Open access peer-reviewed chapter

Autonomic Nervous System and Neurocardiac Physiopathology

Written By

John G. Kingma, Denys Simard and Jacques R. Rouleau

Submitted: 05 February 2018 Reviewed: 09 April 2018 Published: 24 October 2018

DOI: 10.5772/intechopen.77087

From the Edited Volume

Autonomic Nervous System

Edited by Pavol Svorc

Chapter metrics overview

1,509 Chapter Downloads

View Full Metrics

Abstract

The autonomic nervous system regulates multiple physiological functions; how distinct neurons in peripheral autonomic and intrathoracic ganglia communicate remains to be established. Increasing focus is being paid to functionality of the neurocardiac axis and crosstalk between the intrinsic nervous system and diverse organ systems. Current findings indicate that progression of cardiovascular disease comprises peripheral and central aspects of the cardiac nervous system hierarchy. Indeed, autonomic neuronal dysfunction is known to participate in arrhythmogenesis and sudden cardiac death; diverse interventions (pharmacological, non-pharmacological) that affect neuronal remodeling in the heart following injury caused by cardiovascular disease (congestive heart failure, etc.) or acute myocardial infarction are being investigated. Herein we examine recent findings from clinical and animal studies on the role of the intrinsic cardiac nervous system on regulation of myocardial perfusion and the consequences of cardiac injury. We also discuss different interventions that target the autonomic nervous system, stimulate neuronal remodeling and adaptation, and thereby optimize patient outcomes.

Keywords

  • autonomic nervous system
  • sympathetic
  • parasympathetic nerves
  • intrinsic cardiac neurons
  • intrinsic cardiac nervous system
  • ischemia
  • arrhythmias

1. Introduction

Physiological functions (i.e. muscle contraction, glandular function, visceral activity, nerve impulses, etc.) of the body are controlled by the autonomic nervous system (ANS). Innervation to the heart is consistent among species [1, 2, 3]; the ANS comprises central, intrathoracic extracardiac and intrinsic cardiac components (see review by Hanna et al. [4]). The sympathetic and parasympathetic systems interact to stimulate energy expenditure under conditions of stress or return the body to a restful state; these systems comprise pathways that include preganglionic and postganglionic neurons (activated by endogenous chemical neurotransmitters). Increasing attention focuses on the complex anatomy and function of the cardiac neuraxis; how diverse populations of neurons in peripheral autonomic and intrathoracic ganglia communicate with each other and between different organ systems remains the subject of ongoing investigation. Treatment strategies that modulate the ANS are being developed and tested in the setting of cardiac dysfunction, arrhythmias and sudden death with the objective of stimulating or maintaining cardiovascular function. Improved mechanistic understanding of changes that occur within the nervous system hierarchy during pathogenesis of cardiac disease is therefore essential. This chapter examines current scientific literature on the effects of ischemia on the cardiac nervous system; the role of intrinsic cardiac neurons on regulation of myocardial blood flow, cardiac function, pathogenesis of nerve and myocardial tissue injury is discussed. For this review, clinical and basic science reports were searched on MEDLINE, Google Scholar and PubMed with the keywords intrinsic cardiac nervous system (ICNS), ischemia, reperfusion, cellular protection, myocardium, nerves and combinations thereof. In addition, we referred to data from our own research.

Advertisement

2. Cardiac nervous system

The sympathetic (adrenergic) component of the ANS stimulates cardiac conduction and myocardial cells; on the other hand, the parasympathetic (cholinergic) nervous system exerts an inhibitory influence [5, 6]. Regulation of cardiac performance by the ANS involves modulation of heart rate (positive chronotropy), increases in cardiac contractility (positive inotropy) and cardiac relaxation (positive lusitropy), decreased venous capacitance plus constriction of resistance and cutaneous vessels [7].

Sympathetic cardiac nerves originate from stellate, superior, middle cervical and thoracic ganglia [8]; postganglionic sympathetic neurons project efferent axons to the heart [9]. Parasympathetic nerves develop from the cardiac component of the cranial neural crest; preganglionic neurons access to the heart occurs via the vagus nerves [10, 11]. Cardiac ganglia are located in epicardial fat, in ganglionated plexi adjacent to the major cardiac vessels and in the ventricular wall [12, 13, 14]. ANS neurons are classified by chemical phenotyping; cholinergic and adrenergic populations of ganglionic cardiac neurons are readily found in cardiac ganglia [15, 16, 17]. Sensory neurons, interneurons and sensory fibers that develop from the nucleus ambiguus [18, 19, 20] likely play a role in pathogenesis of cardiac disease. In fact, activation of the neuroendocrine system is considered central to pathogenesis of cardiac disease; excess sympathetic activation promotes cardiovascular dysfunction, arrhythmias and sudden death [21]. Of note, is that visualization of the ICNS and the presence of interneuron connections is particularly challenging [22, 23, 24, 25]; however, several immune histochemical techniques which target specific neurotransmitters within parasympathetic and sympathetic neurons have been particularly successful [26, 27, 28, 29, 30]. Neuroimaging techniques, cardioneural optical mapping and optogenetics are also being used to define the complex anatomy of the cardiac nervous system in animals and living humans to evaluate the role of the ANS in normal cardiac function as well as pathogenesis of cardiac disease [4, 31, 32, 33].

Advertisement

3. Vasoregulation

Arteries normally respond to humoral, metabolic, mechanical and neural stimulation; local metabolic control occurs secondarily to myocardial metabolic change [34, 35]. In the heart, blood flow across the ventricular wall is precisely coordinated to metabolic requirements via adjustments in vessel tone; flow is therefore independent of external physical factors since metabolism is the ultimate determinant of regional perfusion over the operative range of autoregulation [36, 37, 38]. The ANS contributes to regulation of myocardial blood flow; sympathetic nerve stimulation produces a biphasic response, which trends to coronary dilatation resulting from increases in myocardial oxygen demand as well as perfusion pressure [39, 40]. Neuropeptide chemicals elevate local catecholamine levels that modulate cardiac dynamics and indirectly increase blood flow across the left ventricular wall [40, 41, 42]. In dogs, with an intact cardiac nervous system, we documented significant increases in myocardial blood flow following application of nicotine or bradykinin to selected ganglionated plexi on the heart [40]; stimulation of nicotine sensitive neurons increases cardiac metabolic demand (i.e. higher heart rate and LV systolic pressure) but stimulation with bradykinin produces a similar result without affecting LV pressure. On the other hand, Vergroesen et al. documented that intact cardiac nerves were not essential for regulation of coronary blood flow [43]; however, they suggested that cardiac nerves essentially alter the speed of response of the coronary vascular bed to changes in heart rate and perfusion pressure. The cardiac nervous reflexes thought to be responsible for these effects has not been established but diverse cardiac afferent fibers and receptor subtypes (i.e. ventricular, coronary artery) have been studied.

Stimulation of ventricular mechanoreceptors causes an increase in arterial perfusion pressure, which results in greater blood volume and reflex coronary vasodilatation [44, 45]; higher perfusion pressures promote vasoconstriction. However, stimulation of coronary artery baroreceptors also promote reflex vasodilatation [46]. These reflex responses following mechanoreceptor stimulation may confer protection against arterial injury and thereby slow progression of coronary artery disease.

Local release of prostaglandins, nitric oxide (NO) and endothelium-derived relaxation factors stimulate activation of select populations of cardiac neurons that contribute to vasoregulation. NO contributes to neuronal mediated vasoregulation; NO induced vasodilatation involves adrenergic, myogenic and hormonal influences [47, 48]. NO in concert with other vasoactive mediators effectively counteracts vasoconstrictor mechanisms [49, 50, 51]; these effects may be gender dependent. Three nitric oxide synthase (NOS) isoforms that synthesize NO from l-arginine have been documented; of these, two are constitutively expressed Ca2+-dependent isoforms—eNOS (endothelial) is localized in cardiocytes as well as vascular and endocardial endothelium while nNOS (neuronal) is found in cardiac neurons [52, 53, 54]. The ubiquitous nature of NO implies a role in regulation of central nervous system, myocardium and vascular function [55]; nNOS and cardiac inhibitory G protein are believed to work in parallel in order to reduce sinus node rate and thereby modulate heart rate variability [56]. NO directly affects intrinsic cardiac neurons; almost 40% of these neurons are NOS positive [57]. Altered neuronal effects of NO may be important in pathogenesis of hypertension, septic shock, diabetes mellitus, etc.

Studies from our laboratory, in dogs subject to acute cardiac decentralization, indicated that intrinsic cardiac neurons function independently of central neuronal inputs. In decentralized and innervated hearts coronary autoregulation was similar (Figure 1) despite substantial reductions in myocardial oxygen demand (in decentralized hearts) [58]. In addition, perfusion across the ventricular wall (in decentralized hearts; Figure 2) was preserved thus confirming earlier findings of Rimoldi and coworkers [59] who reported no change in transmural distribution of myocardial blood flow after regional sympathetic denervation. Stability of perfusion across the ventricular wall was suggested to be due to several factors including neuronal modulation, autoregulation and variations in coronary resistance at the microvessel level (<100 μm). Interestingly, in neuropathy patients the innervation/ventricular perfusion ratio during reactive hyperemia is lower in innervated compared to denervated regions within the ventricular wall [60]. These findings are considerably different from those reported in human subjects after suppression of adenosine-mediated sympathetic activation [61].

Figure 1.

Coronary blood flow versus diastolic coronary artery pressure during autoregulation. Pressure-flow relations in dogs with intact cardiac nerves (closed circles) and after extracardiac nerve ablation (open circles) are shown. Note the similarity between the two curves; reactive hyperemia blood flow was lower in decentralized dogs as shown.

Figure 2.

Myocardial blood flow distribution across the ventricular wall (measured with microspheres) in hearts from dogs with intact cardiac nerves (closed symbols) and after acute cardiac decentralization (open symbols). Data are means ± SEM.

Advertisement

4. Ischemic injury

4.1. Nervous system

Ischemia significantly modulates function of intrinsic cardiac neurons because of local accumulations of metabolic by-products (i.e. reactive oxygen species, purinergic compounds, etc.) [62, 63, 64]. A limited number of animal studies have investigated the overall effects of ischemia on activity of nerves that course over, or through infarcted myocardium [65]; findings indicate that blood supply to these cardiac nerves is not a determining factor for conduction of action potentials [66]. The question of whether, or not, cardiac nerves are more, or less, sensitive to ischemia is less adequately studied; consequently, the injury threshold of cardiac nerves and neurons remains unknown. However, it is possible that activity of cardiac neurons post-ischemia is preserved consequent to stimulation of ventricular sensory neurites that transduce mechanical and chemical milieu in the myocardium [67]. During acute myocardial ischemia, norepinephrine is released from sympathetic nerves; this triggers sympathetic nerve regeneration (i.e. sprouting, budding) and nerve remodeling to promote sympathetic hyperinnervation, which ultimately plays a role in arrhythmogenesis [68, 69, 70, 71, 72]. Function of cardiac sympathetic neurons post-ischemia can also be triggered by the elevated production of intra-neuronal galanin (i.e. promotes regeneration of sympathetic axons) [73]; galanin modifies synaptic transmission and contributes to arrhythmogenesis and sudden cardiac death. Additionally, multiple autacoids (adenosine, bradykinin, NO, reactive oxygen species, etc.) produced during ischemia stimulate the central nervous system, cardiac autonomic ganglia and sympathetic efferent postganglionic axons in coronary vessels [74, 75]. Neuropeptide chemicals released from sensory neurites also modulate intrinsic cardiac neuronal activity [41, 76]. It is interesting to speculate that common survival pathways of cardiac neurons may be shared with cardiocytes but this has not been established.

4.2. Heart

Infarction causes major changes between peripheral and central aspects of the cardiac nervous system; structural and functional alterations at the cardiomyocyte level include; (1) changes in collagen matrix [77], (2) induction of electromechanical dyssynchrony [78], (3) ventricular contractile dysfunction [79], apoptosis [80], etc. In the heart, ischemia affects the ICNS which is the convergence point for cardiac neural control. As such, afferent inputs are modulated along with descending neural inputs [78] (i.e. including reflex-induced sympathoexcitation and reduced central drive from parasympathetic nerves [81, 82]). Heightened sympathetic tone partly mediated by neurotransmission through the stellate ganglia has been linked to cardiac pathogenesis as well as risk of cardiac arrhythmias and sudden death [83, 84].

Acute coronary artery occlusion produces distinct alterations in cardiomyocyte pathology that ultimately contribute to cell death; for cardiac myocytes a transmural gradient of cell death occurs in relation to duration and degree of ischemia [85, 86]. Transmural necrosis is mostly manifest by 6 h after acute coronary occlusion; the potential for tissue salvage after this time is limited (i.e. species dependent). Physiopathology of ischemic injury is generally well-documented [87, 88, 89, 90]; numerous cytoprotective strategies to limit ischemic injury (i.e. pharmacologic, endogenous, etc.) have been tested but none has achieved widespread clinical use [90, 91, 92]. Post-ischemic remodeling of sympathetic neurons in stellate ganglia is not well established; however, a potential relation exists between ganglion inflammation and oxidative stress [93]. A recent study in rodents documented greater oxidative stress (i.e. lipofuscin accumulation, mitochondrial degeneration, etc.), metabolic activity (higher rate of lipid peroxidation) and inflammation in stellate glial cells [94]. These physiopathological mechanisms are believed to contribute to local inflammation (i.e. leukocyte infiltration) within stellate ganglia; this stimulates neuronal activity and oxidative stress, which increases cardiac afferent neurotransmission [95]. Other contributing factors include circulating neurohormonal compounds (i.e. angiotensin II, etc.) and brainstem-mediated increases in efferent sympathetic outflow [96, 97, 98]. Equally, cardiac inflammation and oxidative stress produced by repeated defibrillation are involved in ganglion pathology [99].

The importance of cardiac nerves for the pathogenesis of post-ischemic infarct development and cardiac dysfunction has been investigated in experimental models of ischemia-reperfusion injury. In cardiac decentralized pigs, significant ventricular dysfunction accompanied by patchy subendocardial necrosis occurs after acute coronary occlusion [100]; myocardial perfusion-function relations in these animals were not affected by nerve status. In addition, we reported coronary vascular reserve to be comparable after nerve ablation albeit in a different experimental model [101], which is consistent with most published findings [102, 103, 104, 105]. We also confirmed a trend towards smaller infarcts in dogs subject to extracardiac nerve ablation or pharmacologic decentralization using the autonomic ganglionic blocker hexamethonium bromide (Figure 3) [106]; these findings are also in agreement with earlier studies documenting increased tolerance to ischemic injury and a reduction in ventricular fibrillation threshold post-decentralization [102, 107, 108]. Reduced oxygen demand and improved perfusion of affected tissues could be responsible for increased ischemic tolerance of myocytes [43, 104, 105, 109] in the absence of intact cardiac nerves. Of note, extracardiac surgical ablation of sympathetic and parasympathetic efferent neuronal inputs produces a decentralized (not denervated) heart without complete elimination of parasympathetic involvement [110, 111]; on the other hand, pharmacologic ganglionic blockade with hexamethonium bromide blocks transmission within peripheral autonomic ganglia and vagal cardio-acceleration [112]. Continued research into the identification of endogenous compounds that modulate or activate intrinsic neuronal populations to induce cellular protection remains a priority.

Figure 3.

Histogram of myocardial infarct size (as percent of anatomic area at risk) in dogs undergoing ischemia–reperfusion injury. Three distinct groups are shown: (1) control (CTR); (2) acute cardiac decentralized (DCN) and (3) hexamethonium bromide (HEX). Data are mean ± 1SD; n = 8/group.

4.3. Cardiac arrhythmogenesis

Sudden cardiac death due to ventricular arrhythmias is highly relevant to cardiovascular disease related mortality [113]; autonomic neuronal dysfunction is a major contributor to induction of atrial and ventricular dysrhythmias [114, 115, 116]. Pathologically induced disturbances in neural processing within the cardiac neural hierarchy affect efferent neuronal outputs throughout the myocardium [19, 117] (i.e. intrinsic and extrinsic cardiac ganglia, central reflexes [95, 118, 119, 120, 121]). Cardiac neurons are generally classified as afferent, efferent or convergent on the basis of responses to various cardiovascular stimuli [31, 120]. A study from Ardell’s laboratory examined functional remodeling of neuronal elements within the context of myocardial infarction [120]; they showed that: (1) morphological and phenotypic remodeling of intracardiac ganglia is dependent on the site of injury, (2) attenuation of afferent neural signals to intrinsic cardiac neurons (i.e. within ischemic zone) but preservation of these signals in remote and border regions (i.e. neural sensory border zone), (3) autonomic efferent inputs to intrinsic cardiac neurons are maintained, (4) transduction capacity increases in convergent intrinsic local circuit neurons (of the heart) and (5) connectivity of intrinsic cardiac neurons is reduced. Current findings suggest that neuronal remodeling can occur independently of direct injury to specific neuron subsets; as such, neuronal plasticity within the cardiac neuroaxis is crucial post-infarction and during progression of cardiovascular disease [122, 123]. Indeed, healed myocardium provides a particular challenge to electrical propagation and regulation of cardiac function [124, 125]; abnormal cardiac efferent signaling results in continuous discord between central and peripheral aspects within the neural hierarchy that produces fatal arrhythmias due to excessive sympathoexcitation [122]. The peri-infarct region (i.e. interface between dense scar and surviving myocardium) also has an increased potential for ectopic beats [71, 126]. Ajijola and coworkers recently determined that (1) despite scarring, myocyte loss and ion channel remodeling significant regulation of electrical activation occurs via cardiac sympathetic nerves within the peri-infarct region, and (2) there is significant remodeling of sympathetic innervation within the anteroapical region [127]; additionally, they emphasized the critical role of adrenergic activation in modulating propagation patterns.

Premature ventricular contractions (PVC; contraction of the ventricles caused by abnormal electrical activity) often lead to cardiovascular events, left ventricle dysfunction and sudden cardiac death [128]; multiple mechanisms have been proposed including mechanical dyssynchrony, abnormalities in calcium handling and oxygen consumption and autonomic imbalance [122, 129, 130]. Hamon and coworkers documented (using in vivo cardioneural mapping) that PVCs are a powerful stressor for the ICNS and that PVC-induced neural and electrophysiological changes are critical for arrhythmogenesis and remodeling. PVCs with variable coupling intervals have a more complex impact on cardiac neurons than those with fixed short or long coupling intervals [128]. The unpredictability of coupling intervals could trigger a sympathovagal imbalance that influences cardiomyocyte function and leads to electric instability. Greater neuronal responses (particularly within convergent neurons that are responsible for reflex processing) to variable compared to constant stimulus (i.e. neural adaptation) have been described in sensory neurons of the visual, auditory and olfactory systems [131, 132]. In the heart sympathetic nerve activity is greater during irregular cardiac pacing and is independent of hemodynamic changes [133]. As such, increased variability of PVC coupling interval could play a role in reflex activation of the ANS. Greater understanding of underlying mechanoelectric mechanisms of PVC-induced arrhythmogenesis should help to improve risk stratification in cardiac patients that would allow use of more aggressive pharmacologic and non-pharmacologic therapeutics (i.e. specifically targeting the ANS) in prophylactic management (cf. Table 1).

Pertinent studies
Pharmacological interventions
  • Neuregulin-1

[134, 135, 136, 137, 138]
  • Ghrelin

[139]
  • Vasopressin

[140]
  • Anesthetic preconditioning

[141]
Non-pharmacological interventions
  • Transcutaneous electrical nerve stimulation (TENS)

[142, 143, 144]
  • Bioelectronic block

[145, 146, 147, 148]
  • Spinal cord stimulation (SCS)

[118, 149, 150, 151, 152]
  • Vagal nerve stimulation (VNS)

[78, 153, 154]
  • Renal nerve denervation

[155, 156, 157]
  • Cardiac decentralization and carotid body ablation

[158, 159, 160, 161, 162]
  • Cardiac conditioning (ischemia, exercise)

[21, 163, 164]

Table 1.

Management strategies that target the autonomic nervous system.

Advertisement

5. Therapeutic interventions

5.1. Pharmacological

Pharmacologic interventions can play an important role in post-ischemic nerve repair; though most medications reduce the incidence of arrhythmias some can be proarrhythmic [165]. Significant improvement in acute and chronic ischemic cardiomyopathy, myocarditis and vagus nerve remodeling have recently been reported in clinical and experimental studies with different pharmacological approaches such as epidermal growth factor neuregulin-1 (NRG1) [134, 138, 166, 167, 168]. NRG1 is a key factor for cardiac development [136, 169]; NRG1 activates tyrosine kinase causing a host of cardiovascular effects: (1) regulation of structure and function in cardiomyocytes (i.e. apoptosis, cell proliferation), (2) promotion of angiogenesis and (3) downregulation of sympathetic nerve mRNA and protein expression levels to inhibit nerve remodeling [134, 135, 137, 170].

5.2. Non-pharmacological

Cardiovascular disease is often accompanied by increased activity of carotid body chemoreceptors, which induces an autonomic imbalance [161]; ablation of carotid bodies has been documented to markedly improve post-ischemic cardiovascular end-points in clinical and animal studies [159, 160, 171]. Catheter ablation techniques have been used effectively in patients with ventricular tachyarrhythmias [147]; in addition, bilateral cardiac stellate decentralization (removes excessive sympathetic input to cardiomyocytes) is used in subjects that do not respond to conventional treatments [158]. A drawback of the latter intervention is that it is permanent and generally accompanied by secondary effects [172]. Of note is that the ICNS preserves the ability to coordinate neural activity and electrical stability even after disconnection of inputs from higher central command (i.e. brain) [173].

Specific neuron subpopulations can be targeted for neuromodulation therapy [174, 175, 176]; spinal cord stimulation (SCS), vagus nerve stimulation (VNS) and bioelectronic therapy (i.e. charge-balanced direct current, axonal modulation, kilohertz (kHz) frequency alternating current, etc.) are used in ischemic heart disease patients to abate reflex activation of peripheral ganglia [118, 147, 148, 174, 177, 178]. Application of electric current by stimulation/suppression of action potential propagation along nerves modulates neuron and organ function [145, 179]. Blockade of action potential propagation is produced by either kHz frequency alternating current or direct current; these protocols are used repeatedly and safely in patients [145, 147, 180].

SCS stimulates sympathetic afferents to transduce signals, which initiate from the ischemic myocardium, to spinal cord dorsal horn neurons [121, 181]. In the majority of patients receiving this treatment beneficial effects (i.e. improved exercise capacity, quality of life, etc.) last for more than a year [182, 183]. Additionally, SCS augments resistance to stress in myocytes by modifying myocyte energetics [177, 184]; in our laboratory, we documented that concurrent SCS did not influence post-ischemic ventricular perfusion [150].

VNS, on the other hand, protects myocardium [185, 186, 187] through anti-adrenergic interactions (i.e. higher parasympathetic activity stimulates muscarinic receptor activation that limits excess adrenergic receptor activation [188]) within intrinsic cardiac ganglia [189, 190] combined with reduced release of norepinephrine from presynaptic mechanisms in ischemic myocardium [191]. VNS also influences myocyte energetics due to its regulatory effects on glycogen metabolism [78, 185]; all of these factors can change sensory transduction within the cardiac milieu in the event of disparities between oxygen and nutrient supply and demand.

Salavation and co-workers have examined potential differences between SCS and VNS with regard to their ability to alter cardiac sensory neurons in nodose ganglia to transduce the ischemic myocardium; they reported that these interventions differentially obtund nociceptive-related nodose afferent neuronal inputs to the medulla but do not affect mechanosensitive transduction capabilities [192]. These nerve stimulation techniques are presently being tested in a number of clinical trials in heart failure patients (i.e. NECTAR-HF, ANTHEM-HF, INOVATE-HF) with promising results [153, 193, 194].

Intact neural pathways may be unimportant for protection of ischemic myocardium; this is most apparent in the transplanted heart where autonomic ganglia are disconnected from central neurons [121, 195]. Endogenous compounds released into the bloodstream or locally near nerves, neurons and cardiomyocytes, etc. during ischemia could stimulate intracellular pathways that transduce cytoprotective mechanisms. For instance, cardiac conditioning, which significantly delays development of post-ischemic tissue injury [91, 196, 197, 198], might involve activation of the ICNS (cf. recent review [90]). A variety of conditioning stratagems (both pharmacologic and non-pharmacologic) that trigger cellular transduction pathways (guanylate cyclase, kinases, etc.) mediate cellular protection through end-effectors; significant cross-tolerance exists with regard to the mechanisms involved [106, 199, 200].

Advertisement

6. Conclusions

Neurocardiology involves dynamic exchange between neurohumoral control systems and the cardiac milieu; bi-directional interactions between sympathetic and parasympathetic efferent pathways regulate inter-organ communications at different levels of the neuraxis.This is evident in the cardiac conditioning paradigm (i.e. pre-, per-, post- and remote) where endogenous ligands and catecholamines trigger intracellular transduction pathways to mediate cytoprotective end-effectors that promote cell survival [201, 202]. Strategies that protect against non-lethal ischemic injury could depend on nervous system status the question of how cytoprotective signals are transmitted between organs remains crucial. New findings support the concept that disorders within the ANS contribute to pathogenesis of organ injury, co-morbidities [203, 204] and even survival. Improved comprehension of modifications within the cardiac-neuro axis at the molecular, cellular, organ and whole body levels are critical for development of therapeutic strategies.

Advertisement

Acknowledgments

The authors would like to thank Professor Chantale Simard, Faculty of Pharmacy at Laval University for helpful suggestions during the preparation of this review.

Advertisement

Conflict of interest

The authors have no conflicts of interest to declare.

References

  1. 1. Hopkins DA, Armour JA. Localization of sympathetic postganglionic and parasympathetic preganglionic neurons which innervate different regions of the dog heart. The Journal of Comparative Neurology. 1984;229(2):186-198. DOI: 10.1002/cne.902290205
  2. 2. Janes RD, Brandys JC, Hopkins DA, Johnstone DE, Murphy DA, Armour JA. Anatomy of human extrinsic cardiac nerves and ganglia. The American Journal of Cardiology. 1986;57(4):299-309. PubMed PMID: 3946219
  3. 3. Kawashima T. The autonomic nervous system of the human heart with special reference to its origin, course, and peripheral distribution. Anatomy and Embryology. 2005;209(6):425-438. DOI: 10.1007/s00429-005-0462-1
  4. 4. Hanna P, Rajendran PS, Ajijola OA, Vaseghi M, Andrew Armour J, Ardell JL, et al. Cardiac neuroanatomy—Imaging nerves to define functional control. Autonomic Neuroscience. 2017;207:48-58. DOI: 10.1016/j.autneu.2017.07.008
  5. 5. Pardini BJ, Lund DD, Schmid PG. Organization of the sympathetic postganglionic innervation of the rat heart. Journal of the Autonomic Nervous System. 1989;28(3):193-201
  6. 6. Wallis D, Watson AH, Mo N. Cardiac neurones of autonomic ganglia. Microscopy Research and Technique. 1996;35(1):69-79. DOI: 10.1002/(SICI)1097-0029(19960901)35:1<69::AID-JEMT6>3.0.CO;2-N
  7. 7. Lymperopoulos A, Rengo G, Koch WJ. Adrenergic nervous system in heart failure: Pathophysiology and therapy. Circulation Research. 2013;113(6):739-753. DOI: 10.1161/CIRCRESAHA.113.300308. PubMed PMID: 23989716
  8. 8. Kuder T, Nowak E. Autonomic cardiac nerves: Literature review. Folia Morphologica. 2015;74(1):1-8. DOI: VM/OJS/J/38007 [pii];10.5603/FM.2015.0003
  9. 9. Hopkins DA, MacDonald SE, Murphy DA, Armour JA. Pathology of intrinsic cardiac neurons from ischemic human hearts. The Anatomical Record. 2000;259:424-436
  10. 10. Hasan W. Autonomic cardiac innervation: Development and adult plasticity. Organogenesis. 2013;9(3):176-193. DOI: 24892 [pii];10.4161/org.24892
  11. 11. Kimura K, Ieda M, Fukuda K. Development, maturation, and transdifferentiation of cardiac sympathetic nerves. Circulation Research. 2012;110(2):325-336. DOI: 110/2/325 [pii];10.1161/CIRCRESAHA.111.257253
  12. 12. Ardell JL, Randall WC. Selective vagal innervation of sinoatrial and atrioventricular nodes in canine heart. American Journal of Physiology-Heart and Circulatory Physiology. 1986;251:H764-HH73
  13. 13. Randall WC, Milosavljevic M, Wurster RD, Geis GS, Ardell JL. Selective vagal innervation of the heart. Annals of Clinical and Laboratory Science. 1986;16(3):198-208
  14. 14. Singh S, Johnson PI, Lee RE, Orfei E, Lonchyna VA, Sullivan HJ, et al. Topography of cardiac ganglia in the adult human heart. The Journal of Thoracic and Cardiovascular Surgery. 1996;112(4):943-953. DOI: S0022-5223(96)70094-6
  15. 15. Horackova M, Armour JA, Byczko Z. Distribution of intrinsic cardiac neurons in whole-Mount Guinea pig atria identified by multiple neurochemical coding. A confocal microscope study. Cell and Tissue Research. 1999;297(3):409-421. DOI: 92970409.441
  16. 16. Slavikova J, Kuncova J, Reischig J, Dvorakova M. Catecholaminergic neurons in the rat intrinsic cardiac nervous system. Neurochemical Research. 2003;28(3-4):593-598
  17. 17. Weihe E, Schutz B, Hartschuh W, Anlauf M, Schafer MK, Eiden LE. Coexpression of cholinergic and noradrenergic phenotypes in human and nonhuman autonomic nervous system. The Journal of Comparative Neurology. 2005;492(3):370-379. DOI: 10.1002/cne.20745
  18. 18. Ai J, Epstein PN, Gozal D, Yang B, Wurster R, Cheng ZJ. Morphology and topography of nucleus ambiguus projections to cardiac ganglia in rats and mice. Neuroscience. 2007;149(4):845-860. DOI: S0306-4522(07)00953-0
  19. 19. Armour JA. Myocardial ischaemia and the cardiac nervous system. Cardiovascular Research. 1999;41:41-54
  20. 20. McAllen RM, Salo LM, Paton JF, Pickering AE. Processing of central and reflex vagal drives by rat cardiac ganglion neurones: An intracellular analysis. The Journal of Physiology. 2011;589(Pt 23):5801-5818. DOI: jphysiol.2011.214320
  21. 21. Ardell JL, Andresen MC, Armour JA, Billman GE, Chen PS, Foreman RD, et al. Translational neurocardiology: Preclinical models and cardioneural integrative aspects. The Journal of Physiology. 2016;594(14):3877-3909. DOI: 10.1113/JP271869
  22. 22. Bentivoglio M, Kuypers HG, Catsman-Berrevoets CE, Loewe H, Dann O. Two new fluorescent retrograde neuronal tracers which are transported over long distances. Neuroscience Letters. 1980;18(1):25-30. PubMed PMID: 6189013
  23. 23. Grkovic I, Fernandez K, McAllen RM, Anderson CR. Misidentification of cardiac vagal pre-ganglionic neurons after injections of retrograde tracer into the pericardial space in the rat. Cell and Tissue Research. 2005;321(3):335-340. DOI: 10.1007/s00441-005-1145-1
  24. 24. Kuypers HG, Bentivoglio M, van der Kooy D, Catsman-Berrevoets CE. Retrograde transport of bisbenzimide and propidium iodide through axons to their parent cell bodies. Neuroscience Letters. 1979;12(1):1-7. PubMed PMID: 88694
  25. 25. Tomney PA, Hopkins DA, Armour JA. Axonal branching of canine sympathetic postganglionic cardiopulmonary neurons. A retrograde fluorescent labeling study. Brain Research Bulletin. 1985;14(5):443-452. PubMed PMID: 2411359
  26. 26. Batulevicius D, Pauziene N, Pauza DH. Architecture and age-related analysis of the neuronal number of the Guinea pig intrinsic cardiac nerve plexus. Annals of Anatomy. 2005;187(3):225-243. DOI: 10.1016/j.aanat.2005.01.004
  27. 27. Karnovsky MJ, Roots L. A “direct-coloring” thiocholine method for cholinesterases. The Journal of Histochemistry and Cytochemistry. 1964;12:219-221. DOI: 10.1177/12.3.219
  28. 28. Kawano H, Okada R, Yano K. Histological study on the distribution of autonomic nerves in the human heart. Heart and Vessels. 2003;18(1):32-39. DOI: 10.1007/s003800300005
  29. 29. Pauza DH, Rysevaite-Kyguoliene K, Vismantaite J, Brack KE, Inokaitis H, Pauza AG, et al. A combined acetylcholinesterase and immunohistochemical method for precise anatomical analysis of intrinsic cardiac neural structures. Annals of Anatomy. 2014;196(6):430-440. DOI: 10.1016/j.aanat.2014.08.004
  30. 30. Wang X, Pinol RA, Byrne P, Mendelowitz D. Optogenetic stimulation of locus ceruleus neurons augments inhibitory transmission to parasympathetic cardiac vagal neurons via activation of brainstem alpha1 and beta1 receptors. The Journal of Neuroscience. 2014;34(18):6182-6189. DOI: 10.1523/JNEUROSCI.5093-13.2014
  31. 31. Beaumont E, Salavatian S, Southerland EM, Vinet A, Jacquemet V, Armour JA, et al. Network interactions within the canine intrinsic cardiac nervous system: Implications for reflex control of regional cardiac function. The Journal of Physiology. 2013;591(18):4515-4533. DOI: 10.1113/jphysiol.2013.259382
  32. 32. Bravo PE, Lautamaki R, Carter D, Holt DP, Nekolla SG, Dannals RF, et al. Mechanistic insights into sympathetic neuronal regeneration: Multitracer molecular imaging of catecholamine handling after cardiac transplantation. Circulation. Cardiovascular Imaging. 2015;8(8):e003507. DOI: 10.1161/CIRCIMAGING.115.003507
  33. 33. Calkins H, Lehmann MH, Allman K, Wieland D, Schwaiger M. Scintigraphic pattern of regional cardiac sympathetic innervation in patients with familial long QT syndrome using positron emission tomography. Circulation. 1993;87(5):1616-1621. PubMed PMID: 8491017
  34. 34. Feigl EO, Van Winkle DM, Miyashiro JK. Cholinergic vasodilatation of coronary resistance vessels in dogs, baboons and goats. Blood Vessels. 1990;27:94-105
  35. 35. Feigl EO. Coronary physiology. Physiological Reviews. 1983;63:1-205
  36. 36. Downey JM. Myocardial contractile force as a function of coronary blood flow. The American Journal of Physiology. 1976;230:1-6
  37. 37. Boatwright RB, Downey HF, Bashour FA, Crystal GJ. Transmural variation in autoregulation of coronary blood flow in hyperperfused canine myocardium. Circulation Research. 1980;47(4):599-609
  38. 38. Rouleau JR, Boerboom LE, Surjadhana A, Hoffman JIE. The role of autoregulation and tissue diastolic pressures in the transmural distribution of left ventricular blood flow in anesthetized dogs. Circulation Research. 1979;45(6):804-815
  39. 39. Mohrman DE, Feigl EO. Competition between sympathetic vasoconstriction and metabolic vasodilation in the canine coronary circulation. Circulation Research. 1978;42:79-86
  40. 40. Kingma JG Jr, Armour JA, Rouleau JR. Chemical modulation of in situ intrinsic cardiac neurones influences myocardial blood flow in the anaesthetised dog. Cardiovascular Research. 1994;28(9):1403-1406. PubMed PMID: 7954653
  41. 41. Armour JA, Huang MH, Smith FM. Peptidergic modulation of in situ canine intrinsic cardiac neurons. Peptides. 1993;14(2):191-202
  42. 42. Butler CK, Smith FM, Cardinal R, Murphy DA, Hopkins DA. Cardiac responses to electrical stimulation of discrete loci in atrial and ventricular ganglionated plexi. American Journal of Physiology-Heart and Circulatory Physiology. 1990;259:H1365-H1H73
  43. 43. Vergroesen I, Merkus D, Van Teeffelen JWGE, Dankelman J, Spaan JAE, Van Wezel HB, et al. Chronic cardiac denervation affects the speed of coronary vascular regulation. Cardiovascular Research. 1999;44:615-622
  44. 44. Dankelman J, Vergroesen I, Han Y, Spaan JAE. Dynamic response of coronary regulation to heart rate and perfusion changes in dogs. American Journal of Physiology-Heart and Circulatory Physiology. 1992;263:H447-HH52
  45. 45. Vergroesen I, Noble MI, Spaan JA. Intramyocardial blood volume change in first moments of cardiac arrest in anesthetized goats. The American Journal of Physiology. 1987;253(2 Pt 2):H307-H316. DOI: 10.1152/ajpheart.1987.253.2.H307
  46. 46. McMahon NC, Drinkhill MJ, Hainsworth R. Reflex vascular responses from aortic arch, carotid sinus and coronary baroreceptors in the anaesthetized dog. Experimental Physiology. 1996;81(3):397-408. PubMed PMID: 8737074
  47. 47. Zanzinger J. Role of nitric oxide in the neural control of cardiovascular function. Cardiovascular Research. 1999;43:639-649
  48. 48. Paton JF, Kasparov S, Paterson DJ. Nitric oxide and autonomic control of heart rate: A question of specificity. Trends in Neurosciences. 2002;25(12):626-631. PubMed PMID: 12446130
  49. 49. Toda N, Kitamura Y, Okamura T. Neural mechanism of hypertension by nitric oxide synthase inhibitor in dogs. Hypertension. 1993;21:3-8
  50. 50. Thorin E, Huang PL, Fishman MC, Bevan JA. Nitric oxide inhibits a2-adrenoceptor-mediated endothelium-dependent vasodilation. Circulation Research. 1998;82:1323-1329
  51. 51. Thorin E, Atkinson J. Modulation by the endothelium of sympathetic vasoconstriction in an in vitro preparation of the rat tail artery. British Journal of Pharmacology. 1994;111(1):351-357
  52. 52. Amezcua JL, Palmer RMJ, Souza BM, Moncada S. Nitric oxide synthesized from l-arginine regulates vascular tone in the coronary circulation of the rabbit. British Journal of Pharmacology. 1989;97:1119-1124
  53. 53. Schulz R, Smith JA, Lewis MJ, Moncada S. Nitric oxide synthase in cultured endocardial cells of the pig. British Journal of Pharmacology. 1991;104:21-24
  54. 54. Sosunov AA, Hassall CJS, Loesch A, Turamine M, Burnstock G. Ultrastructural investigation of nitric oxide synthase immunoreactive nerves associated with coronary blood vessels of rat and Guinea-pig. Cell and Tissue Research. 1995;280:575-582
  55. 55. Christopherson KS, Bredt DS. Nitric oxide in excitable tissues: Physiological roles and disease. The Journal of Clinical Investigation. 1997;100:2424-2429
  56. 56. Brown AM. Regulation of heartbeat by G protein-coupled ion channels. The American Journal of Physiology. 1990;259:H1621-H16H8
  57. 57. Armour JA, Smith FM, Losier AM, Ellenberger HH, Hopkins DA. Modulation of intrinsic cardiac neuronal activity by nitric oxide donors induces cardiodynamic changes. The American Journal of Physiology. 1995;268(2 Pt 2):R403-RR13
  58. 58. Rouleau JR, Simard D, Rodrigue N, Blouin A, Kingma JG Jr. Myocardial blood flow after chronic cardiac decentralization in anesthetized dogs: Effects of ACE-inhibition. Autonomic Neuroscience. 2002;97(1):12-18. DOI: S1566-0702(02)00002-4
  59. 59. Rimoldi OE, Drake-Holland AJ, Noble MI, Camici PG. Basal and hyperaemic myocardial blood flow in regionally denervated canine hearts: An in vivo study with positron emission tomography. European Journal of Nuclear Medicine and Molecular Imaging. 2007;34(2):197-205. DOI: 10.1007/s00259-006-0233-0
  60. 60. Stevens MJ, Dayanikli F, Raffel DM, Allman KC, Sandford T, Feldman EL, et al. Scintigraphic assessment of regionalized defects in myocardial sympathetic innervation and blood flow regulation in diabetic patients with autonomic neuropathy. Journal of the American College of Cardiology. 1998;31(7):1575-1584. DOI: S0735-1097(98)00128-4
  61. 61. Kaufmann PA, Rimoldi O, Gnecchi-Ruscone T, Bonser RS, Luscher TF, Camici PG. Systemic inhibition of nitric oxide synthase unmasks neural constraint of maximal myocardial blood flow in humans. Circulation. 2004;110(11):1431-1436. DOI: 10.1161/01.CIR.0000141294.25130.54
  62. 62. Huang H-S, Pan H-L, Stahl GL, Longhurst JC. Ischemia- and reperfusion-sensitive cardiac sympathetic afferents: Influence of H2O2 and hydroxyl radicals. American Journal of Physiology-Heart and Circulatory Physiology. 1995;269:H888-H901
  63. 63. Huang MH, Sylven C, Horackova M, Armour JA. Ventricular sensory neurons in canine dorsal root ganglia: Effects of adenosine and substance P. The American Journal of Physiology. 1995;269(2 Pt 2):R318-RR24
  64. 64. Thompson GW, Horackova M, Armour JA. Sensitivity of canine intrinsic cardiac neurons to H2O2 and hydroxyl radical. The American Journal of Physiology 1998;275(4 Pt 2):H1434-H1H40
  65. 65. Barber MJ, Mueller TM, Henry DP, Felten SY, Zipes DP. Transmural myocardial infarction in the dog produces sympathectomy in noninfarcted myocardium. Circulation. 1983;67:787-796
  66. 66. Janes RD, Johnstone DE, Armour JA. Functional integrity of intrinsic cardiac nerves located over an acute transmural myocardial infarction. Canadian Journal of Physiology and Pharmacology. 1987;65:64-69
  67. 67. Arora RC, Armour JA. Adenosine A1 receptor activation reduces myocardial reperfusion effects on intrinsic cardiac nervous system. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2003;284(5):R1314-R1R21
  68. 68. Holmes JW, Laksman Z, Gepstein L. Making better scar: Emerging approaches for modifying mechanical and electrical properties following infarction and ablation. Progress in Biophysics and Molecular Biology. 2016;120(1-3):134-148. DOI: 10.1016/j.pbiomolbio.2015.11.002
  69. 69. Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: From inflammation to fibrosis. Circulation Research. 2016;119(1):91-112. DOI: 10.1161/CIRCRESAHA.116.303577
  70. 70. Wang Y, Suo F, Liu J, Hu H, Xue M, Cheng W, et al. Myocardial infarction induces sympathetic hyperinnervation via a nuclear factor-kappaB-dependent pathway in rabbit hearts. Neuroscience Letters. 2013;535:128-133. DOI: 10.1016/j.neulet.2012.12.059
  71. 71. Cao JM, Fishbein MC, Han JB, Lai WW, Lai AC, Wu TJ, et al. Relationship between regional cardiac hyperinnervation and ventricular arrhythmia. Circulation. 2000;101(16):1960-1969. PubMed PMID: 10779463
  72. 72. Yu FS, Zhang Y, Feng Y, Zhang L, Ma YH, Song W, et al. Nerve remodeling in a canine model of atrial fibrillation induced by 48 hours right atrial pacing. Zhonghua Xin Xue Guan Bing Za Zhi. 2010;38(7):644-647. PubMed PMID: 21055291
  73. 73. Habecker BA, Gritman KR, Willison BD, Van Winkle DM. Myocardial infarction stimulates galanin expression in cardiac sympathetic neurons. Neuropeptides. 2005;39(2):89-95. DOI: S0143-4179(04)00151-9 [pii];10.1016/j.npep.2004.11.003
  74. 74. Abe T, Morgan DA, Gutterman DD. Role of adenosine receptor subtypes in neural stunning of sympathetic coronary innervation. American Journal of Physiology-Heart and Circulatory Physiology. 1997;272(41):H25-H34
  75. 75. Allen TGJ, Burnstock G. The actions of adenosine 5′-triphosphate on Guinea-pig intracardiac neurones in culture. British Journal of Pharmacology. 1990;100:269-276
  76. 76. Croom JE, Foreman RD, Chandler MJ, Barron KW. Cutaneous vasodilation during dorsal column stimulation is mediated by dorsal roots and CGRP. The American Journal of Physiology. 1997;272:H950-H9H7
  77. 77. Dobaczewski M, de Haan JJ, Frangogiannis NG. The extracellular matrix modulates fibroblast phenotype and function in the infarcted myocardium. Journal of Cardiovascular Translational Research. 2012;5(6):837-847. DOI: 10.1007/s12265-012-9406-3
  78. 78. Beaumont E, Southerland EM, Hardwick JC, Wright GL, Ryan S, Li Y, et al. Vagus nerve stimulation mitigates intrinsic cardiac neuronal and adverse myocyte remodeling postmyocardial infarction. American Journal of Physiology. Heart and Circulatory Physiology. 2015;309(7):H1198-H1206. DOI: 10.1152/ajpheart.00393.2015
  79. 79. Mollema SA, Liem SS, Suffoletto MS, Bleeker GB, van der Hoeven BL, van de Veire NR, et al. Left ventricular dyssynchrony acutely after myocardial infarction predicts left ventricular remodeling. Journal of the American College of Cardiology. 2007;50(16):1532-1540. DOI: 10.1016/j.jacc.2007.07.025
  80. 80. Crow MT, Mani K, Nam YJ, Kitsis RN. The mitochondrial death pathway and cardiac myocyte apoptosis. Circulation Research. 2004;95(10):957-970. DOI: 10.1161/01.RES.0000148632.35500.d9
  81. 81. Armour JA. Potential clinical relevance of the ‘little brain’ on the mammalian heart. Experimental Physiology. 2008;93(2):165-176
  82. 82. Billman GE. A comprehensive review and analysis of 25 years of data from an in vivo canine model of sudden cardiac death: Implications for future anti-arrhythmic drug development. Pharmacology & Therapeutics. 2006;111(3):808-835. DOI: 10.1016/j.pharmthera.2006.01.002
  83. 83. Shivkumar K, Ardell JL. Cardiac autonomic control in health and disease. The Journal of Physiology. 2016;594(14):3851-3852. DOI: 10.1113/JP272580
  84. 84. Shivkumar K, Ajijola OA, Anand I, Armour JA, Chen PS, Esler M, et al. Clinical neurocardiology defining the value of neuroscience-based cardiovascular therapeutics. The Journal of Physiology. 2016;594(14):3911-3954. DOI: 10.1113/JP271870. PubMed PMID: 27114333
  85. 85. Reimer KA, Lowe JE, Rasmussen MM, Jennings RB. The wavefront phenomenon of ischemic cell death: I. Myocardial infarct size vs. duration of coronary occlusion in dogs. Circ. 1977;56:786-794
  86. 86. Jennings RB. Historical perspective on the pathology of myocardial ischemia/reperfusion injury. Circulation Research. 2013;113(4):428-438. DOI: 10.1161/CIRCRESAHA.113.300987
  87. 87. Kloner RA, Jennings RB. Consequences of brief ischemia: Stunning, preconditioning, and their clinical implications: Part 1. Circulation. 2001;104(24):2981-2989
  88. 88. Kloner RA, Jennings RB. Consequences of brief ischemia: Stunning, preconditioning, and their clinical implications: Part 2. Circulation. 2001;104(25):3158-3167
  89. 89. Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. The New England Journal of Medicine. 2007;357(11):1121-1135. DOI: 357/11/1121
  90. 90. Kingma JG, Simard D, Rouleau JR. Influence of cardiac nerve status on cardiovascular regulation and cardioprotection. World Journal of Cardiology. 2017;9(6):508-520. Epub June 26, 2017. DOI: 10.4330/wjc.v9.i6.508
  91. 91. Hausenloy DJ, Barrabes JA, Botker HE, Davidson SM, Di Lisa F, Downey J, et al. Ischaemic conditioning and targeting reperfusion injury: A 30 year voyage of discovery. Basic Research in Cardiology. 2016;111(6):70. DOI: 10.1007/s00395-016-0588-8
  92. 92. Bulluck H, Yellon DM, Hausenloy DJ. Reducing myocardial infarct size: Challenges and future opportunities. Heart. 2016;102(5):341-348. DOI: 10.1136/heartjnl-2015-307855. PubMed PMID: 26674987
  93. 93. James TN, Zipes DP, Finegan RE, Eisele JW, Carter JE. Cardiac ganglionitis associated with sudden unexpected death. Annals of Internal Medicine. 1979;91(5):727-730. PubMed PMID: 496105
  94. 94. Ajijola OA, Hoover DB, Simerly TM, Brown TC, Yanagawa J, Biniwale RM, et al. Inflammation, oxidative stress, and glial cell activation characterize stellate ganglia from humans with electrical storm. JCI Insight. 2017;2(18). DOI: 10.1172/jci.insight.94715
  95. 95. Wang HJ, Wang W, Cornish KG, Rozanski GJ, Zucker IH. Cardiac sympathetic afferent denervation attenuates cardiac remodeling and improves cardiovascular dysfunction in rats with heart failure. Hypertension. 2014;64(4):745-755. DOI: 10.1161/HYPERTENSIONAHA.114.03699
  96. 96. Lambert C, Massillon Y, Meloche S. Upregulation of cardiac angiotensin II AT1 receptors in congenital cardiomyopathic hamsters. Circulation Research. 1995;77(5):1001-1007
  97. 97. Zucker IH, Gao L. The regulation of sympathetic nerve activity by angiotensin II involves reactive oxygen species and MAPK. Circulation Research. 2005;97(8):737-739. DOI: 10.1161/01.RES.0000188261.94569.1f
  98. 98. Lambert GW, Kaye DM, Lefkovits J, Jennings GL, Turner AG, Cox HS, et al. Increased central nervous system monoamine neurotransmitter turnover and its association with sympathetic nervous activity in treated heart failure patients. Circulation. 1995;92(7):1813-1818. PubMed PMID: 7545554
  99. 99. Moss AJ, Ryan DH, Yeaney GA. Ganglionitis and genetic cardiac arrhythmias: More questions than answers. Circulation. Arrhythmia and Electrophysiology. 2014;7(2):190-191. DOI: 10.1161/CIRCEP.114.001589
  100. 100. Huang CH, Vatner SF, Peppas AP, Yang G, Kudej RK. Cardiac nerves affect myocardial stunning through reactive oxygen and nitric oxide mechanisms. Circulation Research. 2003;93(9):866-873. DOI: 10.1161/01.RES.0000097762.64561.D2 [doi];01.RES.0000097762.64561.D2
  101. 101. Kingma JG, Simard D, Voisine P, Rouleau JR. Influence of cardiac decentralization on cardioprotection. PLoS One. 2013;8(11):e79190. DOI: 10.1371/journal.pone.0079190 [doi];PONE-D-13-17924 [pii]
  102. 102. Jones CE, Devous MD Sr, Thomas JX Jr, Dupont E. The effect of chronic cardiac denervation on infarct size following acute coronary occlusion. American Heart Journal. 1978;95(6):738-746
  103. 103. Jones CE, Beck LY, Dupont E, Barnes GE. Effects of coronary ligation of the chronically sympathectomized dog ventricle. The American Journal of Physiology. 1978;235(4):H429-HH34
  104. 104. Dupont E, Jones CE, Luedecke RA, Smith EE. Chronic ventricular sympathectomy: Effect on myocardial perfusion after ligation of the circumflex coronary artery in dogs. Circulatory Shock. 1979;6(4):323-331
  105. 105. Lavallee M, Amano J, Vatner SF, Manders WT, Randall WC, Thomas JXJ. Adverse effects of chronic cardiac denervation in conscious dogs with myocardial ischemia. Circulation Research. 1985;57(3):383-392
  106. 106. Kingma JG Jr, Simard D, Voisine P, Rouleau JR. Role of the autonomic nervous system in cardioprotection by remote preconditioning in isoflurane-anaesthetized dogs. Cardiovascular Research. 2011;89(2):384-391. DOI: 10.1093/cvr/cvq306
  107. 107. Shen YT, Knight DR, Vatner SF, Randall WC, Thomas JX Jr. Responses to coronary artery occlusion in conscious dogs with selective cardiac denervation. The American Journal of Physiology. 1988;255(3 Pt 2):H525-H533. DOI: 10.1152/ajpheart.1988.255.3.H525
  108. 108. Kliks BR, Burgess MJ, Abildskov JA. Influence of sympathetic tone on ventricular fibrillation threshold during experimental coronary occlusion. The American Journal of Cardiology. 1975;36(1):45-49. PubMed PMID: 1146697
  109. 109. Acad B-A, Joselevitz-Goldman J, Scholz PM, Weiss HR. Improved distribution of regional oxygenation in denervated ischemic dog myocardium. Circulation Research. 1988;62(5):1041-1048
  110. 110. Brunsting JR, Schuil HA, Zijlstra WG. Intrinsic heart rate in the dog determined by pharmacologic denervation. The American Journal of Physiology. 1983;245(4):H592-H5H7
  111. 111. Barber MJ, Mueller TM, Davies BG, Zipes DP. Phenol topically applied to canine left ventricular epicardium interrupts sympathetic but not vagal afferents. Circulation Research. 1984;55(4):532-544
  112. 112. Sonoyama K, Tajima K, Fujiwara R, Kasai T. Intravenous infusion of hexamethonium and atropine but not propranolol diminishes apolipoprotein A-IV gene expression in rat ileum. The Journal of Nutrition. 2000;130(3):637-641
  113. 113. Chugh SS, Reinier K, Teodorescu C, Evanado A, Kehr E, Al Samara M, et al. Epidemiology of sudden cardiac death: Clinical and research implications. Progress in Cardiovascular Diseases. 2008;51(3):213-228. DOI: 10.1016/j.pcad.2008.06.003
  114. 114. Armour JA, Hageman GR, Randall WC. Arrhythmias induced by local cardiac nerve stimulation. The American Journal of Physiology. 1972;223(5):1068-1075. DOI: 10.1152/ajplegacy.1972.223.5.1068
  115. 115. Gelband H, Rosen MR, Myerburg RJ, Bush HL, Bassett AL, Hoffman BF. Restorative effect of epinephrine on the electrophysiologic properties of depressed human atrial tissue. Journal of Electrocardiology. 1977;10(4):313-320. PubMed PMID: 915399
  116. 116. Zipes DP. Antiarrhythmic therapy in 2014: Contemporary approaches to treating arrhythmias. Nature Reviews. Cardiology. 2015;12(2):68-69. DOI: 10.1038/nrcardio.2014.211
  117. 117. Kember G, Armour JA, Zamir M. Neural control hierarchy of the heart has not evolved to deal with myocardial ischemia. Physiological Genomics. 2013;45(15):638-644. DOI: physiolgenomics.00027.2013 [pii];10.1152/physiolgenomics.00027.2013
  118. 118. Foreman RD, Linderoth B, Ardell JL, Barron KW, Chandler MJ, Hull SSJ, et al. Modulation of intrinsic cardiac neurons by spinal cord stimulation: Implications for its therapeutic use in angina pectoris. Cardiovascular Research. 2000;47:367-375
  119. 119. Ajijola OA, Yagishita D, Reddy NK, Yamakawa K, Vaseghi M, Downs AM, et al. Remodeling of stellate ganglion neurons after spatially targeted myocardial infarction: Neuropeptide and morphologic changes. Heart Rhythm. 2015;12(5):1027-1035. DOI: 10.1016/j.hrthm.2015.01.045
  120. 120. Rajendran PS, Nakamura K, Ajijola OA, Vaseghi M, Armour JA, Ardell JL, et al. Myocardial infarction induces structural and functional remodelling of the intrinsic cardiac nervous system. The Journal of Physiology. 2016;594(2):321-341. DOI: 10.1113/JP271165
  121. 121. Ardell JL, Cardinal R, Vermeulen M, Armour JA. Dorsal spinal cord stimulation obtunds the capacity of intrathoracic extracardiac neurons to transduce myocardial ischemia. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2009;297(2):R470-R477. DOI: 10.1152/ajpregu.90821.2008
  122. 122. Fukuda K, Kanazawa H, Aizawa Y, Ardell JL, Shivkumar K. Cardiac innervation and sudden cardiac death. Circulation Research. 2015;116(12):2005-2019. DOI: CIRCRESAHA.116.304679 [pii];10.1161/CIRCRESAHA.116.304679
  123. 123. Hardwick JC, Ryan SE, Beaumont E, Ardell JL, Southerland EM. Dynamic remodeling of the Guinea pig intrinsic cardiac plexus induced by chronic myocardial infarction. Autonomic Neuroscience. 2014;181:4-12. DOI: 10.1016/j.autneu.2013.10.008
  124. 124. Rubart M, Zipes DP. Mechanisms of sudden cardiac death. The Journal of Clinical Investigation. 2005;115(9):2305-2315. DOI: 10.1172/JCI26381
  125. 125. Vaseghi M, Shivkumar K. The role of the autonomic nervous system in sudden cardiac death. Progress in Cardiovascular Diseases. 2008;50(6):404-419. DOI: S0033-0620(08)00004-2 [pii];10.1016/j.pcad.2008.01.003
  126. 126. Vracko R, Thorning D, Frederickson RG. Nerve fibers in human myocardial scars. Human Pathology. 1991;22(2):138-146. PubMed PMID: 1705914
  127. 127. Ajijola OA, Lux RL, Khahera A, Kwon O, Aliotta E, Ennis DB, et al. Sympathetic modulation of electrical activation in normal and infarcted myocardium: Implications for arrhythmogenesis. American Journal of Physiology. Heart and Circulatory Physiology. 2017;312(3):H608-H621. DOI: 10.1152/ajpheart.00575.2016
  128. 128. Hamon D, Rajendran PS, Chui RW, Ajijola OA, Irie T, Talebi R, et al. Premature ventricular contraction coupling interval variability destabilizes cardiac neuronal and electrophysiological control: Insights from simultaneous cardioneural mapping. Circulation. Arrhythmia and Electrophysiology. 2017;10(4):e004937. DOI: 10.1161/CIRCEP.116.004937
  129. 129. Hamon D, Blaye-Felice MS, Bradfield JS, Chaachoui N, Tung R, Elayi CS, et al. A new combined parameter to predict premature ventricular complexes induced cardiomyopathy: Impact and recognition of epicardial origin. Journal of Cardiovascular Electrophysiology. 2016;27(6):709-717. DOI: 10.1111/jce.12967
  130. 130. Wang Y, Eltit JM, Kaszala K, Tan A, Jiang M, Zhang M, et al. Cellular mechanism of premature ventricular contraction-induced cardiomyopathy. Heart Rhythm. 2014;11(11):2064-2072. DOI: 10.1016/j.hrthm.2014.07.022
  131. 131. De Palo G, Facchetti G, Mazzolini M, Menini A, Torre V, Altafini C. Common dynamical features of sensory adaptation in photoreceptors and olfactory sensory neurons. Scientific Reports. 2013;3:1251. DOI: 10.1038/srep01251
  132. 132. Grill-Spector K, Henson R, Martin A. Repetition and the brain: Neural models of stimulus-specific effects. Trends in Cognitive Sciences. 2006;10(1):14-23. DOI: 10.1016/j.tics.2005.11.006
  133. 133. Segerson NM, Sharma N, Smith ML, Wasmund SL, Kowal RC, Abedin M, et al. The effects of rate and irregularity on sympathetic nerve activity in human subjects. Heart Rhythm. 2007;4(1):20-26. DOI: 10.1016/j.hrthm.2006.09.017
  134. 134. Lai X, Zhong L, Fu HX, Dang S, Wang X, Zhang N, et al. Effects of neuregulin-1 on autonomic nervous system remodeling post-myocardial infarction in a rat model. Neural Regeneration Research. 2017;12(11):1905-1910. DOI: 10.4103/1673-5374.219054
  135. 135. Odiete O, Hill MF, Sawyer DB. Neuregulin in cardiovascular development and disease. Circulation Research. 2012;111(10):1376-1385. DOI: 10.1161/CIRCRESAHA.112.267286
  136. 136. Mei L, Xiong WC. Neuregulin 1 in neural development, synaptic plasticity and schizophrenia. Nature Reviews. Neuroscience. 2008;9(6):437-452. DOI: 10.1038/nrn2392
  137. 137. Mendes-Ferreira P, De Keulenaer GW, Leite-Moreira AF, Bras-Silva C. Therapeutic potential of neuregulin-1 in cardiovascular disease. Drug Discovery Today. 2013;18(17-18):836-842. DOI: 10.1016/j.drudis.2013.01.010
  138. 138. Jabbour A, Hayward CS, Keogh AM, Kotlyar E, McCrohon JA, England JF, et al. Parenteral administration of recombinant human neuregulin-1 to patients with stable chronic heart failure produces favourable acute and chronic haemodynamic responses. European Journal of Heart Failure. 2011;13(1):83-92. DOI: 10.1093/eurjhf/hfq152
  139. 139. Camargo-Silva G, Turones LC, da Cruz KR, Gomes KP, Mendonca MM, Nunes A, et al. Ghrelin potentiates cardiac reactivity to stress by modulating sympathetic control and beta-adrenergic response. Life Sciences. 2018;196:84-92. DOI: 10.1016/j.lfs.2018.01.019
  140. 140. Lozic M, Sarenac O, Murphy D, Japundzic-Zigon N. Vasopressin, central autonomic control and blood pressure regulation. Current Hypertension Reports. 2018;20(2):11. DOI: 10.1007/s11906-018-0811-0
  141. 141. Pasqualin RC, Mostarda CT, Souza LE, Vane MF, Sirvente R, Otsuki DA, et al. Sevoflurane preconditioning during myocardial ischemia-reperfusion reduces infarct size and preserves autonomic control of circulation in rats. Acta Cirúrgica Brasileira. 2016;31(5):338-345. DOI: 10.1590/S0102-865020160050000008
  142. 142. Jessurun GA, Tio RA, De Jongste MJ, Hautvast RW, Den Heijer P, Crijns HJ. Coronary blood flow dynamics during transcutaneous electrical nerve stimulation for stable angina pectoris associated with severe narrowing of one major coronary artery. The American Journal of Cardiology. 1998;82(8):921-926. PubMed PMID: 9794345
  143. 143. Wang Z, Yu L, Wang S, Huang B, Liao K, Saren G, et al. Chronic intermittent low-level transcutaneous electrical stimulation of auricular branch of vagus nerve improves left ventricular remodeling in conscious dogs with healed myocardial infarction. Circulation. Heart Failure. 2014;7(6):1014-1021. DOI: 10.1161/CIRCHEARTFAILURE.114.001564
  144. 144. Stavrakis S, Humphrey MB, Scherlag BJ, Hu Y, Jackman WM, Nakagawa H, et al. Low-level transcutaneous electrical vagus nerve stimulation suppresses atrial fibrillation. Journal of the American College of Cardiology. 2015;65(9):867-875. DOI: 10.1016/j.jacc.2014.12.026
  145. 145. Vrabec T, Bhadra N, Wainright J, Bhadra N, Franke M, Kilgore K. Characterization of high capacitance electrodes for the application of direct current electrical nerve block. Medical & Biological Engineering & Computing. 2016;54(1):191-203. DOI: 10.1007/s11517-015-1385-5
  146. 146. Vrabec T, Bhadra N, Van Acker G, Bhadra N, Kilgore K. Continuous direct current nerve block using multi contact high capacitance electrodes. IEEE Transactions on Neural Systems and Rehabilitation Engineering. 2017;25(6):517-529. DOI: 10.1109/TNSRE.2016.2589541
  147. 147. Chui RW, Buckley U, Rajendran PS, Vrabec T, Shivkumar K, Ardell JL. Bioelectronic block of paravertebral sympathetic nerves mitigates post-myocardial infarction ventricular arrhythmias. Heart Rhythm. 2017;14(11):1665-1672. DOI: 10.1016/j.hrthm.2017.06.025
  148. 148. Buckley U, Chui RW, Rajendran PS, Vrabec T, Shivkumar K, Ardell JL. Bioelectronic neuromodulation of the paravertebral cardiac efferent sympathetic outflow and its effect on ventricular electrical indices. Heart Rhythm. 2017;14(7):1063-1070. DOI: 10.1016/j.hrthm.2017.02.020
  149. 149. de Vries J, De Jongste MJ, Spincemaille G, Staal MJ. Spinal cord stimulation for ischemic heart disease and peripheral vascular disease. Advances and Technical Standards in Neurosurgery. 2007;32:63-89. PubMed PMID: 17907475
  150. 150. Kingma JG Jr, Linderoth B, Ardell JL, Armour JA, DeJongste MJ, Foreman RD. Neuromodulation therapy does not influence blood flow distribution or left-ventricular dynamics during acute myocardial ischemia. Autonomic Neuroscience. 2001;91(1-2):47-54. DOI: 10.1016/S1566-0702(01)00285-5
  151. 151. de Vries J, Svilaas T, De Jongste MJ, Nieuwland W, Hoekstra-Mars EW, Zijlstra F. Impact of electrical neurostimulation on persistent ST elevation after successful reperfusion by primary percutaneous coronary intervention. Journal of Electrocardiology. 2007;40(6):522-526. DOI: 10.1016/j.jelectrocard.2007.05.014
  152. 152. de Vries J, De Jongste MJ, Durenkamp A, Zijlstra F, Staal MJ. The sustained benefits of long-term neurostimulation in patients with refractory chest pain and normal coronary arteries. European Journal of Pain. 2007;11(3):360-365. DOI: 10.1016/j.ejpain.2006.04.002
  153. 153. Zannad F, De Ferrari GM, Tuinenburg AE, Wright D, Brugada J, Butter C, et al. Chronic vagal stimulation for the treatment of low ejection fraction heart failure: Results of the NEural cardiac TherApy foR heart failure (NECTAR-HF) randomized controlled trial. European Heart Journal. 2015;36(7):425-433. DOI: 10.1093/eurheartj/ehu345
  154. 154. Janig W. Autonomic nervous system and inflammation. Autonomic Neuroscience. 2014;182:1-3. DOI: S1566-0702(14)00032-0 [pii];10.1016/j.autneu.2014.02.002
  155. 155. Bradfield JS, Vaseghi M, Shivkumar K. Renal denervation for refractory ventricular arrhythmias. Trends in Cardiovascular Medicine. 2014;24(5):206-213. DOI: 10.1016/j.tcm.2014.05.006
  156. 156. Chen C, Upadhyay A. Renal denervation for uncontrolled hypertension: Critical review of the evidence. Current Opinion in Nephrology and Hypertension. 2017;26(2):114-122. DOI: 10.1097/MNH.0000000000000300
  157. 157. Nammas W, Airaksinen JK, Paana T, Karjalainen PP. Renal sympathetic denervation for treatment of patients with atrial fibrillation: Reappraisal of the available evidence. Heart Rhythm. 2016;13(12):2388-2394. DOI: 10.1016/j.hrthm.2016.08.043
  158. 158. Vaseghi M, Gima J, Kanaan C, Ajijola OA, Marmureanu A, Mahajan A, et al. Cardiac sympathetic denervation in patients with refractory ventricular arrhythmias or electrical storm: Intermediate and long-term follow-up. Heart Rhythm. 2014;11(3):360-366. DOI: 10.1016/j.hrthm.2013.11.028. PubMed PMID: 24291775
  159. 159. Del Rio R, Marcus NJ, Schultz HD. Carotid chemoreceptor ablation improves survival in heart failure: Rescuing autonomic control of cardiorespiratory function. Journal of the American College of Cardiology. 2013;62(25):2422-2430. DOI: 10.1016/j.jacc.2013.07.079. PubMed PMID: 24013056
  160. 160. Marcus NJ, Del Rio R, Schultz EP, Xia XH, Schultz HD. Carotid body denervation improves autonomic and cardiac function and attenuates disordered breathing in congestive heart failure. The Journal of Physiology. 2014;592(2):391-408. DOI: 10.1113/jphysiol.2013.266221
  161. 161. McBryde FD, Abdala AP, Hendy EB, Pijacka W, Marvar P, Moraes DJ, et al. The carotid body as a putative therapeutic target for the treatment of neurogenic hypertension. Nature Communications. 2013;4:2395. DOI: 10.1038/ncomms3395
  162. 162. Paton JF, Sobotka PA, Fudim M, Engelman ZJ, Hart EC, McBryde FD, et al. The carotid body as a therapeutic target for the treatment of sympathetically mediated diseases. Hypertension. 2013;61(1):5-13. DOI: 10.1161/HYPERTENSIONAHA.111.00064
  163. 163. Kudej RK, Shen YT, Peppas AP, Huang CH, Chen W, Yan L, et al. Obligatory role of cardiac nerves and alpha1-adrenergic receptors for the second window of ischemic preconditioning in conscious pigs. Circulation Research. 2006;99(11):1270-1276
  164. 164. Basalay M, Barsukevich V, Mastitskaya S, Mrochek A, Pernow J, Sjoquist PO, et al. Remote ischaemic pre- and delayed postconditioning—Similar degree of cardioprotection but distinct mechanisms. Experimental Physiology. 2012;97(8):908-917. DOI: expphysiol.2012.064923 [pii];10.1113/expphysiol.2012.064923
  165. 165. Priori SG, Blomstrom-Lundqvist C, Mazzanti A, Blom N, Borggrefe M, Camm J, et al. 2015 ESC guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: The task force for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death of the European Society of Cardiology (ESC)endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC). Europace. 2015;17(11):1601-1687. DOI: 10.1093/europace/euv319
  166. 166. Murphy S, Krainock R, Tham M. Neuregulin signaling via erbB receptor assemblies in the nervous system. Molecular Neurobiology. 2002;25(1):67-77. DOI: 10.1385/MN:25:1:067
  167. 167. Yamada S, Marutsuka M, Inoue M, Zhang J, Abe S, Ishibashi K, et al. The interaction of the ErbB4 intracellular domain p80 with alpha-enolase in the nuclei is associated with the inhibition of the neuregulin1-dependent cell proliferation. International Journal of Biochemistry and Molecular Biology. 2014;5(1):21-29
  168. 168. Gao R, Zhang J, Cheng L, Wu X, Dong W, Yang X, et al. A phase II, randomized, double-blind, multicenter, based on standard therapy, placebo-controlled study of the efficacy and safety of recombinant human neuregulin-1 in patients with chronic heart failure. Journal of the American College of Cardiology. 2010;55(18):1907-1914. DOI: 10.1016/j.jacc.2009.12.044
  169. 169. Pascal D, Giovannelli A, Gnavi S, Hoyng SA, de Winter F, Morano M, et al. Characterization of glial cell models and in vitro manipulation of the neuregulin1/ErbB system. BioMed Research International. 2014;2014:310215. DOI: 10.1155/2014/310215
  170. 170. Rohrbach S, Yan X, Weinberg EO, Hasan F, Bartunek J, Marchionni MA, et al. Neuregulin in cardiac hypertrophy in rats with aortic stenosis. Differential expression of erbB2 and erbB4 receptors. Circulation. 1999;100(4):407-412. PubMed PMID: 10421602
  171. 171. Abdala AP, McBryde FD, Marina N, Hendy EB, Engelman ZJ, Fudim M, et al. Hypertension is critically dependent on the carotid body input in the spontaneously hypertensive rat. The Journal of Physiology. 2012;590(17):4269-4277. DOI: 10.1113/jphysiol.2012.237800
  172. 172. Gossot D, Kabiri H, Caliandro R, Debrosse D, Girard P, Grunenwald D. Early complications of thoracic endoscopic sympathectomy: A prospective study of 940 procedures. The Annals of Thoracic Surgery. 2001;71(4):1116-1119. PubMed PMID: 11308146
  173. 173. Vaseghi M, Lellouche N, Ritter H, Fonarow GC, Patel JK, Moriguchi J, et al. Mode and mechanisms of death after orthotopic heart transplantation. Heart Rhythm. 2009;6(4):503-509. DOI: S1547-5271(09)00013-7
  174. 174. Salavatian S, Beaumont E, Longpre JP, Armour JA, Vinet A, Jacquemet V, et al. Vagal stimulation targets select populations of intrinsic cardiac neurons to control neurally induced atrial fibrillation. American Journal of Physiology. Heart and Circulatory Physiology. 2016;311(5):H1311-H1H20. DOI: 10.1152/ajpheart.00443.2016
  175. 175. Ardell JL, Cardinal R, Beaumont E, Vermeulen M, Smith FM, Andrew Armour J. Chronic spinal cord stimulation modifies intrinsic cardiac synaptic efficacy in the suppression of atrial fibrillation. Autonomic Neuroscience. 2014;186:38-44. DOI: 10.1016/j.autneu.2014.09.017
  176. 176. Stavrakis S, Nakagawa H, Po SS, Scherlag BJ, Lazzara R, Jackman WM. The role of the autonomic ganglia in atrial fibrillation. JACC: Clinical Electrophysiology. 2015;1(1-2):1-13. DOI: 10.1016/j.jacep.2015.01.005
  177. 177. Southerland EM, Milhorn DM, Foreman RD, Linderoth B, DeJongste MJ, Armour JA, et al. Preemptive, but not reactive, spinal cord stimulation mitigates transient ischemia-induced myocardial infarction via cardiac adrenergic neurons. American Journal of Physiology. Heart and Circulatory Physiology. 2007;292(1):H311-H3H7
  178. 178. Lopshire JC, Zhou X, Dusa C, Ueyama T, Rosenberger J, Courtney N, et al. Spinal cord stimulation improves ventricular function and reduces ventricular arrhythmias in a canine postinfarction heart failure model. Circulation. 2009;120(4):286-294. DOI: 10.1161/CIRCULATIONAHA.108.812412
  179. 179. McCreery DB, Agnew WF, Yuen TG, Bullara LA. Comparison of neural damage induced by electrical stimulation with faradaic and capacitor electrodes. Annals of Biomedical Engineering. 1988;16(5):463-481. PubMed PMID: 3189974
  180. 180. Kilgore KL, Bhadra N. Reversible nerve conduction block using kilohertz frequency alternating current. Neuromodulation. 2014;17(3):242-254; discussion 54-5. DOI: 10.1111/ner.12100
  181. 181. Ding X, Ardell JL, Hua F, McAuley RJ, Sutherly K, Daniel JJ, et al. Modulation of cardiac ischemia-sensitive afferent neuron signaling by preemptive C2 spinal cord stimulation: Effect on substance P release from rat spinal cord. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2008;294(1):R93-R101. DOI: 10.1152/ajpregu.00544.2007
  182. 182. Bagger JP, Jensen BS, Johannsen G. Long-term outcome of spinal cord electrical stimulation in patients with refractory chest pain. Clinical Cardiology. 1998;21(4):286-288. PubMed PMID: 9562939
  183. 183. de Jongste MJ, Haaksma J, Hautvast RW, Hillege HL, Meyler PW, Staal MJ, et al. Effects of spinal cord stimulation on myocardial ischaemia during daily life in patients with severe coronary artery disease. A prospective ambulatory electrocardiographic study. British Heart Journal. 1994;71(5):413-418. PubMed PMID: 8011403
  184. 184. Ardell JL. Heart failure: Mechanisms of spinal cord neuromodulation for heart disease. Nature Reviews. Cardiology. 2016;13(3):127-128. DOI: 10.1038/nrcardio.2016.8
  185. 185. Beaumont E, Wright GL, Southerland EM, Li Y, Chui R, KenKnight BH, et al. Vagus nerve stimulation mitigates intrinsic cardiac neuronal remodeling and cardiac hypertrophy induced by chronic pressure overload in Guinea pig. American Journal of Physiology. Heart and Circulatory Physiology. 2016;310(10):H1349-H1359. DOI: 10.1152/ajpheart.00939.2015
  186. 186. Katare RG, Ando M, Kakinuma Y, Arikawa M, Handa T, Yamasaki F, et al. Vagal nerve stimulation prevents reperfusion injury through inhibition of opening of mitochondrial permeability transition pore independent of the bradycardiac effect. The Journal of Thoracic and Cardiovascular Surgery. 2009;137(1):223-231. DOI: S0022-5223(08)01404-9
  187. 187. Shinlapawittayatorn K, Chinda K, Palee S, Surinkaew S, Kumfu S, Kumphune S, et al. Vagus nerve stimulation initiated late during ischemia, but not reperfusion, exerts cardioprotection via amelioration of cardiac mitochondrial dysfunction. Heart Rhythm. 2014;11(12):2278-2287. DOI: 10.1016/j.hrthm.2014.08.001
  188. 188. Olshansky B, Sabbah HN, Hauptman PJ, Colucci WS. Parasympathetic nervous system and heart failure: Pathophysiology and potential implications for therapy. Circulation. 2008;118(8):863-871. DOI: 10.1161/CIRCULATIONAHA.107.760405
  189. 189. Furukawa Y, Hoyano Y, Chiba S. Parasympathetic inhibition of sympathetic effects on sinus rate in anesthetized dogs. American Journal of Physiology-Heart and Circulatory Physiology. 1996;271(40):H44-H50
  190. 190. McGuirt AS, Schmacht DC, Ardell JL. Autonomic interactions for control of atrial rate are maintained after SA nodal parasympathectomy. American Journal of Physiology-Heart and Circulatory Physiology. 1997;272(41):H2525-H2H33
  191. 191. Kawada T, Yamazaki T, Akiyama T, Li M, Ariumi H, Mori H, et al. Vagal stimulation suppresses ischemia-induced myocardial interstitial norepinephrine release. Life Sciences. 2006;78(8):882-887. DOI: 10.1016/j.lfs.2005.05.087
  192. 192. Salavatian S, Beaumont E, Gibbons D, Hammer M, Hoover DB, Armour JA, et al. Thoracic spinal cord and cervical vagosympathetic neuromodulation obtund nodose sensory transduction of myocardial ischemia. Autonomic Neuroscience. 2017;208:57-65. DOI: 10.1016/j.autneu.2017.08.005
  193. 193. Premchand RK, Sharma K, Mittal S, Monteiro R, Dixit S, Libbus I, et al. Extended follow-up of patients with heart failure receiving autonomic regulation therapy in the ANTHEM-HF study. Journal of Cardiac Failure. 2016;22(8):639-642. DOI: 10.1016/j.cardfail.2015.11.002
  194. 194. DiCarlo LA, Libbus I, Kumar HU, Mittal S, Premchand RK, Amurthur B, et al. Autonomic regulation therapy to enhance myocardial function in heart failure patients: The ANTHEM-HFpEF study. ESC Heart Failure. 2018;5(1):95-100. DOI: 10.1002/ehf2.12241
  195. 195. Ardell JL, Butler CK, Smith FM, Hopkins DA, Armour JA. Activity of in vivo atrial and ventricular neurons in chronically decentralized canine hearts. American Journal of Physiology-Heart and Circulatory Physiology. 1991;260(29):H713-HH21
  196. 196. Hausenloy DJ, Yellon DM. The therapeutic potential of ischemic conditioning: An update. Nature Reviews. Cardiology. 2011;8(11):619-629. DOI: 10.1038/nrcardio.2011.85
  197. 197. Cabrera-Fuentes HA, Alba-Alba C, Aragones J, Bernhagen J, Boisvert WA, Botker HE, et al. Meeting report from the 2nd international symposium on new frontiers in cardiovascular research. Protecting the cardiovascular system from ischemia: Between bench and bedside. Basic Research in Cardiology. 2016;111(1):7. DOI: 10.1007/s00395-015-0527-0
  198. 198. Kloner RA, Rezkalla SH. Preconditioning, postconditioning and their application to clinical cardiology. Cardiovascular Research. 2006;70(2):297-307. DOI: 10.1016/j.cardiores.2006.01.012
  199. 199. Lynch C III. Anesthetic preconditioning: Not just for the heart? Anesthesiology. 1999;91(3):606-608
  200. 200. Minguet G, Joris J, Lamy M. Preconditioning and protection against ischaemia-reperfusion in non-cardiac organs: A place for volatile anaesthetics? European Journal of Anaesthesiology. 2007;24(9):733-745. DOI: S0265021507000531 [pii];10.1017/S0265021507000531
  201. 201. Hausenloy DJ, Yellon DM. Preconditioning and postconditioning: New strategies for cardioprotection. Diabetes, Obesity & Metabolism. 2008;10(6):451-459. DOI: DOM762 [pii];10.1111/j.1463-1326.2007.00762.x
  202. 202. Hausenloy DJ, Yellon DM. Preconditioning and postconditioning: Underlying mechanisms and clinical application. Atherosclerosis. 2009;204(2):334-341. DOI: S0021-9150(08)00748-X
  203. 203. Gerritsen J, Dekker JM, TenVoorde BJ, Kostense PJ, Heine RJ, Bouter LM, et al. Impaired autonomic function is associated with increased mortality, especially in subjects with diabetes, hypertension, or a history of cardiovascular disease: The Hoorn study. Diabetes Care. 2001;24(10):1793-1798
  204. 204. Pop-Busui R. Cardiac autonomic neuropathy in diabetes: A clinical perspective. Diabetes Care. 2010;33(2):434-441. DOI: 33/2/434 [pii];10.2337/dc09-1294

Written By

John G. Kingma, Denys Simard and Jacques R. Rouleau

Submitted: 05 February 2018 Reviewed: 09 April 2018 Published: 24 October 2018