1
|
Delgado-Ramírez M, Pacheco-Rojas DO, Villatoro-Gomez K, Moreno-Galindo EG, Rodríguez-Menchaca AA, Navarro-Polanco RA, Sánchez-Chapula JA, Ferrer T. Ancillary subunits KChIP2c and DPP6 differentially modulate the inhibition of Kv4.2 channels by riluzole. Eur J Pharmacol 2025; 986:177146. [PMID: 39586396 DOI: 10.1016/j.ejphar.2024.177146] [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: 05/09/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
In native tissue, Kv4.2 channels associate with the ancillary subunits Kv channels interacting proteins (KChIPs) and dipeptidyl peptidase-related proteins (DPPs) to evoke rapidly activating/inactivating currents in the heart (Ito) and brain (IA). Despite extensive knowledge of Kv4.2 biophysical modulation by auxiliary subunits, the pharmacological effects, especially those related to the co-expressed subunit and the state-dependent drug binding, remain unknown. Here, we investigated the effects of co-expressing KChIP2c or DPP6 on the pharmacological inhibition of Kv4.2 channels by riluzole. Riluzole inhibited Kv4.2, Kv4.2/DPP6, and Kv4.2/KChIP2c channels in a voltage-independent manner, with potency ranked as Kv4.2/DPP6 > Kv4.2 > Kv4.2/KChIP2c. Additionally, to a dissimilar extent, riluzole inhibited the channels from the closed state, left-shifted the inactivation curves, and enhanced the closed-state inactivation (differently modifying the rate constants of this latter). More divergent effects were observed: the inactivation kinetics was accelerated in Kv4.2 and Kv4.2/KChIP2c but not in Kv4.2/DPP6; only in Kv4.2/KChIP2c, the activation curve was left-shifted and the recovery from inactivation was decelerated; and the closed-state inactivation developed faster in Kv4.2 and Kv4.2/DPP6 but was slower in Kv4.2/KChIP2c channels. Notably, inhibition from the closed-inactivated state was more rapid than from the closed state for the three channels. We conclude that riluzole can elicit differential effects on native Kv4.2 channels depending on the presence of distinct ancillary subunits. These findings contribute to our understanding of the interplay between auxiliary subunits and pharmacological regulation of α-subunits of ion channels, highlighting the role of the former by modulating the organ-specific effects of channel-interacting drugs.
Collapse
Affiliation(s)
- Mayra Delgado-Ramírez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965 Col, Villas San Sebastián, Colima, COL, 28045, Mexico
| | - David O Pacheco-Rojas
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965 Col, Villas San Sebastián, Colima, COL, 28045, Mexico
| | - Kathya Villatoro-Gomez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965 Col, Villas San Sebastián, Colima, COL, 28045, Mexico
| | - Eloy G Moreno-Galindo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965 Col, Villas San Sebastián, Colima, COL, 28045, Mexico
| | - Aldo A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza #2405, Col. Los Filtros, San Luis Potosí, SLP, 78210, Mexico
| | - Ricardo A Navarro-Polanco
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965 Col, Villas San Sebastián, Colima, COL, 28045, Mexico
| | - José A Sánchez-Chapula
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965 Col, Villas San Sebastián, Colima, COL, 28045, Mexico
| | - Tania Ferrer
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965 Col, Villas San Sebastián, Colima, COL, 28045, Mexico.
| |
Collapse
|
2
|
Shin J, Hammer MJ, Paul SM, Conley YP, Harris C, Oppegaard K, Morse L, Cooper BA, Levine JD, Miaskowski C. Associations Between Preoperative Shortness of Breath and Potassium Channels Gene Variations in Women With Breast Cancer. Biol Res Nurs 2025; 27:81-90. [PMID: 39137431 PMCID: PMC11788816 DOI: 10.1177/10998004241268088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
OBJECTIVES Shortness of breath is a common symptom in patients with cancer. However, the mechanisms that underlie this troublesome symptom are poorly understood. Therefore, this study aimed to determine the prevalence of and associated risk factors for shortness of breath in women prior to breast cancer surgery and identify associations between shortness of breath and polymorphisms for potassium channel genes. METHODS Patients were recruited prior to breast cancer surgery and completed a self-report questionnaire on the occurrence of shortness of breath. Genotyping of single nucleotides polymorphism (SNPs) in potassium channel genes was performed using a custom array. Multiple logistic regression analyses were done to identify associations between the occurrence of shortness of breath and SNPs in ten candidate genes. RESULTS Of the 398 patients, 11.1% reported shortness of breath. These patients had a lower annual household income, a higher comorbidity burden, and a lower functional status. After controlling for functional status, comorbidity burden, genomic estimates of ancestry and self-reported race and ethnicity, the genetic associations that remained significant in the multiple regression analyses were for potassium voltage-gated channel subfamily D (KCND2) rs12673992, potassium voltage-gated channel modifier subfamily S (KCNS1) rs4499491, and potassium two pore channel subfamily K (KCNK2) rs4411107. CONCLUSIONS While these findings warrant replication, they suggest that alterations in potassium channel function may contribute to the occurrence of shortness of breath in women prior to breast cancer surgery.
Collapse
Affiliation(s)
- Joosun Shin
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Steven M. Paul
- School of Nursing, University of California San Francisco, San Francisco, CA, USA
| | - Yvette P. Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carolyn Harris
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Lisa Morse
- School of Nursing, University of California San Francisco, San Francisco, CA, USA
| | - Bruce A. Cooper
- School of Nursing, University of California San Francisco, San Francisco, CA, USA
| | | | - Christine Miaskowski
- School of Nursing, University of California San Francisco, San Francisco, CA, USA
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Argov CM, Shneyour A, Jubran J, Sabag E, Mansbach A, Sepunaru Y, Filtzer E, Gruber G, Volozhinsky M, Yogev Y, Birk O, Chalifa-Caspi V, Rokach L, Yeger-Lotem E. Tissue-aware interpretation of genetic variants advances the etiology of rare diseases. Mol Syst Biol 2024; 20:1187-1206. [PMID: 39285047 PMCID: PMC11535248 DOI: 10.1038/s44320-024-00061-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
Pathogenic variants underlying Mendelian diseases often disrupt the normal physiology of a few tissues and organs. However, variant effect prediction tools that aim to identify pathogenic variants are typically oblivious to tissue contexts. Here we report a machine-learning framework, denoted "Tissue Risk Assessment of Causality by Expression for variants" (TRACEvar, https://netbio.bgu.ac.il/TRACEvar/ ), that offers two advancements. First, TRACEvar predicts pathogenic variants that disrupt the normal physiology of specific tissues. This was achieved by creating 14 tissue-specific models that were trained on over 14,000 variants and combined 84 attributes of genetic variants with 495 attributes derived from tissue omics. TRACEvar outperformed 10 well-established and tissue-oblivious variant effect prediction tools. Second, the resulting models are interpretable, thereby illuminating variants' mode of action. Application of TRACEvar to variants of 52 rare-disease patients highlighted pathogenicity mechanisms and relevant disease processes. Lastly, the interpretation of all tissue models revealed that top-ranking determinants of pathogenicity included attributes of disease-affected tissues, particularly cellular process activities. Collectively, these results show that tissue contexts and interpretable machine-learning models can greatly enhance the etiology of rare diseases.
Collapse
Affiliation(s)
- Chanan M Argov
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Ariel Shneyour
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Juman Jubran
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Eric Sabag
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Avigdor Mansbach
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Yair Sepunaru
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Emmi Filtzer
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Gil Gruber
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Miri Volozhinsky
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Yuval Yogev
- Morris Kahn Laboratory of Human Genetics and the Genetics Institute at Soroka Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Ohad Birk
- Morris Kahn Laboratory of Human Genetics and the Genetics Institute at Soroka Medical Center, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, 84105, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Vered Chalifa-Caspi
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Lior Rokach
- Department of Software & Information Systems Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
| |
Collapse
|
4
|
Luo S, Zhou X, Wu M, Wang G, Wang L, Feng X, Wu H, Luo R, Lu M, Ju J, Wang W, Yuan L, Luo X, Peng D, Yang L, Zhang Q, Chen M, Liang S, Dong X, Hao G, Zhang Y, Liu Z. Optimizing Nav1.7-Targeted Analgesics: Revealing Off-Target Effects of Spider Venom-Derived Peptide Toxins and Engineering Strategies for Improvement. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406656. [PMID: 39248322 PMCID: PMC11558128 DOI: 10.1002/advs.202406656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/22/2024] [Indexed: 09/10/2024]
Abstract
The inhibition of Nav1.7 is a promising strategy for the development of analgesic treatments. Spider venom-derived peptide toxins are recognized as significant sources of Nav1.7 inhibitors. However, their development has been impeded by limited selectivity. In this study, eight peptide toxins from three distinct spider venom Nav channel families demonstrated robust inhibition of hNav1.7, rKv4.2, and rKv4.3 (rKv4.2/4.3) currents, exhibiting a similar mode of action. The analysis of structure and function relationship revealed a significant overlap in the pharmacophore responsible for inhibiting hNav1.7 and rKv4.2 by HNTX-III, although Lys25 seems to play a more pivotal role in the inhibition of rKv4.2/4.3. Pharmacophore-guided rational design is employed for the development of an mGpTx1 analogue, mGpTx1-SA, which retains its inhibition of hNav1.7 while significantly reducing its inhibition of rKv4.2/4.3 and eliminating cardiotoxicity. Moreover, mGpTx1-SA demonstrates potent analgesic effects in both inflammatory and neuropathic pain models, accompanied by an improved in vivo safety profile. The results suggest that off-target inhibition of rKv4.2/4.3 by specific spider peptide toxins targeting hNav1.7 may arise from a conserved binding motif. This insight promises to facilitate the design of hNav1.7-specific analgesics, aimed at minimizing rKv4.2/4.3 inhibition and associated toxicity, thereby enhancing their suitability for therapeutic applications.
Collapse
|
5
|
Imazio M, Faletra F, Zucco J, Mio C, Carraro M, Gava AM, De Biasio M, Damante G, Collini V. Genetic variants in patients with recurrent pericarditis. J Cardiovasc Med (Hagerstown) 2024; 25:799-804. [PMID: 39347728 PMCID: PMC11581433 DOI: 10.2459/jcm.0000000000001669] [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: 05/21/2024] [Revised: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024]
Abstract
AIMS Presence of family cases and multiple recurrences of pericarditis suggest the existence of a possible genetic background in at least 10% of cases. The aim of the present study is to describe the genetic landscape of a cohort of patients with multiple recurrences (at least two recurrences). METHODS Retrospective cohort study of consecutive adult patients referred for at least two episodes of recurrences in a tertiary referral centre. Genetic testing was performed by whole exome sequencing (WES). RESULTS Our cohort included 108 consecutive patients with recurrent pericarditis [median age 32 years, interquartile range (IQR) 18.5; 67.6% females, all Caucasian, idiopathic aetiology in 71.1%] with a median number of recurrences of 5 (IQR 2). Overall, 16 patients (14.8%) had variants in genes related to the inflammatory response. Eleven variants were located in genes already associated with recurrent pericarditis (NLRP3, TNFRSF1A and MEFV) and five in inflammation/immunodeficiency-related genes (IFIH1, NFKBIA, JAK1, NOD2 and ALPK1). Furthermore, we identified 10 patients with variants located in genes associated with conduction system-related diseases, and 22 variants in 21 patients with genes associated with heart structural-related diseases. CONCLUSION In this first observational study using WES to assess genetic variants in patients with multiple recurrences of pericarditis, about 15% of patients bore at least one variant that may be related to the disease. These findings highlight the importance of addressing the role of genetic predisposition in recurrent pericarditis. Moreover, 28.7% of patients carry variants in different cardiac genes, worthy of a deeper investigation.
Collapse
Affiliation(s)
- Massimo Imazio
- Department of Medicine (DMED), University of Udine
- Cardiology and Cardiothoracic Department
| | - Flavio Faletra
- Institute of Medical Genetics, University Hospital ‘Santa Maria della Misericordia’ (ASUFC), Udine, Italy
| | - Jessica Zucco
- Institute of Medical Genetics, University Hospital ‘Santa Maria della Misericordia’ (ASUFC), Udine, Italy
| | - Catia Mio
- Department of Medicine (DMED), University of Udine
| | | | | | | | - Giuseppe Damante
- Department of Medicine (DMED), University of Udine
- Institute of Medical Genetics, University Hospital ‘Santa Maria della Misericordia’ (ASUFC), Udine, Italy
| | | |
Collapse
|
6
|
Kalm T, Schob C, Völler H, Gardeitchik T, Gilissen C, Pfundt R, Klöckner C, Platzer K, Klabunde-Cherwon A, Ries M, Syrbe S, Beccaria F, Madia F, Scala M, Zara F, Hofstede F, Simon MEH, van Jaarsveld RH, Oegema R, van Gassen KLI, Holwerda SJB, Barakat TS, Bouman A, van Slegtenhorst M, Álvarez S, Fernández-Jaén A, Porta J, Accogli A, Mancardi MM, Striano P, Iacomino M, Chae JH, Jang S, Kim SY, Chitayat D, Mercimek-Andrews S, Depienne C, Kampmeier A, Kuechler A, Surowy H, Bertini ES, Radio FC, Mancini C, Pizzi S, Tartaglia M, Gauthier L, Genevieve D, Tharreau M, Azoulay N, Zaks-Hoffer G, Gilad NK, Orenstein N, Bernard G, Thiffault I, Denecke J, Herget T, Kortüm F, Kubisch C, Bähring R, Kindler S. Etiological involvement of KCND1 variants in an X-linked neurodevelopmental disorder with variable expressivity. Am J Hum Genet 2024; 111:1206-1221. [PMID: 38772379 PMCID: PMC11179411 DOI: 10.1016/j.ajhg.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024] Open
Abstract
Utilizing trio whole-exome sequencing and a gene matching approach, we identified a cohort of 18 male individuals from 17 families with hemizygous variants in KCND1, including two de novo missense variants, three maternally inherited protein-truncating variants, and 12 maternally inherited missense variants. Affected subjects present with a neurodevelopmental disorder characterized by diverse neurological abnormalities, mostly delays in different developmental domains, but also distinct neuropsychiatric signs and epilepsy. Heterozygous carrier mothers are clinically unaffected. KCND1 encodes the α-subunit of Kv4.1 voltage-gated potassium channels. All variant-associated amino acid substitutions affect either the cytoplasmic N- or C-terminus of the channel protein except for two occurring in transmembrane segments 1 and 4. Kv4.1 channels were functionally characterized in the absence and presence of auxiliary β subunits. Variant-specific alterations of biophysical channel properties were diverse and varied in magnitude. Genetic data analysis in combination with our functional assessment shows that Kv4.1 channel dysfunction is involved in the pathogenesis of an X-linked neurodevelopmental disorder frequently associated with a variable neuropsychiatric clinical phenotype.
Collapse
Affiliation(s)
- Tassja Kalm
- Institute for Cellular and Integrative Physiology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Claudia Schob
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hanna Völler
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Thatjana Gardeitchik
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Chiara Klöckner
- Institute of Human Genetics, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Annick Klabunde-Cherwon
- Division of Pediatric Epileptology, Centre for Paediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Markus Ries
- Division of Pediatric Epileptology, Centre for Paediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Steffen Syrbe
- Division of Pediatric Epileptology, Centre for Paediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Francesca Beccaria
- Epilepsy Center, Department of Child Neuropsychiatry, Territorial Social-Health Agency, 46100 Mantova, Italy
| | - Francesca Madia
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Marcello Scala
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16145 Genoa, Italy
| | - Federico Zara
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16145 Genoa, Italy
| | - Floris Hofstede
- Department of General Pediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Marleen E H Simon
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht 3584 EA, the Netherlands
| | - Richard H van Jaarsveld
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht 3584 EA, the Netherlands
| | - Renske Oegema
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht 3584 EA, the Netherlands
| | - Koen L I van Gassen
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht 3584 EA, the Netherlands
| | - Sjoerd J B Holwerda
- Department of Clinical Genetics, University Medical Center Utrecht, Utrecht 3584 EA, the Netherlands
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3000 CA, the Netherlands; ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam 3000 CA, the Netherlands; Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Arjan Bouman
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Marjon van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Sara Álvarez
- Genomics and Medicine, NIMGenetics, 28108 Madrid, Spain
| | - Alberto Fernández-Jaén
- Pediatric Neurology Department, Quironsalud University Hospital Madrid, School of Medicine, European University of Madrid, 28224 Madrid, Spain
| | - Javier Porta
- Genomics, Genologica Medica, 29016 Málaga, Spain
| | - Andrea Accogli
- Division of Medical Genetics, Department of Specialized Medicine, Montreal Children's Hospital, McGill University Health Centre, QC H4A 3J1 Montreal, Canada; Department of Human Genetics, McGill University, QC H4A 3J1 Montreal, Canada
| | | | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16145 Genoa, Italy; Pediatric Neurology and Neuromuscular Diseases Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Michele Iacomino
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Jong-Hee Chae
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea; Department of Genomic Medicine, Rare Disease Center, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - SeSong Jang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea
| | - Soo Y Kim
- Department of Genomic Medicine, Rare Disease Center, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - David Chitayat
- The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto ON M5G 1E2 Toronto, Canada; Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for SickKids, University of Toronto, M5G 1X8 Toronto, Canada
| | - Saadet Mercimek-Andrews
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for SickKids, University of Toronto, M5G 1X8 Toronto, Canada; Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, AB T6G 2H7 Edmonton, Canada
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Antje Kampmeier
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Alma Kuechler
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Harald Surowy
- Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | | | | | - Cecilia Mancini
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Lucas Gauthier
- Department of Molecular Genetics and Cytogenomics, Rare and Autoinflammatory Diseases Unit, University Hospital of Montpellier, 34295 Montpellier, France
| | - David Genevieve
- Montpellier University, Inserm U1183, Montpellier, France; Department of Clinical Genetics, University Hospital of Montpellier, 34295 Montpellier, France
| | - Mylène Tharreau
- Department of Molecular Genetics and Cytogenomics, Rare and Autoinflammatory Diseases Unit, University Hospital of Montpellier, 34295 Montpellier, France
| | - Noy Azoulay
- The Genetic Institute of Maccabi Health Services, Rehovot 7610000, Israel; Raphael Recanati Genetics Institute, Beilinson Hospital, Rabin Medical Center, Petach Tikva 49100, Israel
| | - Gal Zaks-Hoffer
- Raphael Recanati Genetics Institute, Beilinson Hospital, Rabin Medical Center, Petach Tikva 49100, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nesia K Gilad
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petah Tikvah 4920235, Israel
| | - Naama Orenstein
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petah Tikvah 4920235, Israel
| | - Geneviève Bernard
- Division of Medical Genetics, Department of Specialized Medicine, Montreal Children's Hospital, McGill University Health Centre, QC H4A 3J1 Montreal, Canada; Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Isabelle Thiffault
- Genomic Medicine Center, Department of Pediatrics, Children's Mercy Kansas City, Kansas City, MO, USA; UKMC School of Medicine, University of Missouri Kansas City, Kansas City, MO, USA; Department of Pathology and Laboratory Medicine, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Jonas Denecke
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Theresia Herget
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Robert Bähring
- Institute for Cellular and Integrative Physiology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Stefan Kindler
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
7
|
Drabkin M, Jean MM, Noy Y, Halperin D, Yogev Y, Wormser O, Proskorovski-Ohayon R, Dolgin V, Levaot N, Brumfeld V, Ovadia S, Kishner M, Kazenell U, Avraham KB, Shelef I, Birk OS. SMARCA4 mutation causes human otosclerosis and a similar phenotype in mice. J Med Genet 2024; 61:117-124. [PMID: 37399313 PMCID: PMC10756932 DOI: 10.1136/jmg-2023-109264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/09/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Otosclerosis is a common cause of adult-onset progressive hearing loss, affecting 0.3%-0.4% of the population. It results from dysregulation of bone homeostasis in the otic capsule, most commonly leading to fixation of the stapes bone, impairing sound conduction through the middle ear. Otosclerosis has a well-known genetic predisposition including familial cases with apparent autosomal dominant mode of inheritance. While linkage analysis and genome-wide association studies suggested an association with several genomic loci and with genes encoding structural proteins involved in bone formation or metabolism, the molecular genetic pathophysiology of human otosclerosis is yet mostly unknown. METHODS Whole-exome sequencing, linkage analysis, generation of CRISPR mutant mice, hearing tests and micro-CT. RESULTS Through genetic studies of kindred with seven individuals affected by apparent autosomal dominant otosclerosis, we identified a disease-causing variant in SMARCA4, encoding a key component of the PBAF chromatin remodelling complex. We generated CRISPR-Cas9 transgenic mice carrying the human mutation in the mouse SMARCA4 orthologue. Mutant Smarca4+/E1548K mice exhibited marked hearing impairment demonstrated through acoustic startle response and auditory brainstem response tests. Isolated ossicles of the auditory bullae of mutant mice exhibited a highly irregular structure of the incus bone, and their in situ micro-CT studies demonstrated the anomalous structure of the incus bone, causing disruption in the ossicular chain. CONCLUSION We demonstrate that otosclerosis can be caused by a variant in SMARCA4, with a similar phenotype of hearing impairment and abnormal bone formation in the auditory bullae in transgenic mice carrying the human mutation in the mouse SMARCA4 orthologue.
Collapse
Affiliation(s)
- Max Drabkin
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Matan M Jean
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yael Noy
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Halperin
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yuval Yogev
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ohad Wormser
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Regina Proskorovski-Ohayon
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Vadim Dolgin
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Noam Levaot
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vlad Brumfeld
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Ovadia
- Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Mor Kishner
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Udi Kazenell
- Department of Otolaryngology, Head and Neck Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Karen B Avraham
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Shelef
- Department of Radiology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ohad S Birk
- The Morris Kahn Laboratory of Human Genetics, Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Genetics Institute, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
8
|
Simonovsky E, Sharon M, Ziv M, Mauer O, Hekselman I, Jubran J, Vinogradov E, Argov CM, Basha O, Kerber L, Yogev Y, Segrè AV, Im HK, Birk O, Rokach L, Yeger‐Lotem E. Predicting molecular mechanisms of hereditary diseases by using their tissue-selective manifestation. Mol Syst Biol 2023; 19:e11407. [PMID: 37232043 PMCID: PMC10407743 DOI: 10.15252/msb.202211407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/30/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
How do aberrations in widely expressed genes lead to tissue-selective hereditary diseases? Previous attempts to answer this question were limited to testing a few candidate mechanisms. To answer this question at a larger scale, we developed "Tissue Risk Assessment of Causality by Expression" (TRACE), a machine learning approach to predict genes that underlie tissue-selective diseases and selectivity-related features. TRACE utilized 4,744 biologically interpretable tissue-specific gene features that were inferred from heterogeneous omics datasets. Application of TRACE to 1,031 disease genes uncovered known and novel selectivity-related features, the most common of which was previously overlooked. Next, we created a catalog of tissue-associated risks for 18,927 protein-coding genes (https://netbio.bgu.ac.il/trace/). As proof-of-concept, we prioritized candidate disease genes identified in 48 rare-disease patients. TRACE ranked the verified disease gene among the patient's candidate genes significantly better than gene prioritization methods that rank by gene constraint or tissue expression. Thus, tissue selectivity combined with machine learning enhances genetic and clinical understanding of hereditary diseases.
Collapse
Affiliation(s)
- Eyal Simonovsky
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Moran Sharon
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Maya Ziv
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Omry Mauer
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Idan Hekselman
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Juman Jubran
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Ekaterina Vinogradov
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Chanan M Argov
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Omer Basha
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Lior Kerber
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Yuval Yogev
- Morris Kahn Laboratory of Human Genetics and the Genetics Institute at Soroka Medical Center, Faculty of Health SciencesBen Gurion University of the NegevBeer ShevaIsrael
| | - Ayellet V Segrè
- Ocular Genomics Institute, Massachusetts Eye and EarHarvard Medical SchoolBostonMAUSA
- The Broad Institute of MIT and HarvardCambridgeMAUSA
| | - Hae Kyung Im
- Section of Genetic Medicine, Department of MedicineThe University of ChicagoChicagoILUSA
| | | | - Ohad Birk
- Morris Kahn Laboratory of Human Genetics and the Genetics Institute at Soroka Medical Center, Faculty of Health SciencesBen Gurion University of the NegevBeer ShevaIsrael
- The National Institute for Biotechnology in the NegevBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Lior Rokach
- Department of Software & Information Systems EngineeringBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Esti Yeger‐Lotem
- Department of Clinical Biochemistry and PharmacologyBen‐Gurion University of the NegevBeer ShevaIsrael
- The National Institute for Biotechnology in the NegevBen‐Gurion University of the NegevBeer ShevaIsrael
| |
Collapse
|
9
|
Kanemaru K, Cranley J, Muraro D, Miranda AMA, Ho SY, Wilbrey-Clark A, Patrick Pett J, Polanski K, Richardson L, Litvinukova M, Kumasaka N, Qin Y, Jablonska Z, Semprich CI, Mach L, Dabrowska M, Richoz N, Bolt L, Mamanova L, Kapuge R, Barnett SN, Perera S, Talavera-López C, Mulas I, Mahbubani KT, Tuck L, Wang L, Huang MM, Prete M, Pritchard S, Dark J, Saeb-Parsy K, Patel M, Clatworthy MR, Hübner N, Chowdhury RA, Noseda M, Teichmann SA. Spatially resolved multiomics of human cardiac niches. Nature 2023; 619:801-810. [PMID: 37438528 PMCID: PMC10371870 DOI: 10.1038/s41586-023-06311-1] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 06/12/2023] [Indexed: 07/14/2023]
Abstract
The function of a cell is defined by its intrinsic characteristics and its niche: the tissue microenvironment in which it dwells. Here we combine single-cell and spatial transcriptomics data to discover cellular niches within eight regions of the human heart. We map cells to microanatomical locations and integrate knowledge-based and unsupervised structural annotations. We also profile the cells of the human cardiac conduction system1. The results revealed their distinctive repertoire of ion channels, G-protein-coupled receptors (GPCRs) and regulatory networks, and implicated FOXP2 in the pacemaker phenotype. We show that the sinoatrial node is compartmentalized, with a core of pacemaker cells, fibroblasts and glial cells supporting glutamatergic signalling. Using a custom CellPhoneDB.org module, we identify trans-synaptic pacemaker cell interactions with glia. We introduce a druggable target prediction tool, drug2cell, which leverages single-cell profiles and drug-target interactions to provide mechanistic insights into the chronotropic effects of drugs, including GLP-1 analogues. In the epicardium, we show enrichment of both IgG+ and IgA+ plasma cells forming immune niches that may contribute to infection defence. Overall, we provide new clarity to cardiac electro-anatomy and immunology, and our suite of computational approaches can be applied to other tissues and organs.
Collapse
Affiliation(s)
- Kazumasa Kanemaru
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - James Cranley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Daniele Muraro
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Siew Yen Ho
- Cardiac Morphology Unit, Royal Brompton Hospital and Imperial College London, London, UK
| | - Anna Wilbrey-Clark
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Jan Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krzysztof Polanski
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Laura Richardson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Monika Litvinukova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Natsuhiko Kumasaka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Yue Qin
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Zuzanna Jablonska
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Claudia I Semprich
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Lukas Mach
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton Hospital, London, UK
| | - Monika Dabrowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Nathan Richoz
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Lira Mamanova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Rakeshlal Kapuge
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Sam N Barnett
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Carlos Talavera-López
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Würzburg Institute for Systems Immunology, Max Planck Research Group, Julius-Maximilian-Universität, Würzburg, Germany
| | - Ilaria Mulas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, and Cambridge Biorepository for Translational Medicine, NIHR Cambridge Biomedical Centre, Cambridge, UK
| | - Liz Tuck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Lu Wang
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Margaret M Huang
- Department of Surgery, University of Cambridge, and Cambridge Biorepository for Translational Medicine, NIHR Cambridge Biomedical Centre, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Sophie Pritchard
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - John Dark
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, and Cambridge Biorepository for Translational Medicine, NIHR Cambridge Biomedical Centre, Cambridge, UK
| | - Minal Patel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Menna R Clatworthy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | | | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Alrabghi G, Liu Y, Hu W, Hancox JC, Zhang H. Human atrial fibrillation and genetic defects in transient outward currents: mechanistic insights from multi-scale computational models. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220166. [PMID: 37122220 PMCID: PMC10150223 DOI: 10.1098/rstb.2022.0166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/07/2022] [Indexed: 05/02/2023] Open
Abstract
Previous studies have linked dysfunctional Ito arising from mutations to KCND3-encoded Kv4.3 and KCND2-encoded Kv4.2 to atrial fibrillation. Using computational models, this study aimed to investigate the mechanisms underlying pro-arrhythmic effects of the gain-of-function Kv4.3 (T361S, A545P) and Kv4.2 (S447R) mutations. Wild-type and mutant Ito formulations were developed from and validated against experimental data and incorporated into the Colman et al. model of human atrial cells. Single-cell models were incorporated into one- (1D) and two-dimensional (2D) models of atrial tissue, and a three-dimensional (3D) realistic model of the human atria. The three gain-of-function mutations had similar, albeit quantitatively different, effects: shortening of the action potential duration; lowering the plateau membrane potential, abbreviating the effective refractory period (ERP) and the wavelength (WL) of atrial excitation at the tissue level. Restitution curves for the WL, the ERP and the conduction velocity were leftward shifted, facilitating the conduction of atrial excitation waves at high excitation rates. The mutations also increased lifespan and stationarity of re-entry in both 2D and 3D simulations, which further highlighted a mutation-induced increase in spatial dispersion of repolarization. Collectively, these changes account for pro-arrhythmic effects of these Kv4.3 and Kv4.2 mutations in facilitating AF. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Ghadah Alrabghi
- Biological Physics Group, Department of Physics and Astronomy, University of Manchester, Manchester M13 9PL, UK
- Department of Physics, Faculty of Science, University of Jeddah, 21959 Jeddah, Saudi Arabia
| | - Yizhou Liu
- Biological Physics Group, Department of Physics and Astronomy, University of Manchester, Manchester M13 9PL, UK
| | - Wei Hu
- Biological Physics Group, Department of Physics and Astronomy, University of Manchester, Manchester M13 9PL, UK
| | - Jules C. Hancox
- Biological Physics Group, Department of Physics and Astronomy, University of Manchester, Manchester M13 9PL, UK
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, University of Manchester, Manchester M13 9PL, UK
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646099 Luzhou, People's Republic of China
| |
Collapse
|
11
|
Novel preventive effect of isorhamnetin on electrical and structural remodeling in atrial fibrillation. Clin Sci (Lond) 2022; 136:1831-1849. [DOI: 10.1042/cs20220319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Abstract
Isorhamnetin, a natural flavonoid, has strong antioxidant and antifibrotic effects, and a regulatory effect against Ca2+-handling. Atrial remodeling due to fibrosis and abnormal intracellular Ca2+ activities contributes to initiation and persistence of atrial fibrillation (AF). The present study investigated the effect of isorhamnetin on angiotensin II (AngII)-induced AF in mice. Wild-type male mice (C57BL/6J, 8 weeks old) were assigned to three groups: (1) control group, (2) AngII-treated group, and (3) AngII- and isorhamnetin-treated group. AngII (1000 ng/kg/min) and isorhamnetin (5 mg/kg) were administered continuously via an implantable osmotic pump for two weeks and intraperitoneally one week before initiating AngII administration, respectively. AF induction and electrophysiological studies, Ca2+ imaging with isolated atrial myocytes and HL-1 cells, and action potential duration (APD) measurements using atrial tissue and HL-1 cells were performed. AF-related molecule expression was assessed and histopathological examination was performed. Isorhamnetin decreased AF inducibility compared with the AngII group and restored AngII-induced atrial effective refractory period prolongation. Isorhamnetin eliminated abnormal diastolic intracellular Ca2+ activities induced by AngII. Isorhamnetin also abrogated AngII-induced APD prolongation and abnormal Ca2+ loading in HL-1 cells. Furthermore, isorhamnetin strongly attenuated AngII-induced left atrial enlargement and atrial fibrosis. AngII-induced elevated expression of AF-associated molecules, such as ox-CaMKII, p-RyR2, p-JNK, p-ERK, and TRPC3/6, was improved by isorhamnetin treatment. The findings of the present study suggest that isorhamnetin prevents AngII-induced AF vulnerability and arrhythmogenic atrial remodeling, highlighting its therapeutic potential as an anti-arrhythmogenic pharmaceutical or dietary supplement.
Collapse
|
12
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J J M Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
13
|
Guo XJ, Qiu XB, Wang J, Guo YH, Yang CX, Li L, Gao RF, Ke ZP, Di RM, Sun YM, Xu YJ, Yang YQ. PRRX1 Loss-of-Function Mutations Underlying Familial Atrial Fibrillation. J Am Heart Assoc 2021; 10:e023517. [PMID: 34845933 PMCID: PMC9075371 DOI: 10.1161/jaha.121.023517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Atrial fibrillation (AF) is the most common form of clinical cardiac dysrhythmia responsible for thromboembolic cerebral stroke, congestive heart failure, and death. Aggregating evidence highlights the strong genetic basis of AF. Nevertheless, AF is of pronounced genetic heterogeneity, and in an overwhelming majority of patients, the genetic determinants underpinning AF remain elusive. Methods and Results By genome‐wide screening with polymorphic microsatellite markers and linkage analysis in a 4‐generation Chinese family affected with autosomal‐dominant AF, a novel locus for AF was mapped to chromosome 1q24.2–q25.1, a 3.20‐cM (≈4.19 Mbp) interval between markers D1S2851 and D1S218, with the greatest 2‐point logarithm of odds score of 4.8165 for the marker D1S452 at recombination fraction=0.00. Whole‐exome sequencing and bioinformatics analyses showed that within the mapping region, only the mutation in the paired related homeobox 1 (PRRX1) gene, NM_022716.4:c.319C>T;(p.Gln107*), cosegregated with AF in the family. In addition, sequencing analyses of PRRX1 in another cohort of 225 unrelated patients with AF revealed a new mutation, NM_022716.4:c.437G>T; (p.Arg146Ile), in a patient. The 2 mutations were absent in 908 control subjects. Biological analyses in HeLa cells demonstrated that the 2 mutants had significantly diminished transactivation on the target genes ISL1 and SHOX2 and markedly decreased ability to bind the promoters of ISL1 and SHOX2 (2 genes causally linked to AF), although with normal intracellular distribution. Conclusions This study first indicates that PRRX1 loss‐of‐function mutations predispose to AF, which provides novel insight into the molecular pathogenesis underpinning AF, implying potential implications for precisive prophylaxis and management of AF.
Collapse
Affiliation(s)
- Xiao-Juan Guo
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Xing-Biao Qiu
- Department of Cardiology Shanghai Chest HospitalShanghai Jiao Tong University Shanghai China
| | - Jun Wang
- Department of Cardiology Shanghai Jing'an District Central HospitalFudan University Shanghai China
| | - Yu-Han Guo
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Chen-Xi Yang
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Li Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East HospitalTongji University School of Medicine Shanghai China.,Institute of Medical GeneticsTongji University Shanghai China
| | - Ri-Feng Gao
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Zun-Ping Ke
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Ruo-Min Di
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Yu-Min Sun
- Department of Cardiology Shanghai Jing'an District Central HospitalFudan University Shanghai China
| | - Ying-Jia Xu
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China
| | - Yi-Qing Yang
- Department of Cardiology and the Center for Complex Cardiac Arrhythmias of Minhang District Shanghai Fifth People's HospitalFudan University Shanghai China.,Cardiovascular Research Laboratory and Central Laboratory Shanghai Fifth People's HospitalFudan University Shanghai China
| |
Collapse
|
14
|
Velluva A, Radtke M, Horn S, Popp B, Platzer K, Gjermeni E, Lin CC, Lemke JR, Garten A, Schöneberg T, Blüher M, Abou Jamra R, Le Duc D. Phenotype-tissue expression and exploration (PTEE) resource facilitates the choice of tissue for RNA-seq-based clinical genetics studies. BMC Genomics 2021; 22:802. [PMID: 34743696 PMCID: PMC8573933 DOI: 10.1186/s12864-021-08125-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/26/2021] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND RNA-seq emerges as a valuable method for clinical genetics. The transcriptome is "dynamic" and tissue-specific, but typically the probed tissues to analyze (TA) are different from the tissue of interest (TI) based on pathophysiology. RESULTS We developed Phenotype-Tissue Expression and Exploration (PTEE), a tool to facilitate the decision about the most suitable TA for RNA-seq. We integrated phenotype-annotated genes, used 54 tissues from GTEx to perform correlation analyses and identify expressed genes and transcripts between TAs and TIs. We identified skeletal muscle as the most appropriate TA to inquire for cardiac arrhythmia genes and skin as a good proxy to study neurodevelopmental disorders. We also explored RNA-seq limitations and show that on-off switching of gene expression during ontogenesis or circadian rhythm can cause blind spots for RNA-seq-based analyses. CONCLUSIONS PTEE aids the identification of tissues suitable for RNA-seq for a given pathology to increase the success rate of diagnosis and gene discovery. PTEE is freely available at https://bioinf.eva.mpg.de/PTEE/.
Collapse
Affiliation(s)
- Akhil Velluva
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103, Leipzig, Germany.
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, 04103, Leipzig, Germany.
| | - Maximillian Radtke
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Susanne Horn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, 04103, Leipzig, Germany
| | - Bernt Popp
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Konrad Platzer
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Erind Gjermeni
- Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, 04289, Leipzig, Germany
- Department of Cardiology, Median Centre for Rehabilitation Schmannewitz, 04774, Dahlen, Germany
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Johannes R Lemke
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Antje Garten
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103, Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, 04103, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103, Leipzig, Germany
| | - Rami Abou Jamra
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Diana Le Duc
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103, Leipzig, Germany.
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany.
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103, Leipzig, Germany.
| |
Collapse
|
15
|
Crotti L, Odening KE, Sanguinetti MC. Heritable arrhythmias associated with abnormal function of cardiac potassium channels. Cardiovasc Res 2021; 116:1542-1556. [PMID: 32227190 DOI: 10.1093/cvr/cvaa068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiomyocytes express a surprisingly large number of potassium channel types. The primary physiological functions of the currents conducted by these channels are to maintain the resting membrane potential and mediate action potential repolarization under basal conditions and in response to changes in the concentrations of intracellular sodium, calcium, and ATP/ADP. Here, we review the diversity and functional roles of cardiac potassium channels under normal conditions and how heritable mutations in the genes encoding these channels can lead to distinct arrhythmias. We briefly review atrial fibrillation and J-wave syndromes. For long and short QT syndromes, we describe their genetic basis, clinical manifestation, risk stratification, traditional and novel therapeutic approaches, as well as insights into disease mechanisms provided by animal and cellular models.
Collapse
Affiliation(s)
- Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Department of Cardiovascular, Neural and Metabolic Sciences, Istituto Auxologico Italiano, IRCCS, San Luca Hospital, Milan, Italy.,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Katja E Odening
- Department of Cardiology and Angiology I, Heart Center University of Freiburg, Medical Faculty, Freiburg, Germany.,Institute of Experimental Cardiovascular Medicine, Heart Center University of Freiburg, Medical Faculty, Freiburg, Germany.,Department of Cardiology, Translational Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland
| | - Michael C Sanguinetti
- Department of Internal Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
16
|
Xiao Z, Zhao P, Wu X, Kong X, Wang R, Liang S, Tang C, Liu Z. Variation of Two S3b Residues in K V4.1-4.3 Channels Underlies Their Different Modulations by Spider Toxin κ-LhTx-1. Front Pharmacol 2021; 12:692076. [PMID: 34177600 PMCID: PMC8222713 DOI: 10.3389/fphar.2021.692076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
The naturally occurred peptide toxins from animal venoms are valuable pharmacological tools in exploring the structure-function relationships of ion channels. Herein we have identified the peptide toxin κ-LhTx-1 from the venom of spider Pandercetes sp (the Lichen huntsman spider) as a novel selective antagonist of the KV4 family potassium channels. κ-LhTx-1 is a gating-modifier toxin impeded KV4 channels' voltage sensor activation, and mutation analysis has confirmed its binding site on channels' S3b region. Interestingly, κ-LhTx-1 differently modulated the gating of KV4 channels, as revealed by toxin inhibiting KV4.2/4.3 with much more stronger voltage-dependence than that for KV4.1. We proposed that κ-LhTx-1 trapped the voltage sensor of KV4.1 in a much more stable resting state than that for KV4.2/4.3 and further explored the underlying mechanism. Swapping the non-conserved S3b segments between KV4.1(280FVPK283) and KV4.3(275VMTN278) fully reversed their voltage-dependence phenotypes in inhibition by κ-LhTx-1, and intensive mutation analysis has identified P282 in KV4.1, D281 in KV4.2 and N278 in KV4.3 being the key residues. Furthermore, the last two residues in this segment of each KV4 channel (P282/K283 in KV4.1, T280/D281 in KV4.2 and T277/N278 in KV4.3) likely worked synergistically as revealed by our combinatorial mutations analysis. The present study has clarified the molecular basis in KV4 channels for their different modulations by κ-LhTx-1, which have advanced our understanding on KV4 channels' structure features. Moreover, κ-LhTx-1 might be useful in developing anti-arrhythmic drugs given its high affinity, high selectivity and unique action mode in interacting with the KV4.2/4.3 channels.
Collapse
Affiliation(s)
- Zhen Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Piao Zhao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiangyue Wu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiangjin Kong
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ruiwen Wang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
17
|
Targeting of Potassium Channels in Cardiac Arrhythmias. Trends Pharmacol Sci 2021; 42:491-506. [PMID: 33858691 DOI: 10.1016/j.tips.2021.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Cardiomyocytes are endowed with a complex repertoire of ion channels, responsible for the generation of action potentials (APs), travelling waves of electrical excitation, propagating throughout the heart and leading to cardiac contractions. Cardiac AP waveforms are shaped by a striking diversity of K+ channels. The pivotal role of K+ channels in cardiac health and disease is underscored by the dramatic impact that K+ channel dysfunction has on cardiac arrhythmias. The development of drugs targeted to specific K+ channels is expected to provide an optimized approach to antiarrhythmic therapy. Here, we review the functional roles of cardiac potassium channels under normal and diseased states. We survey current antiarrhythmic drugs (AADs) targeted to voltage-gated and Ca2+-activated K+ channels and highlight future research opportunities.
Collapse
|
18
|
Nakajima T, Tamura S, Kurabayashi M, Kaneko Y. Towards Mutation-Specific Precision Medicine in Atypical Clinical Phenotypes of Inherited Arrhythmia Syndromes. Int J Mol Sci 2021; 22:ijms22083930. [PMID: 33920294 PMCID: PMC8069124 DOI: 10.3390/ijms22083930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Most causal genes for inherited arrhythmia syndromes (IASs) encode cardiac ion channel-related proteins. Genotype-phenotype studies and functional analyses of mutant genes, using heterologous expression systems and animal models, have revealed the pathophysiology of IASs and enabled, in part, the establishment of causal gene-specific precision medicine. Additionally, the utilization of induced pluripotent stem cell (iPSC) technology have provided further insights into the pathophysiology of IASs and novel promising therapeutic strategies, especially in long QT syndrome. It is now known that there are atypical clinical phenotypes of IASs associated with specific mutations that have unique electrophysiological properties, which raises a possibility of mutation-specific precision medicine. In particular, patients with Brugada syndrome harboring an SCN5A R1632C mutation exhibit exercise-induced cardiac events, which may be caused by a marked activity-dependent loss of R1632C-Nav1.5 availability due to a marked delay of recovery from inactivation. This suggests that the use of isoproterenol should be avoided. Conversely, the efficacy of β-blocker needs to be examined. Patients harboring a KCND3 V392I mutation exhibit both cardiac (early repolarization syndrome and paroxysmal atrial fibrillation) and cerebral (epilepsy) phenotypes, which may be associated with a unique mixed electrophysiological property of V392I-Kv4.3. Since the epileptic phenotype appears to manifest prior to cardiac events in this mutation carrier, identifying KCND3 mutations in patients with epilepsy and providing optimal therapy will help prevent sudden unexpected death in epilepsy. Further studies using the iPSC technology may provide novel insights into the pathophysiology of atypical clinical phenotypes of IASs and the development of mutation-specific precision medicine.
Collapse
|
19
|
Huang X, Li Y, Zhang J, Wang X, Li Z, Li G. The molecular genetic basis of atrial fibrillation. Hum Genet 2020; 139:1485-1498. [PMID: 32617797 DOI: 10.1007/s00439-020-02203-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022]
Abstract
As the most common cardiac arrhythmia, atrial fibrillation (AF) is a major risk factor for stroke, heart failure, and premature death with considerable associated costs. However, no available treatment options have optimal benefit-harm profiles currently, reflecting an incomplete understanding of the biological mechanisms underlying this complex arrhythmia. Recently, molecular epidemiological studies, especially genome-wide association studies, have emphasized the substantial genetic component of AF etiology. A comprehensive mapping of the genetic underpinnings for AF can expand our knowledge of AF mechanism and further facilitate the process of locating novel therapeutics for AF. Here we provide a state-of-the-art review of the molecular genetics of AF incorporating evidence from linkage analysis and candidate gene, as well as genome-wide association studies of common variations and rare copy number variations; potential epigenetic modifications (e.g., DNA methylation, histone modification, and non-coding RNAs) are also involved. We also outline the challenges in mechanism investigation and potential future directions in this article.
Collapse
Affiliation(s)
- Xin Huang
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Yuhui Li
- Department of Cardiology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Junguo Zhang
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Xiaojie Wang
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Ziyi Li
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Guowei Li
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, 466 Newport Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China. .,Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University Hamilton, 1280 Main St West, Hamilton, ON, L8S 4L8, Canada.
| |
Collapse
|
20
|
Zhang K, Ma Z, Song C, Duan X, Yang Y, Li G. Role of ion channels in chronic intermittent hypoxia-induced atrial remodeling in rats. Life Sci 2020; 254:117797. [PMID: 32417371 DOI: 10.1016/j.lfs.2020.117797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/19/2022]
Abstract
AIMS Atrial remodeling, including structural and electrical remodeling, is considered as the substrate in the development of atrial fibrillation (AF). Structural remodeling mainly involves atrial fibrosis, and electrical remodeling is closely related to the changes of ion channels in atrial myocytes. In this study, we aimed to investigate the changes of ion channels in atrial remodeling induced by CIH in rats, which provide the explication for the mechanisms of AF. MATERIALS AND METHODS 80 male Sprague-Dawley rats were randomized into two groups: Control and CIH group (n = 40). CIH rats were subjected to CIH 8 h/d for 30 days. Atrial epicardial conduction velocity, conduction inhomogeneity and AF inducibility were examined. Masson's trichrome staining was used to evaluate the extent of atrial fibrosis, and the expression levels of ion channel subunits were measured by RT-qPCR, Western blot, and IHC, respectively. The remaining 40 rats were used for whole-cell patch clamp experiments. Action potential, INa, ICa-L, Ito were recorded and compared between two groups. KEY FINDINGS CIH rats showed increased AF inducibility, atrial interstitial collagen deposition, APD, expression levels of RyR2, p-RyR2, CaMKII, p-CaMKII, and decreased atrial epicardial conduction velocity, expression levels of Nav1.5, Cav1.2, Kv1.5, Kv4.2, Kv4.3 compared to the Control rats, and the current density of INa, ICa-L, Ito were significantly decreased in CIH group. SIGNIFICANCE We observed significant atrial remodeling induced by CIH in our rat model, which was characterized by changes in ion channels. These changes may be the mechanisms of CIH promoting AF.
Collapse
Affiliation(s)
- Kai Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zuowang Ma
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Chen Song
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xiaorui Duan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yu Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
21
|
Connell P, Word TA, Wehrens XHT. Targeting pathological leak of ryanodine receptors: preclinical progress and the potential impact on treatments for cardiac arrhythmias and heart failure. Expert Opin Ther Targets 2020; 24:25-36. [PMID: 31869254 DOI: 10.1080/14728222.2020.1708326] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction: Type-2 ryanodine receptor (RyR2) located on the sarcoplasmic reticulum initiate systolic Ca2+ transients within cardiomyocytes. Proper functioning of RyR2 is therefore crucial to the timing and force generated by cardiomyocytes within a healthy heart. Improper intracellular Ca2+ handing secondary to RyR2 dysfunction is associated with a variety of cardiac pathologies including catecholaminergic polymorphic ventricular tachycardia (CPVT), atrial fibrillation (AF), and heart failure (HF). Thus, RyR2 and its associated accessory proteins provide promising drug targets to scientists developing therapeutics for a variety of cardiac pathologies.Areas covered: In this article, we review the role of RyR2 in a variety of cardiac pathologies. We performed a literature search utilizing PubMed and MEDLINE as well as reviewed registries of trials from clinicaltrials.gov from 2010 to 2019 for novel therapeutic approaches that address the cellular mechanisms underlying CPVT, AF, and HF by specifically targeting defective RyR2 channels.Expert opinion: The negative impact of cardiac dysfunction on human health and medical economics are major motivating factors for establishing new and effective therapeutic approaches. Focusing on directly impacting the molecular mechanisms underlying defective Ca2+ handling by RyR2 in HF and arrhythmia has great potential to be translated into novel and innovative therapies.
Collapse
Affiliation(s)
- Patrick Connell
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.,Departments of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Tarah A Word
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.,Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.,Departments of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA.,Medicine (Cardiology, Baylor College of Medicine, Houston, TX, USA.,Neuroscience, Baylor College of Medicine, Houston, TX, USA.,Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
22
|
Jędrychowska J, Korzh V. Kv2.1 voltage-gated potassium channels in developmental perspective. Dev Dyn 2019; 248:1180-1194. [PMID: 31512327 DOI: 10.1002/dvdy.114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 11/11/2022] Open
Abstract
Kv2.1 voltage-gated potassium channels consist of two types of α-subunits: (a) electrically-active Kcnb1 α-subunits and (b) silent or modulatory α-subunits plus β-subunits that, similar to silent α-subunits, also regulate electrically-active subunits. Voltage-gated potassium channels were traditionally viewed, mainly by electrophysiologists, as regulators of the electrical activity of the plasma membrane in excitable cells, a role that is performed by transmembrane protein domains of α-subunits that form the electric pore. Genetic studies revealed a role for this region of α-subunits of voltage-gated potassium channels in human neurodevelopmental disorders, such as epileptic encephalopathy. The N- and C-terminal domains of α-subunits interact to form the cytoplasmic subunit of heterotetrameric potassium channels that regulate electric pores. Subsequent animal studies revealed the developmental functions of Kcnb1-containing voltage-gated potassium channels and illustrated their role during brain development and reproduction. These functions of potassium channels are discussed in this review in the context of regulatory interactions between electrically-active and regulatory subunits.
Collapse
Affiliation(s)
- Justyna Jędrychowska
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| |
Collapse
|
23
|
D’Souza A, Trussell T, Morris GM, Dobrzynski H, Boyett MR. Supraventricular Arrhythmias in Athletes: Basic Mechanisms and New Directions. Physiology (Bethesda) 2019; 34:314-326. [DOI: 10.1152/physiol.00009.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Athletes are prone to supraventricular rhythm disturbances including sinus bradycardia, heart block, and atrial fibrillation. Mechanistically, this is attributed to high vagal tone and cardiac electrical and structural remodeling. Here, we consider the supporting evidence for these three pro-arrhythmic mechanisms in athletic human cohorts and animal models, featuring current controversies, emerging data, and future directions of relevance to the translational research agenda.
Collapse
Affiliation(s)
- Alicia D’Souza
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Tariq Trussell
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Gwilym M. Morris
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Mark R. Boyett
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|