramon J Ayon
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; and Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | | | | |
Collapse
|
40
|
Pharmacologic Approach to Defective Protein Trafficking in the E637K-hERG Mutant with PD-118057 and Thapsigargin. PLoS One 2013; 8:e65481. [PMID: 23840331 PMCID: PMC3686757 DOI: 10.1371/journal.pone.0065481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 04/26/2013] [Indexed: 02/07/2023] Open
Abstract
Background Treatment of LQT2 is inadequate. Many drugs which can pharmacologically rescue defective protein trafficking in LQT2 also result in potent blockade of HERG current, negating their therapeutic benefit. It is reported that PD-118057 and thapsigargin can rescue LQT2 without hERG channel blockade, but the precise mechanism of action is unknown. Furthermore, the effect of PD-118057 and thapsigargin on the dominant negative E637K-hERG mutant has not been previously investigated. Objective In this study, we investigated: (a) the effect of PD-118057 and thapsigargin on the current amplitudes of WT-hERG and WT/E637K-hERG channels; (b) the effect of PD-118057 and thapsigargin on the biophysical properties of WT-hERG and WT/E637K-hERG channels; (c) whether drug treatment can rescue channel processing and trafficking defects of the WT/E637K-hERG mutant. Methods The whole-cell Patch-clamp technique was used to assess the effect of PD-118057 and thapsigargin on the electrophysiological characteristics of the rapidly activating delayed rectifier K+ current (Ikr) of the hERG protein channel. Western blot was done to investigate pharmacological rescue on hERG protein channel function. Results In our study, PD-118057 was shown to significantly enhance both the maximum current amplitude and tail current amplitude, but did not alter the gating and kinetic properties of the WT-hERG channel, with the exception of accelerating steady-state inactivation. Additionally, thapsigargin shows a similar result as PD-118057 for the WT-hERG channel, but with the exception of attenuating steady-state inactivation. However, for the WT/E637K-hERG channel, PD-118057 had no effect on either the current or on the gating and kinetic properties. Furthermore, thapsigargin treatment did not alter the current or the gating and kinetic properties of the WT/E637K-hERG channel, with the exception of opening at more positive voltages. Conclusion Our findings illustrate that neither PD-118057 nor thapsigargin play a role in correcting the dominant-negative effect of the E637K-hERG mutant.
Collapse
|
41
|
Mechanistic basis for type 2 long QT syndrome caused by KCNH2 mutations that disrupt conserved arginine residues in the voltage sensor. J Membr Biol 2013; 246:355-64. [PMID: 23546015 DOI: 10.1007/s00232-013-9539-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 03/19/2013] [Indexed: 01/24/2023]
Abstract
KCNH2 encodes the Kv11.1 channel, which conducts the rapidly activating delayed rectifier K+ current (I Kr) in the heart. KCNH2 mutations cause type 2 long QT syndrome (LQT2), which increases the risk for life-threatening ventricular arrhythmias. LQT2 mutations are predicted to prolong the cardiac action potential (AP) by reducing I Kr during repolarization. Kv11.1 contains several conserved basic amino acids in the fourth transmembrane segment (S4) of the voltage sensor that are important for normal channel trafficking and gating. This study sought to determine the mechanism(s) by which LQT2 mutations at conserved arginine residues in S4 (R531Q, R531W or R534L) alter Kv11.1 function. Western blot analyses of HEK293 cells transiently expressing R531Q, R531W or R534L suggested that only R534L inhibited Kv11.1 trafficking. Voltage-clamping experiments showed that R531Q or R531W dramatically altered Kv11.1 current (I Kv11.1) activation, inactivation, recovery from inactivation and deactivation. Coexpression of wild type (to mimic the patients' genotypes) mostly corrected the changes in I Kv11.1 activation and inactivation, but deactivation kinetics were still faster. Computational simulations using a human ventricular AP model showed that accelerating deactivation rates was sufficient to prolong the AP, but these effects were minimal compared to simply reducing I Kr. These are the first data to demonstrate that coexpressing wild type can correct activation and inactivation dysfunction caused by mutations at a critical voltage-sensing residue in Kv11.1. We conclude that some Kv11.1 mutations might accelerate deactivation to cause LQT2 but that the ventricular AP duration is much more sensitive to mutations that decrease I Kr. This likely explains why most LQT2 mutations are nonsense or trafficking-deficient.
Collapse
|
42
|
Su X, Theberge AB, January CT, Beebe DJ. Effect of microculture on cell metabolism and biochemistry: do cells get stressed in microchannels? Anal Chem 2013; 85:1562-70. [PMID: 23327437 DOI: 10.1021/ac3027228] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Microfluidics is emerging as a promising platform for cell culture, enabling increased microenvironment control and potential for integrated analysis compared to conventional macroculture systems such as well plates and Petri dishes. To advance the use of microfluidic devices for cell culture, it is necessary to better understand how miniaturization affects cell behavior. In particular, microfluidic devices have significantly higher surface-area-to-volume ratios than conventional platforms, resulting in lower volumes of media per cell, which can lead to cell stress. We investigated cell stress under a variety of culture conditions using three cell lines: parental HEK (human embryonic kidney) cells and transfected HEK cells that stably express wild-type (WT) and mutant (G601S) human ether-a-go-go related gene (hERG) potassium channel protein. These three cell lines provide a unique model system through which to study cell-type-specific responses in microculture because mutant hERG is known to be sensitive to environmental conditions, making its expression a particularly sensitive readout through which to compare macro- and microculture. While expression of WT-hERG was similar in microchannel and well culture, the expression of mutant G601S-hERG was reduced in microchannels. Expression of the endoplasmic reticulum (ER) stress marker immunoglobulin binding protein (BiP) was upregulated in all three cell lines in microculture. Using BiP expression, glucose consumption, and lactate accumulation as readouts we developed methods for reducing ER stress including properly increasing the frequency of media replacement, reducing cell seeding density, and adjusting the serum concentration and buffering capacity of culture medium. Indeed, increasing the buffering capacity of culture medium or frequency of media replacement partially restored the expression of the G601S-hERG in microculture. This work illuminates how biochemical properties of cells differ in macro- and microculture and suggests strategies that can be used to modify cell culture protocols for future studies involving miniaturized culture platforms.
Collapse
Affiliation(s)
- Xiaojing Su
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin Institutes for Medical Research, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | | | | | | |
Collapse
|
43
|
Sampson HM, Lam H, Chen PC, Zhang D, Mottillo C, Mirza M, Qasim K, Shrier A, Shyng SL, Hanrahan JW, Thomas DY. Compounds that correct F508del-CFTR trafficking can also correct other protein trafficking diseases: an in vitro study using cell lines. Orphanet J Rare Dis 2013; 8:11. [PMID: 23316740 PMCID: PMC3558398 DOI: 10.1186/1750-1172-8-11] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 01/01/2013] [Indexed: 11/12/2022] Open
Abstract
Background Many genetic diseases are due to defects in protein trafficking where the mutant protein is recognized by the quality control systems, retained in the endoplasmic reticulum (ER), and degraded by the proteasome. In many cases, the mutant protein retains function if it can be trafficked to its proper cellular location. We have identified structurally diverse correctors that restore the trafficking and function of the most common mutation causing cystic fibrosis, F508del-CFTR. Most of these correctors do not act directly as ligands of CFTR, but indirectly on other pathways to promote folding and correction. We hypothesize that these proteostasis regulators may also correct other protein trafficking diseases. Methods To test our hypothesis, we used stable cell lines or transient transfection to express 2 well-studied trafficking disease mutations in each of 3 different proteins: the arginine-vasopressin receptor 2 (AVPR2, also known as V2R), the human ether-a-go-go-related gene (KCNH2, also known as hERG), and finally the sulfonylurea receptor 1 (ABCC8, also known as SUR1). We treated cells expressing these mutant proteins with 9 structurally diverse F508del-CFTR correctors that function through different cellular mechanisms and assessed whether correction occurred via immunoblotting and functional assays. Results were deemed significantly different from controls by a one-way ANOVA (p < 0.05). Results Here we show that F508del-CFTR correctors RDR1, KM60 and KM57 also correct some mutant alleles of other protein trafficking diseases. We also show that one corrector, the cardiac glycoside ouabain, was found to alter the glycosylation of all mutant alleles tested. Conclusions Correctors of F508del-CFTR trafficking might have broader applications to other protein trafficking diseases.
Collapse
Affiliation(s)
- Heidi M Sampson
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, McIntyre Medical Building, Montréal, Québec H3G 1Y6, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Calum A MacRae
- Harvard Medical School, Brigham and Women's Hospital, Cardiovascular Division,
75 Francis Street, Boston, MA 02115, P-857 307 0301, F-857 307 0300, USA
| |
Collapse
|
45
|
Abstract
The abrupt cessation of effective cardiac function due to an aberrant heart rhythm can cause sudden and unexpected death at any age, a syndrome called sudden cardiac death (SCD). Annually, more than 300,000 cases of SCD occur in the United States alone, making this a major public health concern. Our current understanding of the mechanisms responsible for SCD has emerged from decades of basic science investigation into the normal electrophysiology of the heart, the molecular physiology of cardiac ion channels, fundamental cellular and tissue events associated with cardiac arrhythmias, and the molecular genetics of monogenic disorders of heart rhythm. This knowledge has helped shape the current diagnosis and treatment of inherited arrhythmia susceptibility syndromes associated with SCD and has provided a pathophysiological framework for understanding more complex conditions predisposing to this tragic event. This Review presents an overview of the molecular basis of SCD, with a focus on monogenic arrhythmia syndromes.
Collapse
Affiliation(s)
- Alfred L George
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA.
| |
Collapse
|
46
|
Guerriero CJ, Brodsky JL. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev 2012; 92:537-76. [PMID: 22535891 DOI: 10.1152/physrev.00027.2011] [Citation(s) in RCA: 308] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Protein folding is a complex, error-prone process that often results in an irreparable protein by-product. These by-products can be recognized by cellular quality control machineries and targeted for proteasome-dependent degradation. The folding of proteins in the secretory pathway adds another layer to the protein folding "problem," as the endoplasmic reticulum maintains a unique chemical environment within the cell. In fact, a growing number of diseases are attributed to defects in secretory protein folding, and many of these by-products are targeted for a process known as endoplasmic reticulum-associated degradation (ERAD). Since its discovery, research on the mechanisms underlying the ERAD pathway has provided new insights into how ERAD contributes to human health during both normal and diseases states. Links between ERAD and disease are evidenced from the loss of protein function as a result of degradation, chronic cellular stress when ERAD fails to keep up with misfolded protein production, and the ability of some pathogens to coopt the ERAD pathway. The growing number of ERAD substrates has also illuminated the differences in the machineries used to recognize and degrade a vast array of potential clients for this pathway. Despite all that is known about ERAD, many questions remain, and new paradigms will likely emerge. Clearly, the key to successful disease treatment lies within defining the molecular details of the ERAD pathway and in understanding how this conserved pathway selects and degrades an innumerable cast of substrates.
Collapse
Affiliation(s)
- Christopher J Guerriero
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
47
|
Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y, Hill AP. hERG K+ Channels: Structure, Function, and Clinical Significance. Physiol Rev 2012; 92:1393-478. [DOI: 10.1152/physrev.00036.2011] [Citation(s) in RCA: 463] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The human ether-a-go-go related gene (hERG) encodes the pore-forming subunit of the rapid component of the delayed rectifier K+ channel, Kv11.1, which are expressed in the heart, various brain regions, smooth muscle cells, endocrine cells, and a wide range of tumor cell lines. However, it is the role that Kv11.1 channels play in the heart that has been best characterized, for two main reasons. First, it is the gene product involved in chromosome 7-associated long QT syndrome (LQTS), an inherited disorder associated with a markedly increased risk of ventricular arrhythmias and sudden cardiac death. Second, blockade of Kv11.1, by a wide range of prescription medications, causes drug-induced QT prolongation with an increase in risk of sudden cardiac arrest. In the first part of this review, the properties of Kv11.1 channels, including biogenesis, trafficking, gating, and pharmacology are discussed, while the second part focuses on the pathophysiology of Kv11.1 channels.
Collapse
Affiliation(s)
- Jamie I. Vandenberg
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Matthew D. Perry
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Mark J. Perrin
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Stefan A. Mann
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Ying Ke
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Adam P. Hill
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| |
Collapse
|
48
|
Balse E, Steele DF, Abriel H, Coulombe A, Fedida D, Hatem SN. Dynamic of Ion Channel Expression at the Plasma Membrane of Cardiomyocytes. Physiol Rev 2012; 92:1317-58. [DOI: 10.1152/physrev.00041.2011] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac myocytes are characterized by distinct structural and functional entities involved in the generation and transmission of the action potential and the excitation-contraction coupling process. Key to their function is the specific organization of ion channels and transporters to and within distinct membrane domains, which supports the anisotropic propagation of the depolarization wave. This review addresses the current knowledge on the molecular actors regulating the distinct trafficking and targeting mechanisms of ion channels in the highly polarized cardiac myocyte. In addition to ubiquitous mechanisms shared by other excitable cells, cardiac myocytes show unique specialization, illustrated by the molecular organization of myocyte-myocyte contacts, e.g., the intercalated disc and the gap junction. Many factors contribute to the specialization of the cardiac sarcolemma and the functional expression of cardiac ion channels, including various anchoring proteins, motors, small GTPases, membrane lipids, and cholesterol. The discovery of genetic defects in some of these actors, leading to complex cardiac disorders, emphasizes the importance of trafficking and targeting of ion channels to cardiac function. A major challenge in the field is to understand how these and other actors work together in intact myocytes to fine-tune ion channel expression and control cardiac excitability.
Collapse
Affiliation(s)
- Elise Balse
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David F. Steele
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Hugues Abriel
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Alain Coulombe
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David Fedida
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Stéphane N. Hatem
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| |
Collapse
|
49
|
Abstract
As the current paradigms of drug discovery evolve, it has become clear that a more comprehensive understanding of the interactions between small molecules and organismal biology will be vital. The zebrafish is emerging as a complement to existing in vitro technologies and established preclinical in vivo models that can be scaled for high-throughput. In this review, we highlight the current status of zebrafish toxicology studies, identify potential future niches for the model in the drug development pipeline, and define the hurdles that must be overcome as zebrafish technologies are refined for systematic toxicology.
Collapse
Affiliation(s)
- Randall T Peterson
- Harvard Medical School, Massachusetts General Hospital, and Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
50
|
Guo J, Wang T, Yang T, Xu J, Li W, Fridman MD, Fisher JT, Zhang S. Interaction between the cardiac rapidly (IKr) and slowly (IKs) activating delayed rectifier potassium channels revealed by low K+-induced hERG endocytic degradation. J Biol Chem 2011; 286:34664-74. [PMID: 21844197 DOI: 10.1074/jbc.m111.253351] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cardiac repolarization is controlled by the rapidly (I(Kr)) and slowly (I(Ks)) activating delayed rectifier potassium channels. The human ether-a-go-go-related gene (hERG) encodes I(Kr), whereas KCNQ1 and KCNE1 together encode I(Ks). Decreases in I(Kr) or I(Ks) cause long QT syndrome (LQTS), a cardiac disorder with a high risk of sudden death. A reduction in extracellular K(+) concentration ([K(+)](o)) induces LQTS and selectively causes endocytic degradation of mature hERG channels from the plasma membrane. In the present study, we investigated whether I(Ks) compensates for the reduced I(Kr) under low K(+) conditions. Our data show that when hERG and KCNQ1 were expressed separately in human embryonic kidney (HEK) cells, exposure to 0 mM K(+) for 6 h completely eliminated the mature hERG channel expression but had no effect on KCNQ1. When hERG and KCNQ1 were co-expressed, KCNQ1 significantly delayed 0 mM K(+)-induced hERG reduction. Also, hERG degradation led to a significant reduction in KCNQ1 in 0 mM K(+) conditions. An interaction between hERG and KCNQ1 was identified in hERG+KCNQ1-expressing HEK cells. Furthermore, KCNQ1 preferentially co-immunoprecipitated with mature hERG channels that are localized in the plasma membrane. Biophysical and pharmacological analyses indicate that although hERG and KCNQ1 closely interact with each other, they form distinct hERG and KCNQ1 channels. These data extend our understanding of delayed rectifier potassium channel trafficking and regulation, as well as the pathology of LQTS.
Collapse
Affiliation(s)
- Jun Guo
- Department of Physiology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | | | | | | | | | | | | | |
Collapse
|