1
|
Liebich A, Sheikh G, Bundschuh RA, Kircher M, Dierks A, Nittbaur B, Raake P, Rieger M, Higuchi T, Pfob CH, Lapa C. Cardiac risk in recovered Covid-19 patients evaluated by 123I-mIBG. Sci Rep 2025; 15:18039. [PMID: 40410298 PMCID: PMC12102354 DOI: 10.1038/s41598-025-02212-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
To determine whether cardiac sympathetic nervous dysfunction is present, in this single center prospective, non-randomized trial non-invasive SPECT/CT imaging using the radiotracer 123I-metaiodobenzylguanidine was performed in 33 recovered COVID-19 patients without pre-existing cardiac conditions. Increased cardiac sympathetic activity, as indicated by late HMR, was observed in 67.7% of patients. At 6-8 months, 82% of these subjects (27/33) received follow-up, and cardiac sympathetic innervation abnormalities were still present in 70.4% (19/27). Additionally, at 12-15 months post-diagnosis, persistently abnormal HMRs were found in 9 individuals who initially had abnormal sympathetic innervation. Further follow-up is needed to investigate potential long-term cardiovascular consequences of COVID-19.
Collapse
Affiliation(s)
- Alessandro Liebich
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.
- Klinik für Nuklearmedizin, Universitätsklinikum Augsburg, Stenglinstraße 2, 86156, Augsburg, Bavaria, Germany.
| | - Gabriel Sheikh
- Department of Nuclear Medicine, LMU Munich, Munich, Germany
| | - Ralph A Bundschuh
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Malte Kircher
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Alexander Dierks
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Bernd Nittbaur
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Philip Raake
- Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | - Takahiro Higuchi
- Comprehensive Heart Failure Center (CHFC) and Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Christian H Pfob
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
2
|
Ziegler KA, Engelhardt S, Carnevale D, McAlpine CS, Guzik TJ, Dimmeler S, Swirski FK. Neural Mechanisms in Cardiovascular Health and Disease. Circ Res 2025; 136:1233-1261. [PMID: 40403111 DOI: 10.1161/circresaha.125.325580] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 05/24/2025]
Abstract
Although the neurocardiac axis is central to cardiovascular homeostasis, its dysregulation drives heart failure and cardiometabolic diseases. This review examines the bidirectional interplay between the autonomic nervous system and the heart, highlighting the role of this interplay in disease progression and its therapeutic potential. The autonomic nervous system modulates cardiac function and vascular tone through its sympathetic and parasympathetic branches. However, in heart failure, chronic sympathetic overdrive and parasympathetic withdrawal exacerbate myocardial remodeling and metabolic dysfunction, both of which are exacerbated by cardiometabolic conditions such as obesity and diabetes. These conditions are increasingly recognized to impair neurocardiac regulation, thereby promoting inflammation and adverse outcomes. An important emerging area concerns neuroimmune control, in which the brain orchestrates systemic inflammation through circuits involving the bone marrow, spleen, and other organs, thereby amplifying cardiovascular damage. This neuroimmune axis integrates peripheral signals to influence immune responses that contribute to disease progression. Lifestyle factors, such as stress, sleep, exercise, and diet, affect autonomic and immune balance and, thus, cardiovascular disease. Therapeutically, targeting neurocardiac and neuroimmune pathways pharmacologically or via neuromodulation (eg, vagal or splenic nerve stimulation) offers promise although the clinical translation of the latter remains challenging. In this review, we synthesize preclinical and clinical data to highlight the neurocardiac axis as a critical nexus in heart failure and cardiometabolic disease. Harnessing neuroimmune and neurocardiac interactions may inform precision approaches to reduce the burden of these conditions.
Collapse
Affiliation(s)
- Karin A Ziegler
- Institute of Pharmacology and Toxicology, School of Medicine and Health, Technical University of Munich, Germany (K.A.Z., S.E.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (K.A.Z., S.E.)
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, School of Medicine and Health, Technical University of Munich, Germany (K.A.Z., S.E.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (K.A.Z., S.E.)
| | - Daniela Carnevale
- Faculty of Pharmacy and Medicine, Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (D.C.)
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli, Italy (D.C.)
| | - Cameron S McAlpine
- Cardiovascular Research Institute, The Friedman Brain Institute, and Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.S.M., F.K.S.)
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, The University of Edinburgh, United Kingdom (T.J.G.)
- Department of Internal Medicine (T.J.G.), Jagiellonian University Medical College, Kraków, Poland
- Center for Medical Genomics OMICRON (T.J.G.), Jagiellonian University Medical College, Kraków, Poland
| | - Stefanie Dimmeler
- Goethe University Frankfurt, Institute for Cardiovascular Regeneration, Germany (S.D.)
- German Centre for Cardiovascular Research (DZHK), Frankfurt am Main, Germany (S.D.)
- Cardiopulmonary Institute, Goethe University Frankfurt am Main, Germany (S.D.)
| | - Filip K Swirski
- Cardiovascular Research Institute, The Friedman Brain Institute, and Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.S.M., F.K.S.)
| |
Collapse
|
3
|
Velmeden D, Söhne J, Schuch A, Zeid S, Schulz A, Troebs SO, Müller F, Heidorn MW, Buch G, Belanger N, Dinh W, Mondritzki T, Lackner KJ, Gori T, Münzel T, Wild PS, Prochaska JH. Role of Heart Rate Recovery in Chronic Heart Failure: Results From the MyoVasc Study. J Am Heart Assoc 2025; 14:e039792. [PMID: 40371587 DOI: 10.1161/jaha.124.039792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/11/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Cardiac autonomic dysfunction is associated with heart failure (HF). Reduced heart rate recovery (HRR) indicates impaired parasympathetic reactivation after physical activity. Heart rate recovery 60 seconds after peak effort (HRR60) is linked to autonomic dysfunction, but data on its relevance across HF phenotypes are scarce. This study aimed to identify clinical determinants of HRR60 in an HF cohort and assess its relationship with clinical outcomes. METHODS Data from the MyoVasc study (NCT04064450; N=3289) were analyzed. Participants underwent standardized clinical phenotyping including cardiopulmonary exercise testing. HRR60 was defined as the heart rate decline 60 seconds after exercise termination. Clinical determinants of HRR60 were evaluated using multivariate regression, whereas Cox regression analyses assessed all-cause death and worsening of HF. RESULTS The analysis sample comprised 1289 individuals (median age, 66.0 [interquartile range {IQR}, 58.0-73.0] years, 30.4% women) ranging from stage B to stage C/D according to the universal definition of HF. Age, sex, smoking, obesity, peripheral artery disease, and chronic kidney disease were identified as determinants of HRR60. HRR60 showed a strong association with all-cause death (hazard ratio [HR]HRR60 [10 bpm], 1.56 [95% CI, 1.32-1.85]; P<0.0001) and worsening of HF (HRHRR60 [10 bpm], 1.36 [95% CI, 1.10-1.69]; P=0.0052) independent of age, sex, and clinical profile. Sensitivity analysis showed a stronger association with worsening HF in HF with preserved left ventricular ejection fraction (Pinteraction=0.027). CONCLUSIONS HRR60 was associated with clinical outcome in chronic HF. Because it showed a stronger association with outcomes in HF with preserved ejection fraction, future research should consider phenotype-specific differences.
Collapse
Affiliation(s)
- David Velmeden
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Jakob Söhne
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Alexander Schuch
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Silav Zeid
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Sven-Oliver Troebs
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
| | - Felix Müller
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Marc W Heidorn
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Gregor Buch
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI) University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Noémie Belanger
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
| | - Wilfried Dinh
- Bayer AG, Research and Development, Translational Clinical Medicine, Experimental Medicine 1 Wuppertal Germany
- School of Medicine University Witten/Herdecke Witten Germany
| | - Thomas Mondritzki
- Research & Early Development, Clinical Experimentation CV, BAYER AG Wuppertal Germany
| | - Karl J Lackner
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Institute for Clinical Chemistry and Laboratory Medicine University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Tommaso Gori
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Thomas Münzel
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Department of Cardiology - Cardiology I University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH) University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- Institute for Molecular Biology (IMB), Mainz, Working Group Systems Medicine Mainz Germany
| | - Jürgen H Prochaska
- Preventive Cardiology and Preventive Medicine, Department of Cardiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main Mainz Germany
- Clinical Epidemiology and Systems Medicine, Center for Thrombosis and Hemostasis (CTH) University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
| |
Collapse
|
4
|
Hanna P, Hoover DB, Kirkland LG, Smith EH, Poston MD, Peirce SG, Garbe CG, Cha S, Mori S, Brennan JA, Armour JA, Rytkin E, Efimov IR, Ajijola OA, Ardell JL, Shivkumar K. Noradrenergic and cholinergic innervation of the normal human heart and changes associated with cardiomyopathy. Anat Rec (Hoboken) 2025. [PMID: 40365781 DOI: 10.1002/ar.25686] [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: 01/08/2025] [Revised: 04/14/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025]
Abstract
Autonomic nerves are crucial in cardiac function and pathology. However, data on the distribution of cholinergic and noradrenergic nerves in normal and pathologic human hearts is lacking. Nonfailing donor hearts were pressure-perfusion fixed, imaged, and dissected. Left ventricular cardiomyopathy samples were also obtained. Fixed frozen sections were immunostained for nerves, and adjacent tissue underwent clearing for 3D visualization. Cholinergic and noradrenergic nerves were evenly abundant in both atria, except the sinoatrial node, where vesicular acetylcholine transporter (VAChT) nerves were dominant. Noradrenergic consistently outnumbered cholinergic nerves in right (RV) and left ventricular (LV) regions. Noradrenergic innervation of LV regions varied between donors. Cholinergic innervation was higher in RV compared to LV samples, which generally had reduced VAChT nerves. Marked neural remodeling occurred in three cardiomyopathy cases. Tyrosine hydroxylase (TH) nerve density was increased in the right atrial appendage, and all nerves showed a trend to decrease in the left atrial appendage. Cholinergic innervation was reduced in the LV, and TH innervation was heterogeneous. Noradrenergic nerves were present in granulation tissue but absent in regions of dense scar. Some border zone regions had reduced TH innervation but no hyperinnervation. Dual innervation of most atrial regions supports balanced regulation of atrial function. Higher cholinergic input to the sinoatrial node favors vagal dominance in heart rate regulation. Innervation patterns support a significant role of noradrenergic input to the ventricle, especially on the left. Both atrial and ventricular nerves remodel in cardiomyopathy, providing a foundation for asymmetric neural input and dysregulation of cardiac electromechanical function.
Collapse
Affiliation(s)
- Peter Hanna
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Donald B Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee, USA
| | - Logan G Kirkland
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Elizabeth H Smith
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Megan D Poston
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Stanley G Peirce
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Chloe G Garbe
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Steven Cha
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Shumpei Mori
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Jaclyn A Brennan
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - John Andrew Armour
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Eric Rytkin
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
- Department of Medicine (Cardiology), Northwestern University, Chicago, Illinois, USA
| | - Olujimi A Ajijola
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Jeffrey L Ardell
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Kalyanam Shivkumar
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
5
|
Shi P, Liu Y, Sha Y, Wang J, Zhou J, Liu K, Cao Y, Zhang Q, Wang X, Sun H. Wireless, battery-free vagal electrical stimulation: A novel approach to inhibit cardiac hypertrophy via H3K18 lactylation mediated mitophagy. Pharmacol Res 2025; 216:107760. [PMID: 40320225 DOI: 10.1016/j.phrs.2025.107760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
Electrical stimulation (ES) has been established as a reliable and beneficial approach in therapeutic rehabilitation, exhibiting negligible side effects. Nevertheless, research focusing on the application of ES for cardiac hypertrophy remains limited, as it fails to provide an enduring remedy for chronic diseases. In this investigation, vagal ES, characterized by its wireless, battery-free, and fully implantable nature, was utilized to treat cardiac hypertrophy. The vagus nerve at the stimulation site was carefully embedded within an envelope, sealed securely using multiple bioabsorbable sutures. Subsequently, a cardiac hypertrophy model was induced in rats via abdominal aortic coarctation for four weeks. The findings of this investigation demonstrated that ES markedly attenuated cardiac hypertrophy. Metabolomic analysis revealed a notable reduction in lactate levels within myocardial tissue following ES. Proteomic analysis of myocardial tissues indicated a substantial decrease in the expression of autophagy and mitophagy-related proteins after ES. Additionally, ChIP-seq result revealed a specific binding interaction between H3K18 lactylation (H3K18la) and BCL2 interacting protein 3 (Bnip3), while luciferase reporter assays demonstrated that H3K18la directly governed Bnip3 transcriptional activation, exploring its role in modulating mitophagy. Mechanistically, it was shown that ES reduced lactate accumulation through the upregulation of monocarboxylate transporter 4 (MCT4) by decreasing norepinephrine (NE) levels. Furthermore, ES reversed cardiac hypertrophy by diminishing H3K18la levels, thus inhibiting Bnip3 protein expression. This pathway assists in diminishing cardiac hypertrophy, emphasizing the critical involvement of the afferent vagal pathway in regulating cardiac hypertrophy.
Collapse
Affiliation(s)
- Pilong Shi
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Yang Liu
- Department of Basic Nursing, Harbin Medical University, Heilongjiang 163319, China
| | - Yuetong Sha
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Jiaxin Wang
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Jiajun Zhou
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Kai Liu
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Qianhui Zhang
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Xinran Wang
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University, Heilongjiang 163319, China.
| |
Collapse
|
6
|
Heinonen I. Cardiac output limits maximal oxygen consumption, but what limits maximal cardiac output? Exp Physiol 2025; 110:666-674. [PMID: 40193294 PMCID: PMC12053887 DOI: 10.1113/ep091594] [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: 01/15/2024] [Accepted: 12/12/2024] [Indexed: 04/09/2025]
Abstract
Maximal oxygen uptake/consumption is an important variable determining exercise performance. It is generally considered to be limited largely, but not exclusively, by maximal cardiac output (CO), which limits the ability of heart to pump oxygen-rich arterial blood to working muscles. Cardiac output is a product of heart rate and stroke volume, which is the amount of blood ejected from the heart by one heart beat. Exercise training, especially of the endurance type, can increase maximal CO substantially. A straightforward way for the heart to increase maximal CO would be to increase maximal heart rate, but this does not happen; instead, maximal heart rate tends to be reduced after training. This is because heart rate is the most important determinant of myocardial oxygen consumption, and ventricular filling and myocardial blood flow (MBF) would be compromised by further increases in heart rate, given that MBF is blunted by contractions and occurs principally during diastole. Myocardial oxygen extraction is already high at rest and is increased further in endurance-trained athletes, making their hearts even more dependent on increases in MBF. The trained heart therefore also shows reduced MBF, enhanced blood mean transit time and higher myocardial vascular resistance at rest and during submaximal exercise, although MBF reserve is not improved. It follows logically that MBF is an important determinant of myocardial performance, and it is proposed in this review that cardiac afferent sensory nerves might contribute to controlling and limiting heart rate, hence maximal CO, in order to protect the heart from ischaemia.
Collapse
Affiliation(s)
- Ilkka Heinonen
- Turku PET CentreUniversity of Turku and Turku University HospitalTurkuFinland
| |
Collapse
|
7
|
Chen T, Yang Y, Shi K, Si F, Wen Y, Yang X. The developments and emerging trends of Autonomic Nervous System Research in Arrhythmia: a bibliometric study from 2004 to 2024. Front Neurosci 2025; 19:1595253. [PMID: 40356702 PMCID: PMC12066699 DOI: 10.3389/fnins.2025.1595253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
The role of the autonomic nervous system in cardiovascular diseases has increasingly attracted the attention of researchers. This study aims to review research on the autonomic nervous system in arrhythmias from 2004 to 2024, with a focus on understanding the development trends in this field. Data for this study were sourced from the Web of Science Core Collection. We constructed and analyzed bibliometric visualizations related to publication trends, countries/regions, institutions, journals, research categories, themes, references, and keywords. Over the past two decades, academic output related to the autonomic nervous system's role in arrhythmias has grown, although global research distribution remains uneven. The United States leads in publication volume and is home to many high-output institutions, providing it with significant academic influence and fostering international collaboration. By summarizing high-citation literature, clustering keywords, and performing a "burst detection" analysis of keywords, we identified that the mechanisms and assessment methods for autonomic nervous system regulation are major research focuses. Recent hotspots include the psychopathology related to the autonomic nervous system and autonomic regulation therapies. As the biomedical field shifts toward precision medicine, future research trends are likely to focus on identifying precise biomarkers for assessing autonomic nervous system function and developing novel strategies to regulate it. These strategies may include correcting immune dysfunction, psychological interventions, and surgical treatments. This study suggests that ganglionated plexi ablation may represent the most transformative intervention strategy for the Autonomic Nervous System currently available, and highlights electrodermal activity as an evaluation index with considerable potential for widespread application.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Pediatric Cardiology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanfeng Yang
- Department of Pediatric Cardiology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Kun Shi
- Department of Pediatric Cardiology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Feifei Si
- Department of Pediatric Cardiology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yizhou Wen
- Department of Pediatric Cardiology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Yang
- Department of Obstetrics, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
| |
Collapse
|
8
|
Li YL, Li Y, Tu H, Evans AJ, Patel TA, Zheng H, Patel KP. Stellate Ganglia: A Key Therapeutic Target for Malignant Ventricular Arrhythmia in Heart Disease. Circ Res 2025; 136:1049-1069. [PMID: 40273204 PMCID: PMC12026290 DOI: 10.1161/circresaha.124.325384] [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] [Indexed: 04/26/2025]
Abstract
Malignant ventricular arrhythmias (VAs), such as ventricular tachycardia and ventricular fibrillation, are the cause of approximately half a million deaths per year in the United States, which is a common lethal event in heart disease, such as hypertension, catecholaminergic polymorphic ventricular tachycardia, takotsubo cardiomyopathy, long-QT syndrome, and progressing into advanced heart failure. A common characteristic of these heart diseases, and the subsequent development of VAs, is the overactivation of the sympathetic nervous system. Current treatments for VAs in these heart diseases, such as β-adrenergic receptor blockers and cardiac sympathetic ablation, aim at inhibiting cardiac sympathetic overactivation. However, these treatments do not translate into becoming efficacious as long-term suppressors of ventricular tachycardia/ventricular fibrillation events. As a key regulatory component in the heart, cardiac postganglionic sympathetic neurons residing in the stellate ganglia (SGs) release neurotransmitters (such as norepinephrine and NPY [neuropeptide Y]) to perform their regulatory role in dictating cardiac function. Growing evidence from animal experiments and clinical studies has demonstrated that the remodeling of the SG may be intimately involved in malignant arrhythmogenesis. This identifies the SG as a key potential therapeutic target for the treatment of malignant VAs in heart disease. Therefore, this review summarizes the role of SG in ventricular arrhythmogenesis and updates the novel targeting of SG for clinical treatment of VAs in heart disease.
Collapse
Affiliation(s)
- Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yu Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony J. Evans
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tapan A. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Kaushik P. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
9
|
Wang YB, Dow KE, Boychuk CR. GABA AR-δ-subunit mediates increased GABAergic inhibition in cardiac DMV neurons after high-fat diet. iScience 2025; 28:112268. [PMID: 40264791 PMCID: PMC12013407 DOI: 10.1016/j.isci.2025.112268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 12/20/2024] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
Activity of cardiac-projecting neurons in the dorsal motor nucleus of the vagus (CVNDMV) is vital in cardiac reflexes contributing to maintaining cardiovascular health. However, how this population adapts to metabolic challenges, such as high-fat diet (HFD), is unclear. This study aimed to identify neuroplasticity changes induced by HFD in CVNDMV. Using whole-cell patch-clamp electrophysiology, we found that 15-day HFD feeding increased tonic, but not phasic, gamma-aminobutyric acid type A (GABAA) inhibitory neurotransmission, exclusive to CVNDMV. Single-cell quantitative reverse-transcription PCR (scRT-qPCR) analysis revealed a higher number of CVNDMV expressing GABAA receptor δ-subunit (GABAA(δ)R) in HFD compared to normal fat diet (NFD). Deletion of GABAA(δ)R in ChAT-positive motor neurons abolished HFD-induced increased tonic GABAA neurotransmission in CVNDMV. Altogether, this evidence suggests that CVNDMV exhibits early onset HFD-induced increased GABAergic neurotransmission, likely mediated by GABAA(δ)R. This increased inhibitory tone could explain previously reported reduced cardiac vagal motor output, thus contributing to poor cardiometabolic health after HFD.
Collapse
Affiliation(s)
- Yoko Brigitte Wang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Kaylie E. Dow
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Carie R. Boychuk
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
10
|
Zhang D. Cholinergic control of cardiac electrical conduction and ventricular arrhythmia: endogenous or exogenous? Eur Heart J 2025:ehaf011. [PMID: 40200810 DOI: 10.1093/eurheartj/ehaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Affiliation(s)
- Dongze Zhang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
11
|
Refisch A, Papiol S, Schumann A, Malchow B, Bär KJ. Polygenic risk for psychotic disorders in relation to cardiac autonomic dysfunction in unmedicated patients with schizophrenia. Eur Arch Psychiatry Clin Neurosci 2025; 275:863-871. [PMID: 39503783 PMCID: PMC11947016 DOI: 10.1007/s00406-024-01933-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/25/2024] [Indexed: 03/27/2025]
Abstract
Cardiac autonomic dysfunction (CADF), mainly characterized by increased heart rate, decreased heart rate variability, and loss of vagal modulation, has been extensively described in patients with schizophrenia (SCZ) and their healthy first-degree relatives. As such, it represents an apparent physiological link that contributes to the increased cardiovascular mortality in these patients. Common genetic variation is a putative underlying mechanism, along with lifestyle factors and antipsychotic medications. However, the extent to which CADF is associated with genetic factors for SCZ is unknown. A sample of 83 drug-naive SCZ patients and 96 healthy controls, all of European origin, underwent a 30-minute autonomic assessment under resting conditions. We incorporated parameters from several domains into our model, including time and frequency domains (mean heart rate, low/high frequency ratio) and compression entropy, each of which provides different insights into the dynamics of cardiac autonomic function. These parameters were used as outcome variables in linear regression models with polygenic risk scores (PRS) for SCZ as predictors and age, sex, BMI, smoking status, principal components of ancestry and diagnosis as covariates. Of the three CADF parameters, SCZ PRS was significantly associated with mean heart rate in the combined case/control sample. However, this association was was no longer significant after including diagnosis as a covariate (p = 0.29). In contrast, diagnostic status is statistically significant for all three CADF parameters, accounting for a significantly greater proportion of the variance in mean heart rate compared to SCZ PRS (approximately 16% vs. 4%). Despite evidence for a common genetic basis of CADF and SCZ, we were unable to provide further support for an association between the polygenic burden of SCZ and cardiac autonomic function beyond the diagnostic state. This suggests that there are other important characteristics associated with SCZ that lead to CADF that are not captured by SCZ PRS.
Collapse
Affiliation(s)
- Alexander Refisch
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743, Jena, Germany.
| | - Sergi Papiol
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andy Schumann
- Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC), Jena University Hospital, Jena, Germany
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Hospital Göttingen, Göttingen, Germany
| | - Karl-Jürgen Bär
- Department of Psychosomatic Medicine and Psychotherapy, Lab for Autonomic Neuroscience, Imaging and Cognition (LANIC), Jena University Hospital, Jena, Germany
| |
Collapse
|
12
|
Masuyama K, Lee JK, Yokoyama T, Li J, Yasutake H, Kuramoto Y, Hidaka K, Sakata Y. Phase dependent sympathetic dysinnervation in Takotsubo syndrome revealed with transparent heart. Sci Rep 2025; 15:10479. [PMID: 40140703 PMCID: PMC11947107 DOI: 10.1038/s41598-025-94349-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Takotsubo syndrome (TTS) is an acute cardiac syndrome characterized by transient left ventricular dysfunction. Although the wall motion abnormality resolves completely, the prognosis is poor. Defect of 123I-metaiodobenzylguanidine uptake, interpreted as sympathetic impairment, persisted in TTS patients, but the mechanism is not fully understood. We aimed to elucidate morphological sympathetic nerve change in a TTS model mouse using three-dimensional imaging techniques, with a particular focus on the role of factors in these alterations. The TTS model was induced by a single intraperitoneal injection of 2.0 mg/kg adrenaline to C57BL/6 mice, resulting in transient akinesis localized to the inferior apical region of the heart. Three-dimensional morphological assessment revealed that sympathetic nerve length within the inferior apical area of TTS mice reduced during the chronic phase compared with the sham mice. Notably, the study observed a pattern of denervation during the acute phase, followed by re-innervation and subsequent denervation in the chronic phase. The neurotrophic factors expressions changed in a time-dependent manner, corresponding to the phase-specific damage both to cardiomyocytes and sympathetic neurons. The bimodal change in sympathetic nerves and altered neurotrophic factors in TTS mice provide novel insights into the pathophysiological mechanism of TTS to establish therapeutic strategies for TTS.
Collapse
Affiliation(s)
- Kiyoshi Masuyama
- Departments of Cardiovascular Medicine, Graduate School of Medicine, The University of Osaka, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Jong-Kook Lee
- Departments of Cardiovascular Regenerative Medicine and Drug Discovery, Graduate School of Medicine, The University of Osaka, 2-2 Yamadaoka, Suita, 565-0871, Japan.
| | - Teruki Yokoyama
- Departments of Cardiovascular Medicine, Graduate School of Medicine, The University of Osaka, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Jun Li
- Departments of Cardiovascular Regenerative Medicine and Drug Discovery, Graduate School of Medicine, The University of Osaka, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Hideki Yasutake
- Departments of Cardiovascular Regenerative Medicine and Drug Discovery, Graduate School of Medicine, The University of Osaka, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Yuki Kuramoto
- Departments of Cardiovascular Medicine, Graduate School of Medicine, The University of Osaka, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Kyoko Hidaka
- Center for Fundamental Education, The University of Kitakyushu, 4-2-1 Kokura Minami-ku Kitagata, Kitakyushu, 802-8577, Japan
| | - Yasushi Sakata
- Departments of Cardiovascular Medicine, Graduate School of Medicine, The University of Osaka, 2-2 Yamadaoka, Suita, 565-0871, Japan
| |
Collapse
|
13
|
Peng JQ, Zhou MM, Xu L, Wang X, Tang YH, Liu Y. Targeted M-Channel Activation in the Left Stellate Ganglion Protects Against Ischemia-Induced Ventricular Arrhythmias in Canines. J Am Heart Assoc 2025; 14:e039059. [PMID: 40094190 DOI: 10.1161/jaha.124.039059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Acute myocardial ischemia (AMI)-triggered ventricular arrhythmias are closely linked to maladaptive sympathetic hyperactivity mediated via the left stellate ganglion (LSG). Although M-type potassium channels regulate neuronal excitability and hold therapeutic potential for neurological disorders, their role in intrinsic LSG neurons during ischemia remains unexplored. We investigated whether pharmacological M-channel activation in the LSG mitigates sympathetic overdrive and arrhythmogenesis in AMI. METHODS AND RESULTS Twenty-four beagles underwent LSG microinjection of either vehicle (n=12) or retigabine (M-channel activator, 50 μM; n=12) 30 minutes before AMI induction. We assessed (1) neural parameters (LSG electrophysiology, plasma norepinephrine levels, and c-fos+/tyrosine hydroxylase+ neuron expression); (2) cardiac electrophysiological parameters (beat-to-beat repolarization variability, spatial dispersion of effective refractory period and action potential duration, ventricular fibrillation threshold, and spontaneous ventricular arrhythmias incidence); and (3) autonomic and hemodynamic measures (heart rate variability and blood pressure). Retigabine pretreatment significantly suppressed ischemia-induced LSG hyperactivity and reduced sympathetic activation markers compared with controls. Treated animals exhibited attenuated repolarization variability and reduced electrophysiological heterogeneity in ischemic myocardium. The retigabine group demonstrated a higher ventricular fibrillation threshold (26.67±2.61 versus 12.33±1.76 voltage (V), P=0.0008) and a lower incidence of ventricular arrhythmias during AMI, with only negligible effects on baseline cardiac repolarization duration or LSG function before ischemia induction. CONCLUSIONS Targeted activation of LSG M-channels with retigabine stabilizes ischemia-induced sympathetic hyperactivity, promotes cardiac autonomic balance, preserves repolarization homogeneity, and ultimately mitigates arrhythmic susceptibility. These findings highlight ganglionic M-channel modulation as a translatable strategy to suppress neurogenic arrhythmogenesis in AMI.
Collapse
Affiliation(s)
- Jin-Qiang Peng
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Ming-Min Zhou
- Department of Cardiology Sir Run Run Shaw Hospital, Zhejiang University School of Medicine Hangzhou China
| | - Liao Xu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Xi Wang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Yan-Hong Tang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| | - Yu Liu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Cardiovascular Research Institute of Wuhan University Wuhan China
- Hubei Key Laboratory of Cardiology Wuhan China
| |
Collapse
|
14
|
Lee B, Ahmad S, Edling CE, Huang CL, LeBeau FEN, Jeevaratnam K. Age-dependent reduction in voltage-gated inward sodium current and Scn8a gene expression in murine stellate ganglia. Ann N Y Acad Sci 2025; 1545:91-104. [PMID: 39998310 PMCID: PMC11918529 DOI: 10.1111/nyas.15298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Stellate ganglia (SG) provide sympathetic innervation to the heart and may predispose the myocardial conducting system to arrhythmias. However, little is known about age-related changes in the electrophysiology of murine SG. We investigated alterations in the electrophysiological properties of SG with aging. The loose patch clamp technique was adapted to SG tissue to investigate the voltage-gated ionic currents in its neuronal cells. We compared SG and ventricular cells from young (4 months) and aged (13 months) C57BL/6J mice to explore age-related alterations in their voltage-gated ionic currents (n > 30 patches, eight mice in each group). We observed that the voltage-gated inward sodium current (peak INa(Max)) was significantly decreased with aging in the SG, but not in the ventricle. Additionally, Scn8a gene expression, which encodes the Nav 1.6 channel, was decreased with aging in the SG. Application of loose patch clamp electrophysiology thus suggests that ionic current alterations with age in murine SG could contribute to cardiac autonomic dysregulation in geriatric conditions.
Collapse
Affiliation(s)
- Bonn Lee
- School of Veterinary Medicine, Faculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| | - Shiraz Ahmad
- School of Veterinary Medicine, Faculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| | - Charlotte E. Edling
- School of Veterinary Medicine, Faculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| | - Christopher L.‐H. Huang
- Physiological LaboratoryUniversity of CambridgeCambridgeUK
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Fiona E. N. LeBeau
- Biosciences Institute, Faculty of Medical SciencesUniversity of NewcastleNewcastle upon TyneUK
| | - Kamalan Jeevaratnam
- School of Veterinary Medicine, Faculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| |
Collapse
|
15
|
Ajijola OA, Aksu T, Arora R, Biaggioni I, Chen PS, De Ferrari G, Dusi V, Fudim M, Goldberger JJ, Green AL, Herring N, Khalsa SS, Kumar R, Lakatta E, Mehra R, Meyer C, Po S, Stavrakis S, Somers VK, Tan AY, Valderrabano M, Shivkumar K. Clinical neurocardiology: defining the value of neuroscience-based cardiovascular therapeutics - 2024 update. J Physiol 2025; 603:1781-1839. [PMID: 40056025 DOI: 10.1113/jp284741] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 01/28/2025] [Indexed: 04/01/2025] Open
Abstract
The intricate role of the autonomic nervous system (ANS) in regulating cardiac physiology has long been recognized. Aberrant function of the ANS is central to the pathophysiology of cardiovascular diseases. It stands to reason, therefore, that neuroscience-based cardiovascular therapeutics hold great promise in the treatment of cardiovascular diseases in humans. A decade after the inaugural edition, this White Paper reviews the current state of understanding of human cardiac neuroanatomy, neurophysiology and pathophysiology in specific disease conditions, autonomic testing, risk stratification, and neuromodulatory strategies to mitigate the progression of cardiovascular diseases.
Collapse
Affiliation(s)
- Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tolga Aksu
- Division of Cardiology, Yeditepe University Hospital, Istanbul, Türkiye
| | - Rishi Arora
- Division of Cardiology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Italo Biaggioni
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peng-Sheng Chen
- Department of Cardiology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Gaetano De Ferrari
- Department of Medical Sciences, University of Turin, Italy and Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Torino, Italy
| | - Veronica Dusi
- Department of Medical Sciences, University of Turin, Italy and Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Torino, Italy
| | - Marat Fudim
- Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey J Goldberger
- Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander L Green
- Department of Clinical Neurosciences, John Radcliffe Hospital, and Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Neil Herring
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sahib S Khalsa
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rajesh Kumar
- Department of Neurobiology and the Brain Research Institute, University of California, Los Angeles, CA, USA
| | - Edward Lakatta
- National Institute of Aging, National Institutes of Health, Bethesda, MD, USA
| | - Reena Mehra
- Division of Pulmonary Medicine, University of Washington, Seattle, WA, USA
| | - Christian Meyer
- Klinik für Kardiologie, Angiologie, Intensivmedizin, cNEP Research Consortium EVK, Düsseldorf, Germany
- Heart Rhythm Institute, Overland Park, KS, USA
| | - Sunny Po
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stavros Stavrakis
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Virend K Somers
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Alex Y Tan
- Division of Cardiology, Richmond Veterans Affairs Hospital, Richmond, VA, USA
| | - Miguel Valderrabano
- Methodist DeBakey Heart and Vascular Center and Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
16
|
Habecker BA, Bers DM, Birren SJ, Chang R, Herring N, Kay MW, Li D, Mendelowitz D, Mongillo M, Montgomery JM, Ripplinger CM, Tampakakis E, Winbo A, Zaglia T, Zeltner N, Paterson DJ. Molecular and cellular neurocardiology in heart disease. J Physiol 2025; 603:1689-1728. [PMID: 38778747 PMCID: PMC11582088 DOI: 10.1113/jp284739] [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: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
This paper updates and builds on a previous White Paper in this journal that some of us contributed to concerning the molecular and cellular basis of cardiac neurobiology of heart disease. Here we focus on recent findings that underpin cardiac autonomic development, novel intracellular pathways and neuroplasticity. Throughout we highlight unanswered questions and areas of controversy. Whilst some neurochemical pathways are already demonstrating prognostic viability in patients with heart failure, we also discuss the opportunity to better understand sympathetic impairment by using patient specific stem cells that provides pathophysiological contextualization to study 'disease in a dish'. Novel imaging techniques and spatial transcriptomics are also facilitating a road map for target discovery of molecular pathways that may form a therapeutic opportunity to treat cardiac dysautonomia.
Collapse
Affiliation(s)
- Beth A Habecker
- Department of Chemical Physiology & Biochemistry, Department of Medicine Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | - Susan J Birren
- Department of Biology, Volen Center for Complex Systems, Brandeis University, Waltham, MA, USA
| | - Rui Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Johanna M Montgomery
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | | | - Annika Winbo
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Nadja Zeltner
- Departments of Biochemistry and Molecular Biology, Cell Biology, and Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
17
|
Thompson N, Ravagli E, Mastitskaya S, Challita R, Hadaya J, Iacoviello F, Idil AS, Shearing PR, Ajijola OA, Ardell JL, Shivkumar K, Holder D, Aristovich K. Towards spatially selective efferent neuromodulation: anatomical and functional organization of cardiac fibres in the porcine cervical vagus nerve. J Physiol 2025; 603:1983-2004. [PMID: 39183636 PMCID: PMC11955868 DOI: 10.1113/jp286494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024] Open
Abstract
Spatially selective vagus nerve stimulation (sVNS) offers a promising approach for addressing heart disease with enhanced precision. Despite its therapeutic potential, VNS is limited by off-target effects and the need for time-consuming titration. Our research aimed to determine the spatial organization of cardiac afferent and efferent fibres within the vagus nerve of pigs to achieve targeted neuromodulation. Using trial-and-error sVNS in vivo and ex vivo micro-computed tomography fascicle tracing, we found significant spatial separation between cardiac afferent and cardiac efferent fibres at the mid-cervical level and they were localized on average on opposite sides of the nerve cross-section. This was consistent between both in vivo and ex vivo methods. Specifically, cardiac afferent fibres were located near pulmonary fibres, consistent with findings of cardiopulmonary convergent circuits and, notably, cardiac efferent fascicles were exclusive. These cardiac efferent regions were located in close proximity to the recurrent laryngeal regions. This is consistent with the roughly equitable spread across the nerve of the afferent and efferent fibres. Our study demonstrated that targeted neuromodulation via sVNS could achieve scalable heart rate decreases without eliciting cardiac afferent-related reflexes; this is desirable for reducing sympathetic overactivation associated with heart disease. These findings indicate that understanding the spatial organization of cardiac-related fibres within the vagus nerve can lead to more precise and effective VNS therapy, minimizing off-target effects and potentially mitigating the need for titration. KEY POINTS: Spatially selective vagus nerve stimulation (sVNS) presents a promising approach for addressing chronic heart disease with enhanced precision. Our study reveals significant spatial separation between cardiac afferent and efferent fibres in the vagus nerve, particularly at the mid-cervical level. Utilizing trial-and-error sVNS in vivo and micro-computed tomography fascicle tracing, we demonstrate the potential for targeted neuromodulation, achieving therapeutic effects such as scalable heart rate decrease without stimulating cardiac afferent-related reflexes. This spatial understanding opens avenues for more effective VNS therapy, minimizing off-target effects and potentially eliminating the need for titration, thereby expediting therapeutic outcomes in myocardial infarction and related conditions.
Collapse
Affiliation(s)
- Nicole Thompson
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Enrico Ravagli
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Svetlana Mastitskaya
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Ronald Challita
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of ExcellenceDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Joseph Hadaya
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of ExcellenceDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Francesco Iacoviello
- Electrochemical Innovation Lab, Department of Chemical EngineeringUniversity College LondonLondonUK
| | - Ahmad Shah Idil
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Paul R. Shearing
- Electrochemical Innovation Lab, Department of Chemical EngineeringUniversity College LondonLondonUK
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of ExcellenceDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Jeffrey L. Ardell
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of ExcellenceDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of ExcellenceDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - David Holder
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Kirill Aristovich
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| |
Collapse
|
18
|
Vrabec T, Bender S, Chan S, Cha S, Haridas S, Hanna P, Ajijola OA, Shivkumar K, Smith C, Ardell JL. Bioelectronic block of stellate ganglia mitigates pacing-induced heterogeneous release of catecholamine and neuropeptide Y in the infarcted pig heart. J Physiol 2025; 603:2071-2088. [PMID: 39557601 PMCID: PMC11955864 DOI: 10.1113/jp286924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/23/2024] [Indexed: 11/20/2024] Open
Abstract
The sympathetic nervous system modulates cardiac contractile and electrophysiological function and contributes to adverse remodelling following myocardial infarction (MI). Axonal modulation therapy (AMT), directed at the sympathetic chain, blocks efferent sympathetic outflow to the heart and is a strategy to transiently and controllably mitigate chronic MI-associated sympatho-excitation. In porcine models, we evaluated scalable AMT, directed at the paravertebral chain, in blocking reflex-mediated pacing-induced sympatho-excitation post-MI. The level of sympatho-excitation was assessed by dynamic interstitial measurement of noradrenaline (NA) and neuropeptide Y (NPY). In anaesthetized normal (n = 5) and age-matched pigs 6 weeks post-MI induction (n = 10), we electrically stimulated the right sympathetic chain and determined levels of direct current block applied at the T1-T2 level sufficient to reduce the evoked changes in heart rate and/or contractility by 25-75%. Reflex-mediated neural release of NA and NPY into the interstitial space during programmed pacing (PP) was assessed using fast-scanning cyclic voltammetry and capacitive immunoprobes. Normal animals demonstrated homogeneous NA and NPY release profiles during PP. In contrast, for MI animals PP evoked differential NA and NPY release in remote and MI border zones of the left ventricle. Right-sided AMT mitigated NA and NPY pacing-induced release in the remote left ventricle with a positive correlation to increasing AMT levels. Pacing-induced NA and NPY release in the MI border zone was not mitigated by AMT. Differential effects of AMT on NA and NPY may underlie the anti-arrhythmic effects of partial stellate ganglion block in the setting of chronic MI. KEY POINTS: Programmed cardiac pacing evokes homogeneous noradrenaline (NA) and neuropeptide Y (NPY) release in equivalent areas (e.g. medial and lateral aspects) of the normal left ventricle. Programmed cardiac pacing evokes differential NA and NPY release in remote and border zones of the infarcted left ventricle. Axonal modulation therapy (AMT), using a graded direct current block applied to the stellate ganglia, can proportionally modulate cardiac sympathetic reflexes. Unilateral AMT mitigates NA and NPY release in remote left ventricular tissue, with release negatively correlated to increasing AMT levels. Heterogeneities in NA and NPY between the border and remote tissues are reduced by progressive AMT.
Collapse
Affiliation(s)
- Tina Vrabec
- Department of Physical Medicine & RehabilitationMetroHealth Medical CenterClevelandOHUSA
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
| | - Shane Bender
- Department of Physical Medicine & RehabilitationMetroHealth Medical CenterClevelandOHUSA
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
| | - Shyue‐An Chan
- Department of Physiology and BiophysicsCase Western Reserve UniversityClevelandOHUSA
| | - Steven Cha
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Sahil Haridas
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Peter Hanna
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Olujimi A. Ajijola
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Kalyanam Shivkumar
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| | - Corey Smith
- Department of Physiology and BiophysicsCase Western Reserve UniversityClevelandOHUSA
| | - Jeffrey L. Ardell
- David Geffen School of MedicineUniversity of California – Los Angeles (UCLA) Cardiac Arrhythmia CenterLos AngelesCAUSA
- UCLA Neurocardiology Research Program of ExcellenceLos AngelesCAUSA
| |
Collapse
|
19
|
Gillman S, Zucker IH, Wang H. Heartificial intelligence: smart solutions for CHF: An A(I)MT approach. J Physiol 2025; 603:1909-1910. [PMID: 39726247 PMCID: PMC11955859 DOI: 10.1113/jp287953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- Samuel Gillman
- Department of Genetics Cell Biology and AnatomyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Irving H. Zucker
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Han‐Jun Wang
- Department of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| |
Collapse
|
20
|
De Smet MAJ, De Becker B, François C, le Polain de Waroux JB, Knecht S, Duytschaever M, Tavernier R. Atrial Tachyarrhythmias With Ultra-Rapid Ventricular Response and Sudden Death in Patients Without Structural Heart Disease. JACC Clin Electrophysiol 2025; 11:482-495. [PMID: 39708037 DOI: 10.1016/j.jacep.2024.10.025] [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: 07/10/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Sudden cardiac death (SCD) is generally associated with life-threatening ventricular arrhythmias. Supraventricular arrhythmias are an accepted cause of SCD in Wolff-Parkinson-White syndrome and complex congenital heart disease. However, the role of atrial tachyarrhythmias (ATAs) in SCD in patients with structurally normal hearts is unclear. OBJECTIVES The goal of this study was to present data on resuscitated patients without structural heart disease (SHD), experiencing recurrent implantable cardioverter-defibrillator (ICD) shocks, who share common clinical and electrical features suggesting that ATAs can cause SCD. METHODS We describe the clinical characteristics and ICD analysis of syncopal events terminated with shock delivery in 5 young SCD survivors without SHD. Details on the follow-up after ablation of the arrhythmia causing the syncopal episode are also reported. RESULTS In all patients (4 male, 1 female; median age 23 years; age range 15-47 years), a surface electrocardiogram recording in the resuscitation setting suggested ventricular fibrillation. After the index event, all patients exhibited recurrent arrhythmic syncopal episodes in a setting of elevated adrenergic tone, treated with ICD shocks. ICD interrogation suggested ATAs (atrial fibrillation in 4 patients, atrial tachycardia in 1 patient), conducting to the ventricles at rates approaching 300 beats/min, as the underlying arrhythmia leading to the syncopal events. ATA ablation abolished episodes of arrhythmic syncope and shock delivery in all patients after a median follow-up of 34 months. No patient died suddenly during follow-up. CONCLUSIONS Common clinical and electrical features define a distinct entity of SCD caused by ATAs with ultra-rapid ventricular response in otherwise healthy patients. Catheter ablation of the ATA is an effective treatment in these patients.
Collapse
Affiliation(s)
- Maarten A J De Smet
- Department of Cardiology, AZ Sin Jan Bruges, Bruges, Belgium; Department of Cardiology, University Hospital Ghent, Ghent, Belgium
| | | | - Clara François
- Department of Cardiology, AZ Sin Jan Bruges, Bruges, Belgium
| | | | | | - Mattias Duytschaever
- Department of Cardiology, AZ Sin Jan Bruges, Bruges, Belgium; Department of Cardiology, University Hospital Ghent, Ghent, Belgium
| | - Rene Tavernier
- Department of Cardiology, AZ Sin Jan Bruges, Bruges, Belgium.
| |
Collapse
|
21
|
Qu Z, Hanna P, Ajijola OA, Garfinkel A, Shivkumar K. Ultrastructure and cardiac impulse propagation: scaling up from microscopic to macroscopic conduction. J Physiol 2025; 603:1887-1901. [PMID: 39612369 PMCID: PMC11955865 DOI: 10.1113/jp287632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/31/2024] [Indexed: 12/01/2024] Open
Abstract
The standard conception of cardiac conduction is based on the cable theory of nerve conduction, which treats cardiac tissue as a continuous syncytium described by the Hodgkin-Huxley equations. However, cardiac tissue is composed of discretized cells with microscopic and macroscopic heterogeneities and discontinuities, such as subcellular localizations of sodium channels and connexins. In addition to this, there are heterogeneities in the distribution of sympathetic and parasympathetic nerves, which powerfully regulate impulse propagation. In the continuous models, the ultrastructural details, i.e. the microscopic heterogeneities and discontinuities, are ignored by 'coarse graining' or 'smoothing'. However, these ultrastructural components may play crucial roles in cardiac conduction and arrhythmogenesis, particularly in disease states. We discuss the current progress of modelling the effects of ultrastructural components on electrical conduction, the issues and challenges faced by the cardiac modelling community, and how to scale up conduction properties at the subcellular (microscopic) scale to the tissue and whole-heart (macroscopic) scale in future modelling and experimental studies, i.e. how to link the ultrastructure at different scales to impulse conduction and arrhythmogenesis in the heart.
Collapse
Affiliation(s)
- Zhilin Qu
- UCLA Cardiac Arrhythmia Center and Department of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Peter Hanna
- UCLA Cardiac Arrhythmia Center and Department of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Department of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Alan Garfinkel
- UCLA Cardiac Arrhythmia Center and Department of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Department of Medicine, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
22
|
Herring N, Ajijola OA, Foreman RD, Gourine AV, Green AL, Osborn J, Paterson DJ, Paton JFR, Ripplinger CM, Smith C, Vrabec TL, Wang HJ, Zucker IH, Ardell JL. Neurocardiology: translational advancements and potential. J Physiol 2025; 603:1729-1779. [PMID: 39340173 PMCID: PMC11955874 DOI: 10.1113/jp284740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
In our original white paper published in the The Journal of Physiology in 2016, we set out our knowledge of the structural and functional organization of cardiac autonomic control, how it remodels during disease, and approaches to exploit such knowledge for autonomic regulation therapy. The aim of this update is to build on this original blueprint, highlighting the significant progress which has been made in the field since and major challenges and opportunities that exist with regard to translation. Imbalances in autonomic responses, while beneficial in the short term, ultimately contribute to the evolution of cardiac pathology. As our understanding emerges of where and how to target in terms of actuators (including the heart and intracardiac nervous system (ICNS), stellate ganglia, dorsal root ganglia (DRG), vagus nerve, brainstem, and even higher centres), there is also a need to develop sensor technology to respond to appropriate biomarkers (electrophysiological, mechanical, and molecular) such that closed-loop autonomic regulation therapies can evolve. The goal is to work with endogenous control systems, rather than in opposition to them, to improve outcomes.
Collapse
Affiliation(s)
- N. Herring
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - O. A. Ajijola
- UCLA Neurocardiology Research Center of ExcellenceDavid Geffen School of MedicineLos AngelesCAUSA
| | - R. D. Foreman
- Department of Biochemistry and PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - A. V. Gourine
- Centre for Cardiovascular and Metabolic NeuroscienceUniversity College LondonLondonUK
| | - A. L. Green
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | - J. Osborn
- Department of SurgeryUniversity of MinnesotaMinneapolisMNUSA
| | - D. J. Paterson
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - J. F. R. Paton
- Manaaki Manawa – The Centre for Heart Research, Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - C. M. Ripplinger
- Department of PharmacologyUniversity of California DavisDavisCAUSA
| | - C. Smith
- Department of Physiology and BiophysicsCase Western Reserve UniversityClevelandOHUSA
| | - T. L. Vrabec
- Department of Physical Medicine and Rehabilitation, School of MedicineCase Western Reserve UniversityClevelandOHUSA
| | - H. J. Wang
- Department of AnesthesiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - I. H. Zucker
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - J. L. Ardell
- UCLA Neurocardiology Research Center of ExcellenceDavid Geffen School of MedicineLos AngelesCAUSA
| |
Collapse
|
23
|
Paredes-Espinosa MB, Paluh JL. Synthetic embryology of the human heart. Front Cell Dev Biol 2025; 12:1478549. [PMID: 39935786 PMCID: PMC11810959 DOI: 10.3389/fcell.2024.1478549] [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: 08/10/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
The evolution of stem cell-based heart models from cells and tissues to organoids and assembloids and recently synthetic embryology gastruloids, is poised to revolutionize our understanding of cardiac development, congenital to adult diseases, and patient customized therapies. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have already been integrated into transplantable patches and are in preclinical efforts to reverse fibrotic scarring from myocardial infarctions. To inform on the complexity of heart diseases, multi-tissue morphogenic heart models are needed that replicate fundamental components of heart function to heart organogenesis in vitro and which require a deep understanding of heart development. Organoid and assembloid models capture selected multicellular cardiac processes, such as chamber formation and priming events for vascularization. Gastruloid heart models offer deeper insights as synthetic embryology to mimic multi-staged developmental events of in vivo heart organogenesis including established heart fields, crescent formation and heart tube development along with vascular systemic foundation and even further steps. The human Elongating Multi-Lineage Organized Cardiac (EMLOC) gastruloid model captures these stages and additional events including chamber genesis, patterned vascularization, and extrinsic central and intrinsic cardiac nervous system (CNS-ICNS) integration guided by spatiotemporal and morphogenic processes with neural crest cells. Gastruloid synthetic embryology heart models offer new insights into previously hidden processes of development and provide powerful platforms for addressing heart disease that extends beyond cardiomyocytes, such as arrhythmogenic diseases, congenital defects, and systemic injury interactions, as in spinal cord injuries. The holistic view that is emerging will reveal heart development and disease in unprecedented detail to drive transformative state-of-the-art innovative applications for heart health.
Collapse
Affiliation(s)
| | - Janet L. Paluh
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science and Engineering, University at Albany, Albany, NY, United States
| |
Collapse
|
24
|
Katov L, Rostan J, Teumer Y, Diofano F, Bothner C, Rottbauer W, Weinmann-Emhardt K. Antiarrhythmic Effects of SGLT2 Inhibitors on Supraventricular Tachyarrhythmias in Patients with HFrEF. J Clin Med 2025; 14:786. [PMID: 39941457 PMCID: PMC11818141 DOI: 10.3390/jcm14030786] [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: 01/08/2025] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Background: In recent years, sodium-glucose cotransporter-2 (SGLT2) inhibitors have demonstrated significant cardiovascular and renal benefits in patients with heart failure (HF), in addition to their established antidiabetic effects. However, their role in arrhythmia prevention remains unclear. This study aimed to assess the effect of SGLT2 inhibitors on the incidence of supraventricular tachycardia (SVT) and ventricular tachycardia (VT) in patients with HF with reduced ejection fraction (HFrEF) during an extended follow-up period. Methods: This retrospective cohort study was conducted between January 2019 and November 2024 at the Ulm University Heart Center. All patients exhibited severely reduced left ventricular function and underwent primary prophylactic implantable cardioverter-defibrillator (ICD) implantation. Half of the cohort initiated SGLT2 inhibitor therapy alongside optimal medical HF treatment (the SGLT2 group). Patients were followed for approximately three years (846.2 ± 520.0 days) and the incidence of SVT and VT was analyzed using intracardiac Holter records of the ICD. Results: The study population consisted of 78 patients with a mean age of 66.6 ± 12.9 years. Over the follow-up period, a significant prolongation in the time to first occurrence of SVT was observed in the SGLT2 group (Log-Rank p = 0.03), suggesting a potential protective effect of SGLT2 inhibitors. However, regarding VT, additional SGLT2 inhibitor therapy did not show an additional benefit to optimal medical HF treatment. Conclusions: This study suggests that SGLT2 inhibitors may play a beneficial role in reducing the incidence of SVT in patients with HFrEF. These results highlight the importance of further investigating the antiarrhythmic potential of SGLT2 inhibitors through large-scale, prospective studies to better understand their clinical implications and mechanisms of action.
Collapse
|
25
|
Schoffl J, Craig A, McBain C, Pozzato I, Middleton JW, Arora M. The Effect of Non-Invasive, Non-Pharmacological Interventions on Autonomic Regulation of Cardiovascular Function in Adults with Spinal Cord Injury: A Systematic Review with Meta-Analysis. Neurotrauma Rep 2025; 5:1151-1172. [PMID: 40007857 PMCID: PMC11848056 DOI: 10.1089/neur.2024.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
Autonomic regulation of cardiovascular function is often disrupted following a spinal cord injury (SCI). A systematic review was undertaken to evaluate the effect of non-invasive, non-pharmacological (NINP) interventions on cardiovascular autonomic biomarkers in adults with SCI. AMED, CENTRAL, CINAHL EMBASE, and MEDLINE were searched from inception to May 17, 2024. Randomized controlled trials (RCTs) of NINP interventions for cardiovascular autonomic biomarkers (heart rate variability [HRV], systolic blood pressure variability [SBPV], or baroreflex gain) in adults (≥18 years of age) with SCI (>3 months) were included. Primary outcomes included HRV (low-frequency power [HRV-LF], high-frequency power [HRV-HF], root mean square of successive differences [RMSSD]), SBPV (low-frequency power [SBPV-LF]), and baroreflex sensitivity. The quality and certainty of the evidence were assessed using version 2 of the Cochrane risk of bias tool and the Preferred Reporting Items for Systematic Reviews and Meta-Analysis tool, respectively. Of 2651 records identified, six RCTs were included (participants, n = 123). HRV-LF (four studies; participants, n = 69) and HRV-HF (five studies; participants, n = 93) showed no to small changes in favor of NINP interventions ([g = 0.25; 95% confidence interval [CI] = -0.23, 0.73; p = 0.31; I2 = 0%], [g = 0.00; 95% CI = -0.41, 0.42; p = 0.98; I2 = 0%], respectively). Limited evidence was available for RMSSD, SBPV-LF, and baroreflex gain. This review found that the evidence is inconclusive regarding the effect of NINP interventions on the included HRV, BPV, and BRS parameters in adults with SCI. Further research with strong methodological rigor is needed to provide greater insights in this area.
Collapse
Affiliation(s)
- Jacob Schoffl
- John Walsh Centre for Rehabilitation Research, Northern Sydney Local Health District, St Leonards, NSW, Australia
- The Kolling Institute, School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ashley Craig
- John Walsh Centre for Rehabilitation Research, Northern Sydney Local Health District, St Leonards, NSW, Australia
- The Kolling Institute, School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Candice McBain
- John Walsh Centre for Rehabilitation Research, Northern Sydney Local Health District, St Leonards, NSW, Australia
- The Kolling Institute, School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ilaria Pozzato
- John Walsh Centre for Rehabilitation Research, Northern Sydney Local Health District, St Leonards, NSW, Australia
- The Kolling Institute, School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - James W. Middleton
- John Walsh Centre for Rehabilitation Research, Northern Sydney Local Health District, St Leonards, NSW, Australia
- The Kolling Institute, School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mohit Arora
- John Walsh Centre for Rehabilitation Research, Northern Sydney Local Health District, St Leonards, NSW, Australia
- The Kolling Institute, School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
26
|
Danilov A, Frishman WH. Complementary Therapies: Tai Chi in the Prevention and Management of Cardiovascular Disease. Cardiol Rev 2025; 33:54-57. [PMID: 37395587 DOI: 10.1097/crd.0000000000000578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Cardiovascular disease has remained the top contributor to global mortality for decades, necessitating research into the most effective methods of its prevention and treatment. Simultaneous with an immense amount of discovery and innovation in the field of cardiology, certain therapies with traditional Chinese origins have become progressively more popular in the West in recent decades. Specifically, ancient meditative mind-body practices such as Qigong and Tai Chi may lower cardiovascular disease risk and severity through a focus on movement and meditation. Such practices are generally low-cost and modifiable, with few adverse effects. Studies have shown higher quality of life in patients with coronary artery disease and heart failure after participation in Tai Chi, as well as a positive impact on cardiovascular risk factors such as hypertension and waist circumference. Most studies in the field have various limitations, such as small sample size, lack of randomization, and inadequate control; however, these practices show potential as an adjunct in the prevention and treatment of cardiovascular disease. Patients unable or unwilling to partake in traditionally aerobic activities may benefit greatly from such mind-body therapies. Nonetheless, more studies are warranted for more definitive answers to the question of Tai Chi and Qigong's effectiveness. In this narrative review, we discuss the current evidence surrounding the effects of Qigong and Tai Chi on cardiovascular disease, in addition to the limitations and difficulties in conducting such studies.
Collapse
Affiliation(s)
| | - William H Frishman
- From the Department of Medicine, New York Medical College, Valhalla, NY
- Department of Cardiology, Westchester Medical Center, New York Medical College, Valhalla, NY
| |
Collapse
|
27
|
Mehrabi Nasab E, Sadeghian S, Vasheghani Farahani A, Yamini Sharif A, Masoud Kabir F, Bavanpour Karvane H, Zahedi A, Bozorgi A. Determining the recurrence rate of premature ventricular complexes and idiopathic ventricular tachycardia after radiofrequency catheter ablation with the help of designing a machine-learning model. Regen Ther 2024; 27:32-38. [PMID: 38496010 PMCID: PMC10940794 DOI: 10.1016/j.reth.2024.03.001] [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: 01/27/2024] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/19/2024] Open
Abstract
Ventricular arrhythmias increase cardiovascular morbidity and mortality. Recurrent PVCs and IVT are generally considered benign in the absence of structural heart abnormalities. Artificial intelligence is a rapidly growing field. In recent years, medical professionals have shown great interest in the potential use of ML, an integral part of AI, in various disciplines, including diagnostic applications, decision-making, prognostic stratification, and solving complex pathophysiological aspects of diseases from these data at extraordinary complexity, scale, and acquisition rate. The aim of this study was to design an ML model to predict the probability of PVC and IVT recurrence after RF ablation. Data of patients were collected and manipulated using traditional analysis and various artificial intelligence models, namely MLP, Gradient Boosting Machines, Random Forest, and Logistic Regression. Hypertension, male sex, and the use of non-irrigate catheters were associated with less freedom from arrhythmia. All these results were obtained through traditional analytic methods, and according to AI, none of the variables had a clear effect on the recurrence of arrhythmia. Each AI model presents unique strengths and weaknesses, and further optimization and fine-tuning of these models are necessary to increase their clinical utility. By expanding the dataset, improved predictions can be fostered to ultimately increase the clinical utility of AI in predicting PVC erosion outcomes.
Collapse
Affiliation(s)
- Entezar Mehrabi Nasab
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Cardiology, School of Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saeed Sadeghian
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Vasheghani Farahani
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Yamini Sharif
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Masoud Kabir
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ahora Zahedi
- Department of Artificial Intelligence in Medical Sciences, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Bozorgi
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Scalco A, Lee EN, Johnson MA, Sorensen ML, Hilton TN, Omonaka RK, Zeimantz S, Aicher SA, Woodward WR, Habecker BA. Hypertension-induced heart failure disrupts cardiac sympathetic innervation. Am J Physiol Heart Circ Physiol 2024; 327:H1544-H1558. [PMID: 39485300 PMCID: PMC11684885 DOI: 10.1152/ajpheart.00380.2024] [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: 06/06/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024]
Abstract
About 26 million people worldwide live with heart failure (HF), and hypertension is the primary cause in 25% of these cases. Autonomic dysfunction and sympathetic hyperactivity accompany cardiovascular diseases, including HF. However, changes in cardiac sympathetic innervation in HF are not well understood. We hypothesized that cardiac sympathetic innervation is disrupted in hypertension-induced HF. Male and female C57BL6/J mice were infused with angiotensin II (ANG II) for 4 wk to generate hypertension leading to HF; controls were infused with saline. ANG II-treated mice displayed HF phenotype, including reduced cardiac function, hypertrophy, and fibrosis. ANG II-treated mice also had significantly reduced sympathetic nerve density in the left ventricle, intraventricular septum, and right ventricle. In the left ventricle, the subepicardium remained normally innervated, whereas the subendocardium was almost devoid of sympathetic nerves. Loss of sympathetic fibers led to loss of norepinephrine content in the left ventricle. Several potential triggers for axon degeneration were tested and ruled out. ANG II-treated mice had increased premature ventricular contractions after isoproterenol and caffeine injection. Although HF can induce a cholinergic phenotype and neuronal hypertrophy in stellate ganglia, ANG II treatment did not induce a cholinergic phenotype or activation of trophic factors in this study. Cardiac neurons in the left stellate ganglion were significantly smaller in ANG II-treated mice, whereas neurons in the right stellate were unchanged. Our findings show that ANG II-induced HF disrupts sympathetic innervation, particularly in the left ventricle. Further investigations are imperative to unveil the mechanisms of denervation in HF and to develop neuromodulatory therapies for patients with autonomic imbalance.NEW & NOTEWORTHY Angiotensin II (ANG II)-induced hypertension leads to a heart failure phenotype and cardiac sympathetic denervation with the endocardial region of the left ventricle being the most affected. Denervation is accompanied by loss of norepinephrine content in the left ventricle and increased premature ventricular contractions (PVCs) after isoproterenol and caffeine injection. ANG II treatment also causes morphological changes in cardiac-projecting left stellate ganglion neurons.
Collapse
Affiliation(s)
- Arianna Scalco
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Ethan N Lee
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
- Department of Biology, Pomona College, Claremont, California, United States
| | - Morgan A Johnson
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Michelle L Sorensen
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Thomas N Hilton
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Riley K Omonaka
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
- Department of Biology, Linfield University, McMinnville, Oregon, United States
| | - Shae Zeimantz
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Sue A Aicher
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - William R Woodward
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
29
|
Gentile F, Orlando G, Montuoro S, Ferrari Chen YF, Macefield V, Passino C, Giannoni A, Emdin M. Treating heart failure by targeting the vagus nerve. Heart Fail Rev 2024; 29:1201-1215. [PMID: 39117958 PMCID: PMC11455679 DOI: 10.1007/s10741-024-10430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Increased sympathetic and reduced parasympathetic nerve activity is associated with disease progression and poor outcomes in patients with chronic heart failure. The demonstration that markers of autonomic imbalance and vagal dysfunction, such as reduced heart rate variability and baroreflex sensitivity, hold prognostic value in patients with chronic heart failure despite modern therapies encourages the research for neuromodulation strategies targeting the vagus nerve. However, the approaches tested so far have yielded inconclusive results. This review aims to summarize the current knowledge about the role of the parasympathetic nervous system in chronic heart failure, describing the pathophysiological background, the methods of assessment, and the rationale, limits, and future perspectives of parasympathetic stimulation either by drugs or bioelectronic devices.
Collapse
Affiliation(s)
- Francesco Gentile
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy.
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy.
| | - Giulia Orlando
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Sabrina Montuoro
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
| | - Yu Fu Ferrari Chen
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | | | - Claudio Passino
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Piazza Martiri Della Libertà 33, 56127, Pisa, Italy
- Cardiology and Cardiovascular Medicine Division, Fondazione Monasterio, Via G. Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
30
|
Shah AS, Vaccarino V, Moazzami K, Almuwaqqat Z, Garcia M, Ward L, Elon L, Ko YA, Sun YV, Pearce BD, Raggi P, Bremner JD, Lampert R, Quyyumi AA, Shah AJ. Autonomic reactivity to mental stress is associated with cardiovascular mortality. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae086. [PMID: 39588213 PMCID: PMC11588410 DOI: 10.1093/ehjopen/oeae086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/02/2024] [Accepted: 09/19/2024] [Indexed: 11/27/2024]
Abstract
Aims The mechanisms linking acute psychological stress to cardiovascular disease (CVD) mortality are incompletely understood. We studied the relationship of electrocardiographic measures of autonomic dysfunction during acute mental stress provocation and CVD death. Methods and results In a pooled cohort of 765 participants with stable CVD from two related studies, we collected Holter data during standardized laboratory-based mental stress testing with a speech task and followed them for events. We assessed autonomic function using low-frequency (LF) heart rate variability (HRV) in 5-min intervals before, during, and after stress induction, and specifically examined changes from rest to stress. We employed cause-specific survival models to examine its association with CVD and all-cause mortality, controlling for demographic and CVD risk factors. The mean (SD) age was 58 (10) years, 35% were women, and 44% self-identified as Black. After a median follow-up of 5.6 years, 37 (5%) died from CVD causes. A stress-induced LF HRV decrease (67% of sample), vs. increase, was associated with a hazard ratio (HR) of 3.48 (95% confidence interval-3.25, 3.73) for CVD mortality. Low rest LF HRV (bottom quartile) was also independently associated with CVD mortality, HR = 1.75 (1.58, 1.94), vs. normal rest LF HRV (upper three quartiles). The combination of stress-induced LF HRV decrease and low rest LF HRV was associated with HR = 5.73 (5.33, 6.15) vs. the normal stress/rest LF HRV reference. We found similar results with HF HRV. Conclusion Stress-induced LF HRV decrease and low rest LF HRV are both independently and additively associated with a higher CVD mortality risk. Additional research is needed to assess whether targeting autonomic dysfunction may improve CVD outcomes.
Collapse
Affiliation(s)
- Anish S Shah
- Department of Medicine, Division of Cardiology, University of Utah, 30 North Mario Capecchi Dr, 3rd Floor North, Salt Lake City, UT 84112, USA
- Department of Medicine, Division of Cardiology, University of Illinois Chicago, 840 South Wood Street, Suite 1020N, MC 787, Chicago, IL 60612, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
- Emory Clinical Cardiovascular Research Institute, Emory University, 1750 Haygood Dr NE, 2nd Floor, Atlanta, GA 30322, USA
| | - Kasra Moazzami
- Emory Clinical Cardiovascular Research Institute, Emory University, 1750 Haygood Dr NE, 2nd Floor, Atlanta, GA 30322, USA
| | - Zakaria Almuwaqqat
- Emory Clinical Cardiovascular Research Institute, Emory University, 1750 Haygood Dr NE, 2nd Floor, Atlanta, GA 30322, USA
| | - Mariana Garcia
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Laura Ward
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Lisa Elon
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
- Atlanta VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA
| | - Brad D Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Paolo Raggi
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
- Division of Cardiology, Department of Medicine, University of Alberta, 83 Ave NW Edmonton T6G2B7, Canada
| | - J Douglas Bremner
- Atlanta VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, USA
| | - Rachel Lampert
- Section of Cardiology, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Arshed A Quyyumi
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
- Emory Clinical Cardiovascular Research Institute, Emory University, 1750 Haygood Dr NE, 2nd Floor, Atlanta, GA 30322, USA
| | - Amit J Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Rd NE, Atlanta, GA 30322, USA
- Atlanta VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA
| |
Collapse
|
31
|
Varga CR, Cleland JGF, Abraham WT, Lip GYH, Leyva F, Hatamizadeh P. Implantable Cardioverter Defibrillator and Resynchronization Therapy in Patients With Overt Chronic Kidney Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2024; 84:1342-1362. [PMID: 39322329 DOI: 10.1016/j.jacc.2024.05.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/22/2024] [Accepted: 05/30/2024] [Indexed: 09/27/2024]
Abstract
Heart failure and chronic kidney disease are common and clinically important conditions that regularly coexist. Electrophysiologic changes of advanced heart failure often result in abnormal conduction, causing dyssynchronous contraction, and development of ventricular arrhythmias, which can lead to sudden cardiac arrest. In the last 2 decades, implantable cardioverter-defibrillator and cardiac resynchronization therapy devices have been developed to address these complications. However, when the coexisting chronic kidney disease is advanced, the associated pathophysiologic cardiovascular changes can alter the efficacy and safety of those interventions and complicate the management. This review explores the impact of comorbid advanced heart failure and advanced chronic kidney disease on the efficacy and safety of implantable cardioverter-defibrillator and cardiac resynchronization therapy, the currently available evidence, and potential future directions.
Collapse
Affiliation(s)
- Cecilia R Varga
- University of Florida, College of Medicine, Gainesville, Florida, USA
| | - John G F Cleland
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - William T Abraham
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Francisco Leyva
- Aston Medical School, Aston University, Birmingham, United Kingdom
| | - Parta Hatamizadeh
- University of Florida, College of Medicine, Gainesville, Florida, USA; Division of Nephrology, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
32
|
Chakraborty P, Nattel S, Nanthakumar K, Connelly KA, Husain M, Po SS, Ha ACT. Sudden cardiac death due to ventricular arrhythmia in diabetes mellitus: A bench to bedside review. Heart Rhythm 2024; 21:1827-1837. [PMID: 38848857 DOI: 10.1016/j.hrthm.2024.05.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024]
Abstract
Diabetes mellitus (DM) confers an increased risk of sudden cardiac death (SCD) independent of its associated cardiovascular comorbidities. DM induces adverse structural, electrophysiologic, and autonomic cardiac remodeling that can increase one's risk of ventricular arrhythmias and SCD. Although glycemic control and prevention of microvascular and macrovascular complications are cornerstones in the management of DM, they are not adequate for the prevention of SCD. In this narrative review, we describe the contribution of DM to the pathophysiologic mechanism of SCD beyond its role in atherosclerotic cardiovascular disease and heart failure. On the basis of this pathophysiologic framework, we outline potential preventive and therapeutic strategies to mitigate the risk of SCD in this population of high-risk patients.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Kumaraswamy Nanthakumar
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mansoor Husain
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Sunny S Po
- Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew C T Ha
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
33
|
Soomro Q, Mezzano V, Narula N, Rapkiewicz A, Loomis C, Charytan DM. Comparison of Cardiac Autonomic Innervation in Postmortem Tissue from Individuals with Kidney Failure and Preserved Kidney Function. Clin J Am Soc Nephrol 2024; 19:1319-1322. [PMID: 38869950 PMCID: PMC11469776 DOI: 10.2215/cjn.0000000000000477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Affiliation(s)
- Qandeel Soomro
- Nephrology Division, New York Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Valeria Mezzano
- Division of Advanced Research Technologies, Experimental Pathology [RRID:SCR_017928], New York Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Navneet Narula
- Department of Pathology, New York Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Amy Rapkiewicz
- Department of Pathology, New York Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Cynthia Loomis
- Division of Advanced Research Technologies, Experimental Pathology [RRID:SCR_017928], New York Grossman School of Medicine, NYU Langone Health, New York, New York
| | - David M. Charytan
- Nephrology Division, New York Grossman School of Medicine, NYU Langone Health, New York, New York
| |
Collapse
|
34
|
Salavatian S, Wong B, Kuwabara Y, Fritz JR, Varghese CG, Howard-Quijano K, Armour JA, Foreman RD, Ardell JL, Mahajan A. Comparing the Memory Effects of 50-Hz Low-Frequency and 10-kHz High-Frequency Thoracic Spinal Cord Stimulation on Spinal Neural Network in a Myocardial Infarction Porcine Model. Neuromodulation 2024; 27:1177-1186. [PMID: 39078348 DOI: 10.1016/j.neurom.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 07/31/2024]
Abstract
OBJECTIVE This study evaluated the effects of cessation of both conventional low-frequency (50 Hz) and high-frequency (10 kHz) spinal cord stimulation (SCS) on the cardiospinal neural network activity in pigs with myocardial infarction (MI). The objective is to provide an insight into the memory effect of SCS. MATERIALS AND METHODS In nine Yorkshire pigs, chronic MI was created by delivering microspheres to the left circumflex coronary artery. Five weeks after MI, anesthetized pigs underwent sternotomy to expose the heart for performing acute ischemia intervention, and laminectomy to expose the T1-T4 spinal regions for extracellular in vivo neural recording and SCS. Cardiac ischemic-sensitive neurons were identified by selective responsiveness to left anterior descending (LAD) coronary artery occlusion. SCS episodes were delivered in a random order between low- (50 Hz) and high- (10 kHz) frequency, for 1 minute, at 90% of the motor threshold current. Neural firing and synchrony of ischemic-sensitive spinal neurons were evaluated before vs after SCS. RESULTS Using a 64-channel microelectrode array, 2711 spinal neurons were recorded extracellularly. LAD ischemia excited 228 neurons that were labeled as ischemic-responsive neurons. The cessation of 50-Hz SCS caused a higher activation than did inhibition of ischemic-responsive neurons (41 activated vs 19 inhibited), whereas the cessation of 10-kHz SCS caused an opposite response with higher inhibition (11 activated vs 28 inhibited, p < 0.01 vs 50 Hz). Termination of low-frequency SCS caused an increase in ischemic-responsive neuronal firing rate compared with high-frequency SCS (50 Hz: 0.39 Hz ± 0.16 Hz, 10 kHz: -0.11 Hz ± 0.057 Hz, p < 0.01). In addition, SCS delivered at 50 Hz increased the number of synchronized pairs of neurons by 205 pairs, whereas high-frequency SCS decreased the number of synchronized pairs by 345 pairs (p < 0.01). CONCLUSIONS High-frequency (10 kHz) stimulation provides persistent suppression of the ischemia-sensitive neurons after termination of SCS. In contrast, the spinal neural network reverted to excitatory state after termination of low-frequency (50 Hz) stimulation.
Collapse
Affiliation(s)
- Siamak Salavatian
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Medicine, Division of Cardiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin Wong
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuki Kuwabara
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan R Fritz
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher G Varghese
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kimberly Howard-Quijano
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Andrew Armour
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert D Foreman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jeffrey L Ardell
- Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Aman Mahajan
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
35
|
XI H, LI X, ZHANG Z, CUI X, JING X, ZHU B, GAO X. Neuro- and immuno-modulation mediated by the cardiac sympathetic nerve: a novel insight into the anti-ischemic efficacy of acupuncture. J TRADIT CHIN MED 2024; 44:1058-1066. [PMID: 39380238 PMCID: PMC11462539 DOI: 10.19852/j.cnki.jtcm.20240423.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/15/2024] [Indexed: 10/14/2024]
Abstract
Communication between sympathetic nerves and the immune system is a crucial and active process during myocardial ischemia (MI), as myocardial damage and inflammatory stimuli concurrently occur. Sympathetic nerves undergo structural and functional changes after MI, leading to adverse left ventricular (LV) remodeling and heart failure (HF). The complex inflammatory response to MI, including local myocardial anti-inflammatory repair and systemic immune reactions, plays a key role in adverse LV remodeling. Here, we review the progressive structural and electrophysiological remodeling of the LV and the involvement of sympathetic tone in complex and dynamic processes that are susceptible to MI pathological conditions. Acupuncture has been reported to effectively improve cardiac function, eliminate arrhythmia, and mitigate adverse LV remodeling via somatosensory regulation after MI. Moreover, acupuncture has an anti-inflammatory effect on the pathological process of myocardial ischemia. In this Review, we aim to summarize the involvement of sympathetic nerve activation in the neuro-immune modulation of structural and functional cardiac changes after MI. As a noninvasive method for sympathetic regulation, acupuncture is an ideal option because of its anti-ischemic efficacy. A better understanding of the neural circuitry that regulates cardiac function and immune responses following MI could reveal novel targets for acupuncture treatment.
Collapse
Affiliation(s)
- Hanqing XI
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xia LI
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ziyi ZHANG
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiang CUI
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xianghong JING
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bing ZHU
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xinyan GAO
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
36
|
Plott C, Harb T, Arvanitis M, Gerstenblith G, Blumenthal R, Leucker T. Neurocardiac Axis Physiology and Clinical Applications. IJC HEART & VASCULATURE 2024; 54:101488. [PMID: 39224460 PMCID: PMC11367645 DOI: 10.1016/j.ijcha.2024.101488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
The neurocardiac axis constitutes the neuronal circuits between the arteries, heart, brain, and immune organs (including thymus, spleen, lymph nodes, and mucosal associated lymphoid tissue) that together form the cardiovascular brain circuit. This network allows the individual to maintain homeostasis in a variety of environmental situations. However, in dysfunctional states, such as exposure to environments with chronic stressors and sympathetic activation, this axis can also contribute to the development of atherosclerotic vascular disease as well as other cardiovascular pathologies and it is increasingly being recognized as an integral part of the pathogenesis of cardiovascular disease. This review article focuses on 1) the normal functioning of the neurocardiac axis; 2) pathophysiology of the neurocardiac axis; 3) clinical implications of this axis in hypertension, atherosclerotic disease, and heart failure with an update on treatments under investigation; and 4) quantification methods in research and clinical practice to measure components of the axis and future research areas.
Collapse
Affiliation(s)
- Caroline Plott
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Tarek Harb
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Marios Arvanitis
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Gary Gerstenblith
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Roger Blumenthal
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Thorsten Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
37
|
Atabekov TA, Sazonova SI, Khlynin MS, Muslimova EF, Krivolapov SN, Kurlov IO, Rebrova TY, Mishkina AI, Afanasiev SA, Batalov RE, Popov SV. Predictors of appropriate therapies delivered by the implantable cardioverter-defibrillator in patients with coronary artery disease during long-term period. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2024; 40:1863-1874. [PMID: 38963590 DOI: 10.1007/s10554-024-03172-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024]
Abstract
This prospective study aimed to investigate the ability of cardiac autonomic nervous system (CANS) activity assessment to predict appropriate implantable cardioverter-defibrillator (ICD) therapy in patients with coronary artery disease (CAD) during long-term follow-up period. We enrolled patients with CAD and ICD implantation indications that included both secondary and primary prevention of sudden cardiac death. Before ICD implantation CANS was assessed by using heart rate variability (HRV), myocardium scintigraphy with 123I-meta-iodobenzylguanidine (123I-MIBG) and erythrocyte membranes β-adrenoreactivity (EMA). The study's primary endpoint was the documentation of appropriate ICD therapy. Of 45 (100.0%) patients, 15 (33.3%) had appropriate ICD therapy during 36 months follow-up period. Patients with appropriate ICD therapy were likely to have a higher summed 123I-MIBG score delayed (p < 0.001) and lower 123I-MIBG washout rate (p = 0.008) indicators. These parameters were independently associated with endpoint in univariable and multivariable logistic regression. We created a logistic equation and calculated a cut-off value. The resulting ROC curve revealed a discriminative ability with AUC of 0.933 (95% confidence interval 0.817-0.986; sensitivity 100.00%; specificity 93.33%). Combined CANS activity assessment is useful in prediction of appropriate ICD therapy in patients with CAD during long-term follow-up period after device implantation.
Collapse
Affiliation(s)
- Tariel A Atabekov
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia.
| | - Svetlana I Sazonova
- Department of Nuclear Medicine, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Mikhail S Khlynin
- Laboratory of High Technologies for Diagnostics and Treatment of Cardiac Arrhythmias, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Elvira F Muslimova
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Sergey N Krivolapov
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Igor O Kurlov
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Tatiana Yu Rebrova
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Anna I Mishkina
- Department of Nuclear Medicine, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Sergey A Afanasiev
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Roman E Batalov
- Laboratory of High Technologies for Diagnostics and Treatment of Cardiac Arrhythmias, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| | - Sergey V Popov
- Academician of the Russian Academy of Sciences, Director, Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Russian Federation, Kievskaya st., 111a, Tomsk, 634012, Russia
| |
Collapse
|
38
|
Hanna P, Ardell JL. Cardiac Neuroanatomy and Fundamentals of Neurocardiology. Card Electrophysiol Clin 2024; 16:229-237. [PMID: 39084716 DOI: 10.1016/j.ccep.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Cardiac control is mediated via nested-feedback reflex control networks involving the intrinsic cardiac ganglia, intra-thoracic extra-cardiac ganglia, spinal cord, brainstem, and higher centers. This control system is optimized to respond to normal physiologic stressors; however, it can be catastrophically disrupted by pathologic events such as myocardial ischemia. In fact, it is now recognized that cardiac disease progression reflects the dynamic interplay between adverse remodeling of the cardiac substrate coupled with autonomic dysregulation. With advances in understanding of this network dynamic in normal and pathologic states, neuroscience-based neuromodulation therapies can be devised for the management of acute and chronic cardiac pathologies.
Collapse
Affiliation(s)
- Peter Hanna
- University of California Los Angeles (UCLA) Cardiac Arrhythmia Center, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; UCLA Neurocardiology Research Program of Excellence, UCLA, Los Angeles, CA 90095, USA
| | - Jeffrey L Ardell
- University of California Los Angeles (UCLA) Cardiac Arrhythmia Center, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; UCLA Neurocardiology Research Program of Excellence, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
39
|
Li P, Chang Y, Song J. Advances in preclinical surgical therapy of cardiovascular diseases. Int J Surg 2024; 110:4965-4975. [PMID: 38701509 PMCID: PMC11326035 DOI: 10.1097/js9.0000000000001534] [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: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
Cardiovascular disease is the most common cause of death worldwide, resulting in millions of deaths annually. Currently, there are still some deficiencies in the treatment of cardiovascular diseases. Innovative surgical treatments are currently being developed and tested in response to this situation. Large animal models, which are similar to humans in terms of anatomy, physiology, and genetics, play a crucial role in connecting basic research and clinical applications. This article reviews recent preclinical studies and the latest clinical advancements in cardiovascular disease based on large animal models, with a focus on targeted delivery, neural regulation, cardiac remodeling, and hemodynamic regulation. It provides new perspectives and ideas for clinical translation and offers new methods for clinical treatment.
Collapse
Affiliation(s)
- Peiyuan Li
- Department of Cardiac Surgery, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | | | | |
Collapse
|
40
|
Kvitka D, Pauza DH. Anatomy of blood microcirculation in the pig epicardial ganglionated nerve plexus. Ann Anat 2024; 255:152285. [PMID: 38830557 DOI: 10.1016/j.aanat.2024.152285] [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: 03/21/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Embolization of coronary arteries and their terminal arterioles causes ischemia of all tissues distributed within a cardiac wall including the intrinsic cardiac ganglionated nerve plexus (ICGP). The disturbed blood supply to the ICGP causes chronic sympathetic activation with succeeding atrial and ventricular arrhythmias. This study analyses the anatomy of microcirculation of epicardial nerves and ganglia using the hearts of 11 domestic pigs. Our findings demonstrate that thicker epicardial nerves are normally supplied with blood via 12 epineural arterioles penetrating the endoneurium regularly along a nerve, and forming an endoneurial capillary network, which drains the blood into the myocardial blood flow. The mean diameter of intraneural capillaries was 7.2 ± 0.2 µm, while the diameters of arterioles were 25.8 ± 0.7 μm and involved 45 endothelial cells accompanied by circular smooth muscle cells. Usually, two or three arterioles with a mean diameter of 28.9 ± 1.7 μm supplied blood to any epicardial ganglion, in which arterioles proceeded into a network of capillaries with a mean diameter of 6.9 ± 0.3 μm. Both the epicardial nerves and the ganglia distributed near the porta venarum of the heart had tiny arterioles that anastomosed blood vessels from the right and the left coronary arteries. The density of blood vessels in the epicardial nerves was significantly lesser compared with the ganglia. Our electron microscopic observations provided evidence that blood vessels of the pig epicardial nerves and ganglia may be considered as either arterioles or capillaries that have quantitative and qualitative differences comparing to the corresponding blood vessels in humans and, therefore, a pig should not be considered as an animal model of the first choice for further heart functional studies seeking to improve the treatment of cardiac arrhythmias via trans-coronary cardiac neuroablation. STRUCTURED ABSTRACT: This study details the anatomy of microcirculation of epicardial nerves and ganglia, from which intracardiac nerves and bundles of nerve fibers extend into all layers of the atrial and ventricular walls in the most popular animal model of experimental cardiology and cardiac surgery - the domestic pig. Our findings provided evidence that blood vessels of the pig epicardial nerves and ganglia may be considered as either arterioles or capillaries that have quantitative and qualitative differences comparing to the corresponding blood vessels in humans and, therefore, a pig should not be considered as an animal model of the first choice for further heart functional studies seeking to improve the treatment of cardiac arrhythmias via trans-coronary cardiac neuroablation.
Collapse
Affiliation(s)
- Dmitrij Kvitka
- Institute of Anatomy, Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickeviciaus Street 9, Kaunas LT 44307, Lithuania
| | - Dainius H Pauza
- Institute of Anatomy, Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickeviciaus Street 9, Kaunas LT 44307, Lithuania.
| |
Collapse
|
41
|
Zheng K, Hao Y, Xia C, Cheng S, Yu J, Chen Z, Li Y, Niu Y, Ran S, Wang S, Ye W, Luo Z, Li X, Zhao J, Li R, Zong J, Zhang H, Lai L, Huang P, Zhou C, Xia J, Zhang X, Wu J. Effects and mechanisms of the myocardial microenvironment on cardiomyocyte proliferation and regeneration. Front Cell Dev Biol 2024; 12:1429020. [PMID: 39050889 PMCID: PMC11266095 DOI: 10.3389/fcell.2024.1429020] [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: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
The adult mammalian cardiomyocyte has a limited capacity for self-renewal, which leads to the irreversible heart dysfunction and poses a significant threat to myocardial infarction patients. In the past decades, research efforts have been predominantly concentrated on the cardiomyocyte proliferation and heart regeneration. However, the heart is a complex organ that comprises not only cardiomyocytes but also numerous noncardiomyocyte cells, all playing integral roles in maintaining cardiac function. In addition, cardiomyocytes are exposed to a dynamically changing physical environment that includes oxygen saturation and mechanical forces. Recently, a growing number of studies on myocardial microenvironment in cardiomyocyte proliferation and heart regeneration is ongoing. In this review, we provide an overview of recent advances in myocardial microenvironment, which plays an important role in cardiomyocyte proliferation and heart regeneration.
Collapse
Affiliation(s)
- Kexiao Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoxian Cheng
- Jingshan Union Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
42
|
Zafeiropoulos S, Ahmed U, Bikou A, Mughrabi IT, Stavrakis S, Zanos S. Vagus nerve stimulation for cardiovascular diseases: Is there light at the end of the tunnel? Trends Cardiovasc Med 2024; 34:327-337. [PMID: 37506989 DOI: 10.1016/j.tcm.2023.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/12/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023]
Abstract
Autonomic dysfunction and chronic inflammation contribute to the pathogenesis and progression of several cardiovascular diseases (CVD), such as heart failure with preserved ejection fraction, atherosclerotic CVD, pulmonary arterial hypertension, and atrial fibrillation. The vagus nerve provides parasympathetic innervation to the heart, vessels, and lungs, and is also implicated in the neural control of inflammation through a neuroimmune pathway involving the spleen. Stimulation of the vagus nerve (VNS) can in principle restore autonomic balance and suppress inflammation, with potential therapeutic benefits in these diseases. Although VNS ameliorated CVD in several animal models, early human studies have demonstrated variable efficacy. The purpose of this review is to discuss the rationale behind the use of VNS in the treatment of CVD, to critically review animal and human studies of VNS in CVD, and to propose possible means to overcome the challenges in the clinical translation of VNS in CVD.
Collapse
Affiliation(s)
- Stefanos Zafeiropoulos
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, USA; Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Umair Ahmed
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Alexia Bikou
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ibrahim T Mughrabi
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Stavros Stavrakis
- Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stavros Zanos
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, USA; Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
43
|
Connolly E, Knight SP, Duggan E, Scarlett S, Newman L, Cahill M, Kenny RA, Doyle SL, Romero-Ortuno R. Cardiovascular Autonomic Function and Progression of Age-Related Macular Degeneration in The Irish Longitudinal Study of Ageing (TILDA). Invest Ophthalmol Vis Sci 2024; 65:24. [PMID: 38874963 PMCID: PMC11182369 DOI: 10.1167/iovs.65.6.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 05/08/2024] [Indexed: 06/15/2024] Open
Abstract
Purpose To examine if changes in hemodynamic measures during an orthostatic challenge were associated with progression of age-related macular degeneration (AMD) over a 4-year period in The Irish Longitudinal Study on Ageing. Methods Participants with AMD who underwent an active stand (AS) test at wave 1 (2009/2010) and retinal photographs at both wave 1 and wave 3 (2014/2015) were included (N = 159: 121 with no AMD progression and 38 with progression). Beat-to-beat hemodynamic data were non-invasively collected using a Finometer MIDI device during the AS at wave 1, recording systolic blood pressure (sBP), diastolic blood pressure (dBP), mean arterial pressure (MAP), and heart rate. Cardiac output, stroke volume, and total peripheral resistance (TPR) were derived from these measures. Baseline characteristics were compared between groups with and without AMD progression. Mixed-effects linear regression models were used to assess the association between changes in hemodynamic parameters during the AS and AMD progression, controlling for known AMD-associated risk factors. Results At baseline, increasing age and lower dBP were significantly associated with AMD progression. Mixed-effects models for the period between standing and 10 seconds post-stand revealed significant associations with AMD progression with a steeper drop in dBP and a slower drop in TPR. Between 10 and 20 seconds post-stand, AMD progression was significantly associated with less pronounced reduction in heart rate. Conclusions These observational data suggest that impaired hemodynamic responses within the first 20 seconds of orthostasis may be associated with the progression of AMD.
Collapse
Affiliation(s)
- Emma Connolly
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Silvin P. Knight
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Eoin Duggan
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Scarlett
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Louise Newman
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Mark Cahill
- Progressive Vision Research, Dublin, Ireland
| | - Rose Anne Kenny
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sarah L. Doyle
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Roman Romero-Ortuno
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
44
|
Osei J, Vaccarino V, Wang M, Shah AS, Lampert RJ, Li LY, Ko YA, Pearce BD, Kutner M, Garcia E, Piccinelli M, Raggi P, Bremner JD, Quyyumi AA, Sun YV, Ahmed H, Haddad G, Daaboul O, Roberts T, Stefanos L, Correia L, Shah AJ. Stress-Induced Autonomic Dysfunction is Associated With Mental Stress-Induced Myocardial Ischemia in Patients With Coronary Artery Disease. Circ Cardiovasc Imaging 2024; 17:e016596. [PMID: 38868952 PMCID: PMC11187646 DOI: 10.1161/circimaging.124.016596] [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: 01/10/2024] [Accepted: 04/30/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Mental stress-induced myocardial ischemia (MSIMI) is associated with adverse cardiovascular outcomes in individuals with coronary artery disease, but the mechanisms underlying this phenomenon are unknown. We examined the relationship between stress-induced autonomic dysfunction, measured by low heart rate variability (HRV) in response to stress, and MSIMI in patients with stable coronary artery disease. We hypothesized that stress-induced autonomic dysfunction is associated with higher odds of MSIMI. METHODS In 735 participants with stable coronary artery disease, we measured high- and low-frequency HRV in 5-minute intervals before and during a standardized laboratory-based speech stressor using Holter monitoring. HRV at rest and stress were categorized into low HRV (first quartile) versus high HRV (second to fourth quartiles); the low category was used as an indicator of autonomic dysfunction. Multivariable logistic regression models were used to examine the association of autonomic dysfunction with MSIMI. RESULTS The mean age was 58 (SD, ±10) years, 35% were women, 44% were Black participants, and 16% developed MSIMI. Compared with high HRV during stress, low HRV during stress (both high and low frequencies) was associated with higher odds of MSIMI after adjusting for demographic and clinical factors (odds ratio for high-frequency HRV, 2.1 [95% CI, 1.3-3.3]; odds ratio for low-frequency HRV, 2.1 [95% CI, 1.3-3.3]). Low-frequency HRV at rest was also associated with MSIMI but with slightly reduced effect estimates. CONCLUSIONS In individuals with coronary artery disease, mental stress-induced autonomic dysfunction may be a mechanism implicated in the causal pathway of MSIMI.
Collapse
Affiliation(s)
- Jeffery Osei
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maggie Wang
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anish S. Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Rachel J. Lampert
- Division of Cardiology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Louis Y. Li
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Brad D. Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Michael Kutner
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Ernest Garcia
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marina Piccinelli
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Paolo Raggi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - J. Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arshed A. Quyyumi
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yan V. Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Hashir Ahmed
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - George Haddad
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Obada Daaboul
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tatum Roberts
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lewam Stefanos
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Luis Correia
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Amit J. Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Emory Clinical Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
45
|
Depes D, Mennander A, Immonen P, Mäkinen A, Huhtala H, Paavonen T, Kholová I. The autonomic nerves around the vein of Marshall: a postmortem study with clinical implications. APMIS 2024; 132:430-443. [PMID: 38468591 DOI: 10.1111/apm.13400] [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: 03/30/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
This study aims to analyze the vein of Marshall (VOM) in human autopsy hearts and its correlation with clinical data to elucidate the morphological substrates of atrial fibrillation (AF) and other cardiac diseases. Twenty-three adult autopsy hearts were studied, assessing autonomic nerves by immunohistochemistry with tyrosine hydroxylase (sympathetic nerves), choline acetyltransferase (parasympathetic nerves), growth-associated protein 43 (neural growth), and S100 (general neural marker) antibodies. Interstitial fibrosis was assessed by Masson trichrome staining. Measurements were conducted via morphometric software. The results were correlated with clinical data. Sympathetic innervation was abundant in all VOM-adjacent regions. Subjects with a history of AF, cardiovascular cause of death, and histologically verified myocardial infarction had increased sympathetic innervation and neural growth around the VOM at the mitral isthmus. Interstitial fibrosis increased with age and heart weight was associated with AF and cardiovascular cause of death. This study increases our understanding of the cardiac autonomic innervation in the VOM area in various diseases, offering implications for the development of new therapeutic approaches targeting the autonomic nervous system.
Collapse
Affiliation(s)
- Denis Depes
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ari Mennander
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Cardiothoracic Surgery, Tampere University Heart Hospital, Tampere, Finland
| | - Paavo Immonen
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Artturi Mäkinen
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Timo Paavonen
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ivana Kholová
- Department of Pathology, Fimlab Laboratories, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
46
|
Bauer J, Vlcek J, Pauly V, Hesse N, Xia R, Mo L, Chivukula AS, Villgrater H, Dressler M, Hildebrand B, Wolf E, Rizas KD, Bauer A, Kääb S, Tomsits P, Schüttler D, Clauss S. Biomarker Periodic Repolarization Dynamics Indicates Enhanced Risk for Arrhythmias and Sudden Cardiac Death in Myocardial Infarction in Pigs. J Am Heart Assoc 2024; 13:e032405. [PMID: 38639363 PMCID: PMC11179938 DOI: 10.1161/jaha.123.032405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/08/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Periodic repolarization dynamics (PRD) is an electrocardiographic biomarker that captures repolarization instability in the low frequency spectrum and is believed to estimate the sympathetic effect on the ventricular myocardium. High PRD indicates an increased risk for postischemic sudden cardiac death (SCD). However, a direct link between PRD and proarrhythmogenic autonomic remodeling has not yet been shown. METHODS AND RESULTS We investigated autonomic remodeling in pigs with myocardial infarction (MI)-related ischemic heart failure induced by balloon occlusion of the left anterior descending artery (n=17) compared with pigs without MI (n=11). Thirty days after MI, pigs demonstrated enhanced sympathetic innervation in the infarct area, border zone, and remote left ventricle paralleled by altered expression of autonomic marker genes/proteins. PRD was enhanced 30 days after MI compared with baseline (pre-MI versus post-MI: 1.75±0.30 deg2 versus 3.29±0.79 deg2, P<0.05) reflecting pronounced autonomic alterations on the level of the ventricular myocardium. Pigs with MI-related ventricular fibrillation and SCD had significantly higher pre-MI PRD than pigs without tachyarrhythmias, suggesting a potential role for PRD as a predictive biomarker for ischemia-related arrhythmias (no ventricular fibrillation versus ventricular fibrillation: 1.50±0.39 deg2 versus 3.18±0.53 deg2 [P<0.05]; no SCD versus SCD: 1.67±0.32 deg2 versus 3.91±0.63 deg2 [P<0.01]). CONCLUSIONS We demonstrate that ischemic heart failure leads to significant proarrhythmogenic autonomic remodeling. The concomitant elevation of PRD levels in pigs with ischemic heart failure and pigs with MI-related ventricular fibrillation/SCD suggests PRD as a biomarker for autonomic remodeling and as a potential predictive biomarker for ventricular arrhythmias/survival in the context of MI.
Collapse
Affiliation(s)
- Julia Bauer
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Julia Vlcek
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Valerie Pauly
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Nora Hesse
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Ruibing Xia
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Li Mo
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Aparna Sharma Chivukula
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Hannes Villgrater
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Marie Dressler
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Bianca Hildebrand
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU MunichMunichGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU MunichMunichGermany
| | - Konstantinos D. Rizas
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
| | - Axel Bauer
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- University Hospital for Internal Medicine IIIMedical University of InnsbruckInnsbruckAustria
| | - Stefan Kääb
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU MunichMunichGermany
| | - Philipp Tomsits
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Dominik Schüttler
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
| | - Sebastian Clauss
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart AllianceMunichGermany
- Institute of Surgical Research at the Walter‐Brendel‐Centre of Experimental MedicineUniversity Hospital, LMU MunichMunichGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU MunichMunichGermany
| |
Collapse
|
47
|
Mabry SA, Pavon N. Exploring the prospects, advancements, and challenges of in vitro modeling of the heart-brain axis. Front Cell Neurosci 2024; 18:1386355. [PMID: 38766369 PMCID: PMC11099243 DOI: 10.3389/fncel.2024.1386355] [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: 02/15/2024] [Accepted: 04/12/2024] [Indexed: 05/22/2024] Open
Abstract
Research on bidirectional communication between the heart and brain has often relied on studies involving nonhuman animals. Dependance on animal models offer limited applicability to humans and a lack of high-throughput screening. Recently, the field of 3D cell biology, specifically organoid technology, has rapidly emerged as a valuable tool for studying interactions across organ systems, i.e., gut-brain axis. The initial success of organoid models indicates the usefulness of 3D cultures for elucidating the intricate interactivity of the autonomic nervous system and overall health. This perspective aims to explore the potential of advancing in vitro modeling of the heart-brain axis by discussing the benefits, applications, and adaptability of organoid technologies. We closely examine the current state of brain organoids in conjunction with the advancements of cardiac organoids. Moreover, we explore the use of combined organoid systems to investigate pathophysiology and provide a platform for treatment discovery. Finally, we address the challenges that accompany the use of 3D models for studying the heart-brain axis with an emphasis on generating tailored engineering strategies for further refinement of dynamic organ system modeling in vitro.
Collapse
Affiliation(s)
- Senegal Alfred Mabry
- Affect and Cognition Laboratory, Department of Psychology and Human Development, College of Human Ecology, Cornell University, Ithaca, NY, United States
| | - Narciso Pavon
- ChangHui Pak Laboratory, Department of Biochemistry and Molecular Biology, College of Natural Sciences, University of Massachusetts-Amherst, Amherst, MA, United States
| |
Collapse
|
48
|
Peng C, Lu Y, Li R, Zhang L, Liu Z, Xu X, Wang C, Hu R, Tan W, Zhou L, Wang Y, Yu L, Wang Y, Tang B, Jiang H. Neuroimmune modulation mediated by IL-6: A potential target for the treatment of ischemia-induced ventricular arrhythmias. Heart Rhythm 2024; 21:610-619. [PMID: 38160759 DOI: 10.1016/j.hrthm.2023.12.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/09/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Neural remodeling in the left stellate ganglion (LSG), as mediated by neuroimmune reactions, promotes cardiac sympathetic nerve activity (SNA) and thus increases the incidence of ventricular arrhythmias (VAs). Interleukin-6 (IL-6) is an important factor of the neuroimmune interaction. OBJECTIVE The present study explored the effects of IL-6 on LSG hyperactivity and the incidence of VAs. METHODS Eighteen beagles were randomly allocated to a control group (saline with myocardial infarction [MI], n = 6), adeno-associated virus (AAV) group (AAV with MI, n = 6), and IL-6 group (overexpression of IL-6 via AAV vector with MI, n = 6). Ambulatory electrocardiography was performed before and 30 days after AAV microinjection into the LSG. LSG function and ventricular electrophysiology were assessed at 31 days after surgery, and a canine MI model was established. Samples of the LSG were collected for immunofluorescence staining and molecular biological evaluation. Blood samples and 24-hour Holter data were obtained from 24 patients with acute MI on the day after they underwent percutaneous coronary intervention to assess the correlation between IL-6 levels and SNA. RESULTS IL-6 overexpression increased cardiac SNA and worsened postinfarction VAs. Furthermore, sustained IL-6 overexpression enhanced LSG function, promoted expression of nerve growth factor, c-fos, and fos B in the LSG, and activated the signal transducer and activator of transcription 3/regulator of G protein signalling 4 signaling pathway. Clinical sample analysis revealed a correlation between serum IL-6 levels and heart rate variability frequency domain index as well as T-wave alternans. CONCLUSION IL-6 levels are correlated with cardiac SNA. Chronic overexpression of IL-6 mediates LSG neural remodeling through the signal transducer and activator of transcription 3/regulator of G protein signalling 4 signaling pathway, elevating the risk of VA after MI.
Collapse
Affiliation(s)
- Chen Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Yanmei Lu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Rui Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Ling Zhang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Xiao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Changyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Ruijie Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Yuhong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Baopeng Tang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China.
| |
Collapse
|
49
|
Rajendran PS, Hanna P. The irate stellate ganglion: IL-6 in neuroinflammation-induced ventricular arrhythmias. Heart Rhythm 2024; 21:620-621. [PMID: 38286243 DOI: 10.1016/j.hrthm.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/31/2024]
Affiliation(s)
- Pradeep S Rajendran
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Peter Hanna
- University of California Los Angeles (UCLA) Cardiac Arrhythmia Center, David Geffen School of Medicine, UCLA, Los Angeles, California; Neurocardiology Research Program of Excellence, David Geffen School of Medicine, UCLA, Los Angeles, California.
| |
Collapse
|
50
|
Wu LF, Zhou Y, Wang DP, Zhang JJ, Zheng ZF, Guo J, Shen J, Shi JY, Liu QH, Wang XN, Wang HX, Du WJ, Li ML, Cao JM. Nerve growth factor (Ngf) gene-driven semaphorin 3a (Sema3a) expression exacerbates thoracic aortic aneurysm dissection in mice. J Hypertens 2024; 42:816-827. [PMID: 38165021 DOI: 10.1097/hjh.0000000000003647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening disease and currently there is no pharmacological therapy. Sympathetic nerve overactivity plays an important role in the development of TAAD. Sympathetic innervation is mainly controlled by nerve growth factor (NGF, a key neural chemoattractant) and semaphoring 3A (Sema3A, a key neural chemorepellent), while the roles of these two factors in aortic sympathetic innervation and especially TAAD are unknown. We hypothesized that genetically manipulating the NGF/Sema3A ratio by the Ngf -driven Sema3a expression approach may reduce aortic sympathetic nerve innervation and mitigate TAAD progression. A mouse strain of Ngf gene-driven Sema3a expression (namely NgfSema3a/Sema3a mouse) was established by inserting the 2A-Sema3A expression frame to the Ngf terminating codon using CRISPR/Cas9 technology. TAAD was induced by β-aminopropionitrile monofumarate (BAPN) both in NgfSema3a/Sema3a mice and wild type (WT) littermates. Contrary to our expectation, the BAPN-induced TAAD was severer in NgfSema3a/Sema3a mice than in wild-type (WT) mice. In addition, NgfSema3a/Sema3a mice showed higher aortic sympathetic innervation, inflammation and extracellular matrix degradation than the WT mice after BAPN treatment. The aortic vascular smooth muscle cells isolated from NgfSema3a/Sema3a mice and pretreated with BAPN in vivo for two weeks showed stronger capabilities of proliferation and migration than that from the WT mice. We conclude that the strategy of Ngf -driven Sema3a expression cannot suppress but worsens the BAPN-induced TAAD. By investigating the aortic phenotype of NgfSema3a/Sema3a mouse strain, we unexpectedly find a path to exacerbate BAPN-induced TAAD which might be useful in future TAAD studies.
Collapse
Affiliation(s)
- Li-Fei Wu
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Physiology, Shanxi Medical University
- Department of Pathophysiology, Shanxi Medical University
| | - Ying Zhou
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Physiology, Shanxi Medical University
| | - De-Ping Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Physiology, Shanxi Medical University
| | - Jiao-Jiao Zhang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Physiology, Shanxi Medical University
| | - Zhi-Fa Zheng
- Department of Cardiovascular Surgery, Shanxi Bethune Hospital
| | - Jia Guo
- Center for Hypertension Care, Shanxi Medical University First Hospital
| | - Jing Shen
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Physiology, Shanxi Medical University
| | - Jian-Yun Shi
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Physiology, Shanxi Medical University
| | - Qing-Hua Liu
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Pathophysiology, Shanxi Medical University
| | - Xue-Ning Wang
- Department of Cardiovascular Surgery, Shanxi Bethune Hospital
| | - Hai-Xiong Wang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan
| | - Wen-Jing Du
- State Key Laboratory of Medical Molecular Biology, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing
| | - Miao-Ling Li
- Key Laboratory of Medical Electrophysiology at Southwest Medical University, Ministry of Education, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education
- Department of Physiology, Shanxi Medical University
| |
Collapse
|