1
|
Xue B, Xu Y, Huang R, Zhu Q. Novel target identification towards drug repurposing based on biological activity profiles. PLoS One 2025; 20:e0319865. [PMID: 40327632 PMCID: PMC12054903 DOI: 10.1371/journal.pone.0319865] [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: 10/18/2024] [Accepted: 02/09/2025] [Indexed: 05/08/2025] Open
Abstract
Rare diseases affect more than 30 million individuals, with the majority facing limited treatment options, elevating the urgency to innovative therapeutic solutions. Addressing these medical challenges necessitates an exploration of novel treatment modalities. Among these, drug repurposing emerges as a promising avenue, offering both potential and risk mitigation. To achieve this goal, we primarily focused on developing predictive models that harness cutting-edge computational techniques to uncover latent relationships between gene targets and chemical compounds towards drug repurposing. Building upon our previous investigation, where we successfully identified gene targets for compounds from the Tox21 in vitro assays, our endeavor expanded to a systematic prediction of potential targets for drug repurposing employing machine learning models built on diverse algorithms such as Support Vector Classifier, K-Nearest Neighbors, Random Forest, and Extreme Gradient Boosting. These models were trained on comprehensive biological activity profile data to predict the relationship between 143 gene targets and over 6000 compounds. Our models demonstrated high accuracy (>0.75), with predictions further validated by using public experimental datasets. Furthermore, several findings were evaluated via case studies. By elucidating these connections, we aim to streamline the drug repurposing process, ultimately catalyzing the discovery of more effective therapeutic interventions for rare diseases.
Collapse
Affiliation(s)
- Binghan Xue
- Division of Rare Disease Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| | - Yanji Xu
- Division of Rare Disease Research Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| | - Qian Zhu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| |
Collapse
|
2
|
Su X, Shao B, Chen Z, Gu H, Xiong K, Wang G, Zou Q, Cao Y, Zhang C, Xu H, Yuan Y, Zhao X, Liu Y, Shen Y, Xie D, Chen YH. SNAP25-dependent membrane trafficking of the Kv1.5 channel regulates the onset of atrial fibrillation. Nat Commun 2025; 16:3730. [PMID: 40253375 PMCID: PMC12009440 DOI: 10.1038/s41467-025-59096-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 04/09/2025] [Indexed: 04/21/2025] Open
Abstract
Synaptosomal-associated protein 25 kDa (SNAP25) is essential for vesicular trafficking and protein docking at presynaptic membranes in the nervous system, yet its role in the heart remains poorly understood. Here, we show an unrecognized function of SNAP25, which is selectively expressed in the atria, in regulating atrial electrical remodeling and the onset of atrial fibrillation (AF). SNAP25 protein is downregulated in the atria of AF patients. Cardiomyocyte-specific knockout of SNAP25 in male mice significantly shortens the atrial effective refractory period and action potential duration (APD), increasing susceptibility to AF, which is attributed to elevated Kv1.5 current and membrane expression. Blocking Kv1.5 channels effectively restores atrial APD and reduces AF incidence. Mechanistically, SNAP25 deficiency reduces the internalization of Kv1.5 from the cell surface membrane to early endosomes. In human iPSC-derived atrial cardiomyocytes, SNAP25 deficiency similarly elevates arrhythmic activity and accelerates repolarization. In conclusion, this study reveals that SNAP25 regulates AF susceptibility by controlling the trafficking of the atrial-specific Kv1.5 channel, highlighting SNAP25 as a promising therapeutic target for atrial arrhythmias.
Collapse
Affiliation(s)
- Xuling Su
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Beihua Shao
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhiwen Chen
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hongcheng Gu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ke Xiong
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Guanghua Wang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qicheng Zou
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuting Cao
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Caihong Zhang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hongtao Xu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yixin Yuan
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xuxia Zhao
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yi Liu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yunli Shen
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Duanyang Xie
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yi-Han Chen
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, 200092, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China.
- Affiliated Shanghai Blue Cross Brain hospital, School of Medicine, Tongji University, Shanghai, 200020, China.
| |
Collapse
|
3
|
Lei M, Wu L, Terrar DA, Huang CLH. The modernized classification of cardiac antiarrhythmic drugs: Its application to clinical practice. Heart Rhythm 2025:S1547-5271(25)02300-8. [PMID: 40187508 DOI: 10.1016/j.hrthm.2025.03.1997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/19/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Cardiac arrhythmias pose a major public health problem, and pharmacologic intervention remains key to their therapy. The 1970 landmark Vaughan Williams (VW) classification utilizing known actions of then available antiarrhythmic drugs (AADs) became and remains central to management, but it requires revision in response to extensive subsequent advances. Our modernized AAD classification reflected and sought to facilitate such fundamental physiological and clinical development. Here we respond to requests for an adaptation of our scheme specifically focused on clinical practice. (1) This adaptation improves the accessibility of our original scheme to clinical practice, focusing on key AADs in clinical use rather than investigational new drugs (INDs) while conserving and encompassing the classic VW scheme. (2) We preserve a rational conceptual framework based on current understanding of the relevant electrophysiological events, their underlying cellular or molecular cardiomyocyte targets, and the functional mechanisms they mediate. (3) The adopted subclasses within each AAD class parallel clinical practice by including only subclasses containing established AADs, or approved potential off-label drugs, as opposed to those only including INDs. (4) The simplified scheme remains flexible, permitting drugs to be placed in multiple classes where required, and the addition of classes and subclasses in light of future investigations and clinical approvals. Thus, we derive from our comprehensive modernized AAD classification a more focused and simpler scheme for clinical use. This both modernizes yet preserves the classic VW classification and remains flexible, thus accommodating future developments.
Collapse
Affiliation(s)
- Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom.
| | - Lin Wu
- Department of Cardiology, Peking University First Hospital, Beijing, China.
| | - Derek A Terrar
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom; Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
4
|
Huang W, Jia C, Ren C. Artificial Ion Transporters as Potent Therapeutics for Channelopathies. ChemMedChem 2025; 20:e202400811. [PMID: 39572385 DOI: 10.1002/cmdc.202400811] [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: 10/12/2024] [Revised: 11/21/2024] [Indexed: 12/10/2024]
Abstract
Ion channels are essential for the selective transport of ions, playing a fundamental role in critical physiological processes. Dysfunctions in these channels, often arising from genetic mutations or environmental factors, give rise to a class of disorders collectively known as channelopathies. In recent years, artificial ion transporters have been developed to mimic the essential function of natural channels, offering potential therapeutic approaches for these conditions. Although significant progress has been made in improving the activity and selectivity of these synthetic transporters, their application in treating diseases associated with ion transport dysregulation remains in its infancy. This concept provides an overview of recent advancements in artificial ion transporters for treating channelopathies, while highlighting the key challenges and prospects in translating these developments into practical therapies.
Collapse
Affiliation(s)
- Wei Huang
- Department of Respiratory Medicine, The People's Hospital of Gongan County, Gongan, Hubei, 434300, China
- State Key Laboratory of Cellular Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chunyan Jia
- State Key Laboratory of Cellular Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Changliang Ren
- State Key Laboratory of Cellular Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| |
Collapse
|
5
|
Tomar A, Ahluwalia H, Ramkumar S, Pattnaik S, Nandi D, Raturi P. The interplay of heart rate variability and ventricular repolarization parameters in the obese state: a review. Cardiovasc Endocrinol Metab 2025; 14:e00323. [PMID: 39802372 PMCID: PMC11723674 DOI: 10.1097/xce.0000000000000323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
The impact of obesity on heart rate variability (HRV) and ventricular repolarization, both vital indicators of cardiovascular health, is the focus of this review. Obesity, measured by BMI, waist circumference, and waist-to-hip ratio, significantly increases cardiovascular disease (CVD) risk due to structural and autonomic heart changes. Findings show that obese individuals exhibit prolonged QT and Tpeak-to-Tend (Tpe) intervals, suggesting delayed ventricular recovery and greater arrhythmia risk. Additionally, obesity-induced autonomic imbalance favors sympathetic activity over parasympathetic, reducing HRV and raising arrhythmogenic potential. Elevated QT and Tpe intervals reflect extended cardiac recovery phases, which contribute to poor cardiac outcomes. The Tpe interval could serve as an early marker of cardiac dysfunction in obese populations, highlighting the importance of early intervention to reduce CVD risk and enhance treatment strategies for obesity-related cardiovascular changes.
Collapse
Affiliation(s)
- Akash Tomar
- Department of Physiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka
| | - Himani Ahluwalia
- Department of Physiology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi
| | - S Ramkumar
- Department of Physiology, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, Uttarakhand
| | | | - Debarshi Nandi
- Department of Physiology, Lady Hardinge Medical College and Associated Hospitals
| | - Prashant Raturi
- Department of Cardiology, G B Pant Institute of Postgraduate Medical Education and Research, New Delhi, India
| |
Collapse
|
6
|
Zhuang W, Mun SY, Park WS. Direct effects of antipsychotics on potassium channels. Biochem Biophys Res Commun 2025; 749:151344. [PMID: 39842331 DOI: 10.1016/j.bbrc.2025.151344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Schizophrenia (SCZ) and bipolar disorder (BD) and are severe psychiatric conditions that contribute to disability and increased healthcare costs globally. Although first-, second-, and third-generation antipsychotics are available for treating BD and SCZ, most have various side effects unrelated to their unique functions. Many antipsychotics affect K+ channels (Kv, KCa, Kir, K2P, and other channels), which change the functions of various organs. This review summarizes the biological actions of antipsychotics, including off-target side effects involving K+ channels.
Collapse
Affiliation(s)
- Wenwen Zhuang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
7
|
Zhang Y, MacKinnon R. Higher-order transient structures and the principle of dynamic connectivity in membrane signaling. Proc Natl Acad Sci U S A 2025; 122:e2421280121. [PMID: 39739805 PMCID: PMC11725812 DOI: 10.1073/pnas.2421280121] [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: 10/15/2024] [Accepted: 11/22/2024] [Indexed: 01/02/2025] Open
Abstract
We examine the role of higher-order transient structures (HOTS) in M2R regulation of GIRK channels. Electron microscopic membrane protein location maps show that both proteins form HOTS that exhibit a statistical bias to be near each other. Theoretical calculations and electrophysiological measurements suggest that channel activity is isolated near larger M2R HOTS. By invoking weak interactions that permit transient binding of M2R to M2R and GIRK to GIRK (i-i interactions) and M2R to GIRK (i-j interactions), the distribution patterns and electrophysiological properties of HL-1 cells are replicated in a reaction-diffusion simulation. We propose the principle of dynamic connectivity to explain communication between protein components of a membrane signaling pathway. Dynamic connectivity is mediated by weak, transient interactions between proteins. HOTS created by weak i-i interactions, and statistical biases created by weak i-j interactions promoted by the multivalence of HOTS, are the key elements of dynamic connectivity.
Collapse
Affiliation(s)
- Yuxi Zhang
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| |
Collapse
|
8
|
Palmen R, Walton M, Wagner J. Pediatric flecainide pharmacogenomics: a roadmap to delivering precision-based care to pediatrics arrhythmias. Front Pharmacol 2024; 15:1477485. [PMID: 39741635 PMCID: PMC11686437 DOI: 10.3389/fphar.2024.1477485] [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: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Flecainide acetate is a Class 1c anti-arrhythmic with a potent sodium voltage gated channel blockade which is utilized for the second-line treatment of tachyarrhythmias in children and adults. Given its narrow therapeutic index, the individualization of drug therapy is of utmost importance for clinicians. Despite efforts to improve anti-arrhythmic drug therapy, there remain knowledge gaps regarding the impact of variation in the genes relevant to flecainide's disposition and response. This variability is compounded in developing children whose drug disposition and response pathways may remain immature. The purpose of this comprehensive review is to outline flecainide's disposition and response pathways while simultaneously highlighting opportunities for prospective investigation in the pediatric population.
Collapse
Affiliation(s)
- Ronald Palmen
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Mollie Walton
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| | - Jonathan Wagner
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| |
Collapse
|
9
|
Martínez-Hernández L, López-Vera E, Aguilar MB. Peptide Toxins from Marine Conus Snails with Activity on Potassium Channels and/or Currents. Toxins (Basel) 2024; 16:504. [PMID: 39728762 PMCID: PMC11728717 DOI: 10.3390/toxins16120504] [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: 10/24/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Toxins from Conus snails are peptides characterized by a great structural and functional diversity. They have a high affinity for a wide range of membrane proteins such as ion channels, neurotransmitter transporters, and G protein-coupled receptors. Potassium ion channels are integral proteins of cell membranes that play vital roles in physiological processes in muscle and neuron cells, among others, and reports in the literature indicate that perturbation in their function (by mutations or ectopic expression) may result in the development and progression of different ailments in humans. This review aims to gather as much information as possible about Conus toxins (conotoxins) with an effect on potassium channels and/or currents, with a perspective of exploring the possibility of finding or developing a possible drug candidate from these toxins. The research indicates that, among the more than 900 species described for this genus, in only 14 species of the >100 studied to date have such toxins been found (classified according to the most specific evidence for each case), as follows: 17 toxins with activity on two groups of potassium channels (Kv and KCa), 4 toxins with activity on potassium currents, and 5 toxins that are thought to inhibit potassium channels by symptomatology and/or a high sequence similarity.
Collapse
Affiliation(s)
- Luis Martínez-Hernández
- Posgrado en Ciencias Biológicas, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Estuardo López-Vera
- Laboratorio de Toxinología Marina, Unidad Académica de Ecología y Biodiversidad Acuática, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Manuel B. Aguilar
- Laboratorio de Neurofarmacología Marina, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, Mexico
| |
Collapse
|
10
|
Hollenberg SM. Don't-Stop Believing! Crit Care Med 2024; 52:1641-1643. [PMID: 39283205 DOI: 10.1097/ccm.0000000000006377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
|
11
|
Rubinstein J, Pinney SM, Xie C, Wang HS. Association of same-day urinary phenol levels and cardiac electrical alterations: analysis of the Fernald Community Cohort. Environ Health 2024; 23:76. [PMID: 39300535 PMCID: PMC11412060 DOI: 10.1186/s12940-024-01114-x] [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: 05/19/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Exposure to phenols has been linked in animal models and human populations to cardiac function alterations and cardiovascular diseases, although their effects on cardiac electrical properties in humans remains to be established. This study aimed to identify changes in electrocardiographic (ECG) parameters associated with environmental phenol exposure in adults of a midwestern large cohort known as the Fernald Community Cohort (FCC). METHODS During the day of the first comprehensive medical examination, urine samples were obtained, and electrocardiograms were recorded. Cross-sectional linear regression analyses were performed. RESULTS Bisphenol A (BPA) and bisphenol F (BPF) were both associated with a longer PR interval, an indication of delayed atrial-to-ventricle conduction, in females (p < 0.05) but not males. BPA combined with BPF was associated with an increase QRS duration, an indication of delayed ventricular activation, in females (P < 0.05) but not males. Higher triclocarban (TCC) level was associated with longer QTc interval, an indication of delayed ventricular repolarization, in males (P < 0.01) but not females. Body mass index (BMI) was associated with a significant increase in PR and QTc intervals and ventricular rate in females and in ventricular rate in males. In females, the combined effect of being in the top tertile for both BPA urinary concentration and BMI was an estimate of a 10% increase in PR interval. No associations were found with the other phenols. CONCLUSION Higher exposure to some phenols was associated with alterations of cardiac electrical properties in a sex specific manner in the Fernald cohort. Our population-based findings correlate directly with clinically relevant parameters that are associated with known pathophysiologic cardiac conditions in humans.
Collapse
Affiliation(s)
- Jack Rubinstein
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Susan M Pinney
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Changchun Xie
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Hong-Sheng Wang
- Department of Pharmacology, Physiology and Neurobiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
12
|
Hu Y, Qu C, Zou Y, Liu X, Zhang C, Yang B. NBQX mediates ventricular fibrillation susceptibility in rat models of anxiety via the Nrf2/HO-1 pathway. Heliyon 2024; 10:e37358. [PMID: 39296140 PMCID: PMC11408043 DOI: 10.1016/j.heliyon.2024.e37358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/11/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Objective Anxiety disorder (AD) is a common mental disorder related to cardiovascular disease morbidity. Oxidative stress plays a crucial role in the anxiety state and can lead to cardiac remodeling. Over-activation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) in cardiomyocytes and neurons can cause oxidative stress. Additionally, the AMPAR inhibitor-2,3-dihydroxy-6-nitro-7-sulfamoyl-benzoquinoxaline-2,3-dione (NBQX) plays an important role in ameliorating oxidative stress. This study aimed to explore the anti-arrhythmic effects of NBQX in a rat model of anxiety. Methods The AD model was induced using empty bottle stimulation. Male Sprague Dawley rats were randomly divided into four groups: control + saline, control + NBQX, AD + saline, and AD + NBQX. Open field test was conducted to measure anxiety-like behavior. Electrophysiological experiments, histological analysis, biochemical detection and molecular biology were performed to verify the effects of NBQX on the amelioration of electrical remodeling and structural remodeling. Furthermore, the nuclear factor erythroid 2-related factor 2 (Nrf2) inhibitor (ML385) was used in vitro to demonstrate the signaling pathway. Results Oxidative stress levels increased with AMPAR over-activation in AD rats, leading to heightened vulnerability to ventricular fibrillation (VF). NBQX reverses anxiety and VF susceptibility. Our results showed that NBQX activated the Nrf2/heme oxygenase-1 (HO-1) pathway, leading to a decline in oxidative stress levels. Connexin 43 and ion channel expression was upregulated. NBQX treatment attenuated fibrosis and apoptosis. This effect was diminished by ML385 treatment in vitro. Conclusion NBQX can alleviate VF susceptibility in rat models of anxiety by alleviating electrical remodeling, structural remodeling via regulating the Nrf2/HO-1 pathway to some extent.
Collapse
Affiliation(s)
- Yiqian Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Ying Zou
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| |
Collapse
|
13
|
Tao Y, Yao K, Wu J, Xu T, Lin J, Qin Y, Su D, Cai S, Yu W, Chen X. Intravenous anesthetics have differential effects on human potassium channels. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1594-1603. [PMID: 40384046 PMCID: PMC11659785 DOI: 10.3724/abbs.2024151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/02/2024] [Indexed: 05/20/2025] Open
Abstract
General anesthetics are widely used in the clinic and greatly promote the development of surgery. However, the incidence of cardiovascular and respiratory complications caused by general anesthetics is still high, and the underlying mechanisms remain incompletely understood. Potassium channels are widely expressed in the heart and blood vessels and participate in regulating blood pressure, heart rate, and other physiological parameters. Whether they are directly affected by intravenous general anesthetics is unclear. Here, we independently express four classes of potassium channels, TASK-1, TASK-3, Kv1.5, Kv2.1, Kir2.1, SK1 and SK3, in Xenopus oocytes. The effects of propofol, pentobarbital and ketamine on these channels are evaluated by their current change. We find that propofol and ketamine potentiate TASK-3 and SK3 current respectively, while pentobarbital and ketamine inhibit SK1 current. To identify the key residues in TASK-3, SK1 and SK3 that interact with intravenous anesthetics, we predict homology models of the three channels and perform molecular docking simulations. The results show that propofol forms a hydrogen bond with Q126 of TASK-3, ketamine forms a hydrogen bond with S290 of SK1 and S467 of SK3, while pentobarbital forms hydrogen bonds with S330 and T358 of SK1. As these potassium channels are closely related to respiratory system regulation, cardiac rhythm and vasodilation, our study provides a new perspective for further study on the mechanism of general anesthetics-induced respiratory and circulatory side effects.
Collapse
Affiliation(s)
- Ying Tao
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Kejie Yao
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai 200031China; University of Chinese Academy of SciencesBeijing101408China
| | - Jing Wu
- Department of Equipment and MaterialsBiomedical R&D Project TeamZhongshan HospitalFudan UniversityShanghai200031China
| | - Tian Xu
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Junhui Lin
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Yi Qin
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Diansan Su
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Shiqing Cai
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai 200031China; University of Chinese Academy of SciencesBeijing101408China
| | - Weifeng Yu
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| | - Xuemei Chen
- Department of AnesthesiologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghai200127China
| |
Collapse
|
14
|
Zhan G, Wang X, Wang X, Li J, Tang Y, Bi H, Yang X, Xia Y. Dapagliflozin: A sodium-glucose cotransporter 2 inhibitor, attenuates angiotensin II-induced atrial fibrillation by regulating atrial electrical and structural remodeling. Eur J Pharmacol 2024; 978:176712. [PMID: 38906237 DOI: 10.1016/j.ejphar.2024.176712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/23/2024]
Abstract
AIM Atrial fibrillation (AF), the most common arrhythmia, is characterized by atrial electrical and structural remodeling. Previous studies have found that sodium-glucose cotransporter 2 inhibitor (SGLT2i) can protect myocardium in a glucose independent mechanism. But the role of SGLT2i in regulating AF remains largely unknown. This study, we aimed to investigate the effect of Dapagliflozin (DAPA) in reducing AF susceptibility via inhibiting electrical and structural remodeling. METHOD The mouse model was established by Angiotensin II (2000 ng/kg/min) infusion for 3 weeks, and an in vitro model was generated by stimulating HL-1 and primary mouse fibroblast with Ang II (1 μM) for 24 h. Programmed electrical stimulation, ECG and whole-cell patch clamp were used to detect DAPA effect on atrial electrical remodeling induced by Ang II. To observe DAPA effect on atrial structural remodeling induced by Ang II, we used echocardiographic, H&E and Masson staining to evaluate atrial dilation. To further explore the protective mechanism of DAPA, we adopt in silico molecular docking approaches to investigate the binding affinity of Ang II and CaMKII at Met-281 site. Western blot was to detect expression level of CaMKII, ox-CaMKII, Nav1.5, Kv4.3, Kv4.2, Kchip2, Kir2.1 and Cx40. RESULTS Ang II induced AF, atrial dilatation and fibrosis, led to atrial electrical and structural remodeling. However, these effects were markedly abrogated by DAPA treatment, a specific SGLT2i. Our observation of atrial electrical activity in mice revealed that DAPA could rescue the prolonged action potential duration (APD) and the abnormal currents of IK1, Ito and INaL triggered by Ang II infusion. DAPA could reduce the binding affinity of Ang II and CaMKII at Met-281 site, which indicated that DAPA may directly alleviate the activation of CaMKII caused by Ang II. DAPA could reduce the upregulation of ox-CaMKII caused by Ang II infusion in atrial tissues. Moreover, DAPA also ameliorated the aberrant expression levels of electrical activity related proteins (Nav1.5, Kv4.3, Kv4.2, Kchip2, Kir2.1 and Cx40) and fibrosis related signal pathways (TGF-β1, p-smad/smad) caused by Ang II. Furthermore, we confirmed that DAPA, as well as other SGLT2i (EMPA, CANA), could reverse these abnormalities caused by Ang II incubation in HL-1 cells and primary mouse fibroblasts, respectively. CONCLUSION Overall, our study identifies DAPA, a widely used SGLT2i, contributes to inhibiting Ang II-induced ox-CaMKII upregulation and electrical and structural remodeling to reduce AF susceptibility, suggesting that DAPA may be a potential therapy of treating AF.
Collapse
Affiliation(s)
- Ge Zhan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Xinying Wang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Xin Wang
- Department of Ultrasound, The Affiliated Hospital of Innermongolia Medical University, Huhhot 010050, China; Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiatian Li
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Yuqi Tang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Hailian Bi
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China
| | - Xiaolei Yang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China; Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China.
| | - Yunlong Xia
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China; Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, China.
| |
Collapse
|
15
|
Yu M, Li W, Yu Y, Zhao Y, Xiao L, Lauschke VM, Cheng Y, Zhang X, Wang Y. Deep learning large-scale drug discovery and repurposing. NATURE COMPUTATIONAL SCIENCE 2024; 4:600-614. [PMID: 39169261 DOI: 10.1038/s43588-024-00679-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
Large-scale drug discovery and repurposing is challenging. Identifying the mechanism of action (MOA) is crucial, yet current approaches are costly and low-throughput. Here we present an approach for MOA identification by profiling changes in mitochondrial phenotypes. By temporally imaging mitochondrial morphology and membrane potential, we established a pipeline for monitoring time-resolved mitochondrial images, resulting in a dataset comprising 570,096 single-cell images of cells exposed to 1,068 United States Food and Drug Administration-approved drugs. A deep learning model named MitoReID, using a re-identification (ReID) framework and an Inflated 3D ResNet backbone, was developed. It achieved 76.32% Rank-1 and 65.92% mean average precision on the testing set and successfully identified the MOAs for six untrained drugs on the basis of mitochondrial phenotype. Furthermore, MitoReID identified cyclooxygenase-2 inhibition as the MOA of the natural compound epicatechin in tea, which was successfully validated in vitro. Our approach thus provides an automated and cost-effective alternative for target identification that could accelerate large-scale drug discovery and repurposing.
Collapse
Affiliation(s)
- Min Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | | | - Yunru Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yu Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lizhi Xiao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Yiyu Cheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
| | - Xingcai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, China.
- Center for system biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Islam KN, Nguyen ID, Islam R, Pirzadah H, Malik H. Roles of Hydrogen Sulfide (H2S) as a Potential Therapeutic Agent in Cardiovascular Diseases: A Narrative Review. Cureus 2024; 16:e64913. [PMID: 39156383 PMCID: PMC11330631 DOI: 10.7759/cureus.64913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
Cardiovascular disease (CVD) stands as one of the leading causes of morbidity and mortality worldwide, and the continued search for novel therapeutics is vital for addressing this global health challenge. Over the past decade, hydrogen sulfide (H₂S) has garnered significant attention in the field of medical research, as it has been proven to be a cardioprotective gaseous signaling molecule. It joins nitric oxide and carbon monoxide as endogenously produced gasotransmitters. As for its mechanism, H₂S functions through the posttranslational addition of a sulfur group to cysteine residues on target proteins in a process called sulfhydration. As a result, the observed physiological effects of H₂S can include vasodilation, anti-apoptosis, anti-inflammation, antioxidant effects, and regulation of ion channels. Various studies have observed the cardioprotective benefits of H₂S in diseases such as myocardial infarction, ischemia-reperfusion injury, cardiac remodeling, heart failure, arrhythmia, and atherosclerosis. In this review, we discuss the mechanisms and therapeutic potential of H₂S in various CVDs.
Collapse
Affiliation(s)
- Kazi N Islam
- Department of Agricultural Research Development Program, Central State University, Wilberforce, USA
| | - Ivan D Nguyen
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Rahib Islam
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Humza Pirzadah
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Hassan Malik
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| |
Collapse
|
17
|
Rubinstein J, Pinney SM, Xie C, Wang HS. Association of same-day urinary phenol levels and cardiac electrical alterations: analysis of the Fernald Community Cohort. RESEARCH SQUARE 2024:rs.3.rs-4445657. [PMID: 38853936 PMCID: PMC11160919 DOI: 10.21203/rs.3.rs-4445657/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Exposure to phenols has been linked in animal models and human populations to cardiac function alterations and cardiovascular diseases, although their effects on cardiac electrical properties in humans remains to be established. This study aimed to identify changes in electrocardiographic (ECG) parameters associated with environmental phenol exposure in adults of a midwestern large cohort known as the Fernald Community Cohort (FCC). Methods During the day of the first comprehensive medical examination, urine samples were obtained, and electrocardiograms were recorded. Cross-sectional linear regression analyses were performed. Results Bisphenol A (BPA) and bisphenol F (BPF) were both associated with a longer PR interval, an indication of delayed atrial-to-ventricle conduction, in females (p < 0.05) but not males. BPA combined with BPF was associated with an increase QRS duration, an indication of delayed ventricular activation, in females (P < 0.05) but not males. Higher triclocarban (TCC) level was associated with longer QTc interval, an indication of delayed ventricular repolarization, in males (P < 0.01) but not females. Body mass index (BMI) was associated with a significant increase in PR and QTc intervals and ventricular rate in females and in ventricular rate in males. In females, the combined effect of being in the top tertile for both BPA urinary concentration and BMI was an estimate of a 10% increase in PR interval. No associations were found with the other phenols. Conclusion Higher exposure to some phenols was associated with alterations of cardiac electrical properties in a sex specific manner in the Fernald cohort. Our population-based findings correlate directly with clinically relevant parameters that are associated with known pathophysiologic cardiac conditions in humans.
Collapse
|
18
|
Isaksen JL, Sivertsen CB, Jensen CZ, Graff C, Linz D, Ellervik C, Jensen MT, Jørgensen PG, Kanters JK. Electrocardiographic markers in patients with type 2 diabetes and the role of diabetes duration. J Electrocardiol 2024; 84:129-136. [PMID: 38663227 DOI: 10.1016/j.jelectrocard.2024.04.003] [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: 02/05/2024] [Revised: 04/01/2024] [Accepted: 04/14/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND The association between type 2 diabetes and electrocardiographic (ECG) markers are incompletely explored and the dependence on diabetes duration is largely unknown. We aimed to investigate the electrocardiographic (ECG) changes associated with type 2 diabetes over time. METHODS In this cross-sectional study, we matched people with type 2 diabetes 1:1 on sex, age, and body mass index with people without diabetes from the general population. We regressed ECG markers with the presence of diabetes and the duration of clinical diabetes, respectively, adjusted for sex, age, body mass index, smoking, heart rate, diabetes medication, renal function, hypertension, and myocardial infarction. RESULTS We matched 988 people with type 2 diabetes (332, 34% females) with as many controls. Heart rate was 8 bpm higher (p < 0.001) in people with vs. without type 2 diabetes, but the difference declined with increasing diabetes duration. For most depolarization markers, the difference between people with and without type 2 diabetes increased progressively with diabetes duration. On average, R-wave amplitude was 6 mm lower in lead V5 (p < 0.001), P-wave duration was 5 ms shorter (p < 0.001) and QRS duration was 3 ms (p = 0.03). Among repolarization markers, T-wave amplitude (measured in V5) was lower in patients with type 2 diabetes (1 mm lower, p < 0.001) and the QRS-T angle was 10 degrees wider (p = 0.002). We observed no association between diabetes duration and repolarization markers. CONCLUSIONS Type 2 diabetes was independently associated with electrocardiographic depolarization and repolarization changes. Differences in depolarization markers, but not repolarization markers, increased with increasing diabetes duration.
Collapse
Affiliation(s)
- Jonas L Isaksen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Christian B Sivertsen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Zinck Jensen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Graff
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Dominik Linz
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Data and Data Support, Region Zealand, Sorø, Denmark
| | | | - Peter G Jørgensen
- Department of Cardiology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Center of Physiological Research, University of California San Francisco, San Francisco, USA
| |
Collapse
|
19
|
Zavarzina II, Kuzmenkov AI, Dobrokhotov NA, Maleeva EE, Korolkova YV, Peigneur S, Tytgat J, Krylov NA, Vassilevski AA, Chugunov AO. The scorpion toxin BeKm-1 blocks hERG cardiac potassium channels using an indispensable arginine residue. FEBS Lett 2024; 598:889-901. [PMID: 38563123 DOI: 10.1002/1873-3468.14850] [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: 12/28/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 04/04/2024]
Abstract
BeKm-1 is a peptide toxin from scorpion venom that blocks the pore of the potassium channel hERG (Kv11.1) in the human heart. Although individual protein structures have been resolved, the structure of the complex between hERG and BeKm-1 is unknown. Here, we used molecular dynamics and ensemble docking, guided by previous double-mutant cycle analysis data, to obtain an in silico model of the hERG-BeKm-1 complex. Adding to the previous mutagenesis study of BeKm-1, our model uncovers the key role of residue Arg20, which forms three interactions (a salt bridge and hydrogen bonds) with the channel vestibule simultaneously. Replacement of this residue even by lysine weakens the interactions significantly. In accordance, the recombinantly produced BeKm-1R20K mutant exhibited dramatically decreased activity on hERG. Our model may be useful for future drug design attempts.
Collapse
Affiliation(s)
- Iana I Zavarzina
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | | | - Nikita A Dobrokhotov
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | | | | | | | - Jan Tytgat
- Toxicology and Pharmacology, KU Leuven, Belgium
| | - Nikolay A Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Alexander A Vassilevski
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Anton O Chugunov
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
20
|
Ko MY, Chon SH, Park H, Min E, Kim Y, Cha SW, Seo JW, Lee BS, Ka M, Hyun SA. Perfluorooctanoic acid induces cardiac dysfunction in human induced pluripotent stem cell-derived cardiomyocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116170. [PMID: 38452704 DOI: 10.1016/j.ecoenv.2024.116170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/22/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024]
Abstract
Perfluorooctanoic acid (PFOA), commonly found in drinking water, leads to widespread exposure through skin contact, inhalation, and ingestion, resulting in detectable levels of PFOA in the bloodstream. In this study, we found that exposure to PFOA disrupts cardiac function in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). We observed reductions in field and action potentials in hiPSC-CMs exposed to PFOA. Furthermore, PFOA demonstrated a dose-dependent inhibitory effect on various ion channels, including the calcium, sodium, and potassium channels. Additionally, we noted dose-dependent inhibition of the expression of these ion channels in hiPSC-CMs following exposure to PFOA. These findings suggest that PFOA exposure can impair cardiac ion channel function and decrease the transcription of genes associated with these channels, potentially contributing to cardiac dysfunction such as arrhythmias. Our study sheds light on the electrophysiological and epigenetic consequences of PFOA-induced cardiac dysfunction, underscoring the importance of further research on the cardiovascular effects of perfluorinated compounds (PFCs).
Collapse
Affiliation(s)
- Moon Yi Ko
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Sun-Hwa Chon
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea; Graduate School of Pre-Clinical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea
| | - Heejin Park
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Euijun Min
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Younhee Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Sin-Woo Cha
- Department of Nonclinical Studies, Korea Institute of Toxicology, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Joung-Wook Seo
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Byoung-Seok Lee
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| | - Minhan Ka
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| | - Sung-Ae Hyun
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| |
Collapse
|
21
|
Lei M, Salvage SC, Jackson AP, Huang CLH. Cardiac arrhythmogenesis: roles of ion channels and their functional modification. Front Physiol 2024; 15:1342761. [PMID: 38505707 PMCID: PMC10949183 DOI: 10.3389/fphys.2024.1342761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024] Open
Abstract
Cardiac arrhythmias cause significant morbidity and mortality and pose a major public health problem. They arise from disruptions in the normally orderly propagation of cardiac electrophysiological activation and recovery through successive cardiomyocytes in the heart. They reflect abnormalities in automaticity, initiation, conduction, or recovery in cardiomyocyte excitation. The latter properties are dependent on surface membrane electrophysiological mechanisms underlying the cardiac action potential. Their disruption results from spatial or temporal instabilities and heterogeneities in the generation and propagation of cellular excitation. These arise from abnormal function in their underlying surface membrane, ion channels, and transporters, as well as the interactions between them. The latter, in turn, form common regulatory targets for the hierarchical network of diverse signaling mechanisms reviewed here. In addition to direct molecular-level pharmacological or physiological actions on these surface membrane biomolecules, accessory, adhesion, signal transduction, and cytoskeletal anchoring proteins modify both their properties and localization. At the cellular level of excitation-contraction coupling processes, Ca2+ homeostatic and phosphorylation processes affect channel activity and membrane excitability directly or through intermediate signaling. Systems-level autonomic cellular signaling exerts both acute channel and longer-term actions on channel expression. Further upstream intermediaries from metabolic changes modulate the channels both themselves and through modifying Ca2+ homeostasis. Finally, longer-term organ-level inflammatory and structural changes, such as fibrotic and hypertrophic remodeling, similarly can influence all these physiological processes with potential pro-arrhythmic consequences. These normal physiological processes may target either individual or groups of ionic channel species and alter with particular pathological conditions. They are also potentially alterable by direct pharmacological action, or effects on longer-term targets modifying protein or cofactor structure, expression, or localization. Their participating specific biomolecules, often clarified in experimental genetically modified models, thus constitute potential therapeutic targets. The insights clarified by the physiological and pharmacological framework outlined here provide a basis for a recent modernized drug classification. Together, they offer a translational framework for current drug understanding. This would facilitate future mechanistically directed therapeutic advances, for which a number of examples are considered here. The latter are potentially useful for treating cardiac, in particular arrhythmic, disease.
Collapse
Affiliation(s)
- Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samantha C. Salvage
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Antony P. Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
22
|
Lopez-Medina AI, Campos-Staffico AM, A Chahal CA, Volkers I, Jacoby JP, Berenfeld O, Luzum JA. Genetic risk factors for drug-induced long QT syndrome: findings from a large real-world case-control study. Pharmacogenomics 2024; 25:117-131. [PMID: 38506312 PMCID: PMC10964839 DOI: 10.2217/pgs-2023-0229] [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: 12/07/2023] [Accepted: 02/23/2024] [Indexed: 03/21/2024] Open
Abstract
Aim: Drug-induced long QT syndrome (diLQTS), an adverse effect of many drugs, can lead to sudden cardiac death. Candidate genetic variants in cardiac ion channels have been associated with diLQTS, but several limitations of previous studies hamper clinical utility. Materials & methods: Thus, the purpose of this study was to assess the associations of KCNE1-D85N, KCNE2-I57T and SCN5A-G615E with diLQTS in a large observational case-control study (6,083 self-reported white patients treated with 27 different high-risk QT-prolonging medications; 12.0% with diLQTS). Results: KCNE1-D85N significantly associated with diLQTS (adjusted odds ratio: 2.24 [95% CI: 1.35-3.58]; p = 0.001). Given low minor allele frequencies, the study had insufficient power to analyze KCNE2-I57T and SCN5A-G615E. Conclusion: KCNE1-D85N is a risk factor for diLQTS that should be considered in future clinical practice guidelines.
Collapse
Affiliation(s)
- Ana I Lopez-Medina
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | | | - Choudhary Anwar A Chahal
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Cardiology, Barts Heart Centre, London, UK
| | - Isabella Volkers
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Juliet P Jacoby
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Omer Berenfeld
- Center for Arrhythmia Research, Departments of Internal Medicine – Cardiology, Biomedical Engineering, & Applied Physics, University of Michigan, Ann Arbor, MI, USA
| | - Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Sevamontree C, Jintajirapan S, Phakdeekitcharoen P, Vathesatogkit P, Phakdeekitcharoen B. The Factors Associated With Hyperkalemia-Induced Electrocardiogram Changes in the Outpatient Setting. Am J Cardiol 2024; 211:141-142. [PMID: 37944776 DOI: 10.1016/j.amjcard.2023.10.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023]
Affiliation(s)
- Chadapa Sevamontree
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Supreeya Jintajirapan
- Out Patient Intervention and Urgency Care, Department of Nursing, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Prin Vathesatogkit
- Division of Cardiology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Bunyong Phakdeekitcharoen
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
24
|
Kjeldsen ST, Nissen SD, Saljic A, Hesselkilde EM, Carstensen H, Sattler SM, Jespersen T, Linz D, Hopster-Iversen C, Kutieleh R, Sanders P, Buhl R. Structural and electro-anatomical characterization of the equine pulmonary veins: implications for atrial fibrillation. J Vet Cardiol 2024; 52:1-13. [PMID: 38290222 DOI: 10.1016/j.jvc.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION/OBJECTIVES Spontaneous pulmonary vein (PV) activity triggers atrial fibrillation (AF) in humans. Although AF frequently occurs in horses, the origin remains unknown. This study investigated the structural and electro-anatomical properties of equine PVs to determine the potential presence of an arrhythmogenic substrate. ANIMALS, MATERIALS AND METHODS Endocardial three-dimensional electro-anatomical mapping (EnSite Precision) using high-density (HD) catheters was performed in 13 sedated horses in sinus rhythm. Left atrium (LA) access was obtained retrogradely through the carotid artery. Post-mortem, tissue was harvested from the LA, right atrium (RA), and PVs for histological characterization and quantification of ion channel expression using immunohistochemical analysis. RESULTS Geometry, activation maps, and voltage maps of the PVs were created and a median of four ostia were identified. Areas of reduced conduction were found at the veno-atrial junction. The mean myocardial sleeve length varied from 28 ± 13 to 49 ± 22 mm. The PV voltage was 1.2 ± 1.4 mV and lower than the LA (3.4 ± 0.9 mV, P < 0.001). The fibrosis percentage was higher in PV myocardium (26.1 ± 6.6 %) than LA (14.5 ± 5.0 %, P = 0.003). L-type calcium channel (CaV1.2) expression was higher in PVs than LA (P = 0.001). T-type calcium channels (CaV3.3), connexin-43, ryanodine receptor-2, and small conductance calcium-activated potassium channel-3 was expressed in PVs. CONCLUSIONS The veno-atrial junction had lower voltages, increased structural heterogeneity and areas of slower conduction. Myocardial sleeves had variable lengths, and a different ion channel expression compared to the atria. Heterogeneous properties of the PVs interacting with the adjacent LA likely provide the milieu for re-entry and AF initiation.
Collapse
Affiliation(s)
- S T Kjeldsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, 2630 Taastrup, Denmark.
| | - S D Nissen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, 2630 Taastrup, Denmark
| | - A Saljic
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - E M Hesselkilde
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - H Carstensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, 2630 Taastrup, Denmark
| | - S M Sattler
- Department of Cardiology, Herlev and Gentofte University Hospital, Gentofte Hospitalsvej 1, 2900 Hellerup, Denmark
| | - T Jespersen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - D Linz
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 632, 6229 ER Maastricht, Netherlands
| | - C Hopster-Iversen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, 2630 Taastrup, Denmark
| | - R Kutieleh
- Abbott Medical, 214 Greenhill Road, SA 5063, Australia
| | - P Sanders
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital and University of Adelaide, Port Rd, SA 5000, Australia
| | - R Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, 2630 Taastrup, Denmark
| |
Collapse
|
25
|
An X, Cho H. Increased GIRK channel activity prevents arrhythmia in mice with heart failure by enhancing ventricular repolarization. Sci Rep 2023; 13:22479. [PMID: 38110503 PMCID: PMC10728207 DOI: 10.1038/s41598-023-50088-2] [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: 06/26/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023] Open
Abstract
Ventricular arrhythmia causing sudden cardiac death is the leading mode of death in patients with heart failure. Yet, the mechanisms that prevent ventricular arrhythmias in heart failure are not well characterized. Using a mouse model of heart failure created by transverse aorta constriction, we show that GIRK channel, an important regulator of cardiac action potentials, is constitutively active in failing ventricles in contrast to normal cells. Evidence is presented indicating that the tonic activation of M2 muscarinic acetylcholine receptors by endogenously released acetylcholine contributes to the constitutive GIRK activity. This constitutive GIRK activity prevents the action potential prolongation in heart failure ventricles. Consistently, GIRK channel blockade with tertiapin-Q induces QT interval prolongation and increases the incidence of arrhythmia in heart failure, but not in control mice. These results suggest that constitutive GIRK channels comprise a key mechanism to protect against arrhythmia by providing repolarizing currents in heart failure ventricles.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
| |
Collapse
|
26
|
Martínez-Hernández L, López-Vera E, Aguilar MB, Rodriguez-Ruiz XC, Ortíz-Arellano MA. κO-SrVIA Conopeptide, a Novel Inhibitor Peptide for Two Members of the Human EAG Potassium Channel Family. Int J Mol Sci 2023; 24:11513. [PMID: 37511269 PMCID: PMC10380377 DOI: 10.3390/ijms241411513] [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: 06/23/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The first conotoxin affecting the voltage-gated potassium channels of the EAG family was identified and characterized from the venom of the vermivorous species Conus spurius from the Gulf of Mexico. This conopeptide, initially named Cs68 and later designated κO-SrVIA, is extremely hydrophobic and comprises 31 amino acid residues, including six Cysteines in the framework VI/VII, and a free C-terminus. It inhibits the currents mediated by two human EAG subtypes, Kv10.1 (IC50 = 1.88 ± 1.08 µM) and Kv11.1 (IC50 = 2.44 ± 1.06 µM), and also the human subtype Kv1.6 (IC50 = 3.6 ± 1.04 µM). Despite its clear effects on potassium channels, it shares a high sequence identity with δ-like-AtVIA and δ-TsVIA. Also, κO-SrVIA is the third conopeptide from the venom of C. spurius with effects on potassium channels, and the seventh conotoxin that blocks Kv1.6 channels.
Collapse
Affiliation(s)
- Luis Martínez-Hernández
- Posgrado en Ciencias Biológicas, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Estuardo López-Vera
- Laboratorio de Toxinología Marina, Unidad Académica de Ecología y Biodiversidad Acuática, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Manuel B. Aguilar
- Laboratorio de Neurofarmacología Marina, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, Mexico;
| | - Ximena C. Rodriguez-Ruiz
- Laboratorio de Toxinología Marina, Unidad Académica de Ecología y Biodiversidad Acuática, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Mónica A. Ortíz-Arellano
- Laboratorio de Malacología, Facultad de Ciencias del Mar, Universidad Autónoma de Sinaloa, Mazatlán 82000, Mexico;
| |
Collapse
|
27
|
Ma J, Wang NY, Jagani R, Wang HS. Proarrhythmic toxicity of low dose bisphenol A and its analogs in human iPSC-derived cardiomyocytes and human cardiac organoids through delay of cardiac repolarization. CHEMOSPHERE 2023; 328:138562. [PMID: 37004823 PMCID: PMC10121900 DOI: 10.1016/j.chemosphere.2023.138562] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/07/2023] [Accepted: 03/31/2023] [Indexed: 06/19/2023]
Abstract
Bisphenol A (BPA) and its analogs are common environmental chemicals with many potential adverse health effects. The impact of environmentally relevant low dose BPA on human heart, including cardiac electrical properties, is not understood. Perturbation of cardiac electrical properties is a key arrhythmogenic mechanism. In particular, delay of cardiac repolarization can cause ectopic excitation of cardiomyocytes and malignant arrhythmia. This can occur as a result of genetic mutations (i.e., long QT (LQT) syndrome), or cardiotoxicity of drugs and environmental chemicals. To define the impact of low dose BPA on electrical properties of cardiomyocytes in a human-relevant model system, we examined the rapid effects of 1 nM BPA in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using patch-clamp and confocal fluorescence imaging. Acute exposure to BPA delayed repolarization and prolonged action potential duration (APD) in hiPSC-CMs through inhibition of the hERG K+ channel. In nodal-like hiPSC-CMs, BPA acutely increased pacing rate through stimulation of the If pacemaker channel. Existing arrhythmia susceptibility determines the response of hiPSC-CMs to BPA. BPA resulted in modest APD prolongation but no ectopic excitation in baseline condition, while rapidly promoted aberrant excitations and tachycardia-like events in myocytes that had drug-simulated LQT phenotype. In hiPSC-CM-based human cardiac organoids, the effects of BPA on APD and aberrant excitation were shared by its analog chemicals, which are often used in "BPA-free" products, with bisphenol AF having the largest effects. Our results reveal that BPA and its analogs have repolarization delay-associated pro-arrhythmic toxicity in human cardiomyocytes, particularly in myocytes that are prone to arrhythmias. The toxicity of these chemicals depends on existing pathophysiological conditions of the heart, and may be particularly pronounced in susceptible individuals. An individualized approach is needed in risk assessment and protection.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | | | - Ravikumar Jagani
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
28
|
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
|
29
|
Huang CLH, Lei M. Cardiomyocyte electrophysiology and its modulation: current views and future prospects. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220160. [PMID: 37122224 PMCID: PMC10150219 DOI: 10.1098/rstb.2022.0160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/10/2023] [Indexed: 05/02/2023] Open
Abstract
Normal and abnormal cardiac rhythms are of key physiological and clinical interest. This introductory article begins from Sylvio Weidmann's key historic 1950s microelectrode measurements of cardiac electrophysiological activity and Singh & Vaughan Williams's classification of cardiotropic targets. It then proceeds to introduce the insights into cardiomyocyte function and its regulation that subsequently emerged and their therapeutic implications. We recapitulate the resulting view that surface membrane electrophysiological events underlying cardiac excitation and its initiation, conduction and recovery constitute the final common path for the cellular mechanisms that impinge upon this normal or abnormal cardiac electrophysiological activity. We then consider progress in the more recently characterized successive regulatory hierarchies involving Ca2+ homeostasis, excitation-contraction coupling and autonomic G-protein signalling and their often reciprocal interactions with the surface membrane events, and their circadian rhythms. Then follow accounts of longer-term upstream modulation processes involving altered channel expression, cardiomyocyte energetics and hypertrophic and fibrotic cardiac remodelling. Consideration of these developments introduces each of the articles in this Phil. Trans. B theme issue. The findings contained in these articles translate naturally into recent classifications of cardiac electrophysiological targets and drug actions, thereby encouraging future iterations of experimental cardiac electrophysiological discovery, and testing directed towards clinical management. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Christopher L.-H. Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QW, UK
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
30
|
Ahmad FS, Jin Y, Grassam-Rowe A, Zhou Y, Yuan M, Fan X, Zhou R, Mu-u-min R, O'Shea C, Ibrahim AM, Hyder W, Aguib Y, Yacoub M, Pavlovic D, Zhang Y, Tan X, Lei M, Terrar DA. Generation of cardiomyocytes from human-induced pluripotent stem cells resembling atrial cells with ability to respond to adrenoceptor agonists. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220312. [PMID: 37122218 PMCID: PMC10150206 DOI: 10.1098/rstb.2022.0312] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/07/2022] [Indexed: 05/02/2023] Open
Abstract
Atrial fibrillation (AF) is the most common chronic arrhythmia presenting a heavy disease burden. We report a new approach for generating cardiomyocytes (CMs) resembling atrial cells from human-induced pluripotent stem cells (hiPSCs) using a combination of Gremlin 2 and retinoic acid treatment. More than 40% of myocytes showed rod-shaped morphology, expression of CM proteins (including ryanodine receptor 2, α-actinin-2 and F-actin) and striated appearance, all of which were broadly similar to the characteristics of adult atrial myocytes (AMs). Isolated myocytes were electrically quiescent until stimulated to fire action potentials with an AM profile and an amplitude of approximately 100 mV, arising from a resting potential of approximately -70 mV. Single-cell RNA sequence analysis showed a high level of expression of several atrial-specific transcripts including NPPA, MYL7, HOXA3, SLN, KCNJ4, KCNJ5 and KCNA5. Amplitudes of calcium transients recorded from spontaneously beating cultures were increased by the stimulation of α-adrenoceptors (activated by phenylephrine and blocked by prazosin) or β-adrenoceptors (activated by isoproterenol and blocked by CGP20712A). Our new approach provides human AMs with mature characteristics from hiPSCs which will facilitate drug discovery by enabling the study of human atrial cell signalling pathways and AF. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Faizzan S. Ahmad
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
- Cure8bio, Inc, 395 Fulton Street, Westbury, NY 11590, USA
| | - Yongcheng Jin
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | | | - Yafei Zhou
- Key Laboratory of Medical Electrophysiology of the Ministry of Education and Institute of Cardiovascular Research, Southwest Medical University, Luzhou 6400, People's Republic of China
- Shaanxi Institute for Pediatric Diseases, Department of Cardiology, Xi'an Children's Hospital, Xi'an 710003, People's Republic of China
| | - Meng Yuan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education and Institute of Cardiovascular Research, Southwest Medical University, Luzhou 6400, People's Republic of China
| | - Rui Zhou
- Key Laboratory of Medical Electrophysiology of the Ministry of Education and Institute of Cardiovascular Research, Southwest Medical University, Luzhou 6400, People's Republic of China
| | - Razik Mu-u-min
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, College of Medicine and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ayman M. Ibrahim
- Aswan Heart Centre, Aswan 1242770, Egypt
- Department of Zoology, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Wajiha Hyder
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Yasmine Aguib
- Aswan Heart Centre, Aswan 1242770, Egypt
- National Heart and Lung Institute, Heart Science Centre, Imperial College London, Middlesex SW3 6LY, UK
| | - Magdi Yacoub
- Aswan Heart Centre, Aswan 1242770, Egypt
- National Heart and Lung Institute, Heart Science Centre, Imperial College London, Middlesex SW3 6LY, UK
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, College of Medicine and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Yanmin Zhang
- Shaanxi Institute for Pediatric Diseases, Department of Cardiology, Xi'an Children's Hospital, Xi'an 710003, People's Republic of China
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education and Institute of Cardiovascular Research, Southwest Medical University, Luzhou 6400, People's Republic of China
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Derek A. Terrar
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
31
|
Al-Owais MM, Hettiarachchi NT, Dallas ML, Scragg JL, Lippiat JD, Holden AV, Steele DS, Peers C. Inhibition of the voltage-gated potassium channel Kv1.5 by hydrogen sulfide attenuates remodeling through S-nitrosylation-mediated signaling. Commun Biol 2023; 6:651. [PMID: 37336943 PMCID: PMC10279668 DOI: 10.1038/s42003-023-05016-5] [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: 10/11/2022] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
The voltage-gated K+ channel plays a key role in atrial excitability, conducting the ultra-rapid rectifier K+ current (IKur) and contributing to the repolarization of the atrial action potential. In this study, we examine its regulation by hydrogen sulfide (H2S) in HL-1 cardiomyocytes and in HEK293 cells expressing human Kv1.5. Pacing induced remodeling resulted in shorting action potential duration, enhanced both Kv1.5 channel and H2S producing enzymes protein expression in HL-1 cardiomyocytes. H2S supplementation reduced these remodeling changes and restored action potential duration through inhibition of Kv1.5 channel. H2S also inhibited recombinant hKv1.5, lead to nitric oxide (NO) mediated S-nitrosylation and activated endothelial nitric oxide synthase (eNOS) by increased phosphorylation of Ser1177, prevention of NO formation precluded these effects. Regulation of Ikur by H2S has important cardiovascular implications and represents a novel and potential therapeutic target.
Collapse
Affiliation(s)
- Moza M Al-Owais
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Mark L Dallas
- Reading School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Jonathan D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Arun V Holden
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
32
|
Liu T, Li T, Xu D, Wang Y, Zhou Y, Wan J, Huang CLH, Tan X. Small-conductance calcium-activated potassium channels in the heart: expression, regulation and pathological implications. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220171. [PMID: 37122223 PMCID: PMC10150224 DOI: 10.1098/rstb.2022.0171] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/25/2022] [Indexed: 05/02/2023] Open
Abstract
Ca2+-activated K+ channels are critical to cellular Ca2+ homeostasis and excitability; they couple intracellular Ca2+ and membrane voltage change. Of these, the small, 4-14 pS, conductance SK channels include three, KCNN1-3 encoded, SK1/KCa2.1, SK2/KCa2.2 and SK3/KCa2.3, channel subtypes with characteristic, EC50 ∼ 10 nM, 40 pM, 1 nM, apamin sensitivities. All SK channels, particularly SK2 channels, are expressed in atrial, ventricular and conducting system cardiomyocytes. Pharmacological and genetic modification results have suggested that SK channel block or knockout prolonged action potential durations (APDs) and effective refractory periods (ERPs) particularly in atrial, but also in ventricular, and sinoatrial, atrioventricular node and Purkinje myocytes, correspondingly affect arrhythmic tendency. Additionally, mitochondrial SK channels may decrease mitochondrial Ca2+ overload and reactive oxygen species generation. SK channels show low voltage but marked Ca2+ dependences (EC50 ∼ 300-500 nM) reflecting their α-subunit calmodulin (CaM) binding domains, through which they may be activated by voltage-gated or ryanodine-receptor Ca2+ channel activity. SK function also depends upon complex trafficking and expression processes and associations with other ion channels or subunits from different SK subtypes. Atrial and ventricular clinical arrhythmogenesis may follow both increased or decreased SK expression through decreased or increased APD correspondingly accelerating and stabilizing re-entrant rotors or increasing incidences of triggered activity. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Ting Liu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Dandi Xu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yan Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yafei Zhou
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Juyi Wan
- Department of Cardiovascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Christopher L.-H. Huang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
33
|
Vera OD, Wulff H, Braun AP. Endothelial KCa channels: Novel targets to reduce atherosclerosis-driven vascular dysfunction. Front Pharmacol 2023; 14:1151244. [PMID: 37063294 PMCID: PMC10102451 DOI: 10.3389/fphar.2023.1151244] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
Elevated levels of cholesterol in the blood can induce endothelial dysfunction, a condition characterized by impaired nitric oxide production and decreased vasodilatory capacity. Endothelial dysfunction can promote vascular disease, such as atherosclerosis, where macrophages accumulate in the vascular intima and fatty plaques form that impair normal blood flow in conduit arteries. Current pharmacological strategies to treat atherosclerosis mostly focus on lipid lowering to prevent high levels of plasma cholesterol that induce endothelial dysfunction and atherosclerosis. While this approach is effective for most patients with atherosclerosis, for some, lipid lowering is not enough to reduce their cardiovascular risk factors associated with atherosclerosis (e.g., hypertension, cardiac dysfunction, stroke, etc.). For such patients, additional strategies targeted at reducing endothelial dysfunction may be beneficial. One novel strategy to restore endothelial function and mitigate atherosclerosis risk is to enhance the activity of Ca2+-activated K+ (KCa) channels in the endothelium with positive gating modulator drugs. Here, we review the mechanism of action of these small molecules and discuss their ability to improve endothelial function. We then explore how this strategy could mitigate endothelial dysfunction in the context of atherosclerosis by examining how KCa modulators can improve cardiovascular function in other settings, such as aging and type 2 diabetes. Finally, we consider questions that will need to be addressed to determine whether KCa channel activation could be used as a long-term add-on to lipid lowering to augment atherosclerosis treatment, particularly in patients where lipid-lowering is not adequate to improve their cardiovascular health.
Collapse
Affiliation(s)
- O. Daniel Vera
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, United States
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- *Correspondence: Andrew P. Braun,
| |
Collapse
|
34
|
Soda T, Brunetti V, Berra-Romani R, Moccia F. The Emerging Role of N-Methyl-D-Aspartate (NMDA) Receptors in the Cardiovascular System: Physiological Implications, Pathological Consequences, and Therapeutic Perspectives. Int J Mol Sci 2023; 24:ijms24043914. [PMID: 36835323 PMCID: PMC9965111 DOI: 10.3390/ijms24043914] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ligand-gated ion channels that are activated by the neurotransmitter glutamate, mediate the slow component of excitatory neurotransmission in the central nervous system (CNS), and induce long-term changes in synaptic plasticity. NMDARs are non-selective cation channels that allow the influx of extracellular Na+ and Ca2+ and control cellular activity via both membrane depolarization and an increase in intracellular Ca2+ concentration. The distribution, structure, and role of neuronal NMDARs have been extensively investigated and it is now known that they also regulate crucial functions in the non-neuronal cellular component of the CNS, i.e., astrocytes and cerebrovascular endothelial cells. In addition, NMDARs are expressed in multiple peripheral organs, including heart and systemic and pulmonary circulations. Herein, we survey the most recent information available regarding the distribution and function of NMDARs within the cardiovascular system. We describe the involvement of NMDARs in the modulation of heart rate and cardiac rhythm, in the regulation of arterial blood pressure, in the regulation of cerebral blood flow, and in the blood-brain barrier (BBB) permeability. In parallel, we describe how enhanced NMDAR activity could promote ventricular arrhythmias, heart failure, pulmonary artery hypertension (PAH), and BBB dysfunction. Targeting NMDARs could represent an unexpected pharmacological strategy to reduce the growing burden of several life-threatening cardiovascular disorders.
Collapse
Affiliation(s)
- Teresa Soda
- Department of Health Sciences, University of Magna Graecia, 88100 Catanzaro, Italy
| | - Valentina Brunetti
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla 72410, Mexico
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy
- Correspondence: ; Tel.: +39-0382-987613
| |
Collapse
|
35
|
Wang HF, Wang YX, Zhou YP, Wei YP, Yan Y, Zhang ZJ, Jing ZC. Protein O-GlcNAcylation in cardiovascular diseases. Acta Pharmacol Sin 2023; 44:8-18. [PMID: 35817809 PMCID: PMC9813366 DOI: 10.1038/s41401-022-00934-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/30/2022] [Indexed: 01/18/2023]
Abstract
O-GlcNAcylation is a post-translational modification of protein in response to genetic variations or environmental factors, which is controlled by two highly conserved enzymes, i.e. O-GlcNAc transferase (OGT) and protein O-GlcNAcase (OGA). Protein O-GlcNAcylation mainly occurs in the cytoplasm, nucleus, and mitochondrion, and it is ubiquitously implicated in the development of cardiovascular disease (CVD). Alterations of O-GlcNAcylation could cause massive metabolic imbalance and affect cardiovascular function, but the role of O-GlcNAcylation in CVD remains controversial. That is, acutely increased O-GlcNAcylation is an adaptive heart response, which temporarily protects cardiac function. While it is harmful to cardiomyocytes if O-GlcNAcylation levels remain high in chronic conditions or in the long run. The underlying mechanisms include regulation of transcription, energy metabolism, and other signal transduction reactions induced by O-GlcNAcylation. In this review, we will focus on the interactions between protein O-GlcNAcylation and CVD, and discuss the potential molecular mechanisms that may be able to pave a new avenue for the treatment of cardiovascular events.
Collapse
Affiliation(s)
- Hui-Fang Wang
- Department of Medical Laboratory, Weifang Medical University, Weifang, 261053, China
| | - Yi-Xuan Wang
- Department of Medical Laboratory, Weifang Medical University, Weifang, 261053, China
| | - Yu-Ping Zhou
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yun-Peng Wei
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yi Yan
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Ze-Jian Zhang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
36
|
Jin X, Amoni M, Gilbert G, Dries E, Doñate Puertas R, Tomar A, Nagaraju CK, Pradhan A, Yule DI, Martens T, Menten R, Vanden Berghe P, Rega F, Sipido K, Roderick HL. InsP 3R-RyR Ca 2+ channel crosstalk facilitates arrhythmias in the failing human ventricle. Basic Res Cardiol 2022; 117:60. [PMID: 36378362 DOI: 10.1007/s00395-022-00967-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/13/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Dysregulated intracellular Ca2+ handling involving altered Ca2+ release from intracellular stores via RyR channels underlies both arrhythmias and reduced function in heart failure (HF). Mechanisms linking RyR dysregulation and disease are not fully established. Studies in animals support a role for InsP3 receptor Ca2+ channels (InsP3R) in pathological alterations in cardiomyocyte Ca2+ handling but whether these findings translate to the divergent physiology of human cardiomyocytes during heart failure is not determined. Using electrophysiological and Ca2+ recordings in human ventricular cardiomyocytes, we uncovered that Ca2+ release via InsP3Rs facilitated Ca2+ release from RyR and induced arrhythmogenic delayed after depolarisations and action potentials. InsP3R-RyR crosstalk was particularly increased in HF at RyR clusters isolated from the T-tubular network. Reduced SERCA activity in HF further facilitated the action of InsP3. Nanoscale imaging revealed co-localisation of InsP3Rs with RyRs in the dyad, which was increased in HF, providing a mechanism for augmented Ca2+ channel crosstalk. Notably, arrhythmogenic activity dependent on InsP3Rs was increased in tissue wedges from failing hearts perfused with AngII to promote InsP3 generation. These data indicate a central role for InsP3R-RyR Ca2+ signalling crosstalk in the pro-arrhythmic action of GPCR agonists elevated in HF and the potential for their therapeutic targeting.
Collapse
Affiliation(s)
- Xin Jin
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Matthew Amoni
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Guillaume Gilbert
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Eef Dries
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Rosa Doñate Puertas
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Ashutosh Tomar
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Chandan K Nagaraju
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Ankit Pradhan
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, Medical Center School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 711, Rochester, NY, 14642, USA
| | - Tobie Martens
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000, Leuven, Belgium.,Cell and Tissue Imaging Cluster (CIC), KU Leuven, 3000, Leuven, Belgium
| | - Roxane Menten
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000, Leuven, Belgium.,Cell and Tissue Imaging Cluster (CIC), KU Leuven, 3000, Leuven, Belgium
| | - Filip Rega
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.,Department of Cardiology and Department of Cardiac Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Karin Sipido
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - H Llewelyn Roderick
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
37
|
Campos-Ríos A, Rueda-Ruzafa L, Lamas JA. The Relevance of GIRK Channels in Heart Function. MEMBRANES 2022; 12:1119. [PMID: 36363674 PMCID: PMC9698958 DOI: 10.3390/membranes12111119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 06/16/2023]
Abstract
Among the large number of potassium-channel families implicated in the control of neuronal excitability, G-protein-gated inwardly rectifying potassium channels (GIRK/Kir3) have been found to be a main factor in heart control. These channels are activated following the modulation of G-protein-coupled receptors and, although they have been implicated in different neurological diseases in both human and animal studies of the central nervous system, the therapeutic potential of different subtypes of these channel families in cardiac conditions has remained untapped. As they have emerged as a promising potential tool to treat a variety of conditions that disrupt neuronal homeostasis, many studies have started to focus on these channels as mediators of cardiac dynamics, thus leading to research into their implication in cardiovascular conditions. Our aim is to review the latest advances in GIRK modulation in the heart and their role in the cardiovascular system.
Collapse
Affiliation(s)
- Ana Campos-Ríos
- CINBIO, Laboratory of Neuroscience, University of Vigo, 36310 Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), 15706 Vigo, Spain
| | - Lola Rueda-Ruzafa
- Department of Nursing Science, Physiotherapy and Medicine, Faculty of Health Sciences, University of Almeria, 04120 Almeria, Spain
| | - José Antonio Lamas
- CINBIO, Laboratory of Neuroscience, University of Vigo, 36310 Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), 15706 Vigo, Spain
| |
Collapse
|
38
|
Mulla W, Murninkas M, Levi O, Etzion Y. Incorrectly corrected? QT interval analysis in rats and mice. Front Physiol 2022; 13:1002203. [PMID: 36304573 PMCID: PMC9595597 DOI: 10.3389/fphys.2022.1002203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
QT interval, a surrogate measure for ventricular action potential duration (APD) in the surface ECG, is widely used to identify cardiac abnormalities and drug safety. In humans, cardiac APD and QT interval are prominently affected by heart rate (HR), leading to widely accepted formulas to correct the QT interval for HR changes (QT corrected - QTc). While QTc is widely used in the clinic, the proper way to correct the QT interval in small mammals such as rats and mice is not clear. Over the years, empiric correction formulas were developed for rats and mice, which are widely used in the literature. Recent experimental findings obtained from pharmacological and direct pacing experiments in unanesthetized rodents show that the rate-adaptation properties are markedly different from those in humans and the use of existing QTc formulae can lead to major errors in data interpretation. In the present review, these experimental findings are summarized and discussed.
Collapse
Affiliation(s)
- Wesam Mulla
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Wesam Mulla, ; Yoram Etzion,
| | - Michael Murninkas
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Or Levi
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Wesam Mulla, ; Yoram Etzion,
| |
Collapse
|
39
|
Integrated Analysis of the microRNA–mRNA Network Predicts Potential Regulators of Atrial Fibrillation in Humans. Cells 2022; 11:cells11172629. [PMID: 36078037 PMCID: PMC9454849 DOI: 10.3390/cells11172629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Atrial fibrillation (AF) is a form of sustained cardiac arrhythmia and microRNAs (miRs) play crucial roles in the pathophysiology of AF. To identify novel miR–mRNA pairs, we performed RNA-seq from atrial biopsies of persistent AF patients and non-AF patients with normal sinus rhythm (SR). Differentially expressed miRs (11 down and 9 up) and mRNAs (95 up and 82 down) were identified and hierarchically clustered in a heat map. Subsequently, GO, KEGG, and GSEA analyses were run to identify deregulated pathways. Then, miR targets were predicted in the miRDB database, and a regulatory network of negatively correlated miR–mRNA pairs was constructed using Cytoscape. To select potential candidate genes from GSEA analysis, the top-50 enriched genes in GSEA were overlaid with predicted targets of differentially deregulated miRs. Further, the protein–protein interaction (PPI) network of enriched genes in GSEA was constructed, and subsequently, GO and canonical pathway analyses were run for genes in the PPI network. Our analyses showed that TNF-α, p53, EMT, and SYDECAN1 signaling were among the highly affected pathways in AF samples. SDC-1 (SYNDECAN-1) was the top-enriched gene in p53, EMT, and SYDECAN1 signaling. Consistently, SDC-1 mRNA and protein levels were significantly higher in atrial samples of AF patients. Among negatively correlated miRs, miR-302b-3p was experimentally validated to suppress SDC-1 transcript levels. Overall, our results suggested that the miR-302b-3p/SDC-1 axis may be involved in the pathogenesis of AF.
Collapse
|
40
|
Diversification of Potassium Currents in Excitable Cells via Kvβ Proteins. Cells 2022; 11:cells11142230. [PMID: 35883673 PMCID: PMC9317154 DOI: 10.3390/cells11142230] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 12/10/2022] Open
Abstract
Excitable cells of the nervous and cardiovascular systems depend on an assortment of plasmalemmal potassium channels to control diverse cellular functions. Voltage-gated potassium (Kv) channels are central to the feedback control of membrane excitability in these processes due to their activation by depolarized membrane potentials permitting K+ efflux. Accordingly, Kv currents are differentially controlled not only by numerous cellular signaling paradigms that influence channel abundance and shape voltage sensitivity, but also by heteromeric configurations of channel complexes. In this context, we discuss the current knowledge related to how intracellular Kvβ proteins interacting with pore complexes of Shaker-related Kv1 channels may establish a modifiable link between excitability and metabolic state. Past studies in heterologous systems have indicated roles for Kvβ proteins in regulating channel stability, trafficking, subcellular targeting, and gating. More recent works identifying potential in vivo physiologic roles are considered in light of these earlier studies and key gaps in knowledge to be addressed by future research are described.
Collapse
|
41
|
Carrasquel-Ursulaez W, Segura I, Díaz-Franulic I, Márquez-Miranda V, Echeverría F, Lorenzo-Ceballos Y, Espinoza N, Rojas M, Garate JA, Perozo E, Alvarez O, Gonzalez-Nilo FD, Latorre R. Mechanism of voltage sensing in Ca 2+- and voltage-activated K + (BK) channels. Proc Natl Acad Sci U S A 2022; 119:e2204620119. [PMID: 35704760 PMCID: PMC9231616 DOI: 10.1073/pnas.2204620119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/10/2022] [Indexed: 11/18/2022] Open
Abstract
In neurosecretion, allosteric communication between voltage sensors and Ca2+ binding in BK channels is crucially involved in damping excitatory stimuli. Nevertheless, the voltage-sensing mechanism of BK channels is still under debate. Here, based on gating current measurements, we demonstrate that two arginines in the transmembrane segment S4 (R210 and R213) function as the BK gating charges. Significantly, the energy landscape of the gating particles is electrostatically tuned by a network of salt bridges contained in the voltage sensor domain (VSD). Molecular dynamics simulations and proton transport experiments in the hyperpolarization-activated R210H mutant suggest that the electric field drops off within a narrow septum whose boundaries are defined by the gating charges. Unlike Kv channels, the charge movement in BK appears to be limited to a small displacement of the guanidinium moieties of R210 and R213, without significant movement of the S4.
Collapse
Affiliation(s)
- Willy Carrasquel-Ursulaez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Ignacio Segura
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Ignacio Díaz-Franulic
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Valeria Márquez-Miranda
- Centro de Nanotecnología Aplicada, Facultad de Ciencias, Universidad Mayor, Santiago, Chile, 8580745
| | - Felipe Echeverría
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Yenisleidy Lorenzo-Ceballos
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Nicolás Espinoza
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Maximiliano Rojas
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Jose Antonio Garate
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Millennium Nucleus in NanoBioPhysics, Valparaíso 2340000, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7810000, Chile
| | - Eduardo Perozo
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
- Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637
| | - Osvaldo Alvarez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7810000, Chile
| | - Fernando D. Gonzalez-Nilo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| |
Collapse
|
42
|
Lopez-Medina AI, Chahal CAA, Luzum JA. The genetics of drug-induced QT prolongation: evaluating the evidence for pharmacodynamic variants. Pharmacogenomics 2022; 23:543-557. [PMID: 35698903 DOI: 10.2217/pgs-2022-0027] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Drug-induced long QT syndrome (diLQTS) is an adverse effect of many commonly prescribed drugs, and it can increase the risk for lethal ventricular arrhythmias. Genetic variants in pharmacodynamic genes have been associated with diLQTS, but the strength of the evidence for each of those variants has not yet been evaluated. Therefore, the purpose of this review was to evaluate the strength of the evidence for pharmacodynamic genetic variants associated with diLQTS using a novel, semiquantitative scoring system modified from the approach used for congenital LQTS. KCNE1-D85N and KCNE2-T8A had definitive and strong evidence for diLQTS, respectively. The high level of evidence for these variants supports current consideration as risk factors for patients that will be prescribed a QT-prolonging drug.
Collapse
Affiliation(s)
- Ana I Lopez-Medina
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Choudhary Anwar A Chahal
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA.,Barts Heart Centre, St. Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK.,WellSpan Health, Lancaster, PA 17607, USA
| | - Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| |
Collapse
|
43
|
Krahn AD, Laksman Z, Sy RW, Postema PG, Ackerman MJ, Wilde AAM, Han HC. Congenital Long QT Syndrome. JACC Clin Electrophysiol 2022; 8:687-706. [PMID: 35589186 DOI: 10.1016/j.jacep.2022.02.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022]
Abstract
Congenital long QT syndrome (LQTS) encompasses a group of heritable conditions that are associated with cardiac repolarization dysfunction. Since its initial description in 1957, our understanding of LQTS has increased dramatically. The prevalence of LQTS is estimated to be ∼1:2,000, with a slight female predominance. The diagnosis of LQTS is based on clinical, electrocardiogram, and genetic factors. Risk stratification of patients with LQTS aims to identify those who are at increased risk of cardiac arrest or sudden cardiac death. Factors including age, sex, QTc interval, and genetic background all contribute to current risk stratification paradigms. The management of LQTS involves conservative measures such as the avoidance of QT-prolonging drugs, pharmacologic measures with nonselective β-blockers, and interventional approaches such as device therapy or left cardiac sympathetic denervation. In general, most forms of exercise are considered safe in adequately treated patients, and implantable cardioverter-defibrillator therapy is reserved for those at the highest risk. This review summarizes our current understanding of LQTS and provides clinicians with a practical approach to diagnosis and management.
Collapse
Affiliation(s)
- Andrew D Krahn
- Center for Cardiovascular Innovation, Heart Rhythm Services, Division of Cardiology, University of British Columbia, Vancouver, BC, Canada.
| | - Zachary Laksman
- Center for Cardiovascular Innovation, Heart Rhythm Services, Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Raymond W Sy
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Pieter G Postema
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Michael J Ackerman
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, Minnesota, USA; Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota, USA; Departments of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota, USA
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart), Academic University Medical Center, Amsterdam, the Netherlands
| | - Hui-Chen Han
- Center for Cardiovascular Innovation, Heart Rhythm Services, Division of Cardiology, University of British Columbia, Vancouver, BC, Canada; Victorian Heart Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
44
|
Sanghvi S, Szteyn K, Ponnalagu D, Sridharan D, Lam A, Hansra I, Chaudhury A, Majumdar U, Kohut AR, Gururaja Rao S, Khan M, Garg V, Singh H. Inhibition of BK Ca channels protects neonatal hearts against myocardial ischemia and reperfusion injury. Cell Death Dis 2022; 8:175. [PMID: 35393410 PMCID: PMC8989942 DOI: 10.1038/s41420-022-00980-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022]
Abstract
BKCa channels are large-conductance calcium and voltage-activated potassium channels that are heterogeneously expressed in a wide array of cells. Activation of BKCa channels present in mitochondria of adult ventricular cardiomyocytes is implicated in cardioprotection against ischemia-reperfusion (IR) injury. However, the BKCa channel’s activity has never been detected in the plasma membrane of adult ventricular cardiomyocytes. In this study, we report the presence of the BKCa channel in the plasma membrane and mitochondria of neonatal murine and rodent cardiomyocytes, which protects the heart on inhibition but not activation. Furthermore, K+ currents measured in neonatal cardiomyocyte (NCM) was sensitive to iberiotoxin (IbTx), suggesting the presence of BKCa channels in the plasma membrane. Neonatal hearts subjected to IR when post-conditioned with NS1619 during reoxygenation increased the myocardial infarction whereas IbTx reduced the infarct size. In agreement, isolated NCM also presented increased apoptosis on treatment with NS1619 during hypoxia and reoxygenation, whereas IbTx reduced TUNEL-positive cells. In NCMs, activation of BKCa channels increased the intracellular reactive oxygen species post HR injury. Electrophysiological characterization of NCMs indicated that NS1619 increased the beat period, field, and action potential duration, and decreased the conduction velocity and spike amplitude. In contrast, IbTx had no impact on the electrophysiological properties of NCMs. Taken together, our data established that inhibition of plasma membrane BKCa channels in the NCM protects neonatal heart/cardiomyocytes from IR injury. Furthermore, the functional disparity observed towards the cardioprotective activity of BKCa channels in adults compared to neonatal heart could be attributed to their differential localization.
Collapse
Affiliation(s)
- Shridhar Sanghvi
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
| | - Kalina Szteyn
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Devasena Ponnalagu
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Divya Sridharan
- Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Alexander Lam
- Department of Internal Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Inderjot Hansra
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ankur Chaudhury
- Department of Internal Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Uddalak Majumdar
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Andrew R Kohut
- Department of Internal Medicine, Drexel University College of Medicine, Philadelphia, PA, USA.,Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shubha Gururaja Rao
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, Ada, OH, USA
| | - Mahmood Khan
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Vidu Garg
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
45
|
Research Progress on Natural Products’ Therapeutic Effects on Atrial Fibrillation by Regulating Ion Channels. Cardiovasc Ther 2022; 2022:4559809. [PMID: 35387267 PMCID: PMC8964196 DOI: 10.1155/2022/4559809] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 11/18/2022] Open
Abstract
Antiarrhythmic drugs (AADs) have a therapeutic effect on atrial fibrillation (AF) by regulating the function of ion channels. However, several adverse effects and high recurrence rates after drug withdrawal seriously affect patients’ medication compliance and clinical prognosis. Thus, safer and more effective drugs are urgently needed. Active components extracted from natural products are potential choices for AF therapy. Natural products like Panax notoginseng (Burk.) F.H. Chen, Sophora flavescens Ait., Stephania tetrandra S. Moore., Pueraria lobata (Willd.) Ohwi var. thomsonii (Benth.) Vaniot der Maesen., and Coptis chinensis Franch. have a long history in the treatment of arrhythmia, myocardial infarction, stroke, and heart failure in China. Based on the classification of chemical structures, this article discussed the natural product components’ therapeutic effects on atrial fibrillation by regulating ion channels, connexins, and expression of related genes, in order to provide a reference for development of therapeutic drugs for atrial fibrillation.
Collapse
|
46
|
Yang B, Jiang Q, He S, Li T, Ou X, Chen T, Fan X, Jiang F, Zeng X, Huang CLH, Lei M, Tan X. Ventricular SK2 upregulation following angiotensin II challenge: Modulation by p21-activated kinase-1. J Mol Cell Cardiol 2022; 164:110-125. [PMID: 34774547 DOI: 10.1016/j.yjmcc.2021.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/15/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022]
Abstract
Effects of hypertrophic challenge on small-conductance, Ca2+-activated K+(SK2) channel expression were explored in intact murine hearts, isolated ventricular myocytes and neonatal rat cardiomyocytes (NRCMs). An established experimental platform applied angiotensin II (Ang II) challenge in the presence and absence of reduced p21-activated kinase (PAK1) (PAK1cko vs. PAK1f/f, or shRNA-PAK1 interference) expression. SK2 current contributions were detected through their sensitivity to apamin block. Ang II treatment increased such SK2 contributions to optically mapped action potential durations (APD80) and their heterogeneity, and to patch-clamp currents. Such changes were accentuated in PAK1cko compared to PAK1f/f, intact hearts and isolated cardiomyocytes. They paralleled increased histological and echocardiographic hypertrophic indices, reduced cardiac contractility, and increased SK2 protein expression, changes similarly greater with PAK1cko than PAK1f/f. In NRCMs, Ang II challenge replicated such increases in apamin-sensitive SK patch clamp currents as well as in real-time PCR and western blot measures of SK2 mRNA and protein expression and cell hypertrophy. Furthermore, the latter were enhanced by shRNA-PAK1 interference and mitigated by the PAK1 agonist FTY720. Increased CaMKII and CREB phosphorylation accompanied these effects. These were rescued by both FTY720 as well as the CaMKII inhibitor KN93, but not its inactive analogue KN92. Such CREB then specifically bound to the KCNN2 promoter sequence in luciferase assays. These findings associate Ang II induced hypertrophy with increased SK2 expression brought about by a CaMKII/CREB signaling convergent with the PAK1 pathway thence upregulating the KCNN2 promoter activity. SK2 may then influence cardiac electrophysiology under conditions of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Binbin Yang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Oral & Maxillofacial Reconstruction and Regeneration Laboratory, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qin Jiang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shicheng He
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tangting Chen
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Feng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaorong Zeng
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Christopher L-H Huang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
47
|
MicroRNA-365 regulates human cardiac action potential duration. Nat Commun 2022; 13:220. [PMID: 35017523 PMCID: PMC8752767 DOI: 10.1038/s41467-021-27856-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 12/16/2021] [Indexed: 12/21/2022] Open
Abstract
Abnormalities of ventricular action potential cause malignant cardiac arrhythmias and sudden cardiac death. Here, we aim to identify microRNAs that regulate the human cardiac action potential and ask whether their manipulation allows for therapeutic modulation of action potential abnormalities. Quantitative analysis of the microRNA targetomes in human cardiac myocytes identifies miR-365 as a primary microRNA to regulate repolarizing ion channels. Action potential recordings in patient-specific induced pluripotent stem cell-derived cardiac myocytes show that elevation of miR-365 significantly prolongs action potential duration in myocytes derived from a Short-QT syndrome patient, whereas specific inhibition of miR-365 normalizes pathologically prolonged action potential in Long-QT syndrome myocytes. Transcriptome analyses in these cells at bulk and single-cell level corroborate the key cardiac repolarizing channels as direct targets of miR-365, together with functionally synergistic regulation of additional action potential-regulating genes by this microRNA. Whole-cell patch-clamp experiments confirm miR-365-dependent regulation of repolarizing ionic current Iks. Finally, refractory period measurements in human myocardial slices substantiate the regulatory effect of miR-365 on action potential in adult human myocardial tissue. Our results delineate miR-365 to regulate human cardiac action potential duration by targeting key factors of cardiac repolarization.
Collapse
|
48
|
Bae H, Kim T, Lim I. Carbon monoxide activation of delayed rectifier potassium currents of human cardiac fibroblasts through diverse pathways. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:25-36. [PMID: 34965993 PMCID: PMC8723981 DOI: 10.4196/kjpp.2022.26.1.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 06/14/2023]
Abstract
To identify the effect and mechanism of carbon monoxide (CO) on delayed rectifier K+ currents (IK) of human cardiac fibroblasts (HCFs), we used the wholecell mode patch-clamp technique. Application of CO delivered by carbon monoxidereleasing molecule-3 (CORM3) increased the amplitude of outward K+ currents, and diphenyl phosphine oxide-1 (a specific IK blocker) inhibited the currents. CORM3- induced augmentation was blocked by pretreatment with nitric oxide synthase blockers (L-NG-monomethyl arginine citrate and L-NG-nitro arginine methyl ester). Pretreatment with KT5823 (a protein kinas G blocker), 1H-[1,-2,-4] oxadiazolo-[4,-3-a] quinoxalin-1-on (ODQ, a soluble guanylate cyclase blocker), KT5720 (a protein kinase A blocker), and SQ22536 (an adenylate cyclase blocker) blocked the CORM3 stimulating effect on IK. In addition, pretreatment with SB239063 (a p38 mitogen-activated protein kinase [MAPK] blocker) and PD98059 (a p44/42 MAPK blocker) also blocked the CORM3's effect on the currents. When testing the involvement of S-nitrosylation, pretreatment of N-ethylmaleimide (a thiol-alkylating reagent) blocked CO-induced IK activation and DL-dithiothreitol (a reducing agent) reversed this effect. Pretreatment with 5,10,15,20-tetrakis(1-methylpyridinium-4-yl)-21H,23H porphyrin manganese (III) pentachloride and manganese (III) tetrakis (4-benzoic acid) porphyrin chloride (superoxide dismutase mimetics), diphenyleneiodonium chloride (an NADPH oxidase blocker), or allopurinol (a xanthine oxidase blocker) also inhibited CO-induced IK activation. These results suggest that CO enhances IK in HCFs through the nitric oxide, phosphorylation by protein kinase G, protein kinase A, and MAPK, S-nitrosylation and reduction/oxidation (redox) signaling pathways.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, Seoul 06973, Korea
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
49
|
Stukalin I, Olaiya OR, Naik V, Wiebe E, Kekewich M, Kelly M, Wilding L, Halko R, Oczkowski S. Medications and dosages used in medical assistance in dying: a cross-sectional study. CMAJ Open 2022; 10:E19-E26. [PMID: 35042691 PMCID: PMC8920593 DOI: 10.9778/cmajo.20200268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND There is little evidence describing the technical aspects of medical assistance in dying (MAiD) in Canada, such as medications, dosages and complications. Our objective was to describe clinical practice in providing MAiD in Ontario and Vancouver, Canada, and explore relations between medications used, time until death and complications. METHODS We conducted a retrospective cohort study of a sample of adult (age ≥ 18 yr) patients who received MAiD in Ontario between 2016 and 2018, and patients who received MAiD in 1 of 3 Canadian academic hospitals (in Hamilton and Ottawa, Ontario, and Vancouver, British Colombia) between 2019 and 2020. We used de-identified data for 2016-2018 from the Office of the Chief Coroner for Ontario MAiD Database and chart review data for 2019-2020 from the 3 centres. We used multivariable parametric survival analysis to identify relations between medications, dosages and time from procedure start until death. RESULTS The sample included 3557 patients (1786 men [50.2%] and 1770 women [49.8%] with a mean age of 74 [standard deviation 13] yr). The majority of patients (2519 [70.8%]) had a diagnosis of cancer. The medications most often used were propofol (3504 cases [98.5%]), midazolam (3251 [91.4%]) and rocuronium (3228 [90.8%]). The median time from the first injection until death was 9 (interquartile range 6) minutes. Standard-dose lidocaine (40-60 mg) and high-dose propofol (> 1000 mg) were associated with prolonged time until death (prolonged by a median of 1 min and 3 min, respectively). Complications occurred in 41 cases (1.2%), mostly related to venous access or need for administration of a second medication. INTERPRETATION In a large sample of patients who died with medical assistance, certain medications were associated with small differences in time from injection to death, and complications were rare. More research is needed to identify the medication protocols that predict outcomes consistent with patient and family expectations for a medically assisted death.
Collapse
Affiliation(s)
- Igor Stukalin
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Oluwatobi R Olaiya
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Viren Naik
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Ellen Wiebe
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Mike Kekewich
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Michaela Kelly
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Laura Wilding
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Roxanne Halko
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont
| | - Simon Oczkowski
- Department of Medicine (Stukalin), University of Calgary, Calgary, Alta.; Division of Plastic Surgery (Olaiya), Department of Surgery, McMaster University, Hamilton, Ont.; The Ottawa Hospital (Naik, Kekewich, Wilding); Department of Anesthesiology and Pain Medicine (Naik), University of Ottawa, Ottawa, Ont.; Department of Family Practice (Wiebe), University of British Columbia, Vancouver, BC; London School of Hygiene and Tropical Medicine (Kelly), University of London, London, UK; Office of the Chief Coroner for Ontario (Halko), Toronto, Ont.; Departments of Medicine (Oczkowski), and Health Research Methods, Evidence, and Impact (Olaiya, Oczkowski), McMaster University, Hamilton, Ont.
| |
Collapse
|
50
|
González-Sanabria N, Echeverría F, Segura I, Alvarado-Sánchez R, Latorre R. BK in Double-Membrane Organelles: A Biophysical, Pharmacological, and Functional Survey. Front Physiol 2021; 12:761474. [PMID: 34764886 PMCID: PMC8577798 DOI: 10.3389/fphys.2021.761474] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/29/2021] [Indexed: 12/04/2022] Open
Abstract
In the 1970s, calcium-activated potassium currents were recorded for the first time. In 10years, this Ca2+-activated potassium channel was identified in rat skeletal muscle, chromaffin cells and characterized in skeletal muscle membranes reconstituted in lipid bilayers. This calcium- and voltage-activated potassium channel, dubbed BK for “Big K” due to its large ionic conductance between 130 and 300 pS in symmetric K+. The BK channel is a tetramer where the pore-forming α subunit contains seven transmembrane segments. It has a modular architecture containing a pore domain with a highly potassium-selective filter, a voltage-sensor domain and two intracellular Ca2+ binding sites in the C-terminus. BK is found in the plasma membrane of different cell types, the inner mitochondrial membrane (mitoBK) and the nuclear envelope’s outer membrane (nBK). Like BK channels in the plasma membrane (pmBK), the open probability of mitoBK and nBK channels are regulated by Ca2+ and voltage and modulated by auxiliary subunits. BK channels share common pharmacology to toxins such as iberiotoxin, charybdotoxin, paxilline, and agonists of the benzimidazole family. However, the precise role of mitoBK and nBK remains largely unknown. To date, mitoBK has been reported to play a role in protecting the heart from ischemic injury. At the same time, pharmacology suggests that nBK has a role in regulating nuclear Ca2+, membrane potential and expression of eNOS. Here, we will discuss at the biophysical level the properties and differences of mitoBK and nBK compared to those of pmBK and their pharmacology and function.
Collapse
Affiliation(s)
- Naileth González-Sanabria
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Felipe Echeverría
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Ignacio Segura
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Rosangelina Alvarado-Sánchez
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramon Latorre
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|