1
|
Wang T, Wu J, Qin F, Jiang H, Xiao X, Tong Y, Liao C, Huang Z. Physiological modeling of autonomic regulation of cardiac system under graded exercise. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 264:108704. [PMID: 40096784 DOI: 10.1016/j.cmpb.2025.108704] [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: 09/25/2024] [Revised: 12/14/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND AND OBJECTIVE Dysfunction of the autonomic nervous system (ANS) plays a critical role in the progression and assessment of cardiovascular diseases, neurological disorders, and various other pathologies. Therefore, a quantitative assessment of ANS function is vital for personalized medicine in these diseases. However, direct measurements of ANS activity can be costly and invasive, prompting researchers to adopt indirect methods for quantitative evaluation. These methods typically involve mathematical techniques, such as statistical analysis and mathematical modeling, to interpret cardiovascular fluctuations in response to external stimuli.The purpose of this study is to develop a non-invasive mathematical method that quantitatively assesses ANS function during graded exercise. METHODS In this study, we present a physiological mathematical model for autonomic regulation of the cardiac system under graded exercise, which recognizes the crucial role of the ANS in controlling heart rate during physical activity. The model utilizes the metabolic equivalent of walking as the input and heart rate as the output, with model parameters serving as quantitative measures of personalized ANS function. Experimental data were collected from groups with different health statuses and genders. Mann-Whitney U non-parametric tests were conducted on the model parameters to assess performance between individuals who frequently engage in aerobic exercise (15 participants, aerobic exercise frequency of more than 4 times/week) and those who barely exercise (15 participants, aerobic exercise frequency of 1 time per week or less), as well as between male and female participants. RESULTS The experimental results indicate that our model effectively quantitatively assesses ANS function across groups with different health statuses and genders (P < 0.05). Additionally, the model provides precise estimations of heart rate, yielding a Root Mean Square Error of 2.79 beats per minute, a Mean Absolute Error of 2.18 beats per minute, and an R-squared value of 0.93. CONCLUSION Our findings suggest that the proposed physiological mathematical model offers a non-invasive and user-friendly tool for measuring ANS function and monitoring cardiovascular health. This approach is feasible for home application, thereby reducing the need for professional supervision, and supports the early detection and personalized management of cardiovascular diseases. As a result, it enhances clinical decision-making and improves patient outcomes.
Collapse
Affiliation(s)
- Tao Wang
- University of Chinese Academy of Sciences, Beijing 101408, China.
| | - JianKang Wu
- University of Chinese Academy of Sciences, Beijing 101408, China; CAS Institute of Healthcare Technologies, Nanjing, 210000, China.
| | - Fei Qin
- University of Chinese Academy of Sciences, Beijing 101408, China.
| | - Hong Jiang
- Department of Integrative Cardiology, National Center for Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Xiang Xiao
- Department of Integrative Cardiology, National Center for Integrative Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - YongGang Tong
- University of Chinese Academy of Sciences, Beijing 101408, China.
| | - ChuChu Liao
- University of Chinese Academy of Sciences, Beijing 101408, China.
| | - ZhiPei Huang
- University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
2
|
Androshchuk V, Montarello N, Rajani R. Adverse Cardiovascular Effects of Psychotropic Medications. Br J Hosp Med (Lond) 2025; 86:1-18. [PMID: 40265538 DOI: 10.12968/hmed.2024.0773] [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: 04/24/2025]
Abstract
Cardiovascular disease is the leading cause of death in people with serious mental health illnesses, including schizophrenia, major depression and bipolar disorder. The adverse cardiac risk profile of this population is related to the complex interplay between biological, patient-specific and healthcare system factors. A variety of psychotropic medications used to treat these conditions can in themselves produce cardiovascular side effects. This includes autonomic dysfunction, malignant ventricular arrhythmias, heart muscle disorders and vascular thromboembolic events, some of which have been linked with sudden cardiac death. As a result, there is a pressing need for physicians to be aware of the cardio-toxicity associated with psychotropic medication use. In this review, we summarise the main effects of psychotropic drugs on the cardiovascular system and the current recommendations for evaluation and continual monitoring of the many and rapidly increasing number of patients receiving psychotropic pharmacotherapy.
Collapse
Affiliation(s)
- Vitaliy Androshchuk
- School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Natalie Montarello
- Department of Cardiology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ronak Rajani
- Department of Cardiology, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| |
Collapse
|
3
|
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
|
4
|
Ali KA, Kerrigan DLG, Berkman JM. Influence of Primary Neurologic Disease on Cardiovascular Health in Females. Circ Res 2025; 136:618-627. [PMID: 40080534 DOI: 10.1161/circresaha.124.325545] [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: 10/25/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 03/15/2025]
Abstract
Neurocardiology is an interdisciplinary field that examines the complex interactions between the nervous and the cardiovascular systems, exploring how neurological processes, such as autonomic nervous system regulation and brain-heart communication impact heart function and contribute to cardiovascular health and disease. Although much of the focus on cardiovascular health has centered on traditional risk factors, the influence of the nervous system, especially in females, is increasingly recognized as a key determinant of cardiovascular outcomes. This article reviews existing literature on the neurological mechanisms that impact cardiovascular function in females. Specifically, we analyze how primary neurological disorders including cerebrovascular disease, headache disorders, and multiple sclerosis have specific downstream effects on cardiac function. By understanding the complex relationship between neurological and cardiovascular health, this review highlights the need for sex-specific approaches to prevention, diagnosis, and treatment of cardiovascular disease in females, ultimately encouraging the discovery of more effective care strategies and improving health outcomes.
Collapse
Affiliation(s)
- Khadija Awais Ali
- Department of Neurology, Stroke Divison, Vanderbilt University Medical Center, Nashville, TN
| | - Deborah L G Kerrigan
- Department of Neurology, Stroke Divison, Vanderbilt University Medical Center, Nashville, TN
| | - Jillian Molli Berkman
- Department of Neurology, Stroke Divison, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
5
|
Liu Chung Ming C, Patil R, Refaat A, Lal S, Wang X, Gentile C. Acetylcholine-loaded nanoparticles protect against doxorubicin-induced toxicity in in vitrocardiac spheroids. Biofabrication 2025; 17:025023. [PMID: 39965540 DOI: 10.1088/1758-5090/adb7c2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Doxorubicin (DOX) is widely used in chemotherapy, yet it significantly contributes to heart failure-associated death. Acetylcholine (ACh) is cardioprotective by enhancing heart rate variability and reducing mitochondrial dysfunction and inflammation. Nonetheless, the protective role of ACh in countering DOX-induced cardiotoxicity (DIC) remains underexplored as current approaches to increasing ACh levels are invasive and unsafe for patients. In this study, we explore the protective effects of ACh against DIC through three distinct ACh administration strategies: (i) freely-suspended 100µM ACh; (ii) ACh-producing cholinergic neurons (CNs); or (iii) ACh-loaded nanoparticles (ACh-NPs). These are tested inin vitrocardiac spheroids (CSs), which have previously been shown to approximate the complex DIC. We assess ACh's protective effects by measuring the toxicity ratio (cell death/viability), contractile activity, gene expression changes via qPCR and nitric oxide (NO) signaling. Our findings show that ACh effectively attenuates DOX-induced cell death and contractile dysfunction. ACh also counteracts the DOX-induced downregulation of genes controlling myocardial fibrosis, endothelial and cardiomyocyte dysfunction, and autonomic dysregulation. ACh cardioprotection against DOX is dependent on NO signaling in endothelial cells but not in cardiac myocytes or fibroblasts. Altogether, this study shows for the first time that elevating ACh levels showed a promising therapeutic approach for preventing DIC.
Collapse
Affiliation(s)
- Clara Liu Chung Ming
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| | - Runali Patil
- IIT-Bombay Monash Research Academy, IIT Bombay, Powai, Mumbai, Maharashtra 400076, India
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Ahmed Refaat
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Xiaowei Wang
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Tompkins JD, Hoover DB, Havton LA, Patel JC, Cho Y, Smith EH, Biscola NP, Ajijola OA, Shivkumar K, Ardell JL. Comparative specialization of intrinsic cardiac neurons in humans, mice and pigs. J Physiol 2025; 603:2043-2070. [PMID: 39513933 PMCID: PMC11957936 DOI: 10.1113/jp286714] [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: 04/19/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Intrinsic cardiac neurons (ICNs) play a crucial role in the proper functioning of the heart; yet a paucity of data pertaining to human ICNs exist. We took a multidisciplinary approach to complete a detailed cellular comparison of the structure and function of ICNs from mice, pigs and humans. Immunohistochemistry of whole and sectioned ganglia, transmission electron microscopy, intracellular microelectrode recording and dye filling for quantitative morphometry were used to define the neurophysiology, histochemistry and ultrastructure of these neurons across species. The densely packed, smaller ICNs of mouse lacked dendrites, formed axosomatic connections and had high synaptic efficacy constituting an obligatory synapse. At pig ICNs, a convergence of subthreshold cholinergic inputs onto extensive dendritic arbors supported greater summation and integration of synaptic input. Human ICNs were tonically firing, with synaptic stimulation evoking large suprathreshold EPSPs like mouse, and subthreshold potentials like pig. Ultrastructural examination of synaptic terminals revealed conserved architecture, yet small clear vesicles were larger in pigs and humans. The presence and localization of ganglionic neuropeptides was distinct, with abundant vasoactive intestinal polypeptide observed in human but not pig or mouse ganglia, and little substance P or calcitonin gene-related peptide in pig ganglia. Action potential waveforms were similar, but human ICNs had larger after-hyperpolarizations. Intrinsic excitability differed; 95% of human neurons were tonic, all pig neurons were phasic, and both phasic and tonic phenotypes were observed in mouse. In combination, this publicly accessible, multimodal atlas of ICNs from mice, pigs and humans identifies similarities and differences in the evolution of ICNs. KEY POINTS: Intrinsic cardiac neurons (ICNs) are essential to the regulation of cardiac function. We investigated the neurochemistry, morphology, ultrastructure, membrane physiology and synaptic transmission of ICNs from donated human hearts in parallel with identical studies of ICNs from mice and pigs to create a publicly accessible cellular atlas detailing the structure and function of these neurons across species. In addition to presenting foundational data on human ICNs, this comparative study identifies both conserved and derived attributes of these neurons within mammals. The findings have significant implications for understanding the regulation of cardiac autonomic function in humans and may greatly influence strategies for neuromodulation in conditions such as atrial fibrillation and heart failure.
Collapse
Affiliation(s)
- John D. Tompkins
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Donald B. Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Leif A. Havton
- Departments of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Janaki C. Patel
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Youngjin Cho
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Elizabeth H. Smith
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Natalia P. Biscola
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jeffrey L. Ardell
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
9
|
Gupta S, Gee MM, Newton AJH, Kuttippurathu L, Moss A, Tompkins JD, Schwaber JS, Vadigepalli R, Lytton WW. Biophysical modelling of intrinsic cardiac nervous system neuronal electrophysiology based on single-cell transcriptomics. J Physiol 2025; 603:2119-2138. [PMID: 40077928 PMCID: PMC11955872 DOI: 10.1113/jp287595] [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: 08/30/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
The intrinsic cardiac nervous system (ICNS), termed as the heart's 'little brain', is the final point of neural regulation of cardiac function. Studying the dynamic behaviour of these ICNS neurons via multiscale neuronal computer models has been limited by the sparsity of electrophysiological data. We developed and analysed a computational library of neuronal electrophysiological models based on single neuron transcriptomic data obtained from ICNS neurons. Each neuronal genotype was characterized by a unique combination of ion channels identified from the transcriptomic data, using a cycle threshold cutoff that ensured the electrical excitability of the neuronal models. The parameters of the ion channel models were grounded based on passive properties (resting membrane potential, input impedance and rheobase) to avoid biasing the dynamic behaviour of the model. Consistent with experimental observations, the emergent model dynamics showed phasic activity in response to the current clamp stimulus in a majority of neuronal genotypes (61%). Additionally, 24% of the ICNS neurons showed a tonic response, 11% were phasic-to-tonic with increasing current stimulation and 3% showed tonic-to-phasic behaviour. The computational approach and the library of models bridge the gap between widely available molecular-level gene expression and sparse cellular-level electrophysiology for studying the functional role of the ICNS in cardiac regulation and pathology. KEY POINTS: Computational models were developed of neuron electrophysiology from single-cell transcriptomic data from neurons in the heart's 'little brain': the intrinsic cardiac nervous system. The single-cell transcriptomic data were thresholded to select the ion channel combinations in each neuronal model. The library of neuronal models was constrained by the passive electrical properties of the neurons and predicted a distribution of phasic and tonic responses that aligns with experimental observations. The ratios of model-predicted conductance values are correlated with the gene expression ratios from transcriptomic data. These neuron models are a first step towards connecting single-cell transcriptomic data to dynamic, predictive physiology-based models.
Collapse
Affiliation(s)
- Suranjana Gupta
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNYUSA
| | - Michelle M. Gee
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDEUSA
| | - Adam J. H. Newton
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNYUSA
| | - Lakshmi Kuttippurathu
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
| | - Alison Moss
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
| | - John D. Tompkins
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLAUniversity of CaliforniaLos AngelesCAUSA
| | - James S. Schwaber
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDEUSA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDEUSA
| | - William W. Lytton
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNYUSA
- Department of NeurologyKings County HospitalBrooklynNYUSA
| |
Collapse
|
10
|
Paterson DJ, Shivkumar K. Preface Editorial for Special Issue Dedicated to Jeffrey L. Ardell (1952-2025) Cardiac Neurobiology: Concepts to Clinic. J Physiol 2025; 603:1683-1686. [PMID: 40100704 DOI: 10.1113/jp288209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
|
11
|
Mazza M, Veneziani G, Lisci FM, Morini S, Traversi G, Sfratta G, Brisi C, Anesini MB, Bardi F, Benini E, Calderoni C, Chisari L, Crupi A, De Chiara E, Lo Giudice L, Onori L, Sessa I, Balocchi M, Pola R, Gaetani E, Simeoni B, Franceschi F, Sani G, Covino M, Lai C, Romagnoli E, Marano G. Mental Illness Strikes at the Heart: Impact of Psychiatric Diseases on Ventricular Ejection Fraction in Patients with Acute Coronary Syndromes. Life (Basel) 2025; 15:340. [PMID: 40141685 PMCID: PMC11944072 DOI: 10.3390/life15030340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/09/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Mental illnesses can have a significant impact on individuals experiencing acute coronary syndromes (ACS). Mental illnesses are associated with an increased cardiovascular risk profile and early onset of cardiovascular disease. A critical aspect of this interplay is the effect of psychiatric conditions on left ventricular ejection fraction (LVEF), a key parameter in evaluating cardiac function and predicting long-term outcomes in ACS patients. The present single-center, retrospective study investigated the associations between psychiatric conditions and cardiac function, with a focus on LVEF in ACS patients. The inclusion criteria were Italian nationality and 30 years or older. One hundred and sixty-four patients without (Mage = 68.8 ± 10.6, 62 females) and 161 patients with a psychiatric diagnosis (Mage = 68.4 ± 13.7, 63 females) were enrolled. The data collected included sociodemographic variables, psychiatric diagnoses, LVEF, ACS type (STEMI/NSTEMI), smoking status, previous interventions, and pharmacological treatments. Statistical analyses included chi-square, t-tests, ANOVAs, and ANCOVA to assess differences across groups. Findings revealed lower LVEF in patients with a psychiatric diagnosis compared to patients without a psychiatric diagnosis (p = 0.004, d = 0.36). Patients without a psychiatric diagnosis were associated with NSTEMI (p = 0.047, φ = 0.11), hypertension (p = 0.003, φ = -0.16), and dyslipidemia (p = 0.022, φ = -0.13). In contrast, patients with a psychiatric diagnosis were associated with STEMI (p = 0.047, φ = 0.11), neurological dysfunction (p = 0.014, φ = 0.14), and chronic obstructive pulmonary disease (p = 0.010, φ = 0.14). Among psychiatric diagnoses, anxiety disorders were associated with lower LVEF compared to substance abuse disorders (p = 0.012, d = -0.81). The findings underscore the complex relationship between mental illness and cardiac function, emphasising the need to integrate psychiatric evaluations into cardiology care to optimise the management of both mental and cardiovascular health. This study has several limitations, including its design, which prevents causal conclusions, and the use of convenience sampling, which limits the generalizability of the findings.
Collapse
Affiliation(s)
- Marianna Mazza
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Giorgio Veneziani
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University, Via degli Apuli 1, 00185 Rome, Italy
| | - Francesco Maria Lisci
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Sofia Morini
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy (E.R.)
| | - Gianandrea Traversi
- Unit of Medical Genetics, Department of Laboratory Medicine, Ospedale Isola Tiberina-Gemelli Isola, Via di Ponte Quattro Capi 39, 00186 Rome, Italy
| | - Greta Sfratta
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Caterina Brisi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Maria Benedetta Anesini
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Francesca Bardi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Elisabetta Benini
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Claudia Calderoni
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Luca Chisari
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Arianna Crupi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Emanuela De Chiara
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Luca Lo Giudice
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Luca Onori
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Ilenia Sessa
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Marta Balocchi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Roberto Pola
- Section of Internal Medicine and Thromboembolic Diseases, Department of Internal Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Eleonora Gaetani
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
- Unit of Internal Medicine, Cristo Re Hospital, Via delle Calasanziane 25, 00167 Rome, Italy
| | - Benedetta Simeoni
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (B.S.); (M.C.)
| | - Francesco Franceschi
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (B.S.); (M.C.)
| | - Gabriele Sani
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Marcello Covino
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (B.S.); (M.C.)
| | - Carlo Lai
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University, Via degli Apuli 1, 00185 Rome, Italy
| | - Enrico Romagnoli
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy (E.R.)
| | - Giuseppe Marano
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
12
|
Stanciulescu LA, Dorobantu M, Vatasescu R. Targeting Ventricular Arrhythmias in Non-Ischemic Patients: Advances in Diagnosis and Treatment. Diagnostics (Basel) 2025; 15:420. [PMID: 40002571 PMCID: PMC11854509 DOI: 10.3390/diagnostics15040420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Ventricular arrhythmias (VAs) in non-ischemic cardiomyopathy (NICM) present significant clinical challenges due to their diverse etiologies and complex arrhythmogenic substrates, which differ from those in ischemic heart disease. Recent advancements in imaging, electrophysiological mapping, and ablative therapy have improved the management of these arrhythmias. This review examines the spectrum of NICM subtypes, discussing their pathophysiology, prevalence, genetic determinants, and associated arrhythmias. It also explores contemporary ablative techniques, including epicardial, bipolar, and irrigated approaches, as well as emerging modalities such as stereotactic body radiation therapy (SBRT). The role of novel technologies, including high-resolution mapping and artificial intelligence, is considered in refining diagnosis and treatment. This article provides a comprehensive overview of current management strategies and discusses future directions in the treatment of VAs in NICM patients.
Collapse
Affiliation(s)
- Laura Adina Stanciulescu
- Department of Cardiothoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (L.A.S.); (R.V.)
- Cardiology Department, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | | | - Radu Vatasescu
- Department of Cardiothoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania; (L.A.S.); (R.V.)
- Cardiology Department, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
13
|
Samuel M, Arif SG, Afilalo J. Heart rate variability as a digital biomarker for frailty in cardiovascular patients. J Frailty Aging 2025; 14:100007. [PMID: 39855886 DOI: 10.1016/j.tjfa.2024.100007] [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: 04/25/2024] [Accepted: 11/13/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Frailty is a syndrome associated with age-related impairments in multiple organ systems, of which the autonomic nervous system plays a fundamental role. Measurement of heart rate variability (HRV) is a non-invasive method to evaluate the autonomic activity and gain insights into cardiovascular health and potentially, frailty. A few small studies have explored the relationship between HRV and frailty, with promising but conflicting results. OBJECTIVE To investigate the relationship between HRV and frailty among adult patients with cardiovascular disease. DESIGN A cross-sectional study was conducted using clinical data. SETTING Data were collected from an ambulatory cardiology clinic. PARTICIPANTS The cohort comprised 155 patients with a mean age of 67 years (44 % female). MEASUREMENTS HRV was assessed seated at rest for 2.5 min using a finger-based photoplethysmography (PPG) device. Frailty was assessed using the Clinical Frailty Scale (CFS), with a score ≥5 considered frail. Associations between HRV and frailty were examined using a Spearman correlation matrix and multivariable ordinal regression model. The LF/HF ratio (a frequency-domain measure reflecting imbalances between sympathetic and parasympathetic activity) was the primary HRV measure analyzed. RESULTS The prevalence of frailty was 15 %. Among all HRV measures, the LF/HF ratio was most closely correlated with frailty (p < 0.001). In the multivariable model, each 1 standard deviation decrease in LF/HF ratio was associated with a 1.1-point increase in CFS (95 % CI 0.7-1.6, p < 0.001). The optimal ROC cutoff at which the LF/HF ratio was associated with frailty is ≤ 0.37. CONCLUSIONS The LF/HF ratio is inversely correlated with the CFS and independently associated with frailty. Measurement of HRV is a promising technique to enrich existing frailty scales and assist in frailty assessments in an ambulatory cardiology clinic.
Collapse
Affiliation(s)
- Maryia Samuel
- Division of Experimental Medicine, McGill University, Montreal, QC Canada; Division of Cardiology, Jewish General Hospital, Montreal, QC Canada
| | - Saleena Gul Arif
- Division of Experimental Medicine, McGill University, Montreal, QC Canada; Division of Cardiology, Jewish General Hospital, Montreal, QC Canada; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Jonathan Afilalo
- Division of Experimental Medicine, McGill University, Montreal, QC Canada; Division of Cardiology, Jewish General Hospital, Montreal, QC Canada.
| |
Collapse
|
14
|
Goerger K, Abbott K, Larson MK, Holinstat M. Cardiovascular Disease Risk Factors in the Native American Population. J Cardiovasc Dev Dis 2025; 12:27. [PMID: 39852305 PMCID: PMC11765693 DOI: 10.3390/jcdd12010027] [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: 10/28/2024] [Revised: 01/02/2025] [Accepted: 01/12/2025] [Indexed: 01/26/2025] Open
Abstract
Native Americans are disproportionately affected by cardiovascular disease in comparison with other racial and ethnic groups in the United States. Previous research has analyzed risk factors, quantified prevalence rates, and examined outcomes of cardiovascular disease in Native Americans, yet few studies have considered the role of societal and psychological factors on the increased burden of cardiovascular disease in Native Americans. Modifiable risk factors for cardiovascular disease, including poor nutrition, reduced physical activity, obesity, and increased substance use, are exacerbated in Native American communities due to cultural and historical factors. Further, Native Americans have endured historical trauma and continue to experience additional financial and healthcare stressors, resulting in increased levels of chronic stress. Chronic activation of stress responses through the hypothalamic-pituitary-adrenal and autonomic nervous system increases inflammation and cardiovascular dysfunction resulting in an increased risk for cardiovascular disease. Therefore, it is critical to examine the connection between these stressors and the cardiovascular health disparities in Native American communities to create effective strategies to improve health outcomes.
Collapse
Affiliation(s)
- Krista Goerger
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Karla Abbott
- Nursing Department, Augustana University, Sioux Falls, SD 57197, USA;
| | - Mark K. Larson
- Biology Department, Augustana University, Sioux Falls, SD 57197, USA;
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
15
|
Dos Santos Disessa H, Monteiro PHM, da Silva Zacharias V, da Costa Rosa CS, Monteiro HL. A systematic review and meta-analysis investigating the impact of exercise interventions on heart rate variability in hemodialysis patients. Sci Rep 2024; 14:30818. [PMID: 39730561 DOI: 10.1038/s41598-024-81217-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024] Open
Abstract
Patients with chronic kidney disease have a high incidence of cardiovascular diseases, and autonomic dysfunction has a determinant role in the relevant declines. Physical exercise influences heart rate variability and cardiac autonomic modulation. Thus, our objective was to systematically review, with a meta-analysis, the correlation between physical exercise interventions and alterations in cardiac autonomic modulation in hemodialysis patients. A customized research strategy was used across four databases. The search yielded 392 studies, with eight randomized clinical trials included (396 participants), indicating that the investigated indices favor the intervention group by increasing autonomic activity. The exercise training probably increases the standard deviation of all NN intervals (20.71 ms CI 95% [9.55, 31.87], p < 0.001, I²=95%) compared to the control group and showing an moderate certainty, was the most commonly used index (seven studies). Mean RR (35.57 ms CI 95% [14.56, 56.57], p = 0.91, I²=0%), the root mean square sum of squares of differences between NN intervals (10.55 ms CI 95% [6.75, 14.34], p = 0.37, I²=4%), and LF/HF (0.28 ms (n.u) [0.11, 0.44], p = 0.18, I²=39%) were also in favor of the training group. However, based on the GRADE analysis we are uncertain whether Mean RR can increase after an exercise intervention, as well RMSSD and LF/HF may increase slightly, we obtained low certainty of this evidence. The exact magnitude of the impact of physical training on the alteration of cardiac autonomic modulation in this patient population has yet to be conclusively defined.
Collapse
Affiliation(s)
| | | | - Vitor da Silva Zacharias
- Exercise Assessment and Prescription Laboratory, São Paulo State University (UNESP), São Paulo, Brazil
| | - Clara Suemi da Costa Rosa
- Exercise Assessment and Prescription Laboratory, São Paulo State University (UNESP), São Paulo, Brazil
| | - Henrique Luiz Monteiro
- Exercise Assessment and Prescription Laboratory, São Paulo State University (UNESP), São Paulo, Brazil
| |
Collapse
|
16
|
Hao Q, Shi Z, Liang X, Zhang W, Meng Q, Martyniuk CJ. Assessment of the relative cardiotoxicity and behavioral effects of butylated hydroxytoluene and its metabolites in developing zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177518. [PMID: 39551212 DOI: 10.1016/j.scitotenv.2024.177518] [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: 07/30/2024] [Revised: 10/06/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
Butylated hydroxytoluene (BHT) and its transformation products are ubiquitously detected in aquatic environments. Despite studies reporting on the adverse effects of BHT exposure in early-staged zebrafish, the comparative toxicity of its metabolites is not known. To address this, zebrafish embryos were exposed continuously to 0.01-1 μM BHTs (BHT, BHT-Q, BHT-OH, BHT-CHO, and BHT-COOH) for up to 6 days. Each chemical altered the heart rates of developing zebrafish at 72 hpf. When assessing cardiac morphology at 48 and 72 hpf, BHT-COOH showed stronger effects compared to BHT in inducing pericardial edema and myocardial hypertrophy. This effect is hypothesized to be attributed to differences in transcriptional regulation of key genes during cardiac development. BHT predominantly affected transcript expression in the early (15 hpf) or late stages (48-72 hpf) of heart development, but did not impact the transcription during the middle stage (24-36 hpf). Conversely, BHT-COOH altered the expression of early transcription factors (i.e. tbx5, nkx2.5, gata4, hand2) and functional genes (i.e. myh6, nppa, cacna1ab, and atp2a2) at all stages of cardiac development. The effects on the heart may also be related to behavioral changes observed in larval fish. Behavior was most sensitive with low chemical exposures (0.01 μM), while at higher concentrations, heart rate was a more sensitive indicator of BHT-induced toxicity. Taken together, our results indicate that BHTs induce adverse effects on cardiovascular function and BHT metabolites may pose higher risk than the parent compound.
Collapse
Affiliation(s)
- Qiaoqiao Hao
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, Ministry of Education Collaborative Innovation Center for Grassland Ecological Security, Ministry of Education Key Laboratory of Ecology and Resource Use of the Mongolian Plateau, School of Ecology and Environment, Inner Mongolia University, Hohhot 010021, China
| | - Ziyue Shi
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, Ministry of Education Collaborative Innovation Center for Grassland Ecological Security, Ministry of Education Key Laboratory of Ecology and Resource Use of the Mongolian Plateau, School of Ecology and Environment, Inner Mongolia University, Hohhot 010021, China
| | - Xuefang Liang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, Ministry of Education Collaborative Innovation Center for Grassland Ecological Security, Ministry of Education Key Laboratory of Ecology and Resource Use of the Mongolian Plateau, School of Ecology and Environment, Inner Mongolia University, Hohhot 010021, China.
| | - Weiang Zhang
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, Ministry of Education Collaborative Innovation Center for Grassland Ecological Security, Ministry of Education Key Laboratory of Ecology and Resource Use of the Mongolian Plateau, School of Ecology and Environment, Inner Mongolia University, Hohhot 010021, China
| | - Qingjian Meng
- Inner Mongolia Key Laboratory of Environmental Pollution Control & Waste Resource Reuse, Ministry of Education Collaborative Innovation Center for Grassland Ecological Security, Ministry of Education Key Laboratory of Ecology and Resource Use of the Mongolian Plateau, School of Ecology and Environment, Inner Mongolia University, Hohhot 010021, China
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
17
|
Kumar RS, Alqarni AA, Alghamdi MJ, Alharbi SA, Alsharef OL, Ahmed ME. Physiological effects of hand grip and cold pressor tests in young Saudi adults. J Med Life 2024; 17:1067-1071. [PMID: 39877044 PMCID: PMC11771833 DOI: 10.25122/jml-2024-0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 01/31/2025] Open
Abstract
The risk of cardiovascular disease differs among various ethnic groups, highlighting disparities in cardiovascular health among different populations. While multiple studies from other countries have looked at changes in physiological parameters during autonomic function tests like isometric handgrip and cold pressor tests, no correlational research has been done in Saudi Arabia. This lacuna underscores the importance of examining the relationship between cardiorespiratory parameters in young Saudi Arabian individuals during these tests. This study aimed to determine the correlation between the isometric handgrip and cold pressor tests and physiological parameters in healthy young Saudi Arabian college students. A single-arm interventional study was conducted with a cohort of 65 healthy young adult Saudi college students, including male and female participants. A point estimate was calculated with a 95% confidence level. Physiological parameters were analyzed and compared at rest and during isometric handgrip and cold pressor tests. The study involved participants with an average age of 21.12 ± 1.02, predominantly male students. A significant impact was observed only in respiratory rate (P = 0.007) during the isometric handgrip and cold pressor tests. In contrast, blood pressure parameters and arterial oxygen saturation values showed no statistical significance during both tests. This sheds light on their autonomic responses to physiological stressors and contributes to our understanding of cardiovascular health across diverse populations, guiding future interventions for global improvements in cardiorespiratory outcomes.
Collapse
Key Words
- ANS, Autonomic Nervous System
- BMI, Body Mass Index
- BP, Blood Pressure
- CPT, Cold Pressor Test
- DBP, Diastolic Blood Pressure
- HR, Heart Rate
- IHT, Isometric Handgrip Test
- MAP, Mean Arterial Pressure
- RR, Respiratory Rate
- SBP, Systolic Blood Pressure
- SpO2, Peripheral Arterial Oxygen Saturation
- blood pressure
- cold pressor test
- heart rate
- isometric hand grip
- respiratory rate
Collapse
Affiliation(s)
- Raju Suresh Kumar
- Department of Basic Sciences, College of Science and Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
| | - Abdulaziz Abdullah Alqarni
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
- Department of Respiratory Therapy, College of Applied Medical Sciences (COAMS), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Musaad Jamaan Alghamdi
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
- Department of Respiratory Therapy, College of Applied Medical Sciences (COAMS), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Saad Amer Alharbi
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
- Department of Respiratory Therapy, College of Applied Medical Sciences (COAMS), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Omar Loutfi Alsharef
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
- Department of Respiratory Therapy, College of Applied Medical Sciences (COAMS), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
| | - Mohamed Eldigire Ahmed
- Department of Basic Sciences, College of Science and Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Jeddah, Saudi Arabia
| |
Collapse
|
18
|
Gee MM, Hornung E, Gupta S, Newton AJH, Cheng Z(J, Lytton WW, Lenhoff AM, Schwaber JS, Vadigepalli R. Unpacking the multimodal, multi-scale data of the fast and slow lanes of the cardiac vagus through computational modelling. Exp Physiol 2024; 109:1994-2000. [PMID: 37120805 PMCID: PMC10613580 DOI: 10.1113/ep090865] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/12/2023] [Indexed: 05/01/2023]
Abstract
The vagus nerve is a key mediator of brain-heart signaling, and its activity is necessary for cardiovascular health. Vagal outflow stems from the nucleus ambiguus, responsible primarily for fast, beat-to-beat regulation of heart rate and rhythm, and the dorsal motor nucleus of the vagus, responsible primarily for slow regulation of ventricular contractility. Due to the high-dimensional and multimodal nature of the anatomical, molecular and physiological data on neural regulation of cardiac function, data-derived mechanistic insights have proven elusive. Elucidating insights has been complicated further by the broad distribution of the data across heart, brain and peripheral nervous system circuits. Here we lay out an integrative framework based on computational modelling for combining these disparate and multi-scale data on the two vagal control lanes of the cardiovascular system. Newly available molecular-scale data, particularly single-cell transcriptomic analyses, have augmented our understanding of the heterogeneous neuronal states underlying vagally mediated fast and slow regulation of cardiac physiology. Cellular-scale computational models built from these data sets represent building blocks that can be combined using anatomical and neural circuit connectivity, neuronal electrophysiology, and organ/organismal-scale physiology data to create multi-system, multi-scale models that enable in silico exploration of the fast versus slow lane vagal stimulation. The insights from the computational modelling and analyses will guide new experimental questions on the mechanisms regulating the fast and slow lanes of the cardiac vagus toward exploiting targeted vagal neuromodulatory activity to promote cardiovascular health.
Collapse
Affiliation(s)
- Michelle M. Gee
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Pathology and Genomic MedicineDaniel Baugh Institute of Functional Genomics/Computational BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Eden Hornung
- Department of Pathology and Genomic MedicineDaniel Baugh Institute of Functional Genomics/Computational BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Suranjana Gupta
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Adam J. H. Newton
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Zixi (Jack) Cheng
- Burnett School of Biomedical Sciences, College of MedicineUniversity of Central FloridaOrlandoFloridaUSA
| | - William W. Lytton
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Abraham M. Lenhoff
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
| | - James S. Schwaber
- Department of Pathology and Genomic MedicineDaniel Baugh Institute of Functional Genomics/Computational BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Rajanikanth Vadigepalli
- Department of Pathology and Genomic MedicineDaniel Baugh Institute of Functional Genomics/Computational BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
19
|
Toslak D, Erol MK, Parlak AE, Bulut M, Erdem Toslak I. Evaluation of carotid intima-media thickness measurements in patients with central serous chorioretinopathy. RADIOLOGIE (HEIDELBERG, GERMANY) 2024; 64:153-159. [PMID: 39240320 DOI: 10.1007/s00117-024-01358-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Central serous chorioretinopathy (CSC) is a systemic disease more than a disease localized to the eye, and there may be vascular involvement in its pathogenesis. OBJECTIVE This study aimed to evaluate the carotid intima-media thickness (IMT) of patients with CSC, to compare it with that of healthy individuals, and to explore whether there might be an association between CSC and subclinical carotid atherosclerotic disease. MATERIALS AND METHODS Adult patients with CSC (n = 30) and healthy individuals (n = 30) were included in this prospective study. All participants underwent complete ophthalmologic imaging and were then referred to the radiology department. Carotid IMT measurements were performed using ultrasound imaging. Measurements of the two groups were obtained and compared. RESULTS There was no statistically significant difference between patients with CSC and the control group with respect to age, gender, and smoking habits. The mean (±standard deviation, SD) carotid IMT values obtained by ultrasound measurements for the right and left sides in the patient group were 0.71 mm (± 0.19) and 0.71 mm (± 0.21), respectively. The mean (±SD) carotid IMT values for the right and left sides in the control group were 0.61 mm (± 0.15) and 0.60 mm (± 0.15), respectively. The mean carotid IMT values in the patient group were significantly higher than those in the control group for the right and left sides (p = 0.02 and p = 0.03, respectively). CONCLUSION Carotid IMT is increased in patients with CSC compared to healthy individuals. This outcome might reinforce the benefit of carotid artery screening following diagnosis of CSC by ophthalmologists for early detection of subclinical carotid atherosclerotic disease.
Collapse
Affiliation(s)
- Devrim Toslak
- Department of Ophthalmology, Health Sciences University Antalya Training and Research Hospital, Antalya, Turkey.
- Department of Ophthalmology, Health Sciences University Antalya Training and Research Hospital, Varlık mah 100. Yıl cad, 07070, Muratpasa, Antalya, Turkey.
| | - Muhammet Kazım Erol
- Department of Ophthalmology, Health Sciences University Antalya Training and Research Hospital, Antalya, Turkey
| | - Ayşe Eda Parlak
- Department of Radiology, Health Sciences University Antalya Training and Research Hospital, Antalya, Turkey
| | - Mehmet Bulut
- Department of Ophthalmology, Health Sciences University Antalya Training and Research Hospital, Antalya, Turkey
| | - Iclal Erdem Toslak
- Department of Radiology, Health Sciences University Antalya Training and Research Hospital, Antalya, Turkey
| |
Collapse
|
20
|
Lopes Soares L, Portes AMO, Costa SFF, Leite LB, Natali AJ. Autonomic Dysregulation in Pulmonary Hypertension: Role of Physical Exercise. Hypertension 2024; 81:2228-2236. [PMID: 39234679 DOI: 10.1161/hypertensionaha.124.23573] [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: 09/06/2024]
Abstract
Pulmonary hypertension (PH) is a rare and severe condition characterized by increased pressure in the pulmonary circulation, often resulting in right ventricular failure and death. The autonomic nervous system (ANS) plays a crucial role in the cardiovascular and pulmonary controls. Dysfunction of ANS has been implicated in the pathogenesis of cardiopulmonary diseases. Conversely, dysfunctions in ANS can arise from these diseases, impacting cardiac and pulmonary autonomic functions and contributing to disease progression. The complex interaction between ANS dysfunction and PH plays a crucial role in the disease progression, making it essential to explore interventions that modulate ANS, such as physical exercise, to improve the treatment and prognosis of patients with PH. This review addresses autonomic dysfunctions found in PH and their implications for the cardiopulmonary system. Furthermore, we discuss how physical exercise, a significant modulator of ANS, may contribute to the prognosis of PH. Drawing from evidence of aerobic and resistance exercise training in patients and experimental models of PH, potential cardiovascular benefits of exercise are presented. Finally, we highlight emerging therapeutic targets and perspectives to better cope with the complex condition. A comprehensive understanding of the interaction between ANS and PH, coupled with targeted physical exercise interventions, may pave the way for innovative therapeutic strategies and significantly improve the treatment and prognosis of vulnerable patients.
Collapse
Affiliation(s)
- Leôncio Lopes Soares
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| | | | | | - Luciano Bernardes Leite
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| | - Antônio José Natali
- Department of Physical Education, Federal University of Viçosa, Minas Gerais, Brazil (L.L.S., S.F.F.C., L.B.L., A.J.N.)
| |
Collapse
|
21
|
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
|
22
|
Kiss MG, Cohen O, McAlpine CS, Swirski FK. Influence of sleep on physiological systems in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1284-1300. [PMID: 39528718 PMCID: PMC11567060 DOI: 10.1038/s44161-024-00560-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sleep is a fundamental requirement of life and is integral to health. Deviation from optimal sleep associates with numerous diseases including those of the cardiovascular system. Studies, spanning animal models to humans, show that insufficient, disrupted or inconsistent sleep contribute to poor cardiovascular health by disrupting body systems. Fundamental experiments have begun to uncover the molecular and cellular links between sleep and heart health while large-scale human studies have associated sleep with cardiovascular outcomes in diverse populations. Here, we review preclinical and clinical findings that demonstrate how sleep influences the autonomic nervous, metabolic and immune systems to affect atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Máté G Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oren Cohen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Filip K Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
23
|
Franco-Riveros VB, Pividori SM, Martin TI, Nicora FE, Lallana MC, Pontecorvo AA, Flores JC, Tubbs RS, Boezaart AP, Reina MA, Buchholz B. Anatomical study with clinical significance of communicating and visceral branching of the cervical and upper thoracic sympathetic trunk. Clin Anat 2024; 37:886-899. [PMID: 38469730 DOI: 10.1002/ca.24149] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024]
Abstract
Current advances in the management of the autonomic nervous system in various cardiovascular diseases, and in treatments for pain or sympathetic disturbances in the head, neck, or upper limbs, necessitate a thorough understanding of the anatomy of the cervicothoracic sympathetic trunk. Our objective was to enhance our understanding of the origin and distribution of communicating branches and visceral cervicothoracic sympathetic nerves in human fetuses. This was achieved through a comprehensive topographic systematization of the branching patterns observed in the cervical and upper thoracic ganglia, along with the distribution of communicating branches to each cervical spinal nerve. We conducted detailed sub-macroscopic dissections of the cervical and thoracic regions in 20 human fetuses (40 sides). The superior and cervicothoracic ganglia were identified as the cervical sympathetic ganglia that provided the most communicating branches on both sides. The middle and accessory cervical ganglia contributed the fewest branches, with no significant differences between the right and left sides. The cervicothoracic ganglion supplied sympathetic branches to the greatest number of spinal nerves, spanning from C5 to T2. The distribution of communicating branches to spinal nerves was non-uniform. Notably, C3, C4, and C5 received the fewest branches, and more than half of the specimens showed no sympathetic connections. C1 and C2 received sympathetic connections exclusively from the superior ganglion. Spinal nerves that received more branches often did so from multiple ganglia. The vertebral nerve provided deep communicating branches primarily to C6, with lesser contributions to C7, C5, and C8. The vagus nerve stood out as the cranial nerve with the most direct sympathetic connections. The autonomic branching pattern and connections of the cervicothoracic sympathetic trunk are significantly variable in the fetus. A comprehensive understanding of the anatomy of the cervical and upper thoracic sympathetic trunk and its branches is valuable during autonomic interventions and neuromodulation. This knowledge is particularly relevant for addressing various autonomic cardiac diseases and for treating pain and vascular dysfunction in the head, neck, and upper limbs.
Collapse
Affiliation(s)
- Verena B Franco-Riveros
- School of Medicine, Department of Human Anatomy, First Unit, Cardiovascular Anatomy Lab, Buenos Aires University, Buenos Aires, Argentina
- School of Medicine, Department of Pathology, Institute of Cardiovascular Physiopathology (INFICA), Buenos Aires University, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires University School of Medicine, Buenos Aires, Argentina
| | - Sofía M Pividori
- School of Medicine, Department of Human Anatomy, First Unit, Cardiovascular Anatomy Lab, Buenos Aires University, Buenos Aires, Argentina
- Diagnostic Imaging Department, Hospital Británico, Buenos Aires, Argentina
| | - Tomás I Martin
- School of Medicine, Department of Human Anatomy, First Unit, Cardiovascular Anatomy Lab, Buenos Aires University, Buenos Aires, Argentina
| | - Florencia E Nicora
- School of Medicine, Department of Human Anatomy, First Unit, Cardiovascular Anatomy Lab, Buenos Aires University, Buenos Aires, Argentina
| | - María Cecilia Lallana
- School of Medicine, Department of Human Anatomy, First Unit, Cardiovascular Anatomy Lab, Buenos Aires University, Buenos Aires, Argentina
| | - Agustina A Pontecorvo
- School of Medicine, Department of Human Anatomy, First Unit, Cardiovascular Anatomy Lab, Buenos Aires University, Buenos Aires, Argentina
| | - Juan Carlos Flores
- Postgraduate Universitary Training at Interventional Procedures for Chronic Refractory Pain, CAIDBA Comprehensive Pain Center Foundation; and La Plata University School of Medical Sciences, La Plata, Buenos Aires, Argentina
| | - Richard Shane Tubbs
- Department of Neurosurgery, Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Anatomical Sciences, St. George's University, St. George's, West Indies
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Neurosurgery and Ochsner Neuroscience Institute, Ochsner Health System, New Orleans, Louisiana, USA
- Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - André P Boezaart
- Acute and Perioperative Pain Medicine, Department of Anesthesiology, University of Florida College of Medicine, Gainesville, Florida, USA
- Lumina Health Pain Medicine Collaborative, Surrey, UK
| | - Miguel A Reina
- Acute and Perioperative Pain Medicine, Department of Anesthesiology, University of Florida College of Medicine, Gainesville, Florida, USA
- School of Medicine, CEU-San-Pablo University, Madrid, Spain
- Department of Anesthesiology, Madrid-Montepríncipe University Hospital, Madrid, Spain
| | - Bruno Buchholz
- School of Medicine, Department of Human Anatomy, First Unit, Cardiovascular Anatomy Lab, Buenos Aires University, Buenos Aires, Argentina
- School of Medicine, Department of Pathology, Institute of Cardiovascular Physiopathology (INFICA), Buenos Aires University, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL), Buenos Aires University School of Medicine, Buenos Aires, Argentina
| |
Collapse
|
24
|
Boieriu AM, Dumitrel Luca C, Neculoiu CD, Ţînţ D. The impact of inflammatory and oxidative stress biomarkers on the sympathetic nervous system in severe coronary atherosclerosis. Front Cardiovasc Med 2024; 11:1480925. [PMID: 39469122 PMCID: PMC11513282 DOI: 10.3389/fcvm.2024.1480925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
Objective The present study aimed at evaluating the association between sympathetic nervous system activation (SNS) and the severity of coronary artery disease (CAD). In addition, we tested the hypothesis that inflammation and oxidative stress influence the SNS activation. Methods Adult patients with severe CAD scheduled for coronary artery bypass graft (CABG) surgery were enrolled. SYNTAX I score was calculated based on coronary angiography. Systemic activation of the SNS was estimated through circulating levels of norepinephrine (NE). Plasma levels of pro-inflammatory cytokines (IL 1β, IL 6 and HIF 1α) and oxidative stress molecules (SOD-1 and LOX-1) were obtained prior to surgery. Results Circulating NE levels were significantly correlated with the severity of CAD, as assessed by the SYNTAX I score (p 0.002; r 0.329). Elevated levels of circulating pro-inflammatory markers were significantly correlated with increased NE concentrations (for IL-1β: p < 0.001, r = 0.49; for IL-6 and NE: p = 0.003, r = 0.32; for HIF-1α and NE: p = 0.049, r = 0.21). Additionally, oxidative stress molecules were associated with circulating NE levels (for SOD-1 and NE: p = 0.016, r = 0.26; for LOX-1 and NE: p = 0.004, r = 0.31). Conclusion In patients with CAD referred for CABG, SNS activation, indicated by plasma NE levels, was correlated with disease severity as assessed by the SYNTAX I score, as well as with markers of inflammation and oxidative stress. This suggests that inflammation, oxidative stress, and SNS activation form an interconnected network, with each component influencing the others. It might be of interest to develop a scoring system including inflammation and oxidative stress markers to identify patients that require a more aggressive approach to lower inflammation, oxidative stress and modulate the sympathetic nervous system. This could be of use especially in the setting of a scheduled intervention -such as CABG surgery.
Collapse
Affiliation(s)
- Alexandra Maria Boieriu
- Faculty of Medicine, Transilvania University of Brasov, Brasov, Romania
- Department of Cardiology, Emergency County Hospital, Brasov, Romania
| | - Cezar Dumitrel Luca
- Faculty of Medicine, Transilvania University of Brasov, Brasov, Romania
- Cardiovascular Rehabilitation Hospital Covasna, Covasna, Romania
| | | | - Diana Ţînţ
- Faculty of Medicine, Transilvania University of Brasov, Brasov, Romania
- Department of Cardiology, ICCO Clinics, Brasov, Romania
| |
Collapse
|
25
|
Mbikyo MB, Wang A, Ma Q, Miao L, Cui N, Yang Y, Fu H, Sun Y, Li Z. Low-Level Tragus Stimulation Attenuates Blood Pressure in Young Individuals With Hypertension: Results From a Small-Scale Single-Blind Controlled Randomized Clinical Trial. J Am Heart Assoc 2024; 13:e032269. [PMID: 39291497 PMCID: PMC11681467 DOI: 10.1161/jaha.123.032269] [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: 08/16/2023] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Hypertension is a significant risk factor for cardiovascular and chronic kidney diseases. Its management in young people remains limited. Device-based therapies, such as low-level tragus stimulation (LL-TS), a noninvasive method that reduces sympathetic activity, have recently been explored for resistant hypertension. METHODS AND RESULTS This trial involved patients with Grade 1 hypertension with no other medical history. LL-TS (20 Hz, 1 mA, 1 h/day) was applied for 3 months on the tragus (Intervention group [IG]) or earlobe (Control group [CG]). Blood pressure and outcomes were assessed at the first, second, and third months. Among 40 patients, 21 were in IG and 19 in CG. Baseline systolic blood pressure was similar between IG (142.62±8.18 mm Hg) and CG (143.00±8.61 mm Hg), P=0.89. Post-LL-TS, systolic blood pressure showed significant reductions in IG compared with CG at the first (IG: 134.47±5.95 mm Hg, CG: 141.28±6.78 mm Hg, P=0.002), second (IG: 132.50±7.51 mm Hg, CG: 140.62±7.15 mm Hg, P=0.001), and third months (IG: 128.81±7.13 mm Hg, CG: 136.51±7.96 mm Hg, P=0.003). diastolic blood pressure also differed significantly: first month (IG: 85.34±5.81 mm Hg, CG: 89.74±6.32 mm Hg, P=0.03), second month (IG: 82.12±5.22 mm Hg, CG: 88.57±7.11 mm Hg, P=0.002), and third month (IG: 80.71±5.96 mm Hg, CG: 87.55±5.26 mm Hg, P=0.001). Heart rate was unchanged (P>0.05). Only 0.01% of IG subjects reported site irritation, with no serious adverse events. CONCLUSIONS LL-TS led to significant blood pressure reductions in young patients with essential hypertension. Further larger trials are needed to confirm the safety and efficacy of LL-TS. REGISTRATION URL: https://www.chictr.org.cn/; Unique identifier: ChiCTR2000038448.
Collapse
Affiliation(s)
- Muisha B. Mbikyo
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Ai Wang
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
- Department of CardiologyZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Qun Ma
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Linlin Miao
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Nan Cui
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Yiqing Yang
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Haoran Fu
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Yingxian Sun
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Zhao Li
- Department of CardiologyThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
26
|
Guo Q, Liu D, Wang X. Early peripheral perfusion monitoring in septic shock. Eur J Med Res 2024; 29:477. [PMID: 39350276 PMCID: PMC11440805 DOI: 10.1186/s40001-024-02074-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Septic shock is a frequent critical clinical condition and a leading cause of death in critically ill individuals. However, it is challenging to identify affected patients early. In this article, we discuss new perspectives on the methods and uses of peripheral perfusion monitoring, considering the concept of a dysregulated response. Physical examination, and visual and ultrasonographic techniques are used to measure peripheral microcirculatory blood flow to reflect tissue perfusion. Compared with other monitoring techniques, peripheral perfusion monitoring has the benefits of low invasiveness and good repeatability, and allows for quick therapeutic judgments, which have significant practical relevance. Peripheral perfusion monitoring is an effective tool to detect early signs of septic shock, autonomic dysfunction, and organ damage. This method can also be used to evaluate treatment effectiveness, direct fluid resuscitation and the use of vasoactive medications, and monitor vascular reactivity, microcirculatory disorders, and endothelial cell damage. Recent introductions of novel peripheral perfusion monitoring methods, new knowledge of peripheral perfusion kinetics, and multimodal peripheral perfusion evaluation methods have occurred. To investigate new knowledge and therapeutic implications, we examined the methodological attributes and mechanisms of peripheral perfusion monitoring, in this study.
Collapse
Affiliation(s)
- Qirui Guo
- Department of Critical Care Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Dawei Liu
- Department of Critical Care Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, China.
| |
Collapse
|
27
|
Liu Chung Ming C, Wang X, Gentile C. Protective role of acetylcholine and the cholinergic system in the injured heart. iScience 2024; 27:110726. [PMID: 39280620 PMCID: PMC11402255 DOI: 10.1016/j.isci.2024.110726] [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] [Indexed: 09/18/2024] Open
Abstract
This review explores the roles of the cholinergic system in the heart, comprising the neuronal and non-neuronal cholinergic systems. Both systems are essential for maintaining cardiac homeostasis by regulating the release of acetylcholine (ACh). A reduction in ACh release is associated with the early onset of cardiovascular diseases (CVDs), and increasing evidence supports the protective roles of ACh against CVD. We address the challenges and limitations of current strategies to elevate ACh levels, including vagus nerve stimulation and pharmacological interventions such as cholinesterase inhibitors. Additionally, we introduce alternative strategies to increase ACh in the heart, such as stem cell therapy, gene therapy, microRNAs, and nanoparticle drug delivery methods. These findings offer new insights into advanced treatments for regenerating the injured human heart.
Collapse
Affiliation(s)
- Clara Liu Chung Ming
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| | - Xiaowei Wang
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
28
|
Farhat K, Po SS, Stavrakis S. Non-invasive Neuromodulation of Arrhythmias. Card Electrophysiol Clin 2024; 16:307-314. [PMID: 39084723 PMCID: PMC11292161 DOI: 10.1016/j.ccep.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The autonomic nervous system plays a central role in the pathogenesis of arrhythmias. Preclinical and clinical studies have demonstrated the therapeutic effect of neuromodulation at multiple anatomic targets across the neurocardiac axis for the treatment of arrhythmias. In this review, we discuss the rationale and clinical application of noninvasive neuromodulation techniques in treating arrhythmias and explore associated barriers and future directions, including optimization of stimulation parameters and patient selection.
Collapse
Affiliation(s)
| | - Sunny S Po
- University of Oklahoma Health Sciences Center
| | | |
Collapse
|
29
|
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
|
30
|
Sato T, Hanna P, Mori S. Innervation of the coronary arteries and its role in controlling microvascular resistance. J Cardiol 2024; 84:1-13. [PMID: 38346669 DOI: 10.1016/j.jjcc.2024.01.005] [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: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 04/08/2024]
Abstract
The coronary circulation plays a crucial role in balancing myocardial perfusion and oxygen demand to prevent myocardial ischemia. Extravascular compressive forces, coronary perfusion pressure, and microvascular resistance are involved to regulate coronary blood flow throughout the cardiac cycle. Autoregulation of the coronary blood flow through dynamic adjustment of microvascular resistance is maintained by complex interactions among mechanical, endothelial, metabolic, neural, and hormonal mechanisms. This review focuses on the neural mechanism. Anatomy and physiology of the coronary arterial innervation have been extensively investigated using animal models. However, findings in the animal heart have limited applicability to the human heart as cardiac innervation is generally highly variable among species. So far, limited data are available on the human coronary artery innervation, rendering multiple questions unresolved. Recently, the clinical entity of ischemia with non-obstructive coronary arteries has been proposed, characterized by microvascular dysfunction involving abnormal vasoconstriction and impaired vasodilation. Thus, measurement of microvascular resistance has become a standard diagnostic for patients without significant stenosis in the epicardial coronary arteries. Neural mechanism is likely to play a pivotal role, supported by the efficacy of cardiac sympathetic denervation to control symptoms in patients with angina. Therefore, understanding the coronary artery innervation and control of microvascular resistance of the human heart is increasingly important for cardiologists for diagnosis and to select appropriate therapeutic options. Advancement in this field can lead to innovations in diagnostic and therapeutic approaches for coronary artery diseases.
Collapse
Affiliation(s)
- Takanori Sato
- University of California Los Angeles (UCLA) Cardiac Arrhythmia Center, UCLA Health System, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Peter Hanna
- University of California Los Angeles (UCLA) Cardiac Arrhythmia Center, UCLA Health System, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shumpei Mori
- University of California Los Angeles (UCLA) Cardiac Arrhythmia Center, UCLA Health System, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Cook AC, Egli AE, Cohen NE, Bernardi K, Chae MY, Kapalko BA, Coyne SA, Scott R. The Neurophysiological Effects of Craniosacral Treatment on Heart Rate Variability: A Systematic Review of Literature and Meta-Analysis. Cureus 2024; 16:e64807. [PMID: 39156412 PMCID: PMC11329942 DOI: 10.7759/cureus.64807] [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: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
Craniosacral treatment (CST) is an osteopathic technique grounded in the assumption that there is an intrinsic, fine movement of the cerebrospinal fluid. This rhythmic movement can be utilized for diagnostic and therapeutic purposes by palpation and manipulation of the skull, spine, and associated connective tissues. Therapeutic benefit is likely due to action on the autonomic nervous system (ANS), specifically through the vagus nerve. Current literature on the neurophysiological effects of CST is limited, which has contributed to controversy regarding its effectiveness. Heart rate variability (HRV) as a measure of cardiovascular stress and autonomic system activity is thus proposed as a tool to evaluate the neurophysiologic effects of CST. HRV can be analyzed in two different bands, high-frequency (HF) and low-frequency (LF) power associated with a parasympathetic and sympathetic response. In this meta-analysis, we provide a brief introduction to CST, analyze three primary studies, and summarize the therapeutic benefits and pitfalls of this alternative treatment on the ANS. A significant negative HF standardized mean difference after CST was observed; standardized mean difference = -0.46; 95% CI (-0.79,-0.14). No significant effect on LF power was observed. We conclude that CST does provide a moderate short-term increase in parasympathetic activity. These findings suggest that CST may be used to treat patients with an overactive sympathetic state. Further studies should be conducted for comparison against a control group to eliminate the possibility of a placebo effect and to elucidate long-term effects.
Collapse
Affiliation(s)
- Andrew C Cook
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine - Bradenton, Jacksonville, USA
| | - Anna E Egli
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine - Bradenton, St. Augustine, USA
| | - Nathan E Cohen
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine - Bradenton, Jacksonville, USA
| | - Kyrie Bernardi
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine - Bradenton, Jacksonville, USA
| | - Min Y Chae
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine - Bradenton, Jacksonville, USA
| | - Brandon A Kapalko
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine - Bradenton, St. Augustine, USA
| | - Sunni A Coyne
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine - Bradenton, Daytona Beach, USA
| | - Randy Scott
- Regional Dean, Lake Erie College of Osteopathic Medicine - Bradenton, Jacksonville, USA
| |
Collapse
|
32
|
Rai R, Singh V, Ahmad Z, Jain A, Jat D, Mishra SK. Autonomic neuronal modulations in cardiac arrhythmias: Current concepts and emerging therapies. Physiol Behav 2024; 279:114527. [PMID: 38527577 DOI: 10.1016/j.physbeh.2024.114527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
The pathophysiology of atrial fibrillation and ventricular tachycardia that result in cardiac arrhythmias is related to the sustained complicated mechanisms of the autonomic nervous system. Atrial fibrillation is when the heart beats irregularly, and ventricular arrhythmias are rapid and inconsistent heart rhythms, which involves many factors including the autonomic nervous system. It's a complex topic that requires careful exploration. Cultivation of speculative knowledge on atrial fibrillation; the irregular rhythm of the heart and ventricular arrhythmias; rapid oscillating waves resulting from mistakenly inconsistent P waves, and the inclusion of an autonomic nervous system is an inconceivable approach toward clinical intricacies. Autonomic modulation, therefore, acquires new expansions and conceptions of appealing therapeutic intelligence to prevent cardiac arrhythmia. Notably, autonomic modulation uses the neural tissue's flexibility to cause remodeling and, hence, provide therapeutic effects. In addition, autonomic modulation techniques included stimulation of the vagus nerve and tragus, renal denervation, cardiac sympathetic denervation, and baroreceptor activation treatment. Strong preclinical evidence and early human studies support the annihilation of cardiac arrhythmias by sympathetic and parasympathetic systems to transmigrate the cardiac myocytes and myocardium as efficient determinants at the cellular and physiological levels. However, the goal of this study is to draw attention to these promising early pre-clinical and clinical arrhythmia treatment options that use autonomic modulation as a therapeutic modality to conquer the troublesome process of irregular heart movements. Additionally, we provide a summary of the numerous techniques for measuring autonomic tone such as heart rate oscillations and its association with cutaneous sympathetic nerve activity appear to be substitute indicators and predictors of the outcome of treatment.
Collapse
Affiliation(s)
- Ravina Rai
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar 470003 MP, India
| | - Virendra Singh
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005 UP, India
| | - Zaved Ahmad
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar 470003 MP, India
| | - Abhishek Jain
- Sanjeevani Diabetes and Heart Care Centre, Shri Chaitanya Hospital, Sagar, 470002, MP, India
| | - Deepali Jat
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar 470003 MP, India.
| | | |
Collapse
|
33
|
Eisen AM, Bratman GN, Olvera-Alvarez HA. Susceptibility to stress and nature exposure: Unveiling differential susceptibility to physical environments; a randomized controlled trial. PLoS One 2024; 19:e0301473. [PMID: 38630650 PMCID: PMC11023441 DOI: 10.1371/journal.pone.0301473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Emerging epidemiological evidence indicates nature exposure could be associated with greater health benefits among groups in lower versus higher socioeconomic positions. One possible mechanism underpinning this evidence is described by our framework: (susceptibility) adults in low socioeconomic positions face higher exposure to persistent psychosocial stressors in early life, inducing a pro-inflammatory phenotype as a lifelong susceptibility to stress; (differential susceptibility) susceptible adults are more sensitive to the health risks of adverse (stress-promoting) environments, but also to the health benefits of protective (stress-buffering) environments. OBJECTIVE Experimental investigation of a pro-inflammatory phenotype as a mechanism facilitating greater stress recovery from nature exposure. METHODS We determined differences in stress recovery (via heart rate variability) caused by exposure to a nature or office virtual reality environment (10 min) after an acute stressor among 64 healthy college-age males with varying levels of susceptibility (socioeconomic status, early life stress, and a pro-inflammatory state [inflammatory reactivity and glucocorticoid resistance to an in vitro bacterial challenge]). RESULTS Findings for inflammatory reactivity and glucocorticoid resistance were modest but consistently trended towards better recovery in the nature condition. Differences in recovery were not observed for socioeconomic status or early life stress. DISCUSSION Among healthy college-age males, we observed expected trends according to their differential susceptibility when assessed as inflammatory reactivity and glucocorticoid resistance, suggesting these biological correlates of susceptibility could be more proximal indicators than self-reported assessments of socioeconomic status and early life stress. If future research in more diverse populations aligns with these trends, this could support an alternative conceptualization of susceptibility as increased environmental sensitivity, reflecting heightened responses to adverse, but also protective environments. With this knowledge, future investigators could examine how individual differences in environmental sensitivity could provide an opportunity for those who are the most susceptible to experience the greatest health benefits from nature exposure.
Collapse
Affiliation(s)
- Aaron M. Eisen
- School of Nursing, Oregon Health & Science University, Portland, OR, United States of America
| | - Gregory N. Bratman
- School of Environmental and Forest Sciences, University of Washington, Seattle, WA, United States of America
- Department of Psychology, University of Washington, Seattle, WA, United States of America
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
| | | |
Collapse
|
34
|
Tompkins JD, Hoover DB, Havton LA, Patel JC, Cho Y, Smith EH, Biscola NP, Ajijola OA, Shivkumar K, Ardell JL. Comparative specialization of intrinsic cardiac neurons in humans, mice, and pigs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588174. [PMID: 38645175 PMCID: PMC11030249 DOI: 10.1101/2024.04.04.588174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Intrinsic cardiac neurons (ICNs) play a crucial role in the proper functioning of the heart; yet a paucity of data pertaining to human ICNs exists. We took a multidisciplinary approach to complete a detailed cellular comparison of the structure and function of ICNs from mice, pigs, and humans. Immunohistochemistry of whole and sectioned ganglia, transmission electron microscopy, intracellular microelectrode recording and dye filling for quantitative morphometry were used to define the neurophysiology, histochemistry, and ultrastructure of these cells across species. The densely packed, smaller ICNs of mouse lacked dendrites, formed axosomatic connections, and had high synaptic efficacy constituting an obligatory synapse. At Pig ICNs, a convergence of subthreshold cholinergic inputs onto extensive dendritic arbors supported greater summation and integration of synaptic input. Human ICNs were tonically firing, with synaptic stimulation evoking large suprathreshold excitatory postsynaptic potentials like mouse, and subthreshold potentials like pig. Ultrastructural examination of synaptic terminals revealed conserved architecture, yet small clear vesicles (SCVs) were larger in pigs and humans. The presence and localization of ganglionic neuropeptides was distinct, with abundant VIP observed in human but not pig or mouse ganglia, and little SP or CGRP in pig ganglia. Action potential waveforms were similar, but human ICNs had larger after-hyperpolarizations. Intrinsic excitability differed; 93% of human cells were tonic, all pig neurons were phasic, and both phasic and tonic phenotypes were observed in mouse. In combination, this publicly accessible, multimodal atlas of ICNs from mice, pigs, and humans identifies similarities and differences in the evolution of ICNs.
Collapse
Affiliation(s)
- John D. Tompkins
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Donald B. Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Leif A. Havton
- Departments of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Janaki C. Patel
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Youngjin Cho
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Elizabeth H. Smith
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Natalia P. Biscola
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jeffrey L. Ardell
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
35
|
Alrosan AZ, Heilat GB, Alrosan K, Aleikish AA, Rabbaa AN, Shakhatreh AM, Alshalout EM, Al Momany EM. Autonomic brain functioning and age-related health concerns. Curr Res Physiol 2024; 7:100123. [PMID: 38510918 PMCID: PMC10950753 DOI: 10.1016/j.crphys.2024.100123] [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: 01/31/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
The autonomic nervous system (ANS) regulates involuntary bodily functions such as blood pressure, heart rate, breathing, and digestion, in addition to controlling motivation and behavior. In older adults, the ANS is dysregulated, which changes the ability of the ANS to respond to physiological signals, regulate cardiovascular autonomic functionality, diminish gastric motility, and exacerbate sleep problems. For example, a decrease in heart rate variability, or the variation in the interval between heartbeats, is one of the most well-known alterations in the ANS associated with health issues, including cardiovascular diseases and cognitive decline. The inability to perform fundamental activities of daily living and compromising the physiological reactivity or motivational responses of older adults to moving toward or away from specific environmental stimuli are significant negative consequences of chronic and geriatric conditions that pose grave threats to autonomy, health, and well-being. The most updated research has investigated the associations between the action responsiveness of older adults and the maintenance of their physiological and physical health or the development of mental and physical health problems. Once autonomic dysfunction may significantly influence the development of different age-related diseases, including ischemic stroke, cardiovascular disease, and autoimmune diseases, this review aimed to assess the relationship between aging and autonomic functions. The review explored how motivational responses, physiological reactivity, cognitive processes, and lifelong developmental changes associated with aging impact the ANS and contribute to the emergence of health problems.
Collapse
Affiliation(s)
- Amjad Z. Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Ghaith B. Heilat
- Department of General Surgery and Urology, Faculty of Medicine, The Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Khaled Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Abrar A. Aleikish
- Master of Pharmacology, Department of Pharmacology, Faculty of Medicine, The Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Aya N. Rabbaa
- Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Aseel M. Shakhatreh
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ehab M. Alshalout
- Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Enaam M.A. Al Momany
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| |
Collapse
|
36
|
Thompson N, Ravagli E, Mastitskaya S, Challita R, Hadaya J, Iacoviello F, Shah Idil A, Shearing PR, Ajijola OA, Ardell JL, Shivkumar K, Holder D, Aristovich K. Anatomical and functional organization of cardiac fibers in the porcine cervical vagus nerve allows spatially selective efferent neuromodulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574861. [PMID: 38260584 PMCID: PMC10802425 DOI: 10.1101/2024.01.09.574861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Cardiac disease progression reflects the dynamic interaction between adversely remodeled neurohumoral control systems and an abnormal cardiac substrate. Vagal nerve stimulation (VNS) is an attractive neuromodulatory option to dampen this dynamic interaction; however, it is limited by off-target effects. Spatially-selective VNS (sVNS) offers a promising solution to induce cardioprotection while mitigating off-target effects by specifically targeting pre-ganglionic parasympathetic efferent cardiac fibers. This approach also has the potential to enhance therapeutic outcomes by eliminating time-consuming titration required for optimal VNS. Recent studies have demonstrated the independent modulation of breathing rate, heart rate, and laryngeal contraction through sVNS. However, the spatial organization of afferent and efferent cardiac-related fibers within the vagus nerve remains unexplored. By using trial-and-error sVNS in vivo in combination with ex vivo micro-computed tomography fascicle tracing, we show the significant spatial separation of cardiac afferent and efferent fibers (179±55° SD microCT, p<0.05 and 200±137° SD, p<0.05 sVNS - degrees of separation across a cross-section of nerve) at the mid-cervical level. We also show that cardiac afferent fibers are located in proximity to pulmonary fibers consistent with recent findings of cardiopulmonary convergent neurons and circuits. We demonstrate the ability of sVNS to selectively elicit desired scalable heart rate decrease without stimulating afferent-related reflexes. By elucidating the spatial organization of cardiac-related fibers within the vagus nerve, our findings pave the way for more targeted neuromodulation, thereby reducing off-target effects and eliminating the need for titration. This, in turn, will enhance the precision and efficacy of VNS therapy in treating cardiac pathology, allowing for improved therapeutic efficacy.
Collapse
Affiliation(s)
- Nicole Thompson
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Enrico Ravagli
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Svetlana Mastitskaya
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Ronald Challita
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Joseph Hadaya
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Francesco Iacoviello
- Electrochemical Innovation Lab, Department of Chemical Engineering, University College London, London, United Kingdom
| | - Ahmad Shah Idil
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Paul R. Shearing
- Electrochemical Innovation Lab, Department of Chemical Engineering, University College London, London, United Kingdom
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jeffrey L. Ardell
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - David Holder
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Kirill Aristovich
- EIT and Neurophysiology Research Group, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| |
Collapse
|
37
|
Triposkiadis F, Briasoulis A, Kitai T, Magouliotis D, Athanasiou T, Skoularigis J, Xanthopoulos A. The sympathetic nervous system in heart failure revisited. Heart Fail Rev 2024; 29:355-365. [PMID: 37707755 DOI: 10.1007/s10741-023-10345-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 09/15/2023]
Abstract
Several attempts have been made, by the scientific community, to develop a unifying hypothesis that explains the clinical syndrome of heart failure (HF). The currently widely accepted neurohormonal model has substituted the cardiorenal and the cardiocirculatory models, which focused on salt-water retention and low cardiac output/peripheral vasoconstriction, respectively. According to the neurohormonal model, HF with eccentric left ventricular (LV) hypertrophy (LVH) (systolic HF or HF with reduced LV ejection fraction [LVEF] or HFrEF) develops and progresses because endogenous neurohormonal systems, predominantly the sympathetic nervous system (SNS) and the renin-angiotensin-aldosterone system (RAAS), exhibit prolonged activation following the initial heart injury exerting deleterious hemodynamic and direct nonhemodynamic cardiovascular effects. However, there is evidence to suggest that SNS overactivity often preexists HF development due to its association with HF risk factors, is also present in HF with preserved LVEF (diastolic HF or HFpEF), and that it is linked to immune/inflammatory factors. Furthermore, SNS activity in HF may be augmented by coexisting noncardiac morbidities and modified by genetic factors and demographics. The purpose of this paper is to provide a contemporary overview of the complex associations between SNS overactivity and the development and progression of HF, summarize the underlying mechanisms, and discuss the clinical implications as they relate to therapeutic interventions mitigating SNS overactivity.
Collapse
Affiliation(s)
| | - Alexandros Briasoulis
- Department of Therapeutics, Heart Failure and Cardio-Oncology Clinic, National and Kapodistrian Univesity of Athens, 11527, Athens, Greece
| | - Takeshi Kitai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Dimitrios Magouliotis
- Unit of Quality Improvement, Department of Cardiothoracic Surgery, University of Thessaly, Biopolis, 41110, Greece
| | - Thanos Athanasiou
- Department of Surgery and Cancer, Imperial College London, St Mary's Hospital, London, W2 1NY, UK
| | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, 41110, Larissa, Greece
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110, Larissa, Greece
| |
Collapse
|
38
|
Wang Y, Denton KM. Special Issue "Sympathetic Nerves and Cardiovascular Diseases". Int J Mol Sci 2024; 25:2633. [PMID: 38473880 DOI: 10.3390/ijms25052633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Cardiovascular diseases (CVDs) constitute a spectrum of disorders affecting the heart and blood vessels, which include coronary heart disease, cerebrovascular disease, and peripheral artery disease [...].
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Kate M Denton
- Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
39
|
Tendulkar M, Tendulkar R, Dhanda PS, Yadav A, Jain M, Kaushik P. Clinical potential of sensory neurites in the heart and their role in decision-making. Front Neurosci 2024; 17:1308232. [PMID: 38415053 PMCID: PMC10896837 DOI: 10.3389/fnins.2023.1308232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/29/2023] [Indexed: 02/29/2024] Open
Abstract
The process of decision-making is quite complex involving different aspects of logic, emotion, and intuition. The process of decision-making can be summarized as choosing the best alternative among a given plethora of options in order to achieve the desired outcome. This requires establishing numerous neural networks between various factors associated with the decision and creation of possible combinations and speculating their possible outcomes. In a nutshell, it is a highly coordinated process consuming the majority of the brain's energy. It has been found that the heart comprises an intrinsic neural system that contributes not only to the decision-making process but also the short-term and long-term memory. There are approximately 40,000 cells present in the heart known as sensory neurites which play a vital role in memory transfer. The heart is quite a mysterious organ, which functions as a blood-pumping machine and an endocrine gland, as well as possesses a nervous system. There are multiple factors that affect this heart ecosystem, and they directly affect our decision-making capabilities. These interlinked relationships hint toward the sensory neurites which modulate cognition and mood regulation. This review article aims to provide deeper insights into the various roles played by sensory neurites in decision-making and other cognitive functions. The article highlights the pivotal role of sensory neurites in the numerous brain functions, and it also meticulously discusses the mechanisms through which they modulate their effects.
Collapse
Affiliation(s)
- Mugdha Tendulkar
- K. J. Somaiya Medical College and Research Centre, Mumbai, India
| | - Reshma Tendulkar
- Vivekanand Education Society's College of Pharmacy, Mumbai, India
| | | | - Alpa Yadav
- Department of Botany, Indira Gandhi University, Rewari, India
| | - Mukul Jain
- Cell and Developmental Biology Lab, Center of Research for Development, Parul University, Vadodara, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Prashant Kaushik
- Chaudhary Charan Singh Haryana Agricultural University, Hisar, India
| |
Collapse
|
40
|
Kutikuppala LVS, Sharma S, Chavan M, Rangari G, Misra AK, Innamuri SR, Vijayakumar T, Varshitha G. Bromocriptine: does this drug of Parkinson's disease have a role in managing cardiovascular diseases? Ann Med Surg (Lond) 2024; 86:926-929. [PMID: 38333315 PMCID: PMC10849299 DOI: 10.1097/ms9.0000000000001642] [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: 10/07/2023] [Accepted: 12/11/2023] [Indexed: 02/10/2024] Open
Abstract
Cardiovascular disease (CVD) is the most common cause of morbidity and mortality worldwide. Bromocriptine is a partial antagonist for D1 dopamine receptors while also serving as a selective agonist on D2 dopamine receptors as a dopamine receptor agonist. Apart from prolactin inhibiting action, bromocriptine has some beneficial effects on the blood pressure, plasma norepinephrine levels and vascular resistance. Dopamine D2 receptor activation of bromocriptine is associated with the antihypertensive effect, which lowers blood pressure via inhibiting sympathetic nerve activity and Na/K ATPase activity. Plasma levels of the pro-inflammatory cytokines such as interleukin (IL)-1B and IL-18, chemokine CCL2/ MCP-1/, and the pro-inflammatory hormone prolactin, all of which are elevated and linked to accelerated cardiometabolic illness, were decreased because of bromocriptine therapy. The most common side effects of Bromocriptine use are dizziness, nausea, headache, vomiting and hypotension. Bromocriptine is mainly contraindicated in patients with syncope with hypotension, psychosis, and type I diabetes mellitus. The authors suggest that developing therapies directed to increase D2 receptor expression and function by drugs like Bromocriptine can provide practical and novelistic approaches to prevent and manage myocardial and renal injury in the cardiovascular disease patients.
Collapse
Affiliation(s)
| | - Sushil Sharma
- Department of Pharamacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
| | - Madhavrao Chavan
- Department of Pharamacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
| | - Gaurav Rangari
- Department of Pharamacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
| | - Arup Kumar Misra
- Department of Pharamacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
| | - Sai Ram Innamuri
- Department of Pharamacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
| | - Tejus Vijayakumar
- Department of Pharamacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
| | - Golla Varshitha
- Department of General Medicine, International School of Medicine (ISM), Bishkek, Kyrgyzstan
| |
Collapse
|
41
|
Schwarz KG, Vicencio SC, Inestrosa NC, Villaseca P, Del Rio R. Autonomic nervous system dysfunction throughout menopausal transition: A potential mechanism underpinning cardiovascular and cognitive alterations during female ageing. J Physiol 2024; 602:263-280. [PMID: 38064358 DOI: 10.1113/jp285126] [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: 08/02/2023] [Accepted: 11/24/2023] [Indexed: 01/16/2024] Open
Abstract
Cardiovascular diseases (CVD) and neurodegenerative disorders, such as Alzheimer's disease (AD), are highly prevalent conditions in middle-aged women that severely impair quality of life. Recent evidence suggests the existence of an intimate cross-talk between the heart and the brain, resulting from a complex network of neurohumoral circuits. From a pathophysiological perspective, the higher prevalence of AD in women may be explained, at least in part, by sex-related differences in the incidence/prevalence of CVD. Notably, the autonomic nervous system, the main heart-brain axis physiological orchestrator, has been suggested to play a role in the incidence of adverse cardiovascular events in middle-aged women because of decreases in oestrogen-related signalling during transition into menopause. Despite its overt relevance for public health, this hypothesis has not been thoroughly tested. Accordingly, in this review, we aim to provide up to date evidence supporting how changes in circulating oestrogen levels during transition to menopause may trigger autonomic dysfunction, thus promoting cardiovascular and cognitive decline in women. A main focus on the effects of oestrogen-mediated signalling at CNS structures related to autonomic regulation is provided, particularly on the role of oestrogens in sympathoexcitation. Improving the understanding of the contribution of the autonomic nervous system on the development, maintenance and/or progression of both cardiovascular and cognitive dysfunction during the transition to menopause should help improve the clinical management of elderly women, with the outcome being an improved life quality during the natural ageing process.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sinay C Vicencio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Paulina Villaseca
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
42
|
Kattner AA. Colonizing foreign terrain: Insights into bacterial enteropathogens. Biomed J 2023; 46:100681. [PMID: 38042347 PMCID: PMC10774447 DOI: 10.1016/j.bj.2023.100681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023] Open
Abstract
In this present issue of the Biomedical Journal insights into pediatric campylobacteriosis are granted, and a potential path to developing a parenteral vaccine against enterotoxigenic E. coli is demonstrated. Additionally, a study shows how the use of extracorporeal shockwave therapy contributes to countering osteonecrosis of the femoral head. Furthermore, the relation between intimate partner violence and a saliva biomarker is explored. Finally, findings concerning the risk of dementia in patients with autonomic nervous system dysregulation are elucidated; and patterns of non-Alzheimer disease pathophysiology in individuals with depressive disorder are revealed.
Collapse
|
43
|
Salamon RJ, Halbe P, Kasberg W, Bae J, Audhya A, Mahmoud AI. Parasympathetic and sympathetic axons are bundled in the cardiac ventricles and undergo physiological reinnervation during heart regeneration. iScience 2023; 26:107709. [PMID: 37674983 PMCID: PMC10477065 DOI: 10.1016/j.isci.2023.107709] [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: 03/15/2023] [Revised: 07/25/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Sympathetic innervation influences homeostasis, repair, and pathology in the cardiac ventricles; in contrast, parasympathetic innervation is considered to have minimal contribution and influence in the ventricles. Here, we use genetic models, whole-mount imaging, and three-dimensional modeling to define cardiac nerve architecture during development, disease, and regeneration. Our approach reveals that parasympathetic nerves extensively innervate the cardiac ventricles. Furthermore, we identify that parasympathetic and sympathetic axons develop synchronously and are bundled throughout the ventricles. We further investigate cardiac nerve remodeling in the regenerative neonatal and the non-regenerative postnatal mouse heart. Our results show that the regenerating myocardium undergoes a unique process of physiological reinnervation, where proper nerve distribution and architecture is reestablished, in stark contrast to the non-regenerating heart. Mechanistically, we demonstrate that physiological reinnervation during regeneration is dependent on collateral artery formation. Our results reveal clinically significant insights into cardiac nerve plasticity which can identify new therapies for cardiac disease.
Collapse
Affiliation(s)
- Rebecca J. Salamon
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Poorva Halbe
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - William Kasberg
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Jiyoung Bae
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Ahmed I. Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
44
|
Yamaguchi T, Salavatian S, Kuwabara Y, Hellman A, Taylor BK, Howard-Quijano K, Mahajan A. Thoracic Dorsal Root Ganglion Application of Resiniferatoxin Reduces Myocardial Ischemia-Induced Ventricular Arrhythmias. Biomedicines 2023; 11:2720. [PMID: 37893094 PMCID: PMC10604235 DOI: 10.3390/biomedicines11102720] [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: 08/23/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND A myocardial ischemia/reperfusion (IR) injury activates the transient receptor potential vanilloid 1 (TRPV1) dorsal root ganglion (DRG) neurons. The activation of TRPV1 DRG neurons triggers the spinal dorsal horn and the sympathetic preganglionic neurons in the spinal intermediolateral column, which results in sympathoexcitation. In this study, we hypothesize that the selective epidural administration of resiniferatoxin (RTX) to DRGs may provide cardioprotection against ventricular arrhythmias by inhibiting afferent neurotransmission during IR injury. METHODS Yorkshire pigs (n = 21) were assigned to either the sham, IR, or IR + RTX group. A laminectomy and sternotomy were performed on the anesthetized animals to expose the left T2-T4 spinal dorsal root and the heart for IR intervention, respectively. RTX (50 μg) was administered to the DRGs in the IR + RTX group. The activation recovery interval (ARI) was measured as a surrogate for the action potential duration (APD). Arrhythmia risk was investigated by assessing the dispersion of repolarization (DOR), a marker of arrhythmogenicity, and measuring the arrhythmia score and the number of non-sustained ventricular tachycardias (VTs). TRPV1 and calcitonin gene-related peptide (CGRP) expressions in DRGs and CGRP expression in the spinal cord were assessed using immunohistochemistry. RESULTS The RTX mitigated IR-induced ARI shortening (-105 ms ± 13 ms in IR vs. -65 ms ± 11 ms in IR + RTX, p = 0.028) and DOR augmentation (7093 ms2 ± 701 ms2 in IR vs. 3788 ms2 ± 1161 ms2 in IR + RTX, p = 0.020). The arrhythmia score and VT episodes during an IR were decreased by RTX (arrhythmia score: 8.01 ± 1.44 in IR vs. 3.70 ± 0.81 in IR + RTX, p = 0.037. number of VT episodes: 12.00 ± 3.29 in IR vs. 0.57 ± 0.3 in IR + RTX, p = 0.002). The CGRP expression in the DRGs and spinal cord was decreased by RTX (DRGs: 6.8% ± 1.3% in IR vs. 0.6% ± 0.2% in IR + RTX, p < 0.001. Spinal cord: 12.0% ± 2.6% in IR vs. 4.5% ± 0.8% in IR + RTX, p = 0.047). CONCLUSIONS The administration of RTX locally to thoracic DRGs reduces ventricular arrhythmia in a porcine model of IR, likely by inhibiting spinal afferent hyperactivity in the cardio-spinal sympathetic pathways.
Collapse
Affiliation(s)
- Tomoki Yamaguchi
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.Y.); (S.S.); (Y.K.); (A.H.); (B.K.T.); (K.H.-Q.)
| | - Siamak Salavatian
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.Y.); (S.S.); (Y.K.); (A.H.); (B.K.T.); (K.H.-Q.)
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | - Yuki Kuwabara
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.Y.); (S.S.); (Y.K.); (A.H.); (B.K.T.); (K.H.-Q.)
| | - Abigail Hellman
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.Y.); (S.S.); (Y.K.); (A.H.); (B.K.T.); (K.H.-Q.)
| | - Bradley K. Taylor
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.Y.); (S.S.); (Y.K.); (A.H.); (B.K.T.); (K.H.-Q.)
| | - Kimberly Howard-Quijano
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.Y.); (S.S.); (Y.K.); (A.H.); (B.K.T.); (K.H.-Q.)
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | - Aman Mahajan
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.Y.); (S.S.); (Y.K.); (A.H.); (B.K.T.); (K.H.-Q.)
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| |
Collapse
|
45
|
Rawn KP, Keller PS. Child emotion lability is associated with within-task changes of autonomic activity during a mirror-tracing task. Psychophysiology 2023; 60:e14354. [PMID: 37246804 DOI: 10.1111/psyp.14354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 05/30/2023]
Abstract
Adaptive biological and emotional stress responding are both critical for healthy human development. However, the complex associations between the two are not fully understood. The current study addresses this gap in research by studying associations of child emotion regulation and lability with within-task changes in the biological stress response during a mirror-tracing task. Participants were 59 families including two parents and a child between 5 and 12 years old (52.2% female). Parents reported on family demographics and completed the Emotion Regulation Checklist. Child skin conductance level (SCL) and respiratory sinus arrhythmia (RSA) were recorded during a baseline task and during a 3-minute mirror-tracing task. Within-task patterns of SCL and RSA during the task were estimated with multilevel modeling (measures within persons). The emotion regulation subscale was unrelated to any facet of SCL/RSA time courses. However, lower emotion lability was related to SCL patterns that changed less during the task and were overall lower. For RSA, lower emotion lability was related to higher initial RSA that significantly decreased during the task. These findings suggest that higher child emotion lability may promote increased physiological arousal of target organs during challenging activities.
Collapse
Affiliation(s)
- Kyle P Rawn
- Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| | - Peggy S Keller
- Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
46
|
Hadaya J, Dajani AH, Cha S, Hanna P, Challita R, Hoover DB, Ajijola OA, Shivkumar K, Ardell JL. Vagal Nerve Stimulation Reduces Ventricular Arrhythmias and Mitigates Adverse Neural Cardiac Remodeling Post-Myocardial Infarction. JACC Basic Transl Sci 2023; 8:1100-1118. [PMID: 37791302 PMCID: PMC10543930 DOI: 10.1016/j.jacbts.2023.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 10/05/2023]
Abstract
This study sought to evaluate the impact of chronic vagal nerve stimulation (cVNS) on cardiac and extracardiac neural structure/function after myocardial infarction (MI). Groups were control, MI, and MI + cVNS; cVNS was started 2 days post-MI. Terminal experiments were performed 6 weeks post-MI. MI impaired left ventricular mechanical function, evoked anisotropic electrical conduction, increased susceptibility to ventricular tachycardia and fibrillation, and altered neuronal and glial phenotypes in the stellate and dorsal root ganglia, including glial activation. cVNS improved cardiac mechanical function and reduced ventricular tachycardia/ventricular fibrillation post-MI, partly by stabilizing activation/repolarization in the border zone. MI-associated extracardiac neural remodeling, particularly glial activation, was mitigated with cVNS.
Collapse
Affiliation(s)
- Joseph Hadaya
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, California, USA
| | - Al-Hassan Dajani
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Steven Cha
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Peter Hanna
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, California, USA
| | - Ronald Challita
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at 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
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, California, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, California, USA
| | - Jeffrey L. Ardell
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
47
|
Salvini V, Accioli R, Lazzerini PE, Acampa M. Editorial: New challenges and future perspectives in autonomic neuroscience. Front Neurosci 2023; 17:1271499. [PMID: 37680971 PMCID: PMC10482394 DOI: 10.3389/fnins.2023.1271499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023] Open
Affiliation(s)
- Viola Salvini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Riccardo Accioli
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Maurizio Acampa
- Stroke Unit, Department of Emergency-Urgency and Transplants, Azienda Ospedaliera Universitaria Senese, “Santa Maria alle Scotte” General-Hospital, Siena, Italy
| |
Collapse
|
48
|
Nagai M, Dote K, Förster CY. Denervation or stimulation? Role of sympatho-vagal imbalance in HFpEF with hypertension. Hypertens Res 2023; 46:1727-1737. [PMID: 37045971 DOI: 10.1038/s41440-023-01272-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/17/2023] [Accepted: 03/12/2023] [Indexed: 04/14/2023]
Abstract
Heart failure (HF) in the elderly is an increasingly large and complex problem in modern society. Notably, the cause of HF with preserved ejection fraction (HFpEF) is multifactorial and its pathophysiology is not fully understood. Among these, hypertension has emerged as a pivotal factor in the pathophysiology and therapeutic targets of HFpEF. Neuronal elements distributed throughout the cardiac autonomic nervous system, from the level of the central autonomic network including the insular cortex to the intrinsic cardiac nervous system, regulate the human cardiovascular system. Specifically, increased sympathetic nervous system activity due to sympatho-vagal imbalance is suggested to be associated the relationship between hypertension and HFpEF. While several new pharmacological therapies, such as sodium-glucose cotransporter 2 inhibitors, have been shown to be effective in HFpEF, neuromodulatory therapies of renal denervation and vagus nerve stimulation (VNS) have received recent attention. The current review explores the pathophysiology of the brain-heart axis that underlies the relationship between hypertension and HFpEF and the rationale for therapeutic neuromodulation of HFpEF by non-invasive transcutaneous VNS.
Collapse
Affiliation(s)
- Michiaki Nagai
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan.
| | - Keigo Dote
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Carola Yvette Förster
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Würzburg, Germany
| |
Collapse
|
49
|
Grandi E, Navedo MF, Saucerman JJ, Bers DM, Chiamvimonvat N, Dixon RE, Dobrev D, Gomez AM, Harraz OF, Hegyi B, Jones DK, Krogh-Madsen T, Murfee WL, Nystoriak MA, Posnack NG, Ripplinger CM, Veeraraghavan R, Weinberg S. Diversity of cells and signals in the cardiovascular system. J Physiol 2023; 601:2547-2592. [PMID: 36744541 PMCID: PMC10313794 DOI: 10.1113/jp284011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023] Open
Abstract
This white paper is the outcome of the seventh UC Davis Cardiovascular Research Symposium on Systems Approach to Understanding Cardiovascular Disease and Arrhythmia. This biannual meeting aims to bring together leading experts in subfields of cardiovascular biomedicine to focus on topics of importance to the field. The theme of the 2022 Symposium was 'Cell Diversity in the Cardiovascular System, cell-autonomous and cell-cell signalling'. Experts in the field contributed their experimental and mathematical modelling perspectives and discussed emerging questions, controversies, and challenges in examining cell and signal diversity, co-ordination and interrelationships involved in cardiovascular function. This paper originates from the topics of formal presentations and informal discussions from the Symposium, which aimed to develop a holistic view of how the multiple cell types in the cardiovascular system integrate to influence cardiovascular function, disease progression and therapeutic strategies. The first section describes the major cell types (e.g. cardiomyocytes, vascular smooth muscle and endothelial cells, fibroblasts, neurons, immune cells, etc.) and the signals involved in cardiovascular function. The second section emphasizes the complexity at the subcellular, cellular and system levels in the context of cardiovascular development, ageing and disease. Finally, the third section surveys the technological innovations that allow the interrogation of this diversity and advancing our understanding of the integrated cardiovascular function and dysfunction.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Rose E. Dixon
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Ana M. Gomez
- Signaling and Cardiovascular Pathophysiology-UMR-S 1180, INSERM, Université Paris-Saclay, Orsay, France
| | - Osama F. Harraz
- Department of Pharmacology, Larner College of Medicine, and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - David K. Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Trine Krogh-Madsen
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, New York, USA
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Matthew A. Nystoriak
- Department of Medicine, Division of Environmental Medicine, Center for Cardiometabolic Science, University of Louisville, Louisville, KY, 40202, USA
| | - Nikki G. Posnack
- Department of Pediatrics, Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children’s National Heart Institute, Children’s National Hospital, Washington, DC, USA
| | | | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University – Wexner Medical Center, Columbus, OH, USA
| | - Seth Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University – Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
50
|
Li YH, Sun CC, Chen PM, Chen HH. SGK1 Target Genes Involved in Heart and Blood Vessel Functions in PC12 Cells. Cells 2023; 12:1641. [PMID: 37371111 DOI: 10.3390/cells12121641] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is expressed in neuronal cells and involved in the pathogenesis of hypertension and metabolic syndrome, regulation of neuronal function, and depression in the brain. This study aims to identify the cellular mechanisms and signaling pathways of SGK1 in neuronal cells. In this study, the SGK1 inhibitor GSK650394 is used to suppress SGK1 expression in PC12 cells using an in vitro neuroscience research platform. Comparative transcriptomic analysis was performed to investigate the effects of SGK1 inhibition in nervous cells using mRNA sequencing (RNA-seq), differentially expressed genes (DEGs), and gene enrichment analysis. In total, 12,627 genes were identified, including 675 and 2152 DEGs at 48 and 72 h after treatment with GSK650394 in PC12 cells, respectively. Gene enrichment analysis data indicated that SGK1 inhibition-induced DEGs were enriched in 94 and 173 genes associated with vascular development and functional regulation and were validated using real-time PCR, Western blotting, and GEPIA2. Therefore, this study uses RNA-seq, DEG analysis, and GEPIA2 correlation analysis to identify positive candidate genes and signaling pathways regulated by SGK1 in rat nervous cells, which will enable further exploration of the underlying molecular signaling mechanisms of SGK1 and provide new insights into neuromodulation in cardiovascular diseases.
Collapse
Affiliation(s)
- Yu-He Li
- Department of Laboratory Medicine, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| | - Chia-Cheng Sun
- Physical Examination Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Po-Ming Chen
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Hsin-Hung Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| |
Collapse
|