You are on page 1of 18

JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO.

10, 2019

ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION

PUBLISHED BY ELSEVIER

THE PRESENT AND FUTURE

JACC FOCUS SEMINAR

Autonomic Nervous System Dysfunction


JACC Focus Seminar

Jeffrey J. Goldberger, MD,a Rishi Arora, MD,b Una Buckley, MD,c Kalyanam Shivkumar, MD, PHDc

JACC JOURNAL CME/MOC/ECME

This article has been selected as the month’s JACC CME/MOC/ECME Method of Participation and Receipt of CME/MOC/ECME Certificate
activity, available online at http://www.acc.org/jacc-journals-cme by
To obtain credit for JACC CME/MOC/ECME, you must:
selecting the JACC Journals CME/MOC/ECME tab.
1. Be an ACC member or JACC subscriber.
Accreditation and Designation Statement 2. Carefully read the CME/MOC/ECME-designated article available on-
line and in this issue of the Journal.
The American College of Cardiology Foundation (ACCF) is accredited by
3. Answer the post-test questions. A passing score of at least 70% must be
the Accreditation Council for Continuing Medical Education to provide
achieved to obtain credit.
continuing medical education for physicians.
4. Complete a brief evaluation.
The ACCF designates this Journal-based CME activity for a maximum 5. Claim your CME/MOC/ECME credit and receive your certificate electron-
of 1 AMA PRA Category 1 Credit(s). Physicians should claim only the ically by following the instructions given at the conclusion of the activity.
credit commensurate with the extent of their participation in the
activity. CME/MOC/ECME Objective for This Article: Upon completion of this activity,
the learner should be able to: 1) describe different levels of autonomic input
Successful completion of this CME activity, which includes participa-
to the heart and the types of autonomic dysfunction; 2) describe currently
tion in the evaluation component, enables the participant to earn up to
available techniques to assess autonomic function; and 3) describe the role
1 Medical Knowledge MOC point in the American Board of Internal
of autonomic tone and/or dysfunction in cardiac arrhythmias.
Medicine’s (ABIM) Maintenance of Certification (MOC) program. Par-
CME/MOC/ECME Editor Disclosure: JACC CME/MOC/ECME Editor
ticipants will earn MOC points equivalent to the amount of CME credits
Ragavendra R. Baliga, MD, FACC, has reported that he has no financial
claimed for the activity. It is the CME activity provider’s responsibility
relationships or interests to disclose.
to submit participant completion information to ACCME for the pur-
pose of granting ABIM MOC credit. Author Disclosures: Dr. Goldeberger is supported by the National Heart,
Lung, and Blood Institute (NHLBI) (HL70179). Dr. Arora is supported by
Autonomic Nervous System Dysfunction: JACC Focus Seminar will be the NHLBI (HL093490). Dr. Shivkumar is supported by the NHLBI
accredited by the European Board for Accreditation in Cardiology (HL084261), by NHLBI grant R01HL084261, and by National Institutes of
(EBAC) for 1 hour of External CME credits. Each participant should Health grant NIHOT2OD023848. Dr. Arora holds ownership interest in
claim only those hours of credit that have actually been spent in the Rhythm Therapeutics, Inc. All other authors have reported that they have
educational activity. The Accreditation Council for Continuing no relationships relevant to the contents of this paper to disclose.
Medical Education (ACCME) and the European Board for Accredita-
Medium of Participation: Print (article only); online (article and quiz).
tion in Cardiology (EBAC) have recognized each other’s accreditation
systems as substantially equivalent. Apply for credit through the
CME/MOC/ECME Term of Approval
post-course evaluation. While offering the credits noted above, this
program is not intended to provide extensive training or certification Issue Date: March 19, 2019
in the field. Expiration Date: March 18, 2020

Listen to this manuscript’s


audio summary by From the aCardiovascular Division, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida;
Editor-in-Chief b
Feinberg Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Northwestern University-Feinberg
Dr. Valentin Fuster on School of Medicine, Chicago, Illinois; and the cCardiac Arrhythmia Center and Neurocardiology Research Center of Excellence,
JACC.org. University of California-Los Angeles Los Angeles, California. Dr. Goldeberger is supported by the National Heart, Lung, and Blood
Institute (NHLBI) (HL70179). Dr. Arora is supported by the NHLBI (HL093490). Dr. Shivkumar is supported by the NHLBI
(HL084261), by NHLBI grant R01HL084261, and by National Institutes of Health grant NIHOT2OD023848. Dr. Arora holds
ownership interest in Rhythm Therapeutics, Inc. All other authors have reported that they have no relationships relevant to the
contents of this paper to disclose.

Manuscript received April 16, 2018; revised manuscript received December 21, 2018, accepted December 30, 2018.

ISSN 0735-1097/$36.00 https://doi.org/10.1016/j.jacc.2018.12.064


1190 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

Autonomic Nervous System Dysfunction


JACC Focus Seminar
Jeffrey J. Goldberger, MD,a Rishi Arora, MD,b Una Buckley, MD,c Kalyanam Shivkumar, MD, PHDc

ABSTRACT

Autonomic nervous system control of the heart is a dynamic process in both health and disease. A multilevel neural
network is responsible for control of chronotropy, lusitropy, dromotropy, and inotropy. Intrinsic autonomic dysfunction
arises from diseases that directly affect the autonomic nerves, such as diabetes mellitus and the syndromes of primary
autonomic failure. Extrinsic autonomic dysfunction reflects the changes in autonomic function that are secondarily
induced by cardiac or other disease. An array of tests interrogate various aspects of cardiac autonomic control in either
resting conditions or with physiological perturbations from resting conditions. The prognostic significance of these as-
sessments have been well established. Clinical usefulness has not been established, and the precise mechanistic link to
mortality is less well established. Further efforts are required to develop optimal approaches to delineate cardiac
autonomic dysfunction and its adverse effects to develop tools that can be used to guide clinical decision-making.
(J Am Coll Cardiol 2019;73:1189–206) © 2019 by the American College of Cardiology Foundation.

A utonomic nervous system (ANS) control of


the heart is a dynamic process in both health
and disease. ANS dysfunction may result
from primary disorders of the autonomic nerves or
emerging therapies in context. Autonomic control of
the heart is achieved by afferent neural impulses that
are transmitted from the heart to the intrinsic neu-
rons of the heart, to extracardiac intrathoracic ganglia
secondarily in response to cardiac (or other systemic) (e.g., stellate ganglion), to the spinal cord, and to the
disease. Cardiac disease may promote both anatomic brain stem. These afferent neural signals are pro-
(primary) and functional (secondary) changes in car- cessed by various parts of the nervous system to
diac autonomic function. These changes may, in regulate the cardiomotor neural output to the heart
turn, contribute to the progression of disease and/or via the sympathetic and parasympathetic nerves. It is
be involved in arrhythmogenesis. Beta-adrenergic important to emphasize that the anatomical nerve
blockers are the most established autonomic inter- trunks that reach the heart, which are traditionally
vention associated with improved outcomes. Other described as the sympathetic and parasympathetic
interventions (e.g., cardiac sympathetic denervation) trunks, have both afferent and efferent nerve fibers.
have shown promise for the management of refrac-
tory ventricular arrhythmias (1). Much has been ORGANIZATION OF CARDIAC NEURAL CONTROL.
learned about the complex interactions along the This neuroaxis is organized as multiple levels of
neuroaxis and their role in cardiac control. What has integrative centers. At the level of the heart, the
been most challenging is the development of a simple intrinsic cardiac nervous system (ICNS) is a distrib-
method of assessment of the autonomic effects on the uted network system located in the cardiac ganglia
heart and/or autonomic dysfunction. Consequently, that are ganglionated plexi (GPs) that exist in the fat
there have been a plethora of methods that assess pads around the heart, predominantly in the posterior
some aspect of autonomic function. Although many and superior aspects of the atria (2). These connect
measures have been shown to have some prognostic with the intrathoracic extracardiac ganglia (the sym-
significance, none have been adapted or adopted pathetic paravertebral ganglia), the extrathoracic
into clinical practice. This review highlights some of cardiac ganglia (the nodose, dorsal root ganglia), and
the background and newer concepts in autonomic the central nervous system (3,4). At each level, the
control of the heart. system has the ability to modulate cardiac activity
with efferent feedback loops (Figure 1).
NORMAL AUTONOMIC FUNCTION Afferent neural signals are transmitted to the ICNS,
the stellate ganglia, and via the dorsal root ganglia to
A brief description of the advances in our under- the spinal cord and to the nodose ganglia (Figure 2)
standing of the physiology of cardiac autonomic (5,6). The efferent projections to the heart occur
control is in order to place autonomic testing and via short and long feedback loops that occur at
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1191
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

the central nervous system, the spinal cord (inter- The cardiomotor function of this system ABBREVIATIONS

mediolateral columns), the intrathoracic ganglia, and helps slow the heart rate, reduce blood pres- AND ACRONYMS

the ICNS. Sensory neurites associated with the ICNS sure, and balances the system to ensure there
AF = atrial fibrillation
are found in areas of the atrioventricular junction and is a counterbalance to sympathoexcitation.
ANS = autonomic nervous
the adventitia of major vessels (e.g., coronary ar- The parasympathetic effects are coordinated
system
teries). It was previously believed that the different via the cervical vagus nerve, which divides
BRS = baroreflex sensitivity
levels within the ANS were just relay stations, but it is into the superior and inferior cardiac nerves
GP = ganglionated plexus
now well-established that each level processes neural to finally enter the heart via the cardiac
HRR = heart rate recovery
information and coordinates a response by commu- plexus. The parasympathetic nerve fibers are
HRT = heart rate turbulence
nicating with other levels by these feedback loops much more heterogeneously distributed,
(Figure 2). with significant innervation of the sinoatrial HRV = heart rate variability

The ICNS consists of ganglia composed of afferent, node, atrioventricular node (7), and the ven- ICNS = intrinsic cardiac
nervous system
efferent, and interconnecting neurons to other car- tricles (17).
MI = myocardial infarction
diac ganglia. These ganglia coordinate the sympa- The parasympathetic cardiomotor response
thetic and parasympathetic inputs received from the is coordinated by neurons in the ICNS that PV = pulmonary vein

rest of the cardiac ANS. The ICNS has regional control receive pre-ganglionic parasympathetic input from
over different cardiac functions, such as sinus node the cervical vagus and provide a homogeneous or co-
electrical activation and propagation, as well as ordinated response to the atria and ventricles (18,19).
atrioventricular nodal conduction (7,8). Emerging Cervical vagus stimulation can produce different re-
therapies that target these structures have shown the sponses depending on what aspect of the neuroaxis is
importance of emerging research in this area. engaged. In the intact state, stimulation can result
S y m p a t h e t i c a f f e r e n t s a n d e f f e r e n t s . The sym- in both direct and reactive, or reflex initiated re-
pathetic neurons that regulate cardiac function sponses on the intrinsic cardiac nervous system as a
are located in the stellate ganglion. It receives result of central and peripheral interactions initiated
pre-ganglionic sympathetic input from the inter- throughout the cardiac neuronal hierarchy. Low-level
mediolateral column (and other spinal neurons) and stimulation delivered to the cervical vagus can result
coordinates efferent neural responses either directly in tachycardia, probably as a result of engaging cardiac
or via the ansa subclavia to the middle cervical afferent fibers, whereas higher level stimulation re-
ganglia to the heart (9–12). Efferent post-ganglionic sults in bradycardia (20). In the resting state, the car-
fibers travel alongside the coronary vasculature to diac ANS is an intricate balance between sympathetic
penetrate the epicardial regions and travel toward the and parasympathetic inputs. Once the cervical vagus
endocardium. Stimulation of the stellate ganglion is transected from the central nervous system, there is
results in an increase in dromotropy, chronotropy, a significant increase in heart rate that suggests that
lusitropy, and inotropy (13). Cardiac afferent neurons the central cholinergic neuronal drive plays a consid-
are mechanosensory, chemosensory, or multimodal erable role in controlling the basal heart rate (20).
in nature (4). They transduce a variety of chemicals, Integration of cardiac and vascular afferents.
including various neuropeptides, such as substance Another important afferent control system includes
P, bradykinin, and calcitonin generelated peptide. the baroreceptors, which are stretch receptors
These cardiac afferents are also involved in initiating embedded in the adventitia of the aortic wall and
local inflammatory and vascular reactions that may carotid sinus that transduce pressure fluctuations to
play an important role in cardiac remodeling (14). the cardiovascular neural reflex pathways responsible
The sympathetic nerve fibers are located in the for heart rate and blood pressure. There is a contin-
atria and the ventricles. There is a regional response uous strip of mechanosensory neurites along the in-
to the right sympathetic paravertebral ganglia versus ner aortic arch (21). They are believed to transduce
the left sympathetic paravertebral ganglia, with combined mechanosensory information that mea-
predominant effects on the anterior and posterior sures distortion from pulsatile waves in the aorta in
ventricular walls, respectively (15). In addition to space and time. The arterial chemoreceptors are in
increased sinoatrial node firing and enhancement of the carotid arteries, aortic bodies, and medulla, and
atrioventricular nodal conduction, sympathetic respond to hypoxemia and hypercapnia, respectively.
output results in ventricular action potential duration The aortic arch mechanoreceptors can be found in the
shortening (16). nodose ganglion, whereas some of the carotid sinus
P a r a s y m p a t h e t i c a f f e r e n t s a n d e f f e r e n t s . The afferents are found in the petrosal ganglia. These re-
parasympathetic nervous system also has important ceptors then transmit information to the nucleus of
afferent and efferent components (Figures 1 and 2). the tractus solitarius, which modulates sympathetic
1192 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

F I G U R E 1 Autonomic Neural Control of the Heart

Autonomic neural control of the heart. Modified with permission from Shivkumar et al. (4). DRG ¼ dorsal root ganglion; ICNS ¼ intrinsic nervous system; SG ¼ stellate ganglion.
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1193
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

F I G U R E 2 Functional Organization of Cardiac Neural Control

Higher centers Cortex

Carotid body
chemoreceptors
Petrosal Brainstem
Medulla
ganglia
Carotid sinus Aortic
baroreceptors chemoreceptors
Nodose
C1-C2 Aff, soma
Aortic arterial
C1-L5 baroreceptors
Muscle DRG
T1-T4
Aff, soma
Spinal cord

Efferent
LCN Preganglionic
Kidneys Extracardiac
Postganglionic intrathoracic
T6-L2 ganglia (stellate,
Afferent Sympath middle cervical)
soma Efferent soma

LCN
Intrinsic cardiac
nervous system

Afferent soma Sympath Parasym


Efferent soma Efferent soma

Afferent

Neurite Neurite Neurite β1 M2 Neurite

Gs Gi Heart

‘Sympathetic’ ‘Parasympathetic’

The shaded areas (yellow ¼ sympathetic fibers, grey ¼ parasympathetic fibers [vagal trunk]) roughly correspond to the anatomical sympathetic and parasympathetic
nerve fibers that are seen macroscopically. The afferent neural fibers run along with nerves anatomically referred to as sympathetic and parasympathetic trunks.
Modified with permission from Shivkumar et al. (4). Aff ¼ afferent; LCN ¼ local circuit neuron; other abbreviation as Figure 1.

and parasympathetic output to the cardiovascular diseases that directly affect the autonomic nerves,
system. such as diabetes mellitus and the various syndromes
Finally, the regulation of the cardiac ANS is also of primary autonomic failure. Extrinsic autonomic
under central nervous system control. The balance of dysfunction reflects the changes in autonomic func-
activation and inhibitory neurons is finely tuned and tion that are secondarily induced by cardiac or other
is designed to provide dynamic control of the heart disease. Cardiac diseases, such as myocardial infarc-
under a range of physiological conditions ranging tion (MI), can also primarily disrupt the autonomic
from rest to severe exertion. This exquisite multilevel nerves.
neural network is profoundly altered in the presence
PRIMARY AUTONOMIC DYSFUNCTION. P r i m a r y
of cardiac dysfunction and leads to progression of
autonomic dysfunction resulting from diabetic
heart disease (22).
n e u r o p a t h y . Diabetes mellitus is associated with
AUTONOMIC DYSFUNCTION AND ITS ROLE IN the development of both peripheral and autonomic
CARDIOVASCULAR DISEASE. Autonomic dysfunc- neuropathy. Because of its widespread prevalence,
tion may arise from 2 mechanisms—intrinsic or diabetes is the leading cause of primary autonomic
extrinsic. Intrinsic autonomic dysfunction arises from dysfunction. Pathological studies have noted loss of
1194 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

and/or damage to myelinated vagus nerve axons (23– unique challenge in the diagnostic approach to this
25). Although findings in the sympathetic nerves and entity. Although cardiac autonomic reflex tests
ganglia have been disputed (26), they include giant (Ewing’s tests) are generally recommended for diag-
nerve cells, vacuolization, and neuroaxonal dystrophy nosis (47), it is unclear which method, if any, is
(24,26,27). Appenzeller and Richardson (27) noted ab- preferred. It is also unclear how often any assessment
normalities in 4 of 5 patients with diabetes with neu- for cardiac autonomic neuropathy is performed.
ropathy and in 0 of 4 patients without neuropathy. The presence of cardiac autonomic neuropathy
These studies showed significant pathological changes confers an adverse prognosis (48). In a meta-analysis
in the autonomic nerves of patients with diabetes who of 15 studies among subjects with diabetes mellitus
predominantly had advanced disease and peripheral (49), the pooled relative risk for mortality related to
neuropathy. The pathological basis of early diabetic cardiac autonomic neuropathy was 3.45 (95% confi-
autonomic neuropathy has not been elucidated. dence interval: 2.66 to 4.47; p < 0.001). The mecha-
Numerous biochemical mechanisms for hyperglyce- nism for the increased mortality has not been
mic injury have been implicated, including the pro- clarified, but various possibilities have been pro-
duction of advanced glycosylation end products, posed, including QT interval prolongation (50–54),
hyperglycemic activation of the polyol pathway, as renal disease (55–57), and asymptomatic myocardial
well as protein kinase C, immunological processes, ischemia (28,56,58). Some of these potential mecha-
and neurovascular insufficiency that leads to local nisms may arise directly from the primary distur-
ischemia. The proposed mechanisms come largely bance of cardiac autonomic function.
from research on somatic nerves in experimental dia- Other primary disorders of autonomic dysfunction. Other
betic animal models. Although biochemically diverse, primary disorders of autonomic dysfunction and/or
the various degenerative mechanisms have a common failure include a range of disorders—pure autonomic
predisposition for distal axon terminals. The patho- failure, idiopathic orthostatic hypotension, Parkin-
genesis of diabetic cardiac autonomic neuropathy son’s disease with autonomic failure, and multiple
in vivo is likely to be heterogeneous and results from system atrophy (59). These disorders are character-
the interaction of multiple pathways (26,28–30). ized by orthostatic hypotension in combination with
Because the vagus nerve is the longest autonomic other manifestations. Postural orthostatic tachy-
nerve in the body, abnormalities in parasympathetic cardia syndrome is a complex syndrome with a major
innervation of the heart are typically the earliest autonomic component. An autoimmune etiology that
manifestation of cardiac autonomic neuropathy (31). targets ganglionic receptors (nicotinic acetylcholine
The prevalence of cardiac autonomic neuropathy in receptors) may be the underlying etiology for some of
patients with diabetes varies widely depending on the these disorders (60). Extensive pathological studies
cohort studied and the tests and/or criteria used for have not been reported, but in autonomic failure,
assessment, but approximately 15% to 20% of Lewy bodies (abnormal protein aggregates) have
asymptomatic people with diabetes appear to have been identified in the sympathetic ganglia (61,62).
abnormal cardiovascular autonomic function (28,32–34). These disorders are discussed in further detail in
Only later stages of the disease are associated with other studies (63,64).
symptoms. Autonomic abnormalities may even be
detected before the onset of diabetes (35–37). Early AUTONOMIC DYSFUNCTION SECONDARY TO MI,
evidence of cardiac autonomic neuropathy can be CARDIOMYOPATHY, AND HEART FAILURE, AND ITS ROLE
detected in children with type 1 diabetes (38). Many IN THE GENESIS OF VENTRICULAR ARRHYTHMIAS. In
methods are used to diagnose cardiac autonomic contrast to disorders that primarily affect the auto-
neuropathy. Because the heart rate is easily measured nomic nerves, a variety of cardiac pathologies, such
and responds to autonomic stimuli (39), noninvasive as MI, heart failure, and cardiomyopathy result in
studies to assess cardiac autonomic neuropathy focus secondary acute and chronic changes within the ANS
primarily on heart rate or heart rate responses to (65). Direct ischemic damage to cardiac autonomic
physiological manipulations. Cardiac autonomic nerves may result from acute MI (66). In addition, the
neuropathy has been diagnosed based on abnormal- cardiac ANS responds acutely to preserve homeosta-
ities in heart rate variability (HRV), baroreflex sensi- sis. This may involve increased sympathetic and
tivity (BRS), Ewing’s tests (heart rate and/or blood decreased parasympathetic activity to maintain car-
pressure responses to deep breathing, standing, Val- diac contractility and cardiac output in the setting of
salva maneuver, and handgrip), and heart rate recov- a major insult. Hemodynamic changes that occur as a
ery (HRR) (40–44). These parameters are not highly consequence of these conditions cause a cascade
correlated with each other (45,46), which presents a of changes in neurohumoral activity in the
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1195
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

cardiovascular, peripheral vascular, and renal changes as a result of MI (74). The afferent IC
vascular systems. The problem arises when the im- neuronal activity is attenuated in the territory of the
mediate threat has abated, but the imbalance in the MI, whereas those IC neurons in remote regions are
ANS, driven by altered afferent signaling, persists and preserved. As such, this creates a heterogenous
is further modulated by cytokines generated as a neural response within the myocardium (74). This
response to the disease state (65). This can lead to a suggests that the ICNS receives and coordinates the
vicious cycle, with MI, cardiomyopathy, and heart information that is transmitted to the intrathoracic
failure leading to an outpouring of catecholamines and extrathoracic ganglia. In relation to other
(and inflammatory cytokines), and the catechol- ganglionic changes seen as a result of MI, the dorsal
amines, in turn, lead to worsening of cardiomyopa- root ganglia appear not to change morphologically,
thy. This cycle of adrenergic activation and cardiac but they do have a significant change in neurochem-
dysfunction has been well established in congestive ical properties of the neurons present (increased
heart failure with reduced ejection fraction. The neuronal nitric oxide synthase and calcitonin gene-
marked benefit of beta-adrenergic blockers in this en- related peptide) (69). Alterations in neurohumoral
tity, which improve both ejection fraction and out- control, circulating catecholamines, and the renin-
comes, is a testament to the critical role of the angiotensin-aldosterone system also occur as a
secondary changes in the ANS in this disease state and result of cardiac injury.
reversibility of congestive heart failure with treatment. Ventricular arrhythmias that occur in the setting of
Persistent maladaptations within the ANS as a ischemic and nonischemic cardiomyopathy may be
result of cardiac dysfunction have been linked to precipitated as a result of sympathetic overactivity.
atrial and ventricular arrhythmias (4). These changes These occur as a result of an excessive sympatho-
result in a functional reorganization of the ANS and neurohumoral response and a reduced para-
alterations in neural processing at each level of the sympathetic input that results in increased dispersion
cardiac neural hierarchy, which leads to a conflict of electrical activation and repolarization in the ven-
between the central (central nervous system) and tricles from the endocardium to epicardium. In recent
peripheral (intrathoracic cardiac ganglia) control. years, neuromodulation, with the goal of increasing
Cardiac dysfunction can cause short- and long-term parasympathetic tone and suppressing sympathetic
changes in the neural networks, which results in tone, has become an emerging therapeutic strategy
exaggerated reflex responses. Central sensitization for the treatment of ventricular arrhythmias.
of neurons can occur secondarily to constant Emerging therapeutic approaches include left cardiac
afferent signaling from the site of injury, such as the sympathetic denervation, cervical vagal stimulation,
infarct zone, which can elicit a cellular process and spinal cord stimulation (75). Removal of sympa-
that changes the excitability of cell membranes thetic efferent input to the heart by surgical resection
and reduces the inhibitory mechanisms within the or thoracic epidural anesthesia of the stellate gan-
nervous system (67). glion to T4 can significantly reduce ventricular ar-
Morphological changes seen in the stellate gan- rhythmias in the setting of a severe life-threatening
glion secondarily to ischemic and nonischemic car- ventricular electrical storm. Although animal studies
diomyopathy include neuronal enlargement (68), as have clearly shown that vagal nerve stimulation can
well as alterations in neurochemical expression pat- suppress ventricular arrhythmias in heart failure,
terns in the stellate (69). Other changes such as nerve vagal stimulation has only been used in patients with
sprouting around the border zones or periphery of an heart failure in an attempt to improve cardiac
ischemic territory also occur (70). Regional adrenergic remodeling and heart failure (76,75). Similarly, spinal
overexpression with functional denervation may be cord stimulation has been shown to be improve
seen in the border zone after MI in animal models (71) symptoms, functional status, and left ventricular
and humans with ischemic cardiomyopathy (72). function and remodeling in patients with severe
Clinically, it is well appreciated that excess adren- symptomatic heart failure (77); however, spinal cord
ergic activity is a negative prognostic indicator post- stimulation has not been systematically evaluated
MI and in congestive cardiac failure. This forms the for its antiarrhythmic benefits in clinical studies.
basis for the success of beta-blocker therapy in these Despite the potential success of neuromodulation
conditions. in pre-clinical or clinical studies in preventing ven-
There is neuronal enlargement within the stellate tricular arrhythmias, it is important to remember that
ganglion in both the right and left stellate ganglion, cardiac dysfunction results in adverse adaptions of
regardless of the territory of the MI (73). This is in the afferent and efferent inputs at various levels
contrast to the ICNS, where there are regional throughout the cardiac neuroaxis. These adaptions
1196 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

may perpetuate cardiac dysfunction and are generally nerves and a higher density of adrenergic nerves.
pro-arrhythmic. Further in-depth mechanistic un- There was evidence of sympathetic hyperinnervation
derstanding is required to improve the sophistication in patients with persistent AF (96).
of these therapies. Using direct nerve recordings from the stellate
ROLE OF AUTONOMIC DYSFUNCTION IN CREATING A ganglia and the vagus nerve in a canine study, Ogawa
VULNERABLE SUBSTRATE FOR ATRIAL FIBRILLATION. et al. (86) showed increased sympathetic and vagal
Atrial fibrillation (AF) is the most common sustained nerve discharge before the onset of atrial arrhythmias
arrhythmia disturbance and is associated with sig- in pacing-induced heart failure [these atrial arrhyth-
nificant morbidity and mortality. The morbidity and mias were preventable by prophylactic ablation of the
mortality associated with AF are especially increased stellate ganglion and the T2 to T4 thoracic sympa-
in the setting of congestive heart failure, with up to thetic ganglia (85)]. In a canine HF model, Ng et al.
one-half of all patients with heart failure having (78) demonstrated a profound increase in para-
concomitant AF (78). Several mechanisms contribute sympathetic—and to a lesser extent sympathetic—
to the electrophysiological and structural substrate nerves in the left atrium (78), with nerve growth be-
for AF, including fibrosis, stretch, oxidative stress, ing most pronounced in the PVs and posterior left
and altered calcium handling characteristics (79). The atrium. The increase in vagal innervation noted in
ANS has been hypothesized to have a likely role in this model was believed to contribute to the AF sub-
creation of a vulnerable AF substrate (80), with both strate by affecting conduction characteristics of the
sympathetic and parasympathetic nerves believed to PVs and posterior left atrium.
be involved in the genesis of AF (81,82). Armour et al. Taken together, the previous studies indicated an
(2) demonstrated an intricate pattern of autonomic important role for the ANS in the genesis of AF not
innervation in the heart with the atria being inner- only in normal hearts, but also in structural heart
vated by at least 5 major atrial fad pads or GPs (80). disease. These data underscore the potential impor-
Direct nerve recordings from the stellate ganglia, tance of the ANS as a suitable therapeutic target in
vagus nerve, and the cardiac GPs also demonstrated AF. A variety of strategies targeting $1 GPs either
a dynamic interaction between the sympathetic surgically (97,98) or through a transvenous, endo-
and parasympathetic nervous system in creating AF cardial approach (alone or together with PV isolation)
(83–86). Hou et al. (87,88) suggested the presence of have been used in patients with both paroxysmal and
an intricate, interconnecting neural network in the persistent AF with variable success (99–101). Scana-
left atrium that could contribute to a substrate for AF. vacca et al. (102) demonstrated the feasibility of se-
Over the last several years, the pulmonary veins lective atrial vagal denervation, as guided by evoked
(PVs) and adjoining posterior left atrium have been vagal reflexes, to treat patients with paroxysmal AF.
shown to play a significant role in the genesis of AF, Pokushalov et al. (103) reported that regional ablation
with this region demonstrating unique structural, at the anatomic sites of the left atrial GP could be
molecular, and electrophysiological characteristics safely performed and enabled maintenance of sinus
that appear to contribute to the AF substrate. rhythm in 71% of patients with paroxysmal AF. Kar-
Anatomical and physiological studies of the auto- itsis et al (104) and others (105) demonstrated that
nomic innervation of the atria indicate that the pul- when GP ablation was combined with PV isolation, it
monary veins and posterior left atria have a unique yielded better results than PV isolation alone, with
autonomic profile (86,89–91). Chevalier et al. (92) success rates approaching 80% (104). Recent surgical
described several gradients of innervation in and studies also attempted to add GP ablation and/or
around the PVs (92). In a canine study, Arora et al. excision to PV isolation with varying efficacy
(93) showed that the posterior left atrium was more (97,98,106,107). The efficacy and durability of this
richly innervated than the rest of the left atrium, with approach has not been established.
nerve bundles containing both parasympathetic and Renal denervation, which is known to lead to a
sympathetic fibers, with parasympathetic fibers pre- reduction of renal norepinephrine spillover (108) and
dominating over sympathetic fibers. These canine a reduction in firing of single sympathetic vasocon-
studies were in agreement with human studies that strictor fibers (a measure of central sympathetic nerve
demonstrated co-localization of sympathetic and outflow) (109), has been shown to reduce atrial sym-
parasympathetic nerve fibers in the human left pathetic nerve sprouting, structural alterations,
atrium (80,94). Deneke et al. (95) also demonstrated and AF complexity in goats with persistent AF,
co-localization of sympathetic and parasympathetic independent of changes in blood pressure (110).
nerves, with patients in persistent AF that demon- Early stage clinical data suggests that renal denerva-
strated a shift toward a lower density of cholinergic tion may improve the results of pulmonary vein
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1197
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

isolation in patients with persistent AF and/or severe node, atrium, atrioventricular node, and ventricles
resistant hypertension (111). However, the Symplicity differ in normal subjects and may be even more
HTN-3 trial did not demonstrate efficacy of renal accentuated in the presence of cardiac disease with
denervation in reducing blood pressure in patients regional abnormalities in innervation and ganglionic
with resistant hypertension (112). Its role as a function. How closely abnormalities in sinus node
potential therapeutic tool in AF has not been autonomic function parallel abnormalities in other
established. areas of the heart may depend on regional differences
Recent approaches attempted to disrupt auto- in innervation, interruption of feedback loops, and
nomic signaling by using novel pharmacological and the underlying disease state. This conundrum ex-
biological methods. Pokushalov et al. (113,114) re- tends to the prognostic significance of the heart
ported that injection of botulinum toxin in the GPs at ratebased measures. For example, although sym-
the time of open-heart surgery led to a decrease in the pathoexcitation has been considered to be an impor-
incidence of post-operative AF, as well as a decrease tant component in the pathogenesis of life-
of AF burden at 1 year of follow-up. The clinical threatening ventricular arrhythmias, a meta-analysis
usefulness of this approach was not established. In of predictors of sudden cardiac death and
animal studies, Aistrup et al. (115) showed that para- arrhythmic events in patients with nonischemic
sympathetic signaling in the atrium could be selec- dilated cardiomyopathy noted that HRV, BRS, or HRT,
tively disrupted by using G-protein inhibitory which are all tests that focus on autonomic effects on
peptides targeting the C-terminus of the G a i/o sub- the sinus node, were not significant predictors of
units, with the peptides injected either directly (115) ventricular arrhythmic events (117).
or as plasmid expression vectors to obtain constitu- Heart rate can be evaluated under resting and
tive administration of G a i2ctp and G a o1 ctp (116). In steady state, conditions in which parasympathetic
ongoing preclinical studies, the same group of in- effects normally predominate. This manifests as res-
vestigators is actively exploring the efficacy and piratory sinus arrhythmia, the “high-frequency” os-
duration of expression of genes targeting the ANS in cillations of the heart rate that are noted at the
canine models of chronic AF. respiratory frequency due to inspiratory suppression
of vagal nerve discharges. Next, the responsiveness of
ASSESSMENT OF CARDIAC the sinus node to small perturbations from the steady
AUTONOMIC FUNCTION state and resting hemodynamic status can be
assessed. Tests such as BRS and HRT evaluate the
Assessment of normal and abnormal autonomic heart rate response to an acute increase in blood
function is challenging because of the proliferation of pressure and premature ventricular beats, respec-
techniques to assess cardiac autonomic function. tively. Finally, larger perturbations in autonomic ac-
Furthermore, most of the techniques currently tivity can be provoked with exercise. Both the
available to assess autonomic function—whether acceleration of heart rate with exercise and the HRR
direct nerve recordings or indirect assessments of after cessation of exercise have been studied. The
autonomic activity by HRV, heart rate turbulence spectrum of evaluation from steady state to mild
(HRT), and so on—are difficult to use and reliably perturbations from steady state to the large changes
interpret in day-to-day clinical practice. Nonetheless, noted with exercise provides different assessments of
a brief review is presented to provide a historical autonomic function, likely related to the different
perspective on how autonomic testing in cardiovas- multilevel feedback loops that may be engaged in
cular disease has evolved over the years. This review each condition.
largely focuses on the most widely used heart HEART RATE VARIABILITY. HRV represents a mea-
ratebased tests (HRV, HRT, BRS, HRR, and QT-RR sure of the oscillation in the intervals between
slope), with a secondary emphasis on direct nerve consecutive heart beats. The most common methods
recording techniques. Less widely used imaging- for measuring the variation in heart rate can be
based techniques (sympathetic imaging with meta- broadly categorized into either time- or frequency-
iodobenzylguanidine, C-meta-hydroxyephedrine) domain analyses. Various nonlinear analyses have
will not be discussed here. There are many heart also been proposed. Time-domain measures of HRV,
ratebased tests, but the core of these involve eval- such as the SD of normal RR intervals, the root mean
uation of heart rate or heart rate changes. It is critical square of successive RR interval differences, and the
to note that these heart rate evaluations largely rely percentage of normal RR intervals that differ by
on how autonomic input affects the sinus node. >50 ms, are calculated based on statistical and/or
However, regional autonomic effects on the sinus mathematical operations on RR intervals (118).
1198 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

The low-frequency and very–low-frequency compo-


F I G U R E 3 Diagram of the QT-RR Relationship
nents of HRV have a more complex physiology that

390
integrates both sympathetic and parasympathetic
Baseline activities. More specifically, the absolute and
Atropine
370 Double normalized low-frequency powers are influenced by
blockade
sympathetic modulation of the heart rate (121). Up-
350
right tilt-table testing increases sympathetic tone
QT Interval (ms)

β-blockade
330 (39); the low frequency and the low-frequency/high-
frequency ratio (122) increase with upright tilt, and
310
beta-blockade blunts these changes. Although the
290 low-frequency/high-frequency ratio is often consid-
ered an index of sympathovagal balance (123,124), the
270 RR interval may actually be a more precise index of
the net effect of sympathetic and parasympathetic
400 450 500 550 600 650 700 750 effects on the sinus node (125) than the time- or
RR Interval (ms) frequency-domain HRV parameters. Fluctuations in
the renin-angiotensin-aldosterone system and varia-
The QT-RR relationship in the early post-exercise recovery period without autonomic tions in thermoregulatory mechanisms have been
blockade (baseline [orange]), with beta-blockade (green), with parasympathetic speculated to underlie the very–low-frequency
blockade (atropine [grey]), and double blockade with propranolol and atropine (blue).
component. In long-term recordings, the physiolog-
Original data from Sundaram et al. (160).
ical basis for ultralow frequency oscillations has not
been well defined (126).
Nonlinear analysis techniques have been applied
Frequency-domain measures use spectral analysis of to further characterize HRV (127,128). A variety of
a sequence of RR intervals and provide information analysis techniques and parameters have been used
on how power (variance) is distributed as a function to measure nonlinear properties of HRV. Analysis of
of frequency (39), using either short- (2 to 5 min) or 1/f characteristics (i.e., the inverse power–law slope),
long-term (24 h) recordings. which describes the slope of the spectral powers in
Respiratory sinus arrhythmia is a reflection of the ultra-low and very–low-frequency areas has pro-
the oscillatory parasympathetic effects on the vided prognostic information beyond the traditional
sinus node related to the respiratory cycle (119,120), HRV measures (129). None of these techniques have
which yields the high-frequency component of HRV. become widely used. Importantly, autonomic
blockade markedly attenuates all HRV, regardless of
the measure, providing further evidence of the
T A B L E 1 Common Cardiovascular Conditions Linked to the Autonomic Nervous System
complexity of the various different approaches to
Strength of assess autonomic effects on the sinus node.
Conditions Link Evidence
Diminished HRV has been associated with both
Myocardial infarction Imaging: sympathetic denervation þþþþ
Beta-blockers improve survival sudden cardiac death and nonsudden death in MI and
Congestive heart failure Imaging: diminished NE reuptake þþþþ in chronic left ventricular dysfunction, independent
Elevated plasma catecholamines
of the left ventricular ejection fraction (130–132).
Downregulation of beta-adrenergic receptors
Beta-blockers improve survival Large epidemiological studies have also demon-
Hypertension Sympathetic activation þþþ strated the prognostic significance of diminished
Atrial fibrillation Experimental models: changes in innervation; induction of þþþ HRV in the general population (133–135). At present,
AF by vagal stimulation, vagal stimulation plus
sympathetic activation however, measures of HRV by themselves do not
Clinical precipitants: vagal AF, sympathetic AF provide adequate refinement of risk of sudden car-
Long QT syndrome Beta-blockers prevent events þþþ
diac death due to ventricular tachyarrhythmias.
Neurocardiogenic Bezold-Jarisch reflex þþþþ
syncope Furthermore, the pathophysiological link between
Postural orthostatic Sympathetic activation þ reduced HRV and increased mortality is unclear.
tachycardia Although HRV provides a measure of autonomic
syndrome
Diabetic cardiac Abnormalities in autonomic function tests involving þþþþ
modulation, it has not entered the realm of clinical
autonomic parasympathetic and sympathetic reflexes evaluation even after many decades of study.
neuropathy
BAROREFLEX SENSITIVITY. BRS refers to the reflex

AF ¼ atrial fibrillation.
bradycardia that accompanies a transient increase in
systemic blood pressure, and, as such, reflects
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1199
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

C ENTR AL I LL U STRA T I O N Autonomic Nervous System Dysfunction

Goldberger, J.J. et al. J Am Coll Cardiol. 2019;73(10):1189–206.

The complex autonomic changes that may occur secondary to acute myocardial infarction. There is regional denervation in the ischemic zones. Nerve sprouting may
then appear at the border zone. Nerve growth factors and cytokines may also induce remodeling at the stellate ganglion. Afferent and efferent feedback loops to the
brainstem and higher brain centers may modulate the effects of these changes. Stimuli, such as heart failure, will enhance sympathoexcitation and act upon this altered
autonomic substrate. These effects may serve to further enhance heart failure or promote arrhythmias that may be responsible for sudden cardiac death.
DADs ¼ delayed afterdepolarizations; EADs ¼ early afterdepolarizations; NE ¼ norepinephrine; NGF ¼ nerve growth factor; VT/VF ¼ ventricular tachycardia/
ventricular fibrillation.

intrinsic properties of arterial baroreceptors, but is a result, the greater the slope of the regression line,
also attenuated by afferent cardiac sympathetic the stronger the baroreflex.
stimulation (136). BRS is typically measured after a BRS can also be determined by using neck
change in blood pressure has been provoked. This is chamber devices that help activate and/or deactivate
classically done by acute administration of phenyl- carotid baroreceptors by applying positive pressure
ephrine (137). From the simultaneous record of RR or suction to the neck (121). An increase in the
intervals and blood pressure, BRS is calculated as the pressure around the neck is perceived by the baro-
slope of the regression line (to assess the dependency receptors as a decrease in the arterial pressure,
of RR intervals on systolic blood pressure values). As whereas neck suction stimulates an increase in
1200 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

a transient drop in blood pressure that results in


T A B L E 2 Noninvasive Tests of Cardiac Autonomic Function
baroreceptor activation and immediate vagal inhibi-
Test Physiological Information Clinical Usefulness tion, and therefore, an increase in heart rate.
Heart rate Net autonomic effect on the sinus node þþ Augmented myocardial contractility following
Heart rate variability Autonomic modulation of sinus node —
a ventricular premature beat and the subsequent
Heart rate recovery Parasympathetic reactivation after —
cessation of exercise increase in blood pressure lead to an opposite reac-
Baroreflex sensitivity Sinus node response to baroreceptor — tion with a subsequent decrease in sinus node activ-
activation ity; thus, the biphasic HRT curve of acceleration and
Heart rate turbulence Sinus node response to hemodynamic —
perturbation by a PVC
deceleration is created (144).
Autonomic reflex testing Sinus node response to breathing — Several large population studies have shown that
(Ewing’s maneuvers) maneuvers, Valsalva, tilt, handgrip decreased (or abnormal) HRT identifies patients at
Sympathetic nerve recordings Quantify regional sympathetic output — high risk of mortality, including sudden death (147).
Plasma/urinary catecholamines/ Total body spillover to blood/urine þþþ
turnover rates (pheochromocytoma)
HRT is represented by 2 numeric descriptors (144):
Cardiac sympathetic imaging Sympathetic nerve distribution and — turbulence onset, which reflects the initial accelera-
function tion of heart rate after a ventricular premature beat;
and turbulence slope, which describes subsequent
PVC ¼ premature ventricular complex.
deceleration of heart rate following a ventricular
premature beat. La Rovere et al. (148) studied the
blood pressure. Neck suction is typically better relationship between HRT and BRS in 157 heart
tolerated and is therefore more commonly used. failure patients in whom Holter-derived HRT and
Using this method, BRS is typically quantified by phenylephrine-induced BRS were evaluated. Both
the maximum RR interval lengthening taken from turbulence onset and turbulence slope significantly
repeated applications. correlated with phenylephrine-derived BRS. These
Spontaneous fluctuations in arterial pressure and findings strongly support the concept that HRT is
RR interval may also be assessed to determine BRS. mediated by the baroreflex response. HRT was eval-
This is most often done by assessing the coherence of uated in 577 survivors of acute MI in the Multicenter
the power spectra of simultaneously recorded RR and Postinfarction Program and in 614 post-MI patients
blood pressure modulations. The coherence between randomized to the placebo arm in the EMIAT (Euro-
the power spectra is usually estimated in the low- pean Myocardial Infarction Amiodarone Trial).
frequency band and less often in the high-frequency Diminished HRT was a significant predictor of all-
band (121). cause mortality (143). In contrast, HRT was not asso-
Animal studies suggest that diminished BRS after ciated with a significant reduction in all-cause mor-
MI denotes an increased risk of ventricular fibrillation tality in the MADIT-II (Multicenter Automatic
(138). Depressed BRS assessed by the phenylephrine Defibrillator Implantation Trial II) (149).
method was found to be a significant predictor of HEART RATE RECOVERY. During exercise, there is a
5-year mortality in survivors of acute MI with pre- well-described increase in sympathetic activity and
served ventricular function (139). However, data in withdrawal of parasympathetic activity (150,151).
humans after MI are more conflicting (140,141). In the During recovery from exercise, there is an initially
Marburg Cardiomyopathy Study, BRS did not appear rapid and then gradual return of heart rate to its
to be helpful for arrhythmia risk stratification for previous resting level. The dynamic range of changes
patients with idiopathic cardiomyopathy (142). in sympathetic and parasympathetic activity related
HEART RATE TURBULENCE. In 1999, Schmidt et al. to exercise are greater than those assessed with HRV,
described HRT that was manifested by short-term BRS, or HRT. HRR after exercise reflects the sympa-
heart rate changes induced by a premature ventricu- thetic withdrawal and parasympathetic reactivation
lar beat (143). In healthy subjects, a ventricular that occurs. Although some earlier studies (152) sug-
premature beat provokes an early acceleration fol- gested that sympathetic withdrawal is the major
lowed by a late deceleration of heart rate, whereas in autonomic limb contributing to HRR soon after peak
subjects with cardiac dysfunction, such a reaction is exercise cessation (with parasympathetic activation
diminished or even completely nonexistent. Based contributing later in recovery), more recent studies
on data from experimental and clinical studies, indicated that parasympathetic reactivation was the
HRT is most likely mediated via the baroreceptor key factor in early HRR (153). Kannankeril et al. (154)
reflex; however, other mechanisms such as post- studied heart rate and HRR in healthy individuals
extrasystolic potentiation have been proposed (144– during peak exercise and recovery under normal
146). A premature ventricular ectopic beat results in physiological conditions, as well as during selective
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1201
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

parasympathetic blockade with atropine. They noted DIRECT NERVE RECORDINGS. S y m p a t h e t i c


that even during peak exercise, parasympathetic m i c r o n e u r o g r a p h y . Although heart ratebased
withdrawal was not complete. In recovery, para- tests are simple noninvasive tools, they do
sympathetic effects on heart rate appeared rapidly not directly measure autonomic activity. The
within the first minute, increased steadily until 4 min development of microneurography, in which nerve
into recovery, and then plateaued. activity can be recorded directly from intraneural
Another important factor in the analysis of HRR is microelectrodes inserted percutaneously in a
the parameter to use: heart rate or the RR interval. peripheral nerve in awake patients, has provided a
These variables are inversely related and cannot be wealth of information on the control of sympathetic
used interchangeably. In an analysis of heart rate outflow to muscle and skin. Recordings of muscle
and the RR interval after submaximal exercise in sympathetic nerve activity and skin sympathetic
33 healthy subjects at baseline conditions and during nerve activity in different disease states have
selective beta-adrenergic blockade and/or para- increased our understanding of sympathetic
sympathetic blockade (155), it was shown that the function, both in physiological and pathological
heart rate changes provided more physiological in- settings. Several studies have suggested that muscle
formation and should therefore be the preferred sympathetic nerve activity is a reliable marker
variable. of sympathetic response in some internal organs
Abnormal HRR is also associated with an adverse (161). Compared with healthy individuals, muscle
prognosis. Jouven et al. (156) reported a relative risk of sympathetic nerve activity is altered in the setting
2.2 for sudden cardiac death in those with of orthostatic hypotension and syncope, in
1-min HRR <25 beats/min versus those with HRR neurological disorders such as Parkinson’s disease,
>40 beats/min. Cole et al. (157) demonstrated a 4-fold multiple system atrophy, familial dysautonomia,
risk of death in those with abnormal HRR after peak Guillain-Barre syndrome, and in cardiovascular
exercise in a cohort of 2,428 subjects without a history disease states such as hypertension and heart failure
of heart failure, coronary revascularization, coronary (162). Increased muscle sympathetic nerve activity
angiography, or exercise testing. After adjustment for in patients with heart failure is associated with
age, sex, medications, perfusion defects on thallium reduced exercise capacity (163) and also helps
scintigraphy, standard cardiac risk factors, resting predict mortality in this patient population (164).
heart rate, change in heart rate with exercise, and Importantly, interventions, such as exercise training,
workload achieved, there was still a 2-fold risk of appear to significantly decrease sympathetic activity,
death in those with an abnormal HRR. as assessed by muscle sympathetic nerve activity,

QT-RR SLOPE. Ventricular repolarization can be as well as by noninvasive parameters such as HRV

assessed on the electrocardiogram (the QT interval) and HRR (165).

and is subject to autonomic effects. Because the QT Direct recordings from cardiac autonomic nerves.
interval is also strongly influenced by heart rate, Recently, direct recordings of autonomic nerve ac-
determining the independent autonomic effects on tivity have been made from the stellate ganglia,
the QT interval is challenging. A variety of approaches vagus nerve, and from the cardiac GPs. These studies,
have been proposed. Although rate correction for- in large animal models of AF, suggest that sympa-
mulas are widely used to adjust the QT interval for thetic and parasympathetic nerve discharges
the underlying heart rate, these formulas are inade- frequently precede the onset of atrial arrhythmias,
quate to assess autonomic effects on the QT interval. both in the setting of heart failure and in a model of
One approach, assessing the QT-RR slope, has been rapid atrial pacinginduced AF (166). Recent data
shown to provide reliable and reproducible delinea- suggest that it may be possible to record autonomic
tion of cardiac repolarization (158,159). The auto- nerve activity by electrodes implanted in the subcu-
nomic effects on the QT-RR relationship in the first taneous space of the left thorax, and that this sub-
5 min of recovery has been defined (160) with selec- cutaneous nerve activity correlates with the stellage
tive autonomic blockade. Figure 3 provides a diagram ganglia nerve activity (167). Of great interest is the
of the autonomic effects on the QT-RR relationship, possibility of making these types of recordings from
showing a strong parasympathetic effect because skin sympathetic activity in humans (168). Future
atropine steepens the slope dramatically, and a studies are needed to assess the applicability of these
smaller beta-adrenergic effect as propranolol blunts techniques in humans, to better understand the role
the slope. Thus, there are characteristic autonomic of the ANS in the genesis of atrial and ventricular
effects on the QT-RR slope. arrhythmias.
1202 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

SUMMARY inhibitory control. Because of the prominent role the


ANS plays in cardiac disease, other diagnostic ap-
The link between ANS dysfunction and various car- proaches are needed. Potentially useful approaches
diovascular disease states has been clearly estab- include direct nerve recordings and cardiac sympa-
lished (Table 1). The role of exercise and other thetic imaging.
conditions associated with sympathoexcitation as a Therapeutically, the key intervention that has
major precipitant for MI (169,170) and ventricular ar- demonstrated benefit is beta-blocker therapy. Only
rhythmias and/or sudden cardiac death (171,172), and limited data are available for other medical therapies,
the proven usefulness of beta-adrenergic blockers to such as scopolamine, which can improve HRV and
improve survival after MI and in heart failure with BRS (173) but may not affect survival (174). Other in-
reduced ejection fraction highlight the prominent terventions that have been tested include autonomic
role autonomics play in clinical heart disease (Central nerve stimulation and autonomic nerve disruption,
Illustration). with mixed results. It is possible that further efforts
The appreciation of the importance of autonomics to develop optimal approaches to interrogate this
led to the development of many noninvasive di- complex system will enable targeted stimulation and
agnostics that interrogate the ANS, but have limited disruption interventions that can be more specifically
to no clinical applicability at this time (Table 2). This targeted to abnormalities that can be ameliorated
may be due to the fact that the tests are focused on with these approaches.
autonomic effects on the sinus node, whereas the
adverse cardiac effects are generated by autonomic ADDRESS FOR CORRESPONDENCE: Dr. Jeffrey J.
effects on the ventricle. In addition, simple tests of Goldberger, University of Miami, Miller School of
autonomic reflexes may not adequately interrogate Medicine, Cardiovascular Division, 1120 NW 14th
the complex interplay of a multilevel system with Street, Miami, Florida 33136. E-mail: j-goldberger@
multiple levels of feedback and excitatory and miami.edu. Twitter: @DrJGoldberger, @UMiamiHealth.

REFERENCES

1. Vaseghi M, Barwad P, Malavassi Corrales FJ, 9. Irie T, Yamakawa K, Hamon D, Nakamura K, stellate ganglia. Am J Physiol Heart Circ Physiol
et al. Cardiac sympathetic denervation for re- Shivkumar K, Vaseghi M. Cardiac sympathetic 2013;305:H1020–30.
fractory ventricular arrhythmias. J Am Coll Cardiol innervation via middle cervical and stellate ganglia
17. Ulphani JS, Cain JH, Inderyas F, et al. Quanti-
2017;69:3070–80. and antiarrhythmic mechanism of bilateral stel-
tative analysis of parasympathetic innervation of
lectomy. Am J Physiol Heart Circ Physiol 2017;312:
2. Armour JA, Murphy DA, Yuan BX, Macdonald S, the porcine heart. Heart Rhythm 2010;7:1113–9.
H392–405.
Hopkins DA. Gross and microscopic anatomy of 18. Yamakawa K, Rajendran PS, Takamiya T, et al.
the human intrinsic cardiac nervous system. Anat 10. Buckley U, Yamakawa K, Takamiya T, Andrew
Vagal nerve stimulation activates vagal afferent
Rec 1997;247:289–98. Armour J, Shivkumar K, Ardell JL. Targeted stel-
fibers that reduce cardiac efferent para-
late decentralization: implications for sympathetic
3. Buckley U, Shivkumar K, Ardell JL. Autonomic sympathetic effects. Am J Physiol Heart Circ
control of ventricular electrophysiology. Heart
regulation therapy in heart failure. Curr Heart Fail Physiol 2015;309:H1579–90.
Rhythm 2016;13:282–8.
Rep 2015;12:284–93. 19. Yamakawa K, So EL, Rajendran PS, et al.
11. Ellison JP, Williams TH. Sympathetic nerve
4. Shivkumar K, Ajijola OA, Anand I, et al. Clinical pathways to the human heart, and their variations. Electrophysiological effects of right and left vagal
neurocardiology defining the value of Am J Anat 1969;124:149–62. nerve stimulation on the ventricular myocardium.
neuroscience-based cardiovascular therapeutics. Am J Physiol Heart Circ Physiol 2014;307:H722–31.
12. Mizeres NJ. The cardiac plexus in man. Am J
J Physiol 2016;594:3911–54. 20. Ardell JL, Rajendran PS, Nier HA,
Anat 1963;112:141–51.
5. Armour JA. Cardiac neuronal hierarchy in health KenKnight BH, Armour JA. Central-peripheral
13. Ajijola OA, Vaseghi M, Zhou W, et al. Func-
and disease. Am J Physiol Regul Integr Comp neural network interactions evoked by vagus
tional differences between junctional and extra-
Physiol 2004;287:R262–71. nerve stimulation: functional consequences on
junctional adrenergic receptor activation in
control of cardiac function. Am J Physiol Heart Circ
6. Hoover DB, Shepherd AV, Southerland EM, mammalian ventricle. Am J Physiol Heart Circ
Physiol 2015;309:H1740–52.
Armour JA, Ardell JL. Neurochemical diversity of Physiol 2013;304:H579–88.
afferent neurons that transduce sensory signals 21. Kember GC, Armour JA, Zamir M. Mechanism
14. Wang HJ, Wang W, Cornish KG, Rozanski GJ,
from dog ventricular myocardium. Auton Neurosci of smart baroreception in the aortic arch. Phys Rev
Zucker IH. Cardiac sympathetic afferent denerva-
2008;141:38–45. E Stat Nonlin Soft Matter Phys 2006;74:031914.
tion attenuates cardiac remodeling and improves
7. Randall DC, Brown DR, McGuirt AS, cardiovascular dysfunction in rats with heart fail- 22. Kember G, Ardell JL, Shivkumar K, Armour JA.
Thompson GW, Armour JA, Ardell JL. Interactions ure. Hypertension 2014;64:745–55. Recurrent myocardial infarction: mechanisms of
within the intrinsic cardiac nervous system 15. Vaseghi M, Zhou W, Shi J, et al. Sympathetic free-floating adaptation and autonomic derange-
contribute to chronotropic regulation. Am J Physiol innervation of the anterior left ventricular wall by ment in networked cardiac neural control. PLoS
Regul Integr Comp Physiol 2003;285:R1066–75. the right and left stellate ganglia. Heart Rhythm One 2017;12:e0180194.
2012;9:1303–9.
8. Csepe TA, Zhao J, Hansen BJ, et al. Human 23. Guo YP, McLeod JG, Baverstock J. Pathological
sinoatrial node structure: 3D microanatomy of 16. Vaseghi M, Yamakawa K, Sinha A, et al. Mod- changes in the vagus nerve in diabetes and chronic
sinoatrial conduction pathways. Prog Biophys Mol ulation of regional dispersion of repolarization and alcoholism. J Neurol Neurosurg Psychiatry 1987;
Biol 2016;120:164–78. T-peak to T-end interval by the right and left 50:1449–53.
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1203
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

24. Duchen LW, Anjorin A, Watkins PJ, Mackay JD. 40. Vinik AI, Ziegler D. Diabetic cardiovascular 56. Orchard TJ, Lloyd CE, Maser RE, Kuller LH.
Pathology of autonomic neuropathy in diabetes autonomic neuropathy. Circulation 2007;115: Why does diabetic autonomic neuropathy predict
mellitus. Ann Intern Med 1980;92:301–3. 387–97. IDDM mortality? An analysis from the Pittsburgh
Epidemiology of Diabetes Complications Study.
25. Kristensson K, Nordborg C, Olsson Y, 41. Spallone V, Menzinger G. Diagnosis of cardio-
Diabetes Res Clin Pract 1996;34 Suppl 1:S165–71.
Sourander P. Changes in the vagus nerve in dia- vascular autonomic neuropathy in diabetes. Dia-
betes mellitus. Acta Pathol Microbiol Scand A betes 1997;46 Suppl 2:S67–76. 57. Suarez GA, Clark VM, Norell JE, et al. Sudden
1971;79:684–5. cardiac death in diabetes mellitus: risk factors in
42. Ng F, Wong S, Cruz AL, Hernandez MI,
the Rochester Diabetic Neuropathy Study.
26. Schmidt RE. Neuropathology and pathogen- Gomis P, Passariello G. Heart rate recovery in the
J Neurol Neurosurg Psychiatry 2005;76:240–5.
esis of diabetic autonomic neuropathy. Int Rev diagnosis of diabetic cardiovascular autonomic
Neurobiol 2002;50:257–92. neuropathy. Comput Cardiol 2007:681–4. 58. Wackers FJ, Young LH, Inzucchi SE, et al.
43. Tesfaye S, Boulton AJ, Dyck PJ, et al. Diabetic Detection of silent myocardial ischemia in
27. Appenzeller O, Richardson EP Jr. The sympa-
neuropathies: update on definitions, diagnostic asymptomatic diabetic subjects: the DIAD study.
thetic chain in patients with diabetic and alcoholic
criteria, estimation of severity, and treatments. Diabetes Care 2004;27:1954–61.
polyneuropathy. Neurology 1966;16:1205–9.
Diabetes Care 2010;33:2285–93. 59. The Consensus Committee of the American
28. Vinik AI, Maser RE, Mitchell BD, Freeman R.
44. Ewing DJ, Martyn CN, Young RJ, Clarke BF. Autonomic Society, the American Academy of
Diabetic autonomic neuropathy. Diabetes Care
The value of cardiovascular autonomic function Neurology. Consensus statement on the definition
2003;26:1553–79.
tests: 10 years experience in diabetes. Diabetes of orthostatic hypotension, pure autonomic fail-
29. Cameron NE, Cotter MA. Metabolic and Care 1985;8:491–8. ure, and multiple system atrophy. Neurology
vascular factors in the pathogenesis of diabetic 1996;46:1470.
45. Mestivier D, Chau NP, Chanudet X,
neuropathy. Diabetes 1997;46 Suppl 2:S31–7.
Bauduceau B, Larroque P. Relationship between 60. Vernino S, Low PA, Fealey RD, Stewart JD,
30. Schonauer M, Thomas A, Morbach S, diabetic autonomic dysfunction and heart rate Farrugia G, Lennon VA. Autoantibodies to gangli-
Niebauer J, Schonauer U, Thiele H. Cardiac auto- variability assessed by recurrence plot. Am J onic acetylcholine receptors in autoimmune auto-
nomic diabetic neuropathy. Diab Vasc Dis Res Physiol 1997;272:H1094–9. nomic neuropathies. N Engl J Med 2000;343:
2008;5:336–44. 847–55.
46. Khandoker AH, Jelinek HF, Palaniswami M.
31. Pop-Busui R. Cardiac autonomic neuropathy in Identifying diabetic patients with cardiac auto- 61. Hague K, Lento P, Morgello S, Caro S,
diabetes: a clinical perspective. Diabetes Care nomic neuropathy by heart rate complexity anal- Kaufmann H. The distribution of Lewy bodies in
2010;33:434–41. ysis. Biomed Eng Online 2009;8:3. pure autonomic failure: autopsy findings and re-
view of the literature. Acta Neuropathol (Berl)
32. O’Brien IA, O’Hare JP, Lewin IG, Corrall RJ. The 47. American Diabetes Association. Standards of
1997;94:192–6.
prevalence of autonomic neuropathy in insulin- medical care in diabetes–2013. Diabetes Care
dependent diabetes mellitus: a controlled study 2013;36 Suppl 1:S11–66. 62. Hishikawa N, Hashizume Y, Hirayama M, et al.
based on heart rate variability. Q J Med 1986;61: Brainstem-type Lewy body disease presenting
48. Balcıog lu AS, Müderrisog
 lu H. Diabetes and
957–67. with progressive autonomic failure and lethargy.
cardiac autonomic neuropathy: clinical manifesta-
Clin Auton Res 2000;10:139–43.
33. Neil HA, Thompson AV, John S, McCarthy ST, tions, cardiovascular consequences, diagnosis and
Mann JI. Diabetic autonomic neuropathy: the treatment. World J Diabetes 2015;6:80–91. 63. Freeman R, Abuzinadah AR, Gibbons C,
prevalence of impaired heart rate variability in a Jones P, Miglis MG, Sinn DI. Orthostatic hypoten-
49. Maser RE, Mitchell BD, Vinik AI, Freeman R.
geographically defined population. Diabet Med sion: JACC state-of-the-art review. J Am Coll
The association between cardiovascular autonomic
1989;6:20–4. Cardiol 2018;72:1294–309.
neuropathy and mortality in individuals with dia-
34. Ziegler D, Gries FA, Spuler M, Lessmann F. The betes: a meta-analysis. Diabetes Care 2003;26: 64. Bryarly M, Phillips LT, Fu Q, Vernino S,
epidemiology of diabetic neuropathy. Diabetic 1895–901. Levine BD. Postural orthostatic tachycardia syn-
Cardiovascular Autonomic Neuropathy Multicenter drome: JACC focus seminar. J Am Coll Cardiol
50. Naas AA, Davidson NC, Thompson C, et al. QT
Study Group. J Diab Comp 1992;6:49–57. 2019;73:1207–28.
and QTc dispersion are accurate predictors of
35. Carnethon MR, Prineas RJ, Temprosa M, cardiac death in newly diagnosed non-insulin 65. Fukuda K, Kanazawa H, Aizawa Y, Ardell JL,
Zhang ZM, Uwaifo G, Molitch ME. The association dependent diabetes: cohort study. BMJ 1998; Shivkumar K. Cardiac innervation and sudden car-
among autonomic nervous system function, inci- 316:745–6. diac death. Circ Res 2015;116:2005–19.
dent diabetes, and intervention arm in the Dia- 51. Rossing P, Breum L, Major-Pedersen A, et al. 66. Inoue H, Zipes DP. Time course of denervation
betes Prevention Program. Diabetes Care 2006; Prolonged QTc interval predicts mortality in pa- of efferent sympathetic and vagal nerves after
29:914–9. tients with type 1 diabetes mellitus. Diabet Med occlusion of the coronary artery in the canine
36. Carnethon MR, Jacobs DR Jr., Sidney S, 2001;18:199–205. heart. Circ Res 1988;62:1111–20.
Liu K. Influence of autonomic nervous system 52. Sawicki PT, Dahne R, Bender R, Berger M. 67. Costigan M, Moss A, Latremoliere A, et al. T-
dysfunction on the development of type 2 Prolonged QT interval as a predictor of mortality cell infiltration and signaling in the adult dorsal
diabetes: the CARDIA study. Diabetes Care in diabetic nephropathy. Diabetologia 1996;39: spinal cord is a major contributor to neuropathic
2003;26:3035–41. 77–81. pain-like hypersensitivity. J Neurosci 2009;29:
37. Carnethon MR, Golden SH, Folsom AR, 14415–22.
53. Sawicki PT, Kiwitt S, Bender R, Berger M. The
Haskell W, Liao D. Prospective investigation of value of QT interval dispersion for identification of 68. Ajijola OA, Wisco JJ, Lambert HW, et al.
autonomic nervous system function and the total mortality risk in non-insulin-dependent dia- Extracardiac neural remodeling in humans with
development of type 2 diabetes: the Atheroscle- betes mellitus. J Intern Med 1998;243:49–56. cardiomyopathy. Circ Arrhythm Electrophysiol
rosis Risk In Communities study, 1987-1998. Cir- 2012;5:1010–116.
54. Veglio M, Sivieri R, Chinaglia A, Scaglione L,
culation 2003;107:2190–5.
Cavallo-Perin P. QT interval prolongation and 69. Nakamura K, Ajijola OA, Aliotta E, Armour JA,
38. Lucini D, Zuccotti G, Malacarne M, et al. Early mortality in type 1 diabetic patients: a 5-year Ardell JL, Shivkumar K. Pathological effects of
progression of the autonomic dysfunction cohort prospective study. Neuropathy Study chronic myocardial infarction on peripheral neu-
observed in pediatric type 1 diabetes mellitus. Group of the Italian Society of the Study of Dia- rons mediating cardiac neurotransmission. Auton
Hypertension 2009;54:987–94. betes, Piemonte Affiliate. Diabetes Care 2000;23: Neurosci 2016;197:34–40.
1381–3.
39. Lahiri MK, Kannankeril PJ, Goldberger JJ. 70. Li C-Y, Li Y-G. Cardiac sympathetic nerve
Assessment of autonomic function in cardiovas- 55. Ewing DJ, Campbell IW, Clarke BF. Mortality in sprouting and susceptibility to ventricular ar-
cular disease: physiological basis and prognostic diabetic autonomic neuropathy. Lancet 1976;1: rhythmias after myocardial infarction. Cardiol Res
implications. J Am Coll Cardiol 2008;51:1725–33. 601–3. Pract 2015;2015:698368.
1204 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

71. Ajijola OA, Yagishita D, Patel KJ, et al. Focal congestive heart failure. J Am Coll Cardiol 2007; 101. Pokushalov E, Romanov A, Shugayev P, et al.
myocardial infarction induces global remodeling 50:335–43. Selective ganglionated plexi ablation for parox-
of cardiac sympathetic innervation: neural ysmal atrial fibrillation. Heart Rhythm 2009;6:
87. Hou Y, Scherlag BJ, Lin J, et al. Ganglionated
remodeling in a spatial context. Am J Physiol 1257–64.
plexi modulate extrinsic cardiac autonomic nerve
Heart Circ Physiol 2013;305:H1031–40.
input: effects on sinus rate, atrioventricular con- 102. Scanavacca M, Pisani CF, Hachul D, et al.
72. Vaseghi M, Lux RL, Mahajan A, Shivkumar K. duction, refractoriness, and inducibility of atrial Selective atrial vagal denervation guided by
Sympathetic stimulation increases dispersion of fibrillation. J Am Coll Cardiol 2007;50:61–8. evoked vagal reflex to treat patients with parox-
repolarization in humans with myocardial infarc- ysmal atrial fibrillation. Circulation 2006;114:
88. Hou Y, Scherlag BJ, Lin J, et al. Interactive
tion. Am J Physiol Heart Circ Physiol 2012;302: 876–85.
atrial neural network: determining the connections
H1838–46.
between ganglionated plexi. Heart Rhythm 2007; 103. Pokushalov E, Romanov A, Artyomenko S,
73. Ajijola OA, Yagishita D, Reddy NK, et al. 4:56–63. et al. Ganglionated plexi ablation directed by
Remodeling of stellate ganglion neurons after high-frequency stimulation and complex frac-
89. Arora R, Ng J, Ulphani J, et al. Unique auto-
spatially targeted myocardial infarction: neuro- tionated atrial electrograms for paroxysmal atrial
nomic profile of the pulmonary veins and posterior
peptide and morphologic changes. Heart Rhythm fibrillation. Pacing Clin Electrophysiol 2012;35:
left atrium. J Am Coll Cardiol 2007;49:1340–8.
2015;12:1027–35. 776–84.
90. Arora R, Ulphani JS, Villuendas R, et al. Neural
74. Rajendran PS, Nakamura K, Ajijola OA, et al. 104. Katritsis DG, Giazitzoglou E, Zografos T,
substrate for atrial fibrillation: implications for
Myocardial infarction induces structural and Pokushalov E, Po SS, Camm AJ. Rapid pulmonary
targeted parasympathetic blockade in the poste-
functional remodelling of the intrinsic cardiac vein isolation combined with autonomic ganglia
rior left atrium. Am J Physiol 2008;294:H134–44.
nervous system. J Physiol 2016;594:321–41. modification: a randomized study. Heart Rhythm
91. Arora R. Recent insights into the role of the 2011;8:672–8.
75. Hou Y, Zhou Q, Po SS. Neuromodulation for
autonomic nervous system in the creation of
cardiac arrhythmia. Heart Rhythm 2016;13: 105. Zhou Q, Hou Y, Yang S. A meta-analysis of
substrate for atrial fibrillation: implications for
584–92. the comparative efficacy of ablation for atrial
therapies targeting the atrial autonomic nervous
76. Premchand RK, Sharma K, Mittal S, et al. system. Circ Arrhythm Electrophysiol 2012;5: fibrillation with and without ablation of the
Autonomic regulation therapy via left or right 850–9. ganglionated plexi. Pacing Clin Electrophysiol
cervical vagus nerve stimulation in patients with 2011;34:1687–94.
92. Chevalier P, Tabib A, Meyronnet D, et al.
chronic heart failure: results of the ANTHEM-HF 106. Nitta T, Ishii Y, Sakamoto S. Surgery for atrial
Quantitative study of nerves of the human left
trial. J Card Fail 2014;20:808–16. fibrillation: recent progress and future perspec-
atrium. Heart Rhythm 2005;2:518–22.
77. Tse HF, Turner S, Sanders P, et al. Thoracic tive. Gen Thorac Cardiovasc Surg 2012;60:13–20.
93. Arora R, Ulphani JS, Villuendas R, et al. Neural
Spinal Cord Stimulation for Heart Failure as a
substrate for atrial fibrillation: implications for 107. Bagge L, Blomstrom P, Nilsson L,
Restorative Treatment (SCS HEART study): first-
targeted parasympathetic blockade in the poste- Einarsson GM, Jideus L, Blomstrom-Lundqvist C.
in-man experience. Heart Rhythm 2015;12:
rior left atrium. Am J Physiol Heart Circ Physiol Epicardial off-pump pulmonary vein isolation and
588–95.
2008;294:H134–44. vagal denervation improve long-term outcome
78. Ng J, Villuendas R, Cokic I, et al. Autonomic and quality of life in patients with atrial fibrilla-
94. Tan AY, Li H, Wachsmann-Hogiu S, Chen LS,
remodeling in the left atrium and pulmonary veins tion. J Thorac Cardiovasc Surg 2009;137:1265–71.
Chen PS, Fishbein MC. Autonomic innervation and
in heart failure: creation of a dynamic substrate for
segmental muscular disconnections at the human 108. Krum H, Schlaich M, Whitbourn R, et al.
atrial fibrillation. Circ Arrhythm Electrophysiol
pulmonary vein-atrial junction: implications for Catheter-based renal sympathetic denervation for
2011;4:388–96.
catheter ablation of atrial-pulmonary vein junc- resistant hypertension: a multicentre safety and
79. Iwasaki YK, Nishida K, Kato T, Nattel S. Atrial tion. J Am Coll Cardiol 2006;48:132–43. proof-of-principle cohort study. Lancet 2009;373:
fibrillation pathophysiology: implications for 1275–81.
95. Deneke T, Chaar H, de Groot JR, et al. Shift in
management. Circulation 2011;124:2264–74.
the pattern of autonomic atrial innervation in 109. Hering D, Lambert EA, Marusic P, et al.
80. Shen MJ, Choi EK, Tan AY, et al. Neural subjects with persistent atrial fibrillation. Heart Substantial reduction in single sympathetic nerve
mechanisms of atrial arrhythmias. Nat Rev Cardiol Rhythm 2011;8:1357–63. firing after renal denervation in patients with
2011;9:30–9. resistant hypertension. Hypertension 2013;61:
96. Gould PA, Yii M, McLean C, et al. Evidence for
81. Amar D, Zhang H, Miodownik S, Kadish AH. 457–64.
increased atrial sympathetic innervation in persis-
Competing autonomic mechanisms precede the tent human atrial fibrillation. Pacing Clin Electro- 110. Linz D, van Hunnik A, Hohl M, et al. Catheter-
onset of postoperative atrial fibrillation. J Am Coll physiol 2006;29:821–9. based renal denervation reduces atrial nerve
Cardiol 2003;42:1262–8. sprouting and complexity of atrial fibrillation in
97. Edgerton JR, Brinkman WT, Weaver T, et al.
82. Patterson E, Po SS, Scherlag BJ, Lazzara R. goats. Circ Arrhythm Electrophysiol 2015;8:
Pulmonary vein isolation and autonomic denerva-
Triggered firing in pulmonary veins initiated by 466–74.
tion for the management of paroxysmal atrial
in vitro autonomic nerve stimulation.[see fibrillation by a minimally invasive surgical 111. Pokushalov E, Romanov A, Katritsis DG, et al.
comment]. Heart Rhythm 2005;2:624–31. approach. J Thorac Cardiovasc Surg 2010;140: Renal denervation for improving outcomes of
83. Tan AY, Zhou S, Ogawa M, et al. Neural 823–8. catheter ablation in patients with atrial fibrillation
mechanisms of paroxysmal atrial fibrillation and and hypertension: early experience. Heart Rhythm
98. Edgerton JR, Edgerton ZJ, Weaver T, et al.
paroxysmal atrial tachycardia in ambulatory ca- 2014;11:1131–8.
Minimally invasive pulmonary vein isolation and
nines. Circulation 2008;118:916–25. partial autonomic denervation for surgical treat- 112. Bhatt DL, Kandzari DE, O’Neill WW, et al.
84. Choi EK, Shen MJ, Han S, et al. Intrinsic cardiac ment of atrial fibrillation. Ann Thorac Surg 2008; A controlled trial of renal denervation for resistant
nerve activity and paroxysmal atrial tachyar- 86:35–8; discussion 39. hypertension. N Engl J Med 2014;370:1393–401.
rhythmia in ambulatory dogs. Circulation 2010; 113. Pokushalov E, Kozlov B, Romanov A, et al.
99. Pokushalov E, Romanov A, Artyomenko S,
121:2615–23. Long-term suppression of atrial fibrillation by
Turov A, Shirokova N, Katritsis DG. Left atrial
85. Ogawa M, Tan AY, Song J, et al. Cryoablation ablation at the anatomic areas of ganglionated botulinum toxin injection into epicardial fat pads in
of stellate ganglia and atrial arrhythmia in ambu- plexi for paroxysmal atrial fibrillation. Pacing Clin patients undergoing cardiac surgery: one-year
latory dogs with pacing-induced heart failure. Electrophysiol 2010;33:1231–8. follow-up of a randomized pilot study. Circ
Heart Rhythm 2009;6:1772–9. Arrhythm Electrophysiol 2015;8:1334–41.
100. Pokushalov E, Romanov A, Artyomenko S,
86. Ogawa M, Zhou S, Tan AY, et al. Left stellate et al. Ganglionated plexi ablation for longstanding 114. Pokushalov E, Kozlov B, Romanov A, et al.
ganglion and vagal nerve activity and cardiac ar- persistent atrial fibrillation. Europace 2010;12: Botulinum toxin injection in epicardial fat pads can
rhythmias in ambulatory dogs with pacing-induced 342–6. prevent recurrences of atrial fibrillation after
JACC VOL. 73, NO. 10, 2019 Goldberger et al. 1205
MARCH 19, 2019:1189–206 Autonomic Nervous System Dysfunction

cardiac surgery: results of a randomized pilot with increased mortality after acute myocardial heart rate turbulence. Am J Physiol Regul Integr
study. J Am Coll Cardiol 2014;64:628–9. infarction. Am J Cardiol 1987;59:256–62. Comp Physiol 2000;279:R1171–5.

115. Aistrup GL, Villuendas R, Ng J, et al. Targeted 131. La Rovere MT, Pinna GD, Maestri R, et al. 146. Davies LC, Francis DP, Ponikowski P,
G-protein inhibition as a novel approach to Short-term heart rate variability strongly predicts Piepoli MF, Coats AJ. Relation of heart rate and
decrease vagal atrial fibrillation by selective sudden cardiac death in chronic heart failure pa- blood pressure turbulence following premature
parasympathetic attenuation. Cardiovasc Res tients. Circulation 2003;107:565–70. ventricular complexes to baroreflex sensitivity in
2009;83:481–92. chronic congestive heart failure. Am J Cardiol
132. Deyell MW, Krahn AD, Goldberger JJ. Sudden
2001;87:737–42.
116. Aistrup GL, Cokic I, Ng J, et al. Targeted non- cardiac death risk stratification. Circ Res 2015;116:
viral gene-based inhibition of Gai/o-mediated 1907–18. 147. Bauer A, Malik M, Schmidt G, et al. Heart rate
vagal signaling in the posterior left atrium de- turbulence: standards of measurement, physio-
133. Dekker JM, Crow RS, Folsom AR, et al. Low
creases vagal induced AF. Heart Rhythm 2011: logical interpretation, and clinical use: Interna-
heart rate variability in a 2-minute rhythm strip
1722–9. tional Society for Holter and Noninvasive
predicts risk of coronary heart disease and mor-
Electrophysiology Consensus. J Am Coll Cardiol
117. Goldberger JJ, Subacius H, Patel T, tality from several causes: the ARIC Study.
2008;52:1353–65.
Cunnane R, Kadish AH. Sudden cardiac death risk Atherosclerosis Risk In Communities. Circulation
stratification in patients with nonischemic dilated 2000;102:1239–44. 148. La Rovere MT, Maestri R, Pinna GD, Sleight P,
cardiomyopathy. J Am Coll Cardiol 2014;63: Febo O. Clinical and haemodynamic correlates of
134. de Bruyne MC, Kors JA, Hoes AW, et al. Both
1879–89. heart rate turbulence as a non-invasive index of
decreased and increased heart rate variability on
baroreflex sensitivity in chronic heart failure. Clin
118. Kleiger RE, Stein PK, Bosner MS, Rottman JN. the standard 10-second electrocardiogram predict
Sci (Lond) 2011;121:279–84.
Time domain measurements of heart rate vari- cardiac mortality in the elderly: the Rotterdam
ability. Cardiol Clin 1992;10:487–98. Study. Am J Epidemiol 1999;150:1282–8. 149. Berkowitsch A, Zareba W, Neumann T, et al.
Risk stratification using heart rate turbulence and
119. Laude D, Weise F, Girard A, Elghozi JL. 135. Tsuji H, Larson MG, Venditti FJ Jr., et al.
ventricular arrhythmia in MADIT II: usefulness and
Spectral analysis of systolic blood pressure and Impact of reduced heart rate variability on risk for
limitations of a 10-minute Holter recording. Ann
heart rate oscillations related to respiration. Clin cardiac events. The Framingham Heart Study.
Noninvasive Electrocardiol 2004;9:270–9.
Exp Pharmacol Physiol 1995;22:352–7. Circulation 1996;94:2850–5.
150. Robinson BF, Epstein SE, Beiser GD,
120. Elghozi JL, Laude D, Girard A. Effects of 136. Gnecchi Ruscone T, Lombardi F, Malfatto G,
Braunwald E. Control of heart rate by the auto-
respiration on blood pressure and heart rate vari- Malliani A. Attenuation of baroreceptive mecha-
nomic nervous system. Studies in man on the
ability in humans. Clin Exp Pharmacol Physiol nisms by cardiovascular sympathetic afferent fi-
interrelation between baroreceptor mechanisms
1991;18:735–42. bers. Am J Physiol 1987;253:H787–91.
and exercise. Circ Res 1966;19:400–11.
121. Malik MSG. Autonomic testing and cardiac 137. Stein KM. Noninvasive risk stratification for
151. Ekblom B, Goldbarg AN, Kilbom A,
risk. In: Zipes DP, Jalife J, editors. Cardiac Elec- sudden death: signal-averaged electrocardiog- Astrand PO. Effects of atropine and propranolol on
trophysiology: From Cell to Bedside. 6th ed. raphy, nonsustained ventricular tachycardia, heart the oxygen transport system during exercise in
Philadelphia: Elsevier, 2014:649–56. rate variability, baroreflex sensitivity, and QRS man. Scand J Clin Lab Invest 1972;30:35–42.
122. Vybiral T, Bryg RJ, Maddens ME, Boden WE. duration. Prog Cardiovasc Dis 2008;51:106–17.
152. Savin WM, Davidson DM, Haskell WL. Auto-
Effect of passive tilt on sympathetic and para- 138. Billman GE, Schwartz PJ, Stone HL. Barore- nomic contribution to heart rate recovery from
sympathetic components of heart rate variability ceptor reflex control of heart rate: a predictor of exercise in humans. J Appl Physiol Respir Environ
in normal subjects. Am J Cardiol 1989;63:1117–20. sudden cardiac death. Circulation 1982;66: Exerc Physiol 1982;53:1572–5.
123. Pagani M, Lombardi F, Guzzetti S, et al. Power 874–80.
153. Imai K, Sato H, Hori M, et al. Vagally mediated
spectral analysis of heart rate and arterial pressure 139. De Ferrari GM, Sanzo A, Bertoletti A, heart rate recovery after exercise is accelerated in
variabilities as a marker of sympatho-vagal inter- Specchia G, Vanoli E, Schwartz PJ. Baroreflex athletes but blunted in patients with chronic heart
action in man and conscious dog. Circ Res 1986; sensitivity predicts long-term cardiovascular failure. J Am Coll Cardiol 1994;24:1529–35.
59:178–93. mortality after myocardial infarction even in pa-
154. Kannankeril PJ, Le FK, Kadish AH,
124. Chiou CW, Zipes DP. Selective vagal dener- tients with preserved left ventricular function.
Goldberger JJ. Parasympathetic effects on heart
vation of the atria eliminates heart rate variability J Am Coll Cardiol 2007;50:2285–90.
rate recovery after exercise. J Investig Med 2004;
and baroreflex sensitivity while preserving ven- 140. Huikuri HV, Tapanainen JM, Lindgren K, et al. 52:394–401.
tricular innervation. Circulation 1998;98:360–8. Prediction of sudden cardiac death after myocar-
155. Goldberger JJ, Johnson NP, Subacius H, Ng J,
125. Goldberger JJ. Sympathovagal balance: how dial infarction in the beta-blocking era. J Am Coll
Greenland P. Comparison of the physiologic and
should we measure it? Am J Physiol 1999;276: Cardiol 2003;42:652–8.
prognostic implications of the heart rate versus
H1273–80. 141. La Rovere MT, Pinna GD, Hohnloser SH, et al. the RR interval. Heart Rhythm 2014;11:1925–33.
126. Fox K, Borer JS, Camm AJ, et al. Resting heart Baroreflex sensitivity and heart rate variability in
156. Jouven X, Empana JP, Schwartz PJ,
rate in cardiovascular disease. J Am Coll Cardiol the identification of patients at risk for life-
Desnos M, Courbon D, Ducimetiere P. Heart-rate
2007;50:823–30. threatening arrhythmias: implications for clinical
profile during exercise as a predictor of sudden
trials. Circulation 2001;103:2072–7.
127. Huikuri HV, Stein PK. Heart rate variability in death. N Engl J Med 2005;352:1951–8.
risk stratification of cardiac patients. Prog Car- 142. Grimm W, Christ M, Bach J, Muller HH,
157. Cole CR, Blackstone EH, Pashkow FJ,
diovasc Dis 2013;56:153–9. Maisch B. Noninvasive arrhythmia risk stratifica-
Snader CE, Lauer MS. Heart-rate recovery imme-
tion in idiopathic dilated cardiomyopathy: results
128. Huikuri HV, Stein PK. Clinical application of diately after exercise as a predictor of mortality.
of the Marburg Cardiomyopathy Study. Circulation
heart rate variability after acute myocardial N Engl J Med 1999;341:1351–7.
2003;108:2883–91.
infarction. Front Physiol 2012;3:41.
158. Batchvarov VN, Ghuran A, Smetana P, et al.
143. Schmidt G, Malik M, Barthel P, et al. Heart-
129. Huikuri HV, Makikallio TH, Peng CK, QT-RR relationship in healthy subjects exhibits
rate turbulence after ventricular premature beats
Goldberger AL, Hintze U, Moller M. Fractal corre- substantial intersubject variability and high intra-
as a predictor of mortality after acute myocardial
lation properties of R-R interval dynamics and subject stability. Am J Physiol Heart Circ Physiol
infarction. Lancet 1999;353:1390–6.
mortality in patients with depressed left ventric- 2002;282:H2356–63.
ular function after an acute myocardial infarction. 144. Cygankiewicz I. Heart rate turbulence. Prog
159. Fenichel RR, Malik M, Antzelevitch C, et al.
Circulation 2000;101:47–53. Cardiovasc Dis 2013;56:160–71.
Drug-induced torsades de pointes and implications
130. Kleiger RE, Miller JP, Bigger JT Jr., Moss AJ. 145. Mrowka R, Persson PB, Theres H, Patzak A. for drug development. J Cardiovasc Electrophysiol
Decreased heart rate variability and its association Blunted arterial baroreflex causes “pathological” 2004;15:475–95.
1206 Goldberger et al. JACC VOL. 73, NO. 10, 2019

Autonomic Nervous System Dysfunction MARCH 19, 2019:1189–206

160. Sundaram S, Carnethon M, Polito K, atrial fibrillation: pathophysiology and therapy. death from cardiac causes by vigorous exertion.
Kadish AH, Goldberger JJ. Autonomic effects Circ Res 2014;114:1500–15. N Engl J Med 2000;343:1355–61.
on QT-RR interval dynamics after exercise. Am
167. Robinson EA, Rhee KS, Doytchinova A, 172. Lampert R, Joska T, Burg MM, Batsdord WP,
J Physiol Heart Circ Physiol 2008;294:
et al. Estimating sympathetic tone by recording McPherson CA, Jain D. Emotional and physical
H490–7.
subcutaneous nerve activity in ambulatory precipitants of ventricular arrhythmia. Circulation
161. Carter JR, Ray CA. Sympathetic neural adap- dogs. J Cardiovasc Electrophysiol 2015;26: 2002;106:1800–5.
tations to exercise training in humans. Auton 70–8. 173. De Ferrari GM, Mantica M, Vanoli E, Hull SS Jr.,
Neurosci 2015;188:36–43. Schwartz PJ. Scopolamine increases vagal tone and
168. Doytchinova A, Hassel JL, Yuan Y, et al.
162. Macefield VG. Sympathetic micro- vagal reflexes in patients after myocardial infarc-
Simultaneous noninvasive recording of skin sym-
neurography. Handb Clin Neurol 2013;117:353–64. tion. J Am Coll Cardiol 1993;22:1327–34.
pathetic nerve activity and electrocardiogram.
163. Notarius CF, Millar PJ, Floras JS. Muscle Heart Rhythm 2017;14:25–33. 174. Hull SS Jr., Vanoli E, Adamson PB, De
sympathetic activity in resting and exercising Ferrari GM, Foreman RD, Schwartz PJ. Do in-
169. Mittleman MA, Maclure M, Tofler GH,
humans with and without heart failure. Appl creases in markers of vagal activity imply protec-
Sherwood JB, Goldberg RJ, Muller JE. Trig-
Physiol Nutr Metab 2015;40:1107–15. tion from sudden death? The case of scopolamine.
gering of acute myocardial infarction by heavy
Circulation 1995;91:2516–9.
164. Barretto AC, Santos AC, Munhoz R, et al. physical exertion: protection against triggering
Increased muscle sympathetic nerve activity pre- by regular exertion. N Engl J Med 1993;329:
KEY WORDS arrhythmia, autonomic, heart
dicts mortality in heart failure patients. Int J Car- 1677–83.
failure, myocardial infarction
diol 2009;135:302–7.
170. Mittleman MA, Maclure M, Sherwood JB,
165. Pearson MJ, Smart NA. Exercise therapy and et al. Triggering of acute myocardial infarction
autonomic function in heart failure patients: a onset by episodes of anger. Determinants of Go to http://www.acc.org/
systematic review and meta-analysis. Heart Fail Myocardial Infarction Onset Study Investigators. jacc-journals-cme to take
Rev 2018;23:91–108. Circulation 1995;92:1720–5. the CME/MOC/ECME quiz
for this article.
166. Chen PS, Chen LS, Fishbein MC, Lin SF, 171. Albert CM, Mittleman MA, Chae CU, Lee IM,
Nattel S. Role of the autonomic nervous system in Hennekens CH, Manson JE. Triggering of sudden

You might also like