1
|
Bertomeu JB, Fioravanço LP, Ramis TR, Godinho DB, Nascimento AS, Lima GC, Furian AF, Oliveira MS, Fighera MR, Royes LFF. The Role of Ion-Transporting Proteins on Crosstalk Between the Skeletal Muscle and Central Nervous Systems Elicited by Physical Exercise. Mol Neurobiol 2025; 62:5546-5565. [PMID: 39578339 DOI: 10.1007/s12035-024-04613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024]
Abstract
A paradigm shift in the understanding of bidirectional interactions between peripheral and central nervous systems is essential for development of rehabilitation and preventive interventions based on physical exercise. Although a causal relationship has not been completely established, modulation of voltage-dependent ion channels (Ca2+, Cl-, K+, Na+, lactate-, H+) in skeletal and neuronal cells provides opportunities to maintain force production during exercise and reduce the risk of disease. However, there are caveats to consider when interpreting the effects of physical exercise on this bidirectional axis, since exercise protocol details (e.g., duration and intensity) have variable effects on this crosstalk. Therefore, an integrative perspective of the skeletal muscle and brain's communication pathway is discussed, and the role of physical exercise on such communication highway is explained in this review.
Collapse
Affiliation(s)
- Judit Borràs Bertomeu
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Letícia Paiva Fioravanço
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Thiago Rozales Ramis
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Douglas Buchmann Godinho
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Alexandre Seixas Nascimento
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriel Corrêa Lima
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Ana Flavia Furian
- Graduate Program in Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Mauro Schneider Oliveira
- Graduate Program in Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Laboratory of Experimental and Clinical Neuropsychiatry, Department of Neuropsychiatry, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Luiz Fernando Freire Royes
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Department of Sports Methods and Techniques, Center of Physical Education and , Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
2
|
Wu H, Diao H, Zhang F, Jiang W, Pan T, Bian Y. Organelle interplay in cardiovascular diseases: Mechanisms, pathogenesis, and therapeutic perspectives. Biomed Pharmacother 2025; 185:117978. [PMID: 40073746 DOI: 10.1016/j.biopha.2025.117978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/16/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of rising morbidity and mortality among humans worldwide; however, our approach to the pathogenesis, exploration, and management of CVDs still remains limited. As the heart consists of cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, and several types of cells, different types of dysfunction in the interplay between organelles play an important damaging role, resulting in cardiac pathologies. The interplay between cellular organelles is intricate and vital for maintaining cellular homeostasis, as highlighted by multiple diseases linked to the dysfunction of both organelles. Many studies have revealed the potential mechanisms by which organelles communicate with each other and regulate the pathological processes of CVDs together. However, gaps remain in fully understanding the complexity of these interactions and translating these insights into therapeutic approaches. In this review, we summarized how the interplay between cellular organelles in the cardiomyocytes alters in various heart diseases. We find underexplored areas, such as the crucial signaling pathways governing organelle communication, and discuss their implications for disease future progression. Furthermore, we evaluate emerging potential medicines aimed at restoring organelle interactions. Finally, we propose future directions for researching to advance the development of novel medicines and therapies, addressing current gaps and providing a theoretical basis for improved clinical outcomes in CVDs.
Collapse
Affiliation(s)
- Han Wu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hongtao Diao
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Feng Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Weitao Jiang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tengfei Pan
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yu Bian
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
3
|
Zhu C, Li S, Zhang H. Heart Failure and Arrhythmias: Circadian and Epigenetic Interplay in Myocardial Electrophysiology. Int J Mol Sci 2025; 26:2728. [PMID: 40141370 PMCID: PMC11943068 DOI: 10.3390/ijms26062728] [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: 02/12/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Emerging evidence underscores the impact of circadian rhythms on cardiovascular processes, particularly in conditions such as hypertension, myocardial infarction, and heart failure, where circadian rhythm disruptions are linked to disease progression and adverse clinical outcomes. Circadian clock proteins are intricately linked to myocardial electrophysiological remodeling and epigenetic pathways associated with arrhythmias in heart failure. In the context of heart failure, circadian clock dysregulation leads to electrophysiological remodeling in the cardiomyocytes, which can precipitate life-threatening arrhythmias such as ventricular tachycardia (VT) and ventricular fibrillation (VF). This dysregulation may be influenced by environmental factors, such as diet and exercise, as well as genetic factors. Moreover, epigenetic modifications in heart failure have been implicated in the regulation of genes involved in cardiac hypertrophy, fibrosis, and inflammation. The interplay between circadian clock proteins, myocardial electrophysiological remodeling, and epigenetic pathways in heart failure-related arrhythmias is complex and multifaceted. Further research is needed to elucidate how these processes interact and contribute to the development of arrhythmias in heart failure patients. This review aims to explore the connections between circadian rhythms, myocardial electrophysiology, and arrhythmias related to heart failure, with the goal of identifying potential therapeutic targets and interventions that may counteract the adverse effects of circadian disruptions on cardiovascular health.
Collapse
Affiliation(s)
- Chen Zhu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646099, China; (C.Z.); (S.L.)
| | - Shuang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646099, China; (C.Z.); (S.L.)
| | - Henggui Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646099, China; (C.Z.); (S.L.)
- Beijing Academy of Artificial Intelligence, Beijing 100084, China
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
4
|
Makwana R, Patel R, O'Neill R, Marchi E, Lyon GJ. The Cardiovascular Manifestations and Management Recommendations for Ogden Syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25321331. [PMID: 40236393 PMCID: PMC11996587 DOI: 10.1101/2025.02.11.25321331] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The NatA complex is composed of the NAA10, NAA15, and HYPK subunits. It is primarily responsible for N-terminal acetylation, a critical post-translational modification in eukaryotes. Pathogenic variants within NAA10 cause Ogden Syndrome (OS), which is characterized by varying degrees of intellectual disability, hypotonia, developmental delay, and cardiac abnormalities. Although the cardiac manifestations of the disease have been described extensively in case reports, there has not been a study focusing on the cardiac manifestations and their recommended clinical cardiac management. In this study, we describe the cardiac manifestations of OS in a cohort of 85 probands. We found increased incidence of structural and electrophysiologic abnormalities, with particularly high prevalence of QT interval prolongation. Sub-analysis showed that male probands and those with variants within the NAA15-binding domain had more severe phenotypes than females or those with variants outside of the NAA15-binding domain. Our results suggest that an OS diagnosis should be accompanied by full cardiac workup with emphasis on echocardiogram for structural defects and EKG/Holter monitoring for electrophysiologic abnormalities. Additionally, we strongly recommend that the use of QT-prolonging drugs be followed up with routine electrophysiological monitoring or consultation with a pediatric cardiologist. We hope this study guides clinicians and caregivers treating patients with OS and moves the field toward a standardized diagnostic workup for patients with this condition.
Collapse
|
5
|
Li L, Xie S, Deng W. RNA binding proteins: Mechanistic considerations and perspectives in controlling cardiovascular diseases. Eur J Pharmacol 2025; 987:177101. [PMID: 39613174 DOI: 10.1016/j.ejphar.2024.177101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
Cardiovascular diseases (CVDs) are becoming serious disease that endangering human health due to the increasing morbidity and mortality, and many molecular targets are involved in this complex pathologic process. Recently, RNA-binding proteins (RBPs) have received potential attention as a promising targets for preventing CVDs, including myocardial hypertrophy, dilated cardiomyopathy (DCM), myocardial fibrosis, and pulmonary hypertension (PH). As important regulators of RNA metabolism, RBPs play important roles in all steps of the gene expression cascade,and affect the occurrence and development of various diseases. In this review, we discuss the regulatory role of RBPs on various CVDs at the post transcriptional modification level based on current research. We also highlight the existing and potential RNA-based therapeutics that could impact future CVD treatments.
Collapse
Affiliation(s)
- Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
6
|
Sweat ME, Pu WIT. Genetic and Molecular Underpinnings of Atrial Fibrillation. NPJ CARDIOVASCULAR HEALTH 2024; 1:35. [PMID: 39867228 PMCID: PMC11759492 DOI: 10.1038/s44325-024-00035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/02/2024] [Indexed: 01/28/2025]
Abstract
Atrial fibrillation (AF), the most common sustained arrhythmia, increases stroke and heart failure risks. Here we review genes linked to AF and mechanisms by which they alter AF risk. We highlight gene expression differences between atrial and ventricular cardiomyocytes, regulatory mechanisms responsible for these differences, and their potential contribution to AF. Understanding AF mechanisms through the lens of atrial gene regulation is crucial to improving AF treatment.
Collapse
Affiliation(s)
- Mason E. Sweat
- Department of Cardiology, Boston Children’s
Hospital, Boston, MA 02115, USA
| | - WIlliam T. Pu
- Department of Cardiology, Boston Children’s
Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge,
MA 02138, USA
| |
Collapse
|
7
|
Cheng L, Chen H, Maboh RN, Wang H. Effects of Cacna1d D307G Mutation on Blood Pressure and Kidney Function in Rats with Salt Loading. Kidney Blood Press Res 2024; 50:46-60. [PMID: 39622224 PMCID: PMC11844681 DOI: 10.1159/000542828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/20/2024] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Our recent findings revealed that CACNA1D D307G mutation participates in the early-onset hypertension. METHODS We used the CRISPR/Cas9 technique to generate the Cacna1d D307G mutation rat model and investigated the effects of Cacna1d D307G mutation on blood pressure (BP) and renal function. Rats fed normal-salt diet had normal plasma aldosterone levels but higher plasma ET-1 and mildly elevated systolic BP (SBP) in D307G and G307G rats compared with the wild type (WT) until 24 weeks. Renal function and renal histopathology did not significantly differ among the three groups. RESULTS When fed high-salt diet (HSD), D307G and G307G rats showed more sensitivity to HSD. The results showed a further increase in SBP than in WT rats. Plasma and vascular endothelin-1 (ET-1) level and cortex and renal artery endothelin type A (ETA) receptor protein expression were significantly increased. Enhanced renal injury was also noted as indicated by an increased ratio of kidney weight/body weight, elevated urinary protein and albumin/creatinine ratio, higher kidney injury molecule-1 (KIM-1) levels, advanced fibrosis and apoptosis, and inflammation. Further experiments revealed a reduction in urinary sodium excretion and creatinine clearance. Higher protein expression of renal cortex epithelial sodium channel α subunit (αENaC) was confirmed in D307G and G307G rats fed HSD. However, a selective ETA receptor blockade (ABT-627) could partially reverse the increased SBP, increased serum KIM-1 level, upregulated renal cortex protein expression of αENaC, and reduced urinary sodium excretion with reduced creatinine clearance in D307G rats fed HSD. CONCLUSION Activation of the ET-1/ETA system in D307G mutation rats might have contributed to increased sensitivity to salt loading, augmented hypertension, and exacerbated the renal injury.
Collapse
Affiliation(s)
- Lan Cheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Cardiovascular Medicine, Ningbo No.2 Hospital, Ningbo, China
| | - Hui Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Hypertension Laboratory, Fujian Provincial Cardiovascular Disease Institute, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - R. Nfornah Maboh
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Huan Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Hypertension Laboratory, Fujian Provincial Cardiovascular Disease Institute, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| |
Collapse
|
8
|
Elkenani M, Barallobre-Barreiro J, Schnelle M, Mohamed BA, Beuthner BE, Jacob CF, Paul NB, Yin X, Theofilatos K, Fischer A, Puls M, Zeisberg EM, Shah AM, Mayr M, Hasenfuß G, Toischer K. Cellular and extracellular proteomic profiling of paradoxical low-flow low-gradient aortic stenosis myocardium. Front Cardiovasc Med 2024; 11:1398114. [PMID: 39355352 PMCID: PMC11443424 DOI: 10.3389/fcvm.2024.1398114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/22/2024] [Indexed: 10/03/2024] Open
Abstract
Aims Patients with severe aortic stenosis (AS), low transvalvular flow (LF) and low gradient (LG) with normal ejection fraction (EF)-are referred to as paradoxical LF-LG AS (PLF-LG). PLF-LG patients develop more advanced heart failure symptoms and have a worse prognosis than patients with normal EF and high-gradient AS (NEF-HG). Despite its clinical relevance, the mechanisms underlying PLF-LG are still poorly understood. Methods Left ventricular (LV) myocardial biopsies of PLF-LG (n = 5) and NEF-HG patients (n = 6), obtained during transcatheter aortic valve implantation, were analyzed by LC-MS/MS after sequential extraction of cellular and extracellular matrix (ECM) proteins using a three-step extraction method. Proteomic data are available via ProteomeXchange with identifier PXD055391. Results 73 cellular proteins were differentially abundant between the 2 groups. Among these, a network of proteins related to muscle contraction and arrhythmogenic cardiomyopathy (e.g., cTnI, FKBP1A and CACNA2D1) was found in PLF-LG. Extracellularly, upregulated proteins in PLF-LG were related to ATP synthesis and oxidative phosphorylation (e.g., ATP5PF, COX5B and UQCRB). Interestingly, we observed a 1.3-fold increase in cyclophilin A (CyPA), proinflammatory cytokine, in the extracellular extracts of PLF-LG AS patients (p < 0.05). Consistently, immunohistochemical analysis confirmed its extracellular localization in PLF-LG AS LV sections along with an increase in its receptor, CD147, compared to the NEF-HG AS patients. Levels of core ECM proteins, namely collagens and proteoglycans, were comparable between groups. Conclusion Our study pinpointed novel candidates and processes with potential relevance in the pathophysiology of PLF-LG. The role of CyPA in particular warrants further investigation.
Collapse
Affiliation(s)
- Manar Elkenani
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Biochemistry and Molecular Medicine, Medical School OWL, Bielefeld University, Bielefeld, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Javier Barallobre-Barreiro
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, United Kingdom
| | - Moritz Schnelle
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
- Department of Clinical Chemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Belal A. Mohamed
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Bo E. Beuthner
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Christoph Friedemann Jacob
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Niels B. Paul
- Department of Medical Bioinformatics, University Medical Center Goettingen, Goettingen, Germany
| | - Xiaoke Yin
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, United Kingdom
| | - Konstantinos Theofilatos
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, United Kingdom
| | - Andreas Fischer
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
- Department of Clinical Chemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Miriam Puls
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Elisabeth M. Zeisberg
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Ajay M. Shah
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, United Kingdom
| | - Manuel Mayr
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, United Kingdom
| | - Gerd Hasenfuß
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Karl Toischer
- Clinic for Cardiology & Pneumology, University Medical Center Goettingen, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Goettingen, Germany
| |
Collapse
|
9
|
Liu R, Zhao H, Lu Z, Zeng L, Shi H, Wu L, Wang J, Zhong F, Liu C, Zhang Y, Qiu Z. Toxicity profiles of immune checkpoint inhibitors in nervous system cancer: a comprehensive disproportionality analysis using FDA adverse event reporting system. Clin Exp Med 2024; 24:216. [PMID: 39249163 PMCID: PMC11383843 DOI: 10.1007/s10238-024-01403-2] [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: 03/31/2024] [Accepted: 06/12/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Immune-related adverse events (irAEs) always occur during treatment with immune checkpoint inhibitors (ICIs). Patients with nervous system cancer (NSC) may gain clinical benefit from ICIs, but irAEs in NSC patients are rarely examined. Therefore, our study systematically summarized reports of irAEs in NSC. METHODS We obtained information from the FDA adverse event reporting system from the first quarter (Q1) of 2013 to the fourth quarter (Q4) of 2022. We examined use of a combination of ICIs and chemotherapy (ICI_Chemo) or chemotherapy only (ICI_Chemo) for patients with NSC. Multiple disproportionality analyses were applied to assess irAEs. Multiomics data from the gene expression omnibus (GEO) database were analyzed to explore potential molecular mechanisms associated with irAEs in NSC patients. RESULTS Fourteen irAEs were identified in 8,357 NSC patients after removing duplicates; the top five events were seizure, confused state, encephalopathy, muscular weakness and gait disturbance. Older patients were more likely to develop irAEs than were younger patients. From the start of ICIs_Chemo to irAE occurrence, there was a significant difference in the time to onset of irAEs between age groups. irAEs may occur via mechanisms involving the inflammatory response, secretion of inflammatory mediators, and aberrant activation of pathologic pathways. CONCLUSIONS This study helps to characterize irAEs in NSC patients treated with ICIs. We combined GEO database analysis to explore the potential molecular mechanisms of irAEs. The results of this study provide a basis for improving the toxic effects of ICIs in NSC patients.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hui Zhao
- Department of Sleep Medicine, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Zenghong Lu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lingshuai Zeng
- Major of Rehabilitation, Faculty of Medicine, Jinggangshan University, Ji'an, Jiangxi, China
| | - Huaqiu Shi
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Longqiu Wu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jing Wang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Fangjun Zhong
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chuanjian Liu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yu Zhang
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
| | - Zhengang Qiu
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
10
|
Røsand Ø, Wang J, Scrimgeour N, Marwarha G, Høydal MA. Exosomal Preconditioning of Human iPSC-Derived Cardiomyocytes Beneficially Alters Cardiac Electrophysiology and Micro RNA Expression. Int J Mol Sci 2024; 25:8460. [PMID: 39126028 PMCID: PMC11313350 DOI: 10.3390/ijms25158460] [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: 06/13/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Experimental evidence, both in vitro and in vivo, has indicated cardioprotective effects of extracellular vesicles (EVs) derived from various cell types, including induced pluripotent stem cell-derived cardiomyocytes. The biological effects of EV secretion, particularly in the context of ischemia and cardiac electrophysiology, remain to be fully explored. Therefore, the goal of this study was to unveil the effects of exosome (EXO)-mediated cell-cell signaling during hypoxia by employing a simulated preconditioning approach on human-induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CMs). Electrophysiological activity of hIPSC-CMs was measured using a multielectrode array (MEA) system. A total of 16 h of hypoxic stress drastically increased the beat period. Moreover, hIPSC-CMs preconditioned with EXOs displayed significantly longer beat periods compared with non-treated cells after 16 h of hypoxia (+15.7%, p < 0.05). Furthermore, preconditioning with hypoxic EXOs resulted in faster excitation-contraction (EC) coupling compared with non-treated hIPSC-CMs after 16 h of hypoxia (-25.3%, p < 0.05). Additionally, microRNA (miR) sequencing and gene target prediction analysis of the non-treated and pre-conditioned hIPSC-CMs identified 10 differentially regulated miRs and 44 gene targets. These results shed light on the intricate involvement of miRs, emphasizing gene targets associated with cell survival, contraction, apoptosis, reactive oxygen species (ROS) regulation, and ion channel modulation. Overall, this study demonstrates that EXOs secreted by hIPSC-CM during hypoxia beneficially alter electrophysiological properties in recipient cells exposed to hypoxic stress, which could play a crucial role in the development of targeted interventions to improve outcomes in ischemic heart conditions.
Collapse
Affiliation(s)
| | | | | | | | - Morten Andre Høydal
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology (NTNU), 7030 Trondheim, Norway; (Ø.R.); (J.W.); (N.S.); (G.M.)
| |
Collapse
|
11
|
Qauli AI, Danadibrata RZ, Marcellinus A, Lim KM. Development of in-silico drug cardiac toxicity evaluation system with consideration of inter-individual variability. Transl Clin Pharmacol 2024; 32:83-97. [PMID: 38974343 PMCID: PMC11224897 DOI: 10.12793/tcp.2024.32.e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/30/2024] [Accepted: 05/26/2024] [Indexed: 07/09/2024] Open
Abstract
Safety pharmacology examines the potential for new drugs to have unusual, rare side effects such as torsade de pointes (TdP). Recently, as a part of the Comprehensive in vitro Proarrhythmia Assay (CiPA) project, techniques for predicting the development of drug-induced TdP through computer simulations have been proposed and verified. However, CiPA assessment generally does not consider the effect of cardiac cell inter-individual variability, especially related to metabolic status. The study aimed to explore whether rare proarrhythmic effects may be linked to the inter-individual variability of cardiac cells and whether incorporating this variability into computational models could alter the prediction of drugs' TdP risks. This study evaluated the contribution of two biological characteristics to the proarrhythmic effects. The first was spermine concentration, which varies with metabolic status; the second was L-type calcium permeability that could occur due to mutations. Twenty-eight drugs were examined throughout this study, and qNet was analyzed as an essential feature. Even though there were some discrepancies of TdP risk predictions from the baseline model, we found that considering the inter-individual variability might change the TdP risk of drugs. Several drugs in the high-risk drugs group were predicted to affect as intermediate and low-risk drugs in some individuals and vice versa. Also, most intermediate-risk drugs were expected to act as low-risk drugs. When compared, the effects of inter-individual variability of L-type calcium were more significant than spermine in altering the TdP risk of compounds. These results emphasize the importance of considering inter-individual variability to assess drugs.
Collapse
Affiliation(s)
- Ali Ikhsanul Qauli
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
- Department of Engineering, Faculty of Advanced Technology and Multidiscipline, Universitas Airlangga, Surabaya 60115, Indonesia
| | | | - Aroli Marcellinus
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Ki Moo Lim
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
- Meta Heart Inc., Gumi 39177, Korea
| |
Collapse
|
12
|
Xu F, Cai W, Liu B, Qiu Z, Zhang X. Natural L-type calcium channels antagonists from Chinese medicine. Chin Med 2024; 19:72. [PMID: 38773596 PMCID: PMC11107034 DOI: 10.1186/s13020-024-00944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
L-type calcium channels (LTCCs), the largest subfamily of voltage-gated calcium channels (VGCCs), are the main channels for Ca2+ influx during extracellular excitation. LTCCs are widely present in excitable cells, especially cardiac and cardiovascular smooth muscle cells, and participate in various Ca2+-dependent processes. LTCCs have been considered as worthy drug target for cardiovascular, neurological and psychological diseases for decades. Natural products from Traditional Chinese medicine (TCM) have shown the potential as new drugs for the treatment of LTCCs related diseases. In this review, the basic structure, function of LTCCs, and the related human diseases caused by structural or functional abnormalities of LTCCs, and the natural LTCCs antagonist and their potential usages were summarized.
Collapse
Affiliation(s)
- Fangfang Xu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Wanna Cai
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Bo Liu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Zhenwen Qiu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| | - Xiaoqi Zhang
- Guangdong Provincial Engineering Research Center for Modernization of TCM, NMPA Key Laboratory for Quality Evaluation of TCM, Jinan University, Guangzhou, 510632, People's Republic of China.
| |
Collapse
|
13
|
Sartorius AM, Rokicki J, Birkeland S, Bettella F, Barth C, de Lange AMG, Haram M, Shadrin A, Winterton A, Steen NE, Schwarz E, Stein DJ, Andreassen OA, van der Meer D, Westlye LT, Theofanopoulou C, Quintana DS. An evolutionary timeline of the oxytocin signaling pathway. Commun Biol 2024; 7:471. [PMID: 38632466 PMCID: PMC11024182 DOI: 10.1038/s42003-024-06094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Oxytocin is a neuropeptide associated with both psychological and somatic processes like parturition and social bonding. Although oxytocin homologs have been identified in many species, the evolutionary timeline of the entire oxytocin signaling gene pathway has yet to be described. Using protein sequence similarity searches, microsynteny, and phylostratigraphy, we assigned the genes supporting the oxytocin pathway to different phylostrata based on when we found they likely arose in evolution. We show that the majority (64%) of genes in the pathway are 'modern'. Most of the modern genes evolved around the emergence of vertebrates or jawed vertebrates (540 - 530 million years ago, 'mya'), including OXTR, OXT and CD38. Of those, 45% were under positive selection at some point during vertebrate evolution. We also found that 18% of the genes in the oxytocin pathway are 'ancient', meaning their emergence dates back to cellular organisms and opisthokonta (3500-1100 mya). The remaining genes (18%) that evolved after ancient and before modern genes were classified as 'medium-aged'. Functional analyses revealed that, in humans, medium-aged oxytocin pathway genes are highly expressed in contractile organs, while modern genes in the oxytocin pathway are primarily expressed in the brain and muscle tissue.
Collapse
Affiliation(s)
- Alina M Sartorius
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Jaroslav Rokicki
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre of Research and Education in Forensic Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Siri Birkeland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
- Natural History Museum, University of Oslo, Oslo, Norway
| | - Francesco Bettella
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Ann-Marie G de Lange
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Marit Haram
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Mental Health and Suicide, Norwegian Institute of Public Health, Oslo, Norway
| | - Alexey Shadrin
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Adriano Winterton
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Emanuel Schwarz
- Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dan J Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Daniel S Quintana
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine and Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway.
- Department of Psychology, University of Oslo, Oslo, Norway.
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway.
- NevSom, Department of Rare Disorders, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
14
|
Wei Y, Yu Z, Wang L, Li X, Li N, Bai Q, Wang Y, Li R, Meng Y, Xu H, Wang X, Dong Y, Huang Z, Zhang XC, Zhao Y. Structural bases of inhibitory mechanism of Ca V1.2 channel inhibitors. Nat Commun 2024; 15:2772. [PMID: 38555290 PMCID: PMC10981686 DOI: 10.1038/s41467-024-47116-8] [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: 12/05/2022] [Accepted: 03/19/2024] [Indexed: 04/02/2024] Open
Abstract
The voltage-gated calcium channel CaV1.2 is essential for cardiac and vessel smooth muscle contractility and brain function. Accumulating evidence demonstrates that malfunctions of CaV1.2 are involved in brain and heart diseases. Pharmacological inhibition of CaV1.2 is therefore of therapeutic value. Here, we report cryo-EM structures of CaV1.2 in the absence or presence of the antirheumatic drug tetrandrine or antihypertensive drug benidipine. Tetrandrine acts as a pore blocker in a pocket composed of S6II, S6III, and S6IV helices and forms extensive hydrophobic interactions with CaV1.2. Our structure elucidates that benidipine is located in the DIII-DIV fenestration site. Its hydrophobic sidechain, phenylpiperidine, is positioned at the exterior of the pore domain and cradled within a hydrophobic pocket formed by S5DIII, S6DIII, and S6DIV helices, providing additional interactions to exert inhibitory effects on both L-type and T-type voltage gated calcium channels. These findings provide the structural foundation for the rational design and optimization of therapeutic inhibitors of voltage-gated calcium channels.
Collapse
Affiliation(s)
- Yiqing Wei
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhuoya Yu
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lili Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiaojing Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Na Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Qinru Bai
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuhang Wang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Renjie Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yufei Meng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Xu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xianping Wang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanli Dong
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Xuejun Cai Zhang
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yan Zhao
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
15
|
Khan M, Chen XXL, Dias M, Santos JR, Kour S, You J, van Bruggen R, Youssef MMM, Wan YW, Liu Z, Rosenfeld JA, Tan Q, Pandey UB, Yalamanchili HK, Park J. MATR3 pathogenic variants differentially impair its cryptic splicing repression function. FEBS Lett 2024; 598:415-436. [PMID: 38320753 DOI: 10.1002/1873-3468.14806] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 02/28/2024]
Abstract
Matrin-3 (MATR3) is an RNA-binding protein implicated in neurodegenerative and neurodevelopmental diseases. However, little is known regarding the role of MATR3 in cryptic splicing within the context of functional genes and how disease-associated variants impact this function. We show that loss of MATR3 leads to cryptic exon inclusion in many transcripts. We reveal that ALS-linked S85C pathogenic variant reduces MATR3 solubility but does not impair RNA binding. In parallel, we report a novel neurodevelopmental disease-associated M548T variant, located in the RRM2 domain, which reduces protein solubility and impairs RNA binding and cryptic splicing repression functions of MATR3. Altogether, our research identifies cryptic events within functional genes and demonstrates how disease-associated variants impact MATR3 cryptic splicing repression function.
Collapse
Affiliation(s)
- Mashiat Khan
- Department of Molecular Genetics, University of Toronto, Canada
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Xiao Xiao Lily Chen
- Department of Molecular Genetics, University of Toronto, Canada
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Michelle Dias
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Jhune Rizsan Santos
- Department of Molecular Genetics, University of Toronto, Canada
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Sukhleen Kour
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Justin You
- Department of Molecular Genetics, University of Toronto, Canada
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Rebekah van Bruggen
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Mohieldin M M Youssef
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Ying-Wooi Wan
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Zhandong Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Baylor Genetics Laboratories, Houston, TX, USA
| | - Qiumin Tan
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Udai Bhan Pandey
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, School of Public Health, Pittsburgh, PA, USA
| | - Hari Krishna Yalamanchili
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jeehye Park
- Department of Molecular Genetics, University of Toronto, Canada
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
16
|
Kanithi M, Kumari L, Yalakaturi K, Munjal K, Jimitreddy S, Kandamuri M, Veeramachineni P, Chopra H, Junapudi S. Nanoparticle Polymers Influence on Cardiac Health: Good or Bad for Cardiac Physiology? Curr Probl Cardiol 2024; 49:102145. [PMID: 37852559 DOI: 10.1016/j.cpcardiol.2023.102145] [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: 10/06/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Cardiovascular diseases (CVD) are one of the leading causes of death and morbidity worldwide. Lifestyle modifications, medications, and addressing epidemiological factors have long been at the forefront of targeting therapeutics for CVD. Treatments can be further complicated given the intersection of gender, age, unique comorbidities, and healthcare access, among many other factors. Therefore, expanding treatment and diagnostic modalities for CVD is absolutely necessary. Nanoparticles and nanomaterials are increasingly being used as therapeutic and diagnostic modalities in various disciplines of biomedicine. Nanoparticles have multiple ways of interacting with the cardiovascular system. Some of them alter cardiac physiology by impacting ion channels, whereas others influence ions directly or indirectly, improving cellular death via decreasing oxidative stress. While embedding nanoparticles into therapeutics can help enhance healthy cardiovascular function in other scenarios, they can also impair physiology by increasing reactive oxidative species and leading to cardiotoxicity. This review explores different types of nanoparticles, their effects, and the applicable dosages to create a better foundation for understanding the current research findings.
Collapse
Affiliation(s)
- Manasa Kanithi
- Michigan State University College of Osteopathic Medicine, East Lansing, MI
| | - Lata Kumari
- People University of Medical and Health Sciences, Nawab Shah, Sindh, Pakistan
| | | | - Kavita Munjal
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | | | | | | | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Sunil Junapudi
- Geethanjali College of Pharmacy, Hyderabad, Telangana, India.
| |
Collapse
|
17
|
Yang J, Ye K, Zhang R, Fan X, Xiong R, Zhang S, Liu Q, Lin M, Wang B, Tan X, Wen Q, Ou X. The characteristics and molecular targets of antiarrhythmic natural products. Biomed Pharmacother 2023; 168:115762. [PMID: 37897974 DOI: 10.1016/j.biopha.2023.115762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023] Open
Abstract
Arrhythmia is one of the most common cardiovascular diseases. The search for new drugs to suppress various types of cardiac arrhythmias has always been the focus of attention. In the past decade, the screening of antiarrhythmic active substances from plants has received extensive attention. These natural compounds have obvious antiarrhythmic effects, and chemical modifications based on natural compounds have greatly increased their pharmacological properties. The chemical modification of botanical antiarrhythmic drugs is closely related to the development of new and promising drugs. Therefore, the structural characteristics and action targets of natural compounds with antiarrhythmic effects are reviewed in this paper, so that pharmacologists can select antiarrhythmic lead compounds from natural compounds based on the disease target - chemical structural characteristics.
Collapse
Affiliation(s)
- Jun Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China; Department of Pharmacy, Santai County People's Hospital of Sichuan Province, Mianyang 621100, China
| | - Kejun Ye
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China; Pharmacy Department, Chongqing Armed Police Corps Hospital, Chongqing 400061, China
| | - Rui Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Xinrong Fan
- The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Rui Xiong
- Department of Pharmacy of the 958 Hospital of Chinese PLA/Jiangbei Campus, The First Affiliated Hospital of Army Medical University, Chongqing 400020, China
| | - Shiyu Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Qiming Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Miao Lin
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Bin Wang
- The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Qiang Wen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, Guangxi Province, China.
| |
Collapse
|
18
|
Li P, Qin D, Chen T, Hou W, Song X, Yin S, Song M, Fernando WCHA, Chen X, Sun Y, Wang J. Dysregulated Rbfox2 produces aberrant splicing of Ca V1.2 calcium channel in diabetes-induced cardiac hypertrophy. Cardiovasc Diabetol 2023; 22:168. [PMID: 37415128 PMCID: PMC10324275 DOI: 10.1186/s12933-023-01894-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND L-type Ca2+ channel CaV1.2 is essential for cardiomyocyte excitation, contraction and gene transcription in the heart, and abnormal functions of cardiac CaV1.2 channels are presented in diabetic cardiomyopathy. However, the underlying mechanisms are largely unclear. The functions of CaV1.2 channels are subtly modulated by splicing factor-mediated alternative splicing (AS), but whether and how CaV1.2 channels are alternatively spliced in diabetic heart remains unknown. METHODS Diabetic rat models were established by using high-fat diet in combination with low dose streptozotocin. Cardiac function and morphology were assessed by echocardiography and HE staining, respectively. Isolated neonatal rat ventricular myocytes (NRVMs) were used as a cell-based model. Cardiac CaV1.2 channel functions were measured by whole-cell patch clamp, and intracellular Ca2+ concentration was monitored by using Fluo-4 AM. RESULTS We find that diabetic rats develop diastolic dysfunction and cardiac hypertrophy accompanied by an increased CaV1.2 channel with alternative exon 9* (CaV1.2E9*), but unchanged that with alternative exon 8/8a or exon 33. The splicing factor Rbfox2 expression is also increased in diabetic heart, presumably because of dominate-negative (DN) isoform. Unexpectedly, high glucose cannot induce the aberrant expressions of CaV1.2 exon 9* and Rbfox2. But glycated serum (GS), the mimic of advanced glycation end-products (AGEs), upregulates CaV1.2E9* channels proportion and downregulates Rbfox2 expression in NRVMs. By whole-cell patch clamp, we find GS application hyperpolarizes the current-voltage curve and window currents of cardiac CaV1.2 channels. Moreover, GS treatment raises K+-triggered intracellular Ca2+ concentration ([Ca2+]i), enlarges cell surface area of NRVMs and induces hypertrophic genes transcription. Consistently, siRNA-mediated knockdown of Rbfox2 in NRVMs upregulates CaV1.2E9* channel, shifts CaV1.2 window currents to hyperpolarization, increases [Ca2+]i and induces cardiomyocyte hypertrophy. CONCLUSIONS AGEs, not glucose, dysregulates Rbfox2 which thereby increases CaV1.2E9* channels and hyperpolarizes channel window currents. These make the channels open at greater negative potentials and lead to increased [Ca2+]i in cardiomyocytes, and finally induce cardiomyocyte hypertrophy in diabetes. Our work elucidates the underlying mechanisms for CaV1.2 channel regulation in diabetic heart, and targeting Rbfox2 to reset the aberrantly spliced CaV1.2 channel might be a promising therapeutic approach in diabetes-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Pengpeng Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dongxia Qin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Tiange Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Wei Hou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xinyu Song
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Shumin Yin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Miaomiao Song
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - W C Hewith A Fernando
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiaojie Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yu Sun
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Juejin Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
19
|
Chaudhry-Waterman N, Dara B, Bucholz E, Londono Obregon C, Grenier M, Snyder K, Cuneo BF. Fetal Heart Rate < 3rd Percentile for Gestational Age Can Be a Marker of Inherited Arrhythmia Syndromes. J Clin Med 2023; 12:4464. [PMID: 37445499 DOI: 10.3390/jcm12134464] [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: 04/30/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Repeated fetal heart rates (FHR) < 3rd percentile for gestational age (GA) with 1:1 atrioventricular conduction (sinus bradycardia) can be a marker for long QT syndrome. We hypothesized that other inherited arrhythmia syndromes might present with fetal sinus bradycardia. METHODS We reviewed pregnancies referred with sinus bradycardia to the Colorado Fetal Care Center between 2013 and 2023. FHR/GA data, family history, medication exposure, normalized isovolumic contraction times (n-IVRT), postnatal genetic testing, and ECGs at 4-6 weeks after birth were reviewed. RESULTS Twenty-nine bradycardic subjects were evaluated by fetal echocardiography. Five were lost to follow-up, one refused genetic testing, and one had negative genetic testing for any inherited arrhythmia. Six had non-genetic causes of fetal bradycardia with normal prenatal n-IVRT and postnatal QTc. Thirteen carried pathogenic variants in RYR2 (n = 2), HCN4 (n = 2), KCNQ1 (6), and other LQTS genes (n = 4). The postnatal QTc was <470 ms in subjects with RYR2, HCN4, and two of those with KCNQ1 mutations, and >470 ms in subjects with CALM 2, KCNH2, SCN5A, and four of those with KCNQ1 mutations. LQTS and RYR2 mutations were associated with prolonged n-IVRT, but HCN4 was not. Two fetuses died in utero with variants of uncertain significance (CACNA1 and KCNE1). Cascade testing uncovered six affected but undiagnosed parents and confirmed familial inheritance in five. CONCLUSION In addition to heralding LQTS, repeated FHR < 3rd percentile for GA is a risk factor for other inherited arrhythmia syndromes. These findings suggest that genetic testing should be offered to infants with a history of FHR < 3rd percentile for GA even if the postnatal ECG demonstrates a normal QTc interval.
Collapse
Affiliation(s)
- Nadia Chaudhry-Waterman
- The Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Denver, CO 80045, USA
| | - Bharat Dara
- Presbyterian Hospital, Albuquerque, NM 87106, USA
| | - Emily Bucholz
- The Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Denver, CO 80045, USA
- The Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Camila Londono Obregon
- The Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Denver, CO 80045, USA
- The Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Michelle Grenier
- The Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Denver, CO 80045, USA
- The Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Kristen Snyder
- The Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Bettina F Cuneo
- The Division of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Denver, CO 80045, USA
- The Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO 80045, USA
- The Department of Obstetrics, University of Colorado School of Medicine, Denver, CO 80045, USA
| |
Collapse
|
20
|
Pharmacological mechanism of natural drugs and their active ingredients in the treatment of arrhythmia via calcium channel regulation. Biomed Pharmacother 2023; 160:114413. [PMID: 36805187 DOI: 10.1016/j.biopha.2023.114413] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/11/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Arrhythmia is characterized by abnormal heartbeat rhythms and frequencies caused by heart pacing and conduction dysfunction. Arrhythmia is the leading cause of death in patients with cardiovascular disease, with high morbidity and mortality rates, posing a serious risk to human health. Natural drugs and their active ingredients, such as matrine(MAT), tetrandrine(TET), dehydroevodiamine, tanshinone IIA, and ginsenosides, have been widely used for the treatment of atrial fibrillation, ventricular ectopic beats, sick sinus syndrome, and other arrhythmia-like diseases owing to their unique advantages. This review summarizes the mechanism of action of natural drugs and their active ingredients in the treatment of arrhythmia via the regulation of Ca2+, such as alkaloids, quinones, saponins, terpenoids, flavonoids, polyphenols, and lignan compounds, to provide ideas for the innovative development of natural drugs with potential antiarrhythmic efficacy.
Collapse
|
21
|
Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol 2023; 20:109-125. [PMID: 35931887 PMCID: PMC9362470 DOI: 10.1038/s41569-022-00741-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 01/21/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as a gaseous signalling molecule with crucial implications for cardiovascular health. H2S is involved in many biological functions, including interactions with nitric oxide, activation of molecular signalling cascades, post-translational modifications and redox regulation. Various preclinical and clinical studies have shown that H2S and its synthesizing enzymes - cystathionine γ-lyase, cystathionine β-synthase and 3-mercaptosulfotransferase - can protect against cardiovascular pathologies, including arrhythmias, atherosclerosis, heart failure, myocardial infarction and ischaemia-reperfusion injury. The bioavailability of H2S and its metabolites, such as hydropersulfides and polysulfides, is substantially reduced in cardiovascular disease and has been associated with single-nucleotide polymorphisms in H2S synthesis enzymes. In this Review, we highlight the role of H2S, its synthesizing enzymes and metabolites, their roles in the cardiovascular system, and their involvement in cardiovascular disease and associated pathologies. We also discuss the latest clinical findings from the field and outline areas for future study.
Collapse
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Paari Dominic
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
22
|
Kainer D, Templeton AR, Prates ET, Jacboson D, Allan ER, Climer S, Garvin MR. Structural variants identified using non-Mendelian inheritance patterns advance the mechanistic understanding of autism spectrum disorder. HGG ADVANCES 2023; 4:100150. [PMID: 36340933 PMCID: PMC9634371 DOI: 10.1016/j.xhgg.2022.100150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
The heritability of autism spectrum disorder (ASD), based on 680,000 families and five countries, is estimated to be nearly 80%, yet heritability reported from SNP-based studies are consistently lower, and few significant loci have been identified with genome-wide association studies. This gap in genomic information may reside in rare variants, interaction among variants (epistasis), or cryptic structural variation (SV) and may provide mechanisms that underlie ASD. Here we use a method to identify potential SVs based on non-Mendelian inheritance patterns in pedigrees using parent-child genotypes from ASD families and demonstrate that they are enriched in ASD-risk genes. Most are in non-coding genic space and are over-represented in expression quantitative trait loci, suggesting that they affect gene regulation, which we confirm with their overlap of differentially expressed genes in postmortem brain tissue of ASD individuals. We then identify an SV in the GRIK2 gene that alters RNA splicing and a regulatory region of the ACMSD gene in the kynurenine pathway as significantly associated with a non-verbal ASD phenotype, supporting our hypothesis that these currently excluded loci can provide a clearer mechanistic understanding of ASD. Finally, we use an explainable artificial intelligence approach to define subgroups demonstrating their use in the context of precision medicine.
Collapse
Affiliation(s)
- David Kainer
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Alan R. Templeton
- Department of Biology, Washington University – St Louis, St. Louis, MO, USA
| | - Erica T. Prates
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Daniel Jacboson
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | - Sharlee Climer
- Department of Computer Science, University of Missouri, St. Louis, MO, USA
| | - Michael R. Garvin
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
- Williwaw Biosciences, LLC, Clarkston, MI, USA
| |
Collapse
|
23
|
Sallam M, Mysara M, Benotmane MA, Tamarat R, Santos SCR, Crijns APG, Spoor D, Van Nieuwerburgh F, Deforce D, Baatout S, Guns PJ, Aerts A, Ramadan R. DNA Methylation Alterations in Fractionally Irradiated Rats and Breast Cancer Patients Receiving Radiotherapy. Int J Mol Sci 2022; 23:16214. [PMID: 36555856 PMCID: PMC9783664 DOI: 10.3390/ijms232416214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Radiation-Induced CardioVascular Disease (RICVD) is an important concern in thoracic radiotherapy with complex underlying pathophysiology. Recently, we proposed DNA methylation as a possible mechanism contributing to RICVD. The current study investigates DNA methylation in heart-irradiated rats and radiotherapy-treated breast cancer (BC) patients. Rats received fractionated whole heart X-irradiation (0, 0.92, 6.9 and 27.6 Gy total doses) and blood was collected after 1.5, 3, 7 and 12 months. Global and gene-specific methylation of the samples were evaluated; and gene expression of selected differentially methylated regions (DMRs) was validated in rat and BC patient blood. In rats receiving an absorbed dose of 27.6 Gy, DNA methylation alterations were detected up to 7 months with differential expression of cardiac-relevant DMRs. Of those, SLMAP showed increased expression at 1.5 months, which correlated with hypomethylation. Furthermore, E2F6 inversely correlated with a decreased global longitudinal strain. In BC patients, E2F6 and SLMAP exhibited differential expression directly and 6 months after radiotherapy, respectively. This study describes a systemic radiation fingerprint at the DNA methylation level, elucidating a possible association of DNA methylation to RICVD pathophysiology, to be validated in future mechanistic studies.
Collapse
Affiliation(s)
- Magy Sallam
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
- Laboratory of Physiopharmacology, University of Antwerp, 2610 Wilrijk, Belgium;
| | - Mohamed Mysara
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| | - Radia Tamarat
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, LR2I, 92260 Fontenay-aux-Roses, France;
| | - Susana Constantino Rosa Santos
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Lisbon School of Medicine of the Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Anne P. G. Crijns
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.P.G.C.); (D.S.)
| | - Daan Spoor
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.P.G.C.); (D.S.)
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Ghent University, 9000 Ghent, Belgium; (F.V.N.); (D.D.)
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Ghent University, 9000 Ghent, Belgium; (F.V.N.); (D.D.)
| | - Sarah Baatout
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
- Department of Molecular Biotechnology, Ghent University, 9000 Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, 2610 Wilrijk, Belgium;
| | - An Aerts
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| | - Raghda Ramadan
- Radiobiology Unit, Interdisciplinary Biosciences, Belgian Nuclear Research Centre, SCK CEN, 2400 Mol, Belgium; (M.S.); (M.M.); (M.A.B.); (S.B.); (A.A.)
| |
Collapse
|
24
|
Zhong R, Zhang F, Yang Z, Li Y, Xu Q, Lan H, Cyganek L, El-Battrawy I, Zhou X, Akin I, Borggrefe M. Epigenetic mechanism of L-type calcium channel β-subunit downregulation in short QT human induced pluripotent stem cell-derived cardiomyocytes with CACNB2 mutation. Europace 2022; 24:2028-2036. [PMID: 35894107 DOI: 10.1093/europace/euac091] [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: 12/30/2021] [Accepted: 05/15/2022] [Indexed: 12/14/2022] Open
Abstract
AIMS A loss-of-function mutation in L-type calcium (Ca2+) channel subunit gene CACNB2 has been reported to cause short QT syndrome subtype 5 (SQT5). However, the mechanism underlying the loss-of-function of the Ca2+ channel has not been clarified. In the present study, we aim to explore the DNA methylation mechanism of L-type Ca2+ channel downregulation in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) of SQT5. METHODS AND RESULTS The hiPSC-CMs were generated from a healthy donor and a SQT5 patient carrying the CACNB2 variant c.1439C > T/p.S480L. The variant was genetically corrected using ribonucleoprotein-based CRISPR/Cas9 technique to obtain an isogenic control cell line. The action potential (AP) and Ca2+ current were measured by patch clamp. Protein expression levels were determined by western blotting. Dot blotting and bisulfite sequence were performed for epigenetic study. Our results showed that AP durations at 10% repolarization (APD10) and 50% repolarization (APD50) were significantly shortened in SQT5 cells and both the expression level of the β-subunit and channel current of L-type Ca2+ channel were reduced. Besides, an increased level of whole-genome DNA methylation and DNA methylation of CpG island in the promoter region of CACNB2 gene was detected. Overexpression of demethylation enzyme could rescue the decreased expression of CACNB2 and the L-type Ca2+ current. CONCLUSION In SQT5 hiPSC-CMs carrying the CACNB2-S480L variant, the decreased L-type Ca2+ current resulting from decreased CACNB2 protein expression was caused by enhanced methylation in the promoter region of the CACNB2 gene and upregulation of DNA methyltransferases might be one of the mechanisms.
Collapse
Affiliation(s)
- Rujia Zhong
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Feng Zhang
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Zhen Yang
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Yingrui Li
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Qiang Xu
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Huan Lan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen 37075, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen 37075, Germany
| | - Ibrahim El-Battrawy
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim 68167, Germany
| | - Xiaobo Zhou
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim 68167, Germany.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Ibrahim Akin
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim 68167, Germany
| | - Martin Borggrefe
- First Department of Medicine (Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care), Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim 68167, Germany
| |
Collapse
|
25
|
Morgenstern TJ, Nirwan N, Hernández-Ochoa EO, Bibollet H, Choudhury P, Laloudakis YD, Ben Johny M, Bannister RA, Schneider MF, Minor DL, Colecraft HM. Selective posttranslational inhibition of Ca Vβ 1-associated voltage-dependent calcium channels with a functionalized nanobody. Nat Commun 2022; 13:7556. [PMID: 36494348 PMCID: PMC9734117 DOI: 10.1038/s41467-022-35025-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
Ca2+ influx through high-voltage-activated calcium channels (HVACCs) controls diverse cellular functions. A critical feature enabling a singular signal, Ca2+ influx, to mediate disparate functions is diversity of HVACC pore-forming α1 and auxiliary CaVβ1-CaVβ4 subunits. Selective CaVα1 blockers have enabled deciphering their unique physiological roles. By contrast, the capacity to post-translationally inhibit HVACCs based on CaVβ isoform is non-existent. Conventional gene knockout/shRNA approaches do not adequately address this deficit owing to subunit reshuffling and partially overlapping functions of CaVβ isoforms. Here, we identify a nanobody (nb.E8) that selectively binds CaVβ1 SH3 domain and inhibits CaVβ1-associated HVACCs by reducing channel surface density, decreasing open probability, and speeding inactivation. Functionalizing nb.E8 with Nedd4L HECT domain yielded Chisel-1 which eliminated current through CaVβ1-reconstituted CaV1/CaV2 and native CaV1.1 channels in skeletal muscle, strongly suppressed depolarization-evoked Ca2+ influx and excitation-transcription coupling in hippocampal neurons, but was inert against CaVβ2-associated CaV1.2 in cardiomyocytes. The results introduce an original method for probing distinctive functions of ion channel auxiliary subunit isoforms, reveal additional dimensions of CaVβ1 signaling in neurons, and describe a genetically-encoded HVACC inhibitor with unique properties.
Collapse
Affiliation(s)
- Travis J. Morgenstern
- grid.239585.00000 0001 2285 2675Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY USA
| | - Neha Nirwan
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| | - Erick O. Hernández-Ochoa
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Hugo Bibollet
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Papiya Choudhury
- grid.239585.00000 0001 2285 2675Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY USA
| | - Yianni D. Laloudakis
- grid.239585.00000 0001 2285 2675Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY USA
| | - Manu Ben Johny
- grid.239585.00000 0001 2285 2675Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY USA
| | - Roger A. Bannister
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Martin F. Schneider
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Daniel L. Minor
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811Department of Biochemistry and Biophysics, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA USA ,grid.184769.50000 0001 2231 4551Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Henry M. Colecraft
- grid.239585.00000 0001 2285 2675Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY USA ,grid.239585.00000 0001 2285 2675Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
26
|
Akerberg AA, Trembley M, Butty V, Schwertner A, Zhao L, Beerens M, Liu X, Mahamdeh M, Yuan S, Boyer L, MacRae C, Nguyen C, Pu WT, Burns CE, Burns CG. RBPMS2 Is a Myocardial-Enriched Splicing Regulator Required for Cardiac Function. Circ Res 2022; 131:980-1000. [PMID: 36367103 PMCID: PMC9770155 DOI: 10.1161/circresaha.122.321728] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND RBPs (RNA-binding proteins) perform indispensable functions in the post-transcriptional regulation of gene expression. Numerous RBPs have been implicated in cardiac development or physiology based on gene knockout studies and the identification of pathogenic RBP gene mutations in monogenic heart disorders. The discovery and characterization of additional RBPs performing indispensable functions in the heart will advance basic and translational cardiovascular research. METHODS We performed a differential expression screen in zebrafish embryos to identify genes enriched in nkx2.5-positive cardiomyocytes or cardiopharyngeal progenitors compared to nkx2.5-negative cells from the same embryos. We investigated the myocardial-enriched gene RNA-binding protein with multiple splicing (variants) 2 [RBPMS2)] by generating and characterizing rbpms2 knockout zebrafish and human cardiomyocytes derived from RBPMS2-deficient induced pluripotent stem cells. RESULTS We identified 1848 genes enriched in the nkx2.5-positive population. Among the most highly enriched genes, most with well-established functions in the heart, we discovered the ohnologs rbpms2a and rbpms2b, which encode an evolutionarily conserved RBP. Rbpms2 localizes selectively to cardiomyocytes during zebrafish heart development and strong cardiomyocyte expression persists into adulthood. Rbpms2-deficient embryos suffer from early cardiac dysfunction characterized by reduced ejection fraction. The functional deficit is accompanied by myofibril disarray, altered calcium handling, and differential alternative splicing events in mutant cardiomyocytes. These phenotypes are also observed in RBPMS2-deficient human cardiomyocytes, indicative of conserved molecular and cellular function. RNA-sequencing and comparative analysis of genes mis-spliced in RBPMS2-deficient zebrafish and human cardiomyocytes uncovered a conserved network of 29 ortholog pairs that require RBPMS2 for alternative splicing regulation, including RBFOX2, SLC8A1, and MYBPC3. CONCLUSIONS Our study identifies RBPMS2 as a conserved regulator of alternative splicing, myofibrillar organization, and calcium handling in zebrafish and human cardiomyocytes.
Collapse
Affiliation(s)
- Alexander A. Akerberg
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Michael Trembley
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Vincent Butty
- BioMicroCenter, Department of Biology (V.B.), Massachusetts Institute of Technology, Cambridge‚ MA
- Department of Biology (V.B., L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
| | - Asya Schwertner
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Long Zhao
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Manu Beerens
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (M.B., C.M.)
| | - Xujie Liu
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Mohammed Mahamdeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Shiaulou Yuan
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| | - Laurie Boyer
- Department of Biology (V.B., L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
- Department of Biological Engineering (L.B.), Massachusetts Institute of Technology, Cambridge‚ MA
| | - Calum MacRae
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (M.B., C.M.)
| | - Christopher Nguyen
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Innovation Research Center, Heart Vascular & Thoracic Institute, Cleveland Clinic‚ Cleveland‚ OH (C.N.)
| | - William T. Pu
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Harvard Stem Cell Institute, Cambridge, MA (W.T.P., C.E.B.)
| | - Caroline E. Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
- Harvard Stem Cell Institute, Cambridge, MA (W.T.P., C.E.B.)
| | - C. Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children’s Hospital, Boston‚ MA (A.A.A., M.T., X.L., W.T.P., C.E.B., C.G.B.)
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown‚ MA (A.A.A., A.S., L.Z., M.M., S.Y., C.N., C.E.B., C.G.B.)
- Harvard Medical School, Boston, MA (A.A.A., M.T., A.S., L.Z., M.B., X.L., M.M., S.Y., C.M., C.N., W.T.P., C.E.B., C.G.B.)
| |
Collapse
|
27
|
Guo X, Sha Y, Pu X, Xu Y, Yao L, Liu X, He Y, Hu J, Wang J, Li S, Chen G. Coevolution of Rumen Epithelial circRNAs with Their Microbiota and Metabolites in Response to Cold-Season Nutritional Stress in Tibetan Sheep. Int J Mol Sci 2022; 23:ijms231810488. [PMID: 36142400 PMCID: PMC9499677 DOI: 10.3390/ijms231810488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
This study explores the effects of the coevolution of the host genome (the first genome) and gut microbiome (the second genome) on nutrition stress in Tibetan sheep during the cold season. The rumen epithelial tissue of six Tibetan sheep (Oula-type) was collected as experimental samples during the cold and warm seasons and the study lasted for half a year. The cDNA library was constructed and subjected to high-throughput sequencing. The circRNAs with significant differential expression were identified through bioinformatics analysis and functional prediction, and verified by real-time quantitative PCR (qRT-PCR). The results showed that a total of 56 differentially expressed (DE) circRNAs of rumen epithelial tissue were identified using RNA-seq technology, among which 29 were significantly upregulated in the cold season. The circRNA-miRNA regulatory network showed that DE circRNAs promoted the adaptation of Tibetan sheep in the cold season by targeting miR-150 and oar-miR-370-3p. The results of correlation analysis among circRNAs, microbiota, and metabolites showed that the circRNA NC_040275.1:28680890|28683112 had a very significant positive correlation with acetate, propionate, butyrate, and total volatile fatty acid (VFA) (p < 0.01), and had a significant positive correlation with Ruminococcus-1 (p < 0.05). In addition, circRNA NC_040256.1:78451819|78454934 and metabolites were enriched in the same KEGG pathway biosynthesis of amino acids (ko01230). In conclusion, the host genome and rumen microbiome of Tibetan sheep co-encoded a certain glycoside hydrolase (β-glucosidase) and coevolved efficient VFA transport functions and amino acid anabolic processes; thus, helping Tibetan sheep adapt to nutrient stress in the cold season in high-altitude areas.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiu Liu
- Correspondence: (X.L.); (G.C.)
| | | | | | | | | | | |
Collapse
|
28
|
Nakajima T, Kawabata-Iwakawa R, Tamura S, Hasegawa H, Kobari T, Itoh H, Horie M, Nishiyama M, Kurabayashi M, Kaneko Y, Ishii H. Novel CACNA1C R511Q mutation, located in domain Ⅰ-Ⅱ linker, causes non-syndromic type-8 long QT syndrome. PLoS One 2022; 17:e0271796. [PMID: 35862440 PMCID: PMC9302756 DOI: 10.1371/journal.pone.0271796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 07/07/2022] [Indexed: 11/26/2022] Open
Abstract
Background Gain-of-function mutations in CACNA1C encoding Cav1.2 cause syndromic or non-syndromic type-8 long QT syndrome (LQTS) (sLQT8 or nsLQT8). The cytoplasmic domain (D)Ⅰ-Ⅱ linker in Cav1.2 plays a pivotal role in calcium channel inactivation, and mutations in this site have been associated with sLQT8 (such as Timothy syndrome) but not nsLQT8. Objective Since we identified a novel CACNA1C mutation, located in the DⅠ-Ⅱ linker, associated with nsLQTS, we sought to reveal its biophysical defects. Methods Target panel sequencing was employed in 24 genotype-negative nsLQTS probands (after Sanger sequencing) and three family members. Wild-type (WT) or R511Q Cav1.2 was transiently expressed in tsA201 cells, then whole-cell Ca2+ or Ba2+ currents (ICa or IBa) were recorded using whole-cell patch-clamp techniques. Results We identified two CACNA1C mutations, a previously reported R858H mutation and a novel R511Q mutation located in the DⅠ-Ⅱ linker. Four members of one nsLQTS family harbored the CACNA1C R511Q mutation. The current density and steady-state activation were comparable to those of WT-ICa. However, persistent currents in R511Q-ICa were significantly larger than those of WT-ICa (WT at +20 mV: 3.3±0.3%, R511Q: 10.8±0.8%, P<0.01). The steady-state inactivation of R511Q-ICa was weak in comparison to that of WT-ICa at higher prepulse potentials, resulting in increased window currents in R511Q-ICa. Slow component of inactivation of R511Q-ICa was significantly delayed compared to that of WT-ICa (WT-tau at +20 mV: 81.3±3.3 ms, R511Q-tau: 125.1±5.0 ms, P<0.01). Inactivation of R511Q-IBa was still slower than that of WT-IBa, indicating that voltage-dependent inactivation (VDI) of R511Q-ICa was predominantly delayed. Conclusions Delayed VDI, increased persistent currents, and increased window currents of R511Q-ICa cause nsLQT8. Our data provide novel insights into the structure-function relationships of Cav1.2 and the pathophysiological roles of the DⅠ-Ⅱ linker in phenotypic manifestations.
Collapse
Affiliation(s)
- Tadashi Nakajima
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- * E-mail:
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Maebashi, Gunma, Japan
| | - Shuntaro Tamura
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hiroshi Hasegawa
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Takashi Kobari
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hideki Itoh
- Division of Patient Safety, Hiroshima University Hospital, Hiroshima, Hiroshima, Japan
| | - Minoru Horie
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Ohtsu, Shiga, Japan
| | | | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yoshiaki Kaneko
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hideki Ishii
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
29
|
Wang M, Tu X. The Genetics and Epigenetics of Ventricular Arrhythmias in Patients Without Structural Heart Disease. Front Cardiovasc Med 2022; 9:891399. [PMID: 35783865 PMCID: PMC9240357 DOI: 10.3389/fcvm.2022.891399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/25/2022] [Indexed: 12/19/2022] Open
Abstract
Ventricular arrhythmia without structural heart disease is an arrhythmic disorder that occurs in structurally normal heart and no transient or reversible arrhythmia factors, such as electrolyte disorders and myocardial ischemia. Ventricular arrhythmias without structural heart disease can be induced by multiple factors, including genetics and environment, which involve different genetic and epigenetic regulation. Familial genetic analysis reveals that cardiac ion-channel disorder and dysfunctional calcium handling are two major causes of this type of heart disease. Genome-wide association studies have identified some genetic susceptibility loci associated with ventricular tachycardia and ventricular fibrillation, yet relatively few loci associated with no structural heart disease. The effects of epigenetics on the ventricular arrhythmias susceptibility genes, involving non-coding RNAs, DNA methylation and other regulatory mechanisms, are gradually being revealed. This article aims to review the knowledge of ventricular arrhythmia without structural heart disease in genetics, and summarizes the current state of epigenetic regulation.
Collapse
|
30
|
Park J, Kim Y, Jeon HJ, Kim K, Kim C, Lee S, Son J, Lee SE. Acute and developmental toxic effects of mono-halogenated and halomethyl naphthalenes on zebrafish (Danio rerio) embryos: Cardiac malformation after 2-bromomethyl naphthalene exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 297:118786. [PMID: 34990738 DOI: 10.1016/j.envpol.2021.118786] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/04/2021] [Accepted: 12/31/2021] [Indexed: 06/14/2023]
Abstract
Polyhalogenated polycyclic aromatic hydrocarbons (HPAHs) represent a major environmental concern due to their persistency and toxicity. Among them, mono-halogenated (HNs) and halomethyl naphthalenes (HMNs) are not well-studied, and the toxicity of many HNs to fishes has not been reported. In this study, we exposed zebrafish (Danio rerio) embryos to naphthalene and five HNs at concentrations ranging from 0.25 to 2.0 mg L-1 to assess acute toxicities and developmental effects. Among them, 2-bromomethyl naphthalene (2-BMN) produced moderate lethal effects (96-h LC50 = 1.4 mg L-1) and significantly reduced hatchability. Abnormal phenotypes, including pericardial edema, spine curvature, and shortened body length, were also induced by 2-BMN (96-h EC50 = 0.45 mg L-1). Treatments of 0.5-2.0 mg L-1 2-BMN evoked cardiac malformations via significant down-regulation of the cacna1c gene, which codes the voltage-dependent calcium channel, at 72 hpf and up-regulation of the nppa gene, responsible for the expression of natriuretic peptides, at 96 hpf in zebrafish. One presumable toxic photo-dissociated metabolite of 2-BMN, the 2-naphthylmethyl radical, may be responsible for the toxic effect on zebrafish embryos. HPAHs must be monitored and managed due to their adverse effects on living organisms at low concentrations.
Collapse
Affiliation(s)
- Jungeun Park
- Department of Integrative Biology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yurim Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hwang-Ju Jeon
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kyeongnam Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Chaeeun Kim
- Department of Integrative Biology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Seungki Lee
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea
| | - Jino Son
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea
| | - Sung-Eun Lee
- Department of Integrative Biology, Kyungpook National University, Daegu, 41566, Republic of Korea; Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
31
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
32
|
Thyroid hormones regulate cardiac repolarization and QT-interval related gene expression in hiPSC cardiomyocytes. Sci Rep 2022; 12:568. [PMID: 35022468 PMCID: PMC8755773 DOI: 10.1038/s41598-021-04659-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022] Open
Abstract
Prolongation of cardiac repolarization (QT interval) represents a dangerous and potentially life-threatening electrical event affecting the heart. Thyroid hormones (THs) are critical for cardiac development and heart function. However, little is known about THs influence on ventricular repolarization and controversial effects on QT prolongation are reported. Human iPSC-derived cardiomyocytes (hiPSC-CMs) and multielectrode array (MEA) systems were used to investigate the influence of 3,3',5-triiodo-L-Thyronine (T3) and 3,3',5,5'-tetraiodo-L-Thyronine (T4) on corrected Field Potential Duration (FPDc), the in vitro analog of QT interval, and on local extracellular Action Potential Duration (APD). Treatment with high THs doses induces a significant prolongation of both FPDc and APD, with the strongest increase reached after 24 h exposure. Preincubation with reverse T3 (rT3), a specific antagonist for nuclear TH receptor binding, significantly reduces T3 effects on FPDc, suggesting a TRs-mediated transcriptional mechanism. RNA-seq analysis showed significant deregulation in genes involved in cardiac repolarization pathways, including several QT-interval related genes. In conclusion, long-time administration of high THs doses induces FPDc prolongation in hiPSC-CMs probably through the modulation of genes linked to QT-interval regulation. These results open the way to investigate new potential diagnostic biomarkers and specific targeted therapies for cardiac repolarization dysfunctions.
Collapse
|
33
|
Nakagawa-Tamagawa N, Kirino E, Sugao K, Nagata H, Tagawa Y. Involvement of Calcium-Dependent Pathway and β Subunit-Interaction in Neuronal Migration and Callosal Projection Deficits Caused by the Cav1.2 I1166T Mutation in Developing Mouse Neocortex. Front Neurosci 2021; 15:747951. [PMID: 34955712 PMCID: PMC8692569 DOI: 10.3389/fnins.2021.747951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Gain-of-function mutations in the L-type Ca2+ channel Cav1.2 cause Timothy syndrome (TS), a multisystem disorder associated with neurologic symptoms, including autism spectrum disorder (ASD), seizures, and intellectual disability. Cav1.2 plays key roles in neural development, and its mutation can affect brain development and connectivity through Ca2+-dependent and -independent mechanisms. Recently, a gain-of-function mutation, I1166T, in Cav1.2 was identified in patients with TS-like disorder. Its channel properties have been analyzed in vitro but in vivo effects of this mutation on brain development remain unexplored. Methods:In utero electroporation was performed on ICR mice at embryonic day 15 to express GFP, wild-type, and mutant Cav1.2 channels into cortical layer 2/3 excitatory neurons in the primary somatosensory area. The brain was fixed at postnatal days 14–16, sliced, and scanned using confocal microscopy. Neuronal migration of electroporated neurons was examined in the cortex of the electroporated hemisphere, and callosal projection was examined in the white matter and contralateral hemisphere. Results: Expression of the I1166T mutant in layer 2/3 neurons caused migration deficits in approximately 20% of electroporated neurons and almost completely diminished axonal arborization in the contralateral hemisphere. Axonal projection in the white matter was not affected. We introduced second mutations onto Cav1.2 I1166T; L745P mutation blocks Ca2+ influx through Cav1.2 channels and inhibits the Ca2+-dependent pathway, and the W440A mutation blocks the interaction of the Cav1.2 α1 subunit to the β subunit. Both second mutations recovered migration and projection. Conclusion: This study demonstrated that the Cav1.2 I1166T mutation could affect two critical steps during cerebrocortical development, migration and axonal projection, in the mouse brain. This is mediated through Ca2+-dependent pathway downstream of Cav1.2 and β subunit-interaction.
Collapse
Affiliation(s)
- Nao Nakagawa-Tamagawa
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.,Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Saitama, Japan
| | - Emi Kirino
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kohtaroh Sugao
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Hidetaka Nagata
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Yoshiaki Tagawa
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
34
|
Jiang F, Xu H, Shi X, Han B, Chu Z, Xu B, Liu X. Dynamic Electrocardiogram under P Wave Detection Algorithm Combined with Low-Dose Betaloc in Diagnosis and Treatment of Patients with Arrhythmia after Hepatocarcinoma Resection. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6034180. [PMID: 34697566 PMCID: PMC8541856 DOI: 10.1155/2021/6034180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023]
Abstract
This work aimed to study the diagnostic value of dynamic electrocardiogram (ECG) based on P wave detection algorithm for arrhythmia after hepatectomy in patients with primary liver cancer, and to compare the therapeutic effect of different doses of Betaloc. P wave detection algorithm was introduced for ECG automatic detection and analysis, which can be used for early diagnosis of arrhythmia. Sixty patients with arrhythmia after hepatectomy for primary liver cancer were selected as the research objects. They were randomly divided into control group, SD group, MD group, and HD group, with 15 cases in each group. No Betaloc, low-dose (≤47.5 mg), medium-dose (47.5-95 mg), and high-dose (142.5-190 mg) Betaloc were used for treatment. As a result, P wave detection algorithms can mark P waves that may be submerged in strong interference. P waves from arrhythmia database were used to verify the performance of the proposed algorithm. The prediction precision (Pp) of ventricular arrhythmia and atrial arrhythmia was 98.53% and 98.76%, respectively. Systolic blood pressure (117.35 ± 7.33, 126.44 ± 9.38, and 116.02 ± 8.2) mmHg in SD group, MD group, and HD group was significantly lower than that in control group (140.3 ± 7.21) mmHg after two weeks of treatment. Moreover, those of SD group and HD group were significantly lower than MD group (P < 0.05). The effective rate of cardiac function improvement in SD group (72.35 ± 1.21%) was significantly higher than that in control group, MD group, and HD group (38.2 ± 0.98%, 65.12 ± 1.33%, and 60.43 ± 1.25%; P < 0.05). In short, dynamic ECG based on P wave detection algorithm had high diagnostic value for arrhythmia after hepatectomy in patients with primary liver cancer. It was safe and effective for patients to choose small dose of Betaloc.
Collapse
Affiliation(s)
- Fenfen Jiang
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Haokai Xu
- Department of Surgery, Ningbo Yinzhou No. 2 Hospital, Ningbo 315199, Zhejiang, China
| | - Xiaowen Shi
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Bingjiang Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Zhenliang Chu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Bin Xu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Xiaorong Liu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| |
Collapse
|
35
|
A comparative review on heart ion channels, action potentials and electrocardiogram in rodents and human: extrapolation of experimental insights to clinic. Lab Anim Res 2021; 37:25. [PMID: 34496976 PMCID: PMC8424989 DOI: 10.1186/s42826-021-00102-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/26/2021] [Indexed: 11/10/2022] Open
Abstract
Electrocardiogram (ECG) is a non-invasive valuable diagnostic tool that is used in clinics for investigation and monitoring of heart electrical rhythm/conduction, ischemia/injury of heart, electrolyte disturbances and agents/drugs induced cardiac toxicity. Nowadays using animal models to study heart diseases such as electrical and mechanical disturbance is common. In addition, given to ethical consideration and availability, the use of small rodents has been a top priority for cardiovascular researchers. However, extrapolation of experimental findings from the lab to the clinic needs sufficient basic knowledge of similarities and differences between heart action potential and ECG of rodents and humans in normal and disease conditions. This review compares types of human action potentials, the dominant ion currents during action potential phases, alteration in ion channels activities in channelopathies-induced arrhythmias and the ECG appearance of mouse, rat, guinea pig, rabbit and human. Also, it briefly discusses the responsiveness and alterations in ECG following some interventions such as cardiac injury and arrhythmia induction. Overall, it provides a roadmap for researchers in selecting the best animal model/species whose studies results can be translated into clinical practice. In addition, this study will also be useful to biologists, physiologists, pharmacologists, veterinarians and physicians working in the fields of comparative physiology, pharmacology, toxicology and diseases.
Collapse
|
36
|
Mesirca P, Nakao S, Nissen SD, Forte G, Anderson C, Trussell T, Li J, Cox C, Zi M, Logantha S, Yaar S, Cartensen H, Bidaud I, Stuart L, Soattin L, Morris GM, da Costa Martins PA, Cartwright EJ, Oceandy D, Mangoni ME, Jespersen T, Buhl R, Dobrzynski H, Boyett MR, D'Souza A. Intrinsic Electrical Remodeling Underlies Atrioventricular Block in Athletes. Circ Res 2021; 129:e1-e20. [PMID: 33849278 DOI: 10.1161/circresaha.119.316386] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Pietro Mesirca
- IGF, Université de Montpellier, CNRS, INSERM, France (P.M., I.B., M.E.M.)
| | - Shu Nakao
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
- Department of Biomedical Sciences, Ritsumeikan University, Japan (S.N.)
| | - Sarah Dalgas Nissen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences (S.D.N., H.C., R.B.), University of Copenhagen, Denmark
| | - Gabriella Forte
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Cali Anderson
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Tariq Trussell
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Jue Li
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Charlotte Cox
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Min Zi
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Sunil Logantha
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
- Liverpool Centre for Cardiovascular Sciences, University of Liverpool, United Kingdom (S.L.)
| | - Sana Yaar
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Helena Cartensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences (S.D.N., H.C., R.B.), University of Copenhagen, Denmark
| | - Isabelle Bidaud
- IGF, Université de Montpellier, CNRS, INSERM, France (P.M., I.B., M.E.M.)
| | - Luke Stuart
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | | | - Gwilym M Morris
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | | | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| | - Matteo E Mangoni
- IGF, Université de Montpellier, CNRS, INSERM, France (P.M., I.B., M.E.M.)
| | - Thomas Jespersen
- Department of Biomedical Sciences (T.J., M.R.B.), University of Copenhagen, Denmark
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences (S.D.N., H.C., R.B.), University of Copenhagen, Denmark
| | - Halina Dobrzynski
- Department of Anatomy, Jagiellonian University Medical College, Poland (H.D.)
| | - Mark R Boyett
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
- Department of Biomedical Sciences (T.J., M.R.B.), University of Copenhagen, Denmark
| | - Alicia D'Souza
- Division of Cardiovascular Sciences, University of Manchester, United Kingdom (S.N., G.F., C.A., T.T., J.L., C.C., M.Z., S.L., S.Y., L. Stuart, L. Soattin, G.M.M., E.J.C., D.O., H.D., M.R.B., A.D.)
| |
Collapse
|
37
|
Striessnig J. Voltage-Gated Ca 2+-Channel α1-Subunit de novo Missense Mutations: Gain or Loss of Function - Implications for Potential Therapies. Front Synaptic Neurosci 2021; 13:634760. [PMID: 33746731 PMCID: PMC7966529 DOI: 10.3389/fnsyn.2021.634760] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
This review summarizes our current knowledge of human disease-relevant genetic variants within the family of voltage gated Ca2+ channels. Ca2+ channelopathies cover a wide spectrum of diseases including epilepsies, autism spectrum disorders, intellectual disabilities, developmental delay, cerebellar ataxias and degeneration, severe cardiac arrhythmias, sudden cardiac death, eye disease and endocrine disorders such as congential hyperinsulinism and hyperaldosteronism. A special focus will be on the rapidly increasing number of de novo missense mutations identified in the pore-forming α1-subunits with next generation sequencing studies of well-defined patient cohorts. In contrast to likely gene disrupting mutations these can not only cause a channel loss-of-function but can also induce typical functional changes permitting enhanced channel activity and Ca2+ signaling. Such gain-of-function mutations could represent therapeutic targets for mutation-specific therapy of Ca2+-channelopathies with existing or novel Ca2+-channel inhibitors. Moreover, many pathogenic mutations affect positive charges in the voltage sensors with the potential to form gating-pore currents through voltage sensors. If confirmed in functional studies, specific blockers of gating-pore currents could also be of therapeutic interest.
Collapse
Affiliation(s)
- Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
38
|
Changes in Ion Selectivity Following the Asymmetrical Addition of Charge to the Selectivity Filter of Bacterial Sodium Channels. ENTROPY 2020; 22:e22121390. [PMID: 33316962 PMCID: PMC7764494 DOI: 10.3390/e22121390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/30/2022]
Abstract
Voltage-gated sodium channels (NaVs) play fundamental roles in eukaryotes, but their exceptional size hinders their structural resolution. Bacterial NaVs are simplified homologues of their eukaryotic counterparts, but their use as models of eukaryotic Na+ channels is limited by their homotetrameric structure at odds with the asymmetric Selectivity Filter (SF) of eukaryotic NaVs. This work aims at mimicking the SF of eukaryotic NaVs by engineering radial asymmetry into the SF of bacterial channels. This goal was pursued with two approaches: the co-expression of different monomers of the NaChBac bacterial channel to induce the random assembly of heterotetramers, and the concatenation of four bacterial monomers to form a concatemer that can be targeted by site-specific mutagenesis. Patch-clamp measurements and Molecular Dynamics simulations showed that an additional gating charge in the SF leads to a significant increase in Na+ and a modest increase in the Ca2+ conductance in the NavMs concatemer in agreement with the behavior of the population of random heterotetramers with the highest proportion of channels with charge -5e. We thus showed that charge, despite being important, is not the only determinant of conduction and selectivity, and we created new tools extending the use of bacterial channels as models of eukaryotic counterparts.
Collapse
|
39
|
Varró A, Tomek J, Nagy N, Virág L, Passini E, Rodriguez B, Baczkó I. Cardiac transmembrane ion channels and action potentials: cellular physiology and arrhythmogenic behavior. Physiol Rev 2020; 101:1083-1176. [PMID: 33118864 DOI: 10.1152/physrev.00024.2019] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electrophysiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the first part of this review, current knowledge on the differences in ion channel expression and properties of the ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynamics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed. The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult. Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
40
|
Fochi S, Lorenzi P, Galasso M, Stefani C, Trabetti E, Zipeto D, Romanelli MG. The Emerging Role of the RBM20 and PTBP1 Ribonucleoproteins in Heart Development and Cardiovascular Diseases. Genes (Basel) 2020; 11:genes11040402. [PMID: 32276354 PMCID: PMC7230170 DOI: 10.3390/genes11040402] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022] Open
Abstract
Alternative splicing is a regulatory mechanism essential for cell differentiation and tissue organization. More than 90% of human genes are regulated by alternative splicing events, which participate in cell fate determination. The general mechanisms of splicing events are well known, whereas only recently have deep-sequencing, high throughput analyses and animal models provided novel information on the network of functionally coordinated, tissue-specific, alternatively spliced exons. Heart development and cardiac tissue differentiation require thoroughly regulated splicing events. The ribonucleoprotein RBM20 is a key regulator of the alternative splicing events required for functional and structural heart properties, such as the expression of TTN isoforms. Recently, the polypyrimidine tract-binding protein PTBP1 has been demonstrated to participate with RBM20 in regulating splicing events. In this review, we summarize the updated knowledge relative to RBM20 and PTBP1 structure and molecular function; their role in alternative splicing mechanisms involved in the heart development and function; RBM20 mutations associated with idiopathic dilated cardiovascular disease (DCM); and the consequences of RBM20-altered expression or dysfunction. Furthermore, we discuss the possible application of targeting RBM20 in new approaches in heart therapies.
Collapse
|
41
|
Nam JM, Lim JE, Ha TW, Oh B, Kang JO. Cardiac-specific inactivation of Prdm16 effects cardiac conduction abnormalities and cardiomyopathy-associated phenotypes. Am J Physiol Heart Circ Physiol 2020; 318:H764-H777. [PMID: 32083975 DOI: 10.1152/ajpheart.00647.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A variant in the PRDM16 locus has been correlated with QRS duration in an electrocardiogram genome-wide association study, and the deletion of PRDM16 has been implicated as a causal factor of the dilated cardiomyopathy that is linked to 1p36 deletion syndrome. We aimed to determine how a null mutation of Prdm16 affects cardiac function and study the underlying mechanism of the resulting phenotype in an appropriate mouse model. We used cardiac-specific Prdm16 conditional knockout mice to examine cardiac function by electrocardiography. QRS duration and QTc interval increased significantly in cardiac-specific Prdm16 knockout animals compared with wild-type mice. Further, we assessed cardiomyopathy-associated features by trichrome staining, densitometry, and hydroxyproline assay. Prdm16-null hearts showed greater fibrosis and cardiomyocyte hypertrophy. By quantitative real-time PCR, Prdm16-null hearts upregulated extracellular matrix-related genes (Ctgf, Timp1) and α-smooth muscle actin (Acta2), a myofibroblast marker. Moreover, TGF-β signaling was activated in Prdm16-null hearts, as evidenced by increased Tgfb1-3 transcript levels and phosphorylated Smad2. However, the inhibition of TGF-β receptor did not reverse the aberrations in conduction in cardiac-specific Prdm16 knockout mice. To determine the underlying mechanisms, we performed RNA-seq using mouse left ventricular tissue. By functional analysis, Prdm16-null hearts experienced dysregulated expression of ion channel genes, including Kcne1, Scn5a, Cacna1h, and Cacna2d2. Mice with Prdm16-null hearts develop abnormalities in cardiac conduction and cardiomyopathy-associated phenotypes, including fibrosis and cellular hypertrophy. Further, the RNA-seq findings suggest that impairments in ion homeostasis (Ca2+, K+, and Na+) may at least partially underlie the abnormal conduction in cardiac-specific Prdm16 knockout mice.NEW & NOTEWORTHY This is the first study that describes aberrant cardiac function and cardiomyopathy-associated phenotypes in an appropriate murine genetic model with cardiomyocyte-specific Prdm16-null mutation. It is noteworthy that the correlation of PRDM16 with QRS duration is replicated in a murine animal model and the potential underlying mechanism may be the impairment of ion homeostasis.
Collapse
Affiliation(s)
- Jeong Min Nam
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Ji Eun Lim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Tae Woong Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Bermseok Oh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Ji-One Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
42
|
Ragab AAY, Sitorus GDS, Brundel BBJJM, de Groot NMS. The Genetic Puzzle of Familial Atrial Fibrillation. Front Cardiovasc Med 2020; 7:14. [PMID: 32118049 PMCID: PMC7033574 DOI: 10.3389/fcvm.2020.00014] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/28/2020] [Indexed: 12/17/2022] Open
Abstract
Atrial fibrillation (AF) is the most common clinical tachyarrhythmia. In Europe, AF is expected to reach a prevalence of 18 million by 2060. This estimate will increase hospitalization for AF to 4 million and 120 million outpatient visits. Besides being an independent risk factor for mortality, AF is also associated with an increased risk of morbidities. Although there are many well-defined risk factors for developing AF, no identifiable risk factors or cardiac pathology is seen in up to 30% of the cases. The heritability of AF has been investigated in depth since the first report of familial atrial fibrillation (FAF) in 1936. Despite the limited value of animal models, the advances in molecular genetics enabled identification of many common and rare variants related to FAF. The importance of AF heritability originates from the high prevalence of lone AF and the lack of clear understanding of the underlying pathophysiology. A better understanding of FAF will facilitate early identification of people at high risk of developing FAF and subsequent development of more effective management options. In this review, we reviewed FAF epidemiological studies, identified common and rare variants, and discussed their clinical implications and contributions to developing new personalized therapeutic strategies.
Collapse
Affiliation(s)
- Ahmed A Y Ragab
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Gustaf D S Sitorus
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Bianca B J J M Brundel
- Department of Physiology, Institute for Cardiovascular Research, VU Medical Center, Amsterdam, Netherlands
| | - Natasja M S de Groot
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
43
|
Li Q, Zhai Z, Li J. Fibroblast growth factor homologous factors are potential ion channel modifiers associated with cardiac arrhythmias. Eur J Pharmacol 2020; 871:172920. [PMID: 31935396 DOI: 10.1016/j.ejphar.2020.172920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/10/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022]
Abstract
Stable electrical activity in cardiac myocytes is the basis of maintaining normal myocardial systolic and diastolic function. Cardiac ionic currents and their associated regulatory proteins are crucial to myocyte excitability and heart function. Fibroblast growth factor homologous factors (FHFs) are intracellular noncanonical fibroblast growth factors (FGFs) that are incapable of activating FGF receptors. The main functions of FHFs are to regulate ion channels and influence excitability, which are processes involved in sustaining normal cardiac function. In addition to their regulatory effect on ion channels, FHFs can be regulators of cardiac hypertrophic signaling and alter signaling pathways, including the protein kinase, NF<kappa>B, and p53 pathways, which are related to the pathological processes of heart diseases. This review emphasizes FHF-mediated regulation of cardiac excitability and the association of FHFs with cardiac arrhythmias and explores the idea that abnormal FHFs may be an unrecognized cause of cardiac disorders.
Collapse
Affiliation(s)
- Qing Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhenyu Zhai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
44
|
Dystrophin and calcium current are decreased in cardiomyocytes expressing Cre enzyme driven by αMHC but not TNT promoter. Sci Rep 2019; 9:19422. [PMID: 31857666 PMCID: PMC6923407 DOI: 10.1038/s41598-019-55950-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
The Cre/lox system is a potent technology to control gene expression in mouse tissues. However, cardiac-specific Cre recombinase expression alone can lead to cardiac alterations when no loxP sites are present, which is not well understood. Many loxP-like sites have been identified in the mouse genome that might be Cre sensitive. One of them is located in the Dmd gene encoding dystrophin, a protein important for the function and stabilization of voltage-gated calcium (Cav1.2) and sodium (Nav1.5) channels, respectively. Here, we investigate whether Cre affects dystrophin expression and function in hearts without loxP sites in the genome. In mice expressing Cre under the alpha-myosin heavy chain (MHC-Cre) or Troponin T (TNT-Cre) promoter, we investigated dystrophin expression, Nav1.5 expression, and Cav1.2 function. Compared to age-matched MHC-Cre- mice, dystrophin protein level was significantly decreased in hearts from MHC-Cre+ mice of more than 12-weeks-old. Quantitative RT-PCR revealed decreased mRNA levels of Dmd gene. Unexpectedly, calcium current (ICaL), but not Nav1.5 protein expression was altered in those mice. Surprisingly, in hearts from 12-week-old and older TNT-Cre+ mice, neither ICaL nor dystrophin and Nav1.5 protein content were altered compared to TNT-Cre-. Cre recombinase unpredictably affects cardiac phenotype, and Cre-expressing mouse models should be carefully investigated before experimental use.
Collapse
|
45
|
Kushner J, Ferrer X, Marx SO. Roles and Regulation of Voltage-gated Calcium Channels in Arrhythmias. J Innov Card Rhythm Manag 2019; 10:3874-3880. [PMID: 32494407 PMCID: PMC7252866 DOI: 10.19102/icrm.2019.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 03/04/2019] [Indexed: 12/25/2022] Open
Abstract
Calcium flowing through voltage-dependent calcium channels into cardiomyocytes mediates excitation–contraction coupling, controls action-potential duration and automaticity in nodal cells, and regulates gene expression. Proper surface targeting and basal and hormonal regulation of calcium channels are vital for normal cardiac physiology. In this review, we discuss the roles of voltage-gated calcium channels in the heart and the mechanisms by which these channels are regulated by physiological signaling pathways in health and disease.
Collapse
Affiliation(s)
- Jared Kushner
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Xavier Ferrer
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
46
|
Stanley CE, Mauss AS, Borst A, Cooper RL. The Effects of Chloride Flux on Drosophila Heart Rate. Methods Protoc 2019; 2:mps2030073. [PMID: 31443492 PMCID: PMC6789470 DOI: 10.3390/mps2030073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 11/16/2022] Open
Abstract
Approaches are sought after to regulate ionotropic and chronotropic properties of the mammalian heart. Electrodes are commonly used for rapidly exciting cardiac tissue and resetting abnormal pacing. With the advent of optogenetics and the use of tissue-specific expression of light-activated channels, cardiac cells cannot only be excited but also inhibited with ion-selective conductance. As a proof of concept for the ability to slow down cardiac pacing, anion-conducting channelrhodopsins (GtACR1/2) and the anion pump halorhodopsin (eNpHR) were expressed in hearts of larval Drosophila and activated by light. Unlike body wall muscles in most animals, the equilibrium potential for Cl− is more positive as compared to the resting membrane potential in larval Drosophila. As a consequence, upon activating the two forms of GtACR1 and 2 with low light intensity the heart rate increased, likely due to depolarization and opening of voltage-gated Ca2+ channels. However, with very intense light activation the heart rate ceases, which may be due to Cl– shunting to the reversal potential for chloride. Activating eNpHR hyperpolarizes body wall and cardiac muscle in larval Drosophila and rapidly decreases heart rate. The decrease in heart rate is related to light intensity. Intense light activation of eNpHR stops the heart from beating, whereas lower intensities slowed the rate. Even with upregulation of the heart rate with serotonin, the pacing of the heart was slowed with light. Thus, regulation of the heart rate in Drosophila can be accomplished by activating anion-conducting channelrhodopsins using light. These approaches are demonstrated in a genetically amenable insect model.
Collapse
Affiliation(s)
- Catherine E Stanley
- Department of Biology, Center for Muscle Biology. University of Kentucky, Lexington, KY 40506-0225, USA
| | - Alex S Mauss
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Alexander Borst
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Robin L Cooper
- Department of Biology, Center for Muscle Biology. University of Kentucky, Lexington, KY 40506-0225, USA.
| |
Collapse
|
47
|
Qu YS, Lazzerini PE, Capecchi PL, Laghi-Pasini F, El Sherif N, Boutjdir M. Autoimmune Calcium Channelopathies and Cardiac Electrical Abnormalities. Front Cardiovasc Med 2019; 6:54. [PMID: 31119135 PMCID: PMC6507622 DOI: 10.3389/fcvm.2019.00054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/16/2019] [Indexed: 12/24/2022] Open
Abstract
Patients with autoimmune diseases are at increased risk for developing cardiovascular diseases, and abnormal electrocardiographic findings are common. Voltage-gated calcium channels play a major role in the cardiovascular system and regulate cardiac excitability and contractility. Particularly, by virtue of their localization and expression in the heart, calcium channels modulate pace making at the sinus node, conduction at the atrioventricular node and cardiac repolarization in the working myocardium. Consequently, emerging evidence suggests that calcium channels are targets to autoantibodies in autoimmune diseases. Autoimmune-associated cardiac calcium channelopathies have been recognized in both sinus node dysfunction atrioventricular block in patients positive for anti-Ro/La antibodies, and ventricular arrhythmias in patients with dilated cardiomyopathy. In this review, we discuss mechanisms of autoimmune-associated calcium channelopathies and their relationship with the development of cardiac electrical abnormalities.
Collapse
Affiliation(s)
- Yongxia Sarah Qu
- Department of Cardiology, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY, United States.,VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Nabil El Sherif
- VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Mohamed Boutjdir
- VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States.,NYU School of Medicine, New York, NY, United States
| |
Collapse
|
48
|
Dehghani-Samani A, Madreseh-Ghahfarokhi S, Dehghani-Samani A. Mutations of Voltage-Gated Ionic Channels and Risk of Severe Cardiac Arrhythmias. ACTA CARDIOLOGICA SINICA 2019; 35:99-110. [PMID: 30930557 PMCID: PMC6434417 DOI: 10.6515/acs.201903_35(2).20181028a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 10/28/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Arrhythmias as important malfunctions of heart are known as abnormal rhythm of heart. Several causes can make arrhythmias and most of them are related to generation and/or conduction of action potential in heart. Action potential in myocytes results from the sequential opening and closing of ion channel proteins that span the plasma membrane of individual myocytes. Action potential's conduction through the heart is depended on electrical coupling between myocytes, which is mediated by gap junctions. Generation and conduction of action potentials are related to perfect action of ionic channels in heart. OBJECTIVES This novel review comprehensively addressed the ionic mechanisms of the arrhythmogenic mutations in cardiac voltage-gated ionic channels including: CACNA1C, CACNA1D, KCNA5, KCND2, KCND3, KCNE1, KCNE2, KCNE5, KCNH2, KCNJ2, KCNJ5, KCNQ1, SCN4A, SCN5A, SCN1B, SCN2B, SCN3B and SCN4B. METHODS Current study, for the first time, review and discuses about relation between cardiac arrhythmias and whole of important voltage gated ionic channels from different families, altogether and at the same time. RESULTS This review clears that mutations in voltage-gated ionic channels play important roles in generation of severe cardiac arrhythmias, and among them it is looked that mutations in voltage-gated potassium channels are more important. CONCLUSIONS Most of induced arrhythmias due to voltage-gated ionic channels mutations result in action potentials prolongation and long QT syndromes. Study on ionic channel regulators can be considered as a subject for future research.
Collapse
Affiliation(s)
- Amir Dehghani-Samani
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord
| | - Samin Madreseh-Ghahfarokhi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad
| | | |
Collapse
|