+ All Categories
Home > Documents > ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin...

ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin...

Date post: 04-Jun-2020
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
18
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, PHD c JACC JOURNAL CME/MOC/ECME This article has been selected as the months JACC CME/MOC/ECME activity, available online at http://www.acc.org/jacc-journals-cme by selecting the JACC Journals CME/MOC/ECME tab. Accreditation and Designation Statement The American College of Cardiology Foundation (ACCF) is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians. The ACCF designates this Journal-based CME activity for a maximum of 1 AMA PRA Category 1 Credit(s)Ô. Physicians should claim only the credit commensurate with the extent of their participation in the activity. Successful completion of this CME activity, which includes participa- tion in the evaluation component, enables the participant to earn up to 1 Medical Knowledge MOC point in the American Board of Internal Medicines (ABIM) Maintenance of Certication (MOC) program. Par- ticipants will earn MOC points equivalent to the amount of CME credits claimed for the activity. It is the CME activity providers responsibility to submit participant completion information to ACCME for the pur- pose of granting ABIM MOC credit. Autonomic Nervous System Dysfunction: JACC Focus Seminar will be accredited by the European Board for Accreditation in Cardiology (EBAC) for 1 hour of External CME credits. Each participant should claim only those hours of credit that have actually been spent in the educational activity. The Accreditation Council for Continuing Medical Education (ACCME) and the European Board for Accredita- tion in Cardiology (EBAC) have recognized each others accreditation systems as substantially equivalent. Apply for credit through the post-course evaluation. While offering the credits noted above, this program is not intended to provide extensive training or certication in the eld. Method of Participation and Receipt of CME/MOC/ECME Certicate To obtain credit for JACC CME/MOC/ECME, you must: 1. Be an ACC member or JACC subscriber. 2. Carefully read the CME/MOC/ECME-designated article available on- line and in this issue of the Journal. 3. Answer the post-test questions. A passing score of at least 70% must be achieved to obtain credit. 4. Complete a brief evaluation. 5. Claim your CME/MOC/ECME credit and receive your certicate electron- ically by following the instructions given at the conclusion of 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 to the heart and the types of autonomic dysfunction; 2) describe currently available techniques to assess autonomic function; and 3) describe the role of autonomic tone and/or dysfunction in cardiac arrhythmias. CME/MOC/ECME Editor Disclosure: JACC CME/MOC/ECME Editor Ragavendra R. Baliga, MD, FACC, has reported that he has no nancial relationships or interests to disclose. Author Disclosures: 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. Medium of Participation: Print (article only); online (article and quiz). CME/MOC/ECME Term of Approval Issue Date: March 19, 2019 Expiration Date: March 18, 2020 ISSN 0735-1097/$36.00 https://doi.org/10.1016/j.jacc.2018.12.064 From the a Cardiovascular Division, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida; b Feinberg Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Northwestern University-Feinberg School of Medicine, Chicago, Illinois; and the c Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, 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. Listen to this manuscripts audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10, 2019 ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION PUBLISHED BY ELSEVIER
Transcript
Page 1: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

Listen to this manuscript’s

audio summary by

Editor-in-Chief

Dr. Valentin Fuster on

JACC.org.

J O U R N A L O F T H E AM E R I C A N C O L L E G E O F C A R D I O L O G Y V O L . 7 3 , N O . 1 0 , 2 0 1 9

ª 2 0 1 9 B Y T H E AM E R I C A N C O L L E G E O F C A R D I O L O G Y F O UN DA T I O N

P U B L I S H E D B Y E L S E V I E R

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

activity, available online at http://www.acc.org/jacc-journals-cme by

selecting the JACC Journals CME/MOC/ECME tab.

Accreditation and Designation Statement

The American College of Cardiology Foundation (ACCF) is accredited by

the Accreditation Council for Continuing Medical Education to provide

continuing medical education for physicians.

The ACCF designates this Journal-based CME activity for a maximum

of 1 AMA PRA Category 1 Credit(s)�. Physicians should claim only the

credit commensurate with the extent of their participation in the

activity.

Successful completion of this CME activity, which includes participa-

tion in the evaluation component, enables the participant to earn up to

1 Medical Knowledge MOC point in the American Board of Internal

Medicine’s (ABIM) Maintenance of Certification (MOC) program. Par-

ticipants will earn MOC points equivalent to the amount of CME credits

claimed for the activity. It is the CME activity provider’s responsibility

to submit participant completion information to ACCME for the pur-

pose of granting ABIM MOC credit.

Autonomic Nervous System Dysfunction: JACC Focus Seminar will be

accredited by the European Board for Accreditation in Cardiology

(EBAC) for 1 hour of External CME credits. Each participant should

claim only those hours of credit that have actually been spent in the

educational activity. The Accreditation Council for Continuing

Medical Education (ACCME) and the European Board for Accredita-

tion in Cardiology (EBAC) have recognized each other’s accreditation

systems as substantially equivalent. Apply for credit through the

post-course evaluation. While offering the credits noted above, this

program is not intended to provide extensive training or certification

in the field.

ISSN 0735-1097/$36.00

From the aCardiovascular Division, Department of Medicine, University ofbFeinberg Cardiovascular Research Institute, Division of Cardiology, Departm

School of Medicine, Chicago, Illinois; and the cCardiac Arrhythmia Center a

University of California-Los Angeles Los Angeles, California. Dr. Goldeberger

Institute (NHLBI) (HL70179). Dr. Arora is supported by the NHLBI (HL09

(HL084261), by NHLBI grant R01HL084261, and by National Institutes o

ownership interest in Rhythm Therapeutics, Inc. All other authors have repo

contents of this paper to disclose.

Manuscript received April 16, 2018; revised manuscript received December 2

Method of Participation and Receipt of CME/MOC/ECME Certificate

To obtain credit for JACC CME/MOC/ECME, you must:

1. Be an ACC member or JACC subscriber.

2. Carefully read the CME/MOC/ECME-designated article available on-

line and in this issue of the Journal.

3. Answer the post-test questions. A passing score of at least 70%must be

achieved to obtain credit.

4. Complete a brief evaluation.

5. Claim your CME/MOC/ECME credit and receive your certificate electron-

ically by following the instructions given at the conclusion of the activity.

CME/MOC/ECME Objective for This Article: Uponcompletionof thisactivity,

the learner should be able to: 1) describe different levels of autonomic input

to the heart and the types of autonomic dysfunction; 2) describe currently

available techniques to assess autonomic function; and 3) describe the role

of autonomic tone and/or dysfunction in cardiac arrhythmias.

CME/MOC/ECME Editor Disclosure: JACC CME/MOC/ECME Editor

Ragavendra R. Baliga, MD, FACC, has reported that he has no financial

relationships or interests to disclose.

Author Disclosures: 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.

Medium of Participation: Print (article only); online (article and quiz).

CME/MOC/ECME Term of Approval

Issue Date: March 19, 2019

Expiration Date: March 18, 2020

https://doi.org/10.1016/j.jacc.2018.12.064

Miami Miller School of Medicine, Miami, Florida;

ent of Medicine, Northwestern University-Feinberg

nd Neurocardiology Research Center of Excellence,

is supported by the National Heart, Lung, and Blood

3490). Dr. Shivkumar is supported by the NHLBI

f Health grant NIHOT2OD023848. Dr. Arora holds

rted that they have no relationships relevant to the

1, 2018, accepted December 30, 2018.

Page 2: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1190

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 ofthe heart is a dynamic process in both healthand disease. ANS dysfunction may result

from primary disorders of the autonomic nerves orsecondarily in response to cardiac (or other systemic)disease. Cardiac disease may promote both anatomic(primary) and functional (secondary) changes in car-diac autonomic function. These changes may, inturn, contribute to the progression of disease and/orbe involved in arrhythmogenesis. Beta-adrenergicblockers are the most established autonomic inter-vention associated with improved outcomes. Otherinterventions (e.g., cardiac sympathetic denervation)have shown promise for the management of refrac-tory ventricular arrhythmias (1). Much has beenlearned about the complex interactions along theneuroaxis and their role in cardiac control. What hasbeen most challenging is the development of a simplemethod of assessment of the autonomic effects on theheart and/or autonomic dysfunction. Consequently,there have been a plethora of methods that assesssome aspect of autonomic function. Although manymeasures have been shown to have some prognosticsignificance, none have been adapted or adoptedinto clinical practice. This review highlights some ofthe background and newer concepts in autonomiccontrol of the heart.

NORMAL AUTONOMIC FUNCTION

A brief description of the advances in our under-standing of the physiology of cardiac autonomiccontrol is in order to place autonomic testing and

emerging therapies in context. Autonomic control ofthe heart is achieved by afferent neural impulses thatare transmitted from the heart to the intrinsic neu-rons of the heart, to extracardiac intrathoracic ganglia(e.g., stellate ganglion), to the spinal cord, and to thebrain stem. These afferent neural signals are pro-cessed by various parts of the nervous system toregulate the cardiomotor neural output to the heartvia the sympathetic and parasympathetic nerves. It isimportant to emphasize that the anatomical nervetrunks that reach the heart, which are traditionallydescribed as the sympathetic and parasympathetictrunks, have both afferent and efferent nerve fibers.

ORGANIZATION OF CARDIAC NEURAL CONTROL.

This neuroaxis is organized as multiple levels ofintegrative centers. At the level of the heart, theintrinsic cardiac nervous system (ICNS) is a distrib-uted network system located in the cardiac gangliathat are ganglionated plexi (GPs) that exist in the fatpads around the heart, predominantly in the posteriorand superior aspects of the atria (2). These connectwith the intrathoracic extracardiac ganglia (the sym-pathetic paravertebral ganglia), the extrathoraciccardiac ganglia (the nodose, dorsal root ganglia), andthe central nervous system (3,4). At each level, thesystem has the ability to modulate cardiac activitywith efferent feedback loops (Figure 1).

Afferent neural signals are transmitted to the ICNS,the stellate ganglia, and via the dorsal root ganglia tothe spinal cord and to the nodose ganglia (Figure 2)(5,6). The efferent projections to the heart occurvia short and long feedback loops that occur at

Page 3: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

AB BR E V I A T I O N S

AND ACRONYM S

AF = atrial fibrillation

ANS = autonomic nervous

system

BRS = baroreflex sensitivity

GP = ganglionated plexus

HRR = heart rate recovery

HRT = heart rate turbulence

HRV = heart rate variability

ICNS = intrinsic cardiac

nervous system

MI = myocardial infarction

PV = pulmonary vein

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1191

the central nervous system, the spinal cord (inter-mediolateral columns), the intrathoracic ganglia, andthe ICNS. Sensory neurites associated with the ICNSare found in areas of the atrioventricular junction andthe adventitia of major vessels (e.g., coronary ar-teries). It was previously believed that the differentlevels within the ANS were just relay stations, but it isnow well-established that each level processes neuralinformation and coordinates a response by commu-nicating with other levels by these feedback loops(Figure 2).

The ICNS consists of ganglia composed of afferent,efferent, and interconnecting neurons to other car-diac ganglia. These ganglia coordinate the sympa-thetic and parasympathetic inputs received from therest of the cardiac ANS. The ICNS has regional controlover different cardiac functions, such as sinus nodeelectrical activation and propagation, as well asatrioventricular nodal conduction (7,8). Emergingtherapies that target these structures have shown theimportance of emerging research in this area.Sympathet ic af ferents and efferents . The sym-pathetic neurons that regulate cardiac functionare located in the stellate ganglion. It receivespre-ganglionic sympathetic input from the inter-mediolateral column (and other spinal neurons) andcoordinates efferent neural responses either directlyor via the ansa subclavia to the middle cervicalganglia to the heart (9–12). Efferent post-ganglionicfibers travel alongside the coronary vasculature topenetrate the epicardial regions and travel toward theendocardium. Stimulation of the stellate ganglionresults in an increase in dromotropy, chronotropy,lusitropy, and inotropy (13). Cardiac afferent neuronsare mechanosensory, chemosensory, or multimodalin nature (4). They transduce a variety of chemicals,including various neuropeptides, such as substanceP, bradykinin, and calcitonin gene�related peptide.These cardiac afferents are also involved in initiatinglocal inflammatory and vascular reactions that mayplay an important role in cardiac remodeling (14).

The sympathetic nerve fibers are located in theatria and the ventricles. There is a regional responseto the right sympathetic paravertebral ganglia versusthe left sympathetic paravertebral ganglia, withpredominant effects on the anterior and posteriorventricular walls, respectively (15). In addition toincreased sinoatrial node firing and enhancement ofatrioventricular nodal conduction, sympatheticoutput results in ventricular action potential durationshortening (16).Parasympathet i c a f ferents and efferents . Theparasympathetic nervous system also has importantafferent and efferent components (Figures 1 and 2).

The cardiomotor function of this systemhelps slow the heart rate, reduce blood pres-sure, and balances the system to ensure thereis a counterbalance to sympathoexcitation.The parasympathetic effects are coordinatedvia the cervical vagus nerve, which dividesinto the superior and inferior cardiac nervesto finally enter the heart via the cardiacplexus. The parasympathetic nerve fibers aremuch more heterogeneously distributed,with significant innervation of the sinoatrialnode, atrioventricular node (7), and the ven-tricles (17).

The parasympathetic cardiomotor responseis coordinated by neurons in the ICNS that

receive pre-ganglionic parasympathetic input fromthe cervical vagus and provide a homogeneous or co-ordinated response to the atria and ventricles (18,19).Cervical vagus stimulation can produce different re-sponses depending on what aspect of the neuroaxis isengaged. In the intact state, stimulation can resultin both direct and reactive, or reflex initiated re-sponses on the intrinsic cardiac nervous system as aresult of central and peripheral interactions initiatedthroughout the cardiac neuronal hierarchy. Low-levelstimulation delivered to the cervical vagus can resultin tachycardia, probably as a result of engaging cardiacafferent fibers, whereas higher level stimulation re-sults in bradycardia (20). In the resting state, the car-diac ANS is an intricate balance between sympatheticand parasympathetic inputs. Once the cervical vagusis transected from the central nervous system, there isa significant increase in heart rate that suggests thatthe central cholinergic neuronal drive plays a consid-erable role in controlling the basal heart rate (20).Integrat ion of card iac and vascu la r a fferents .Another important afferent control system includesthe baroreceptors, which are stretch receptorsembedded in the adventitia of the aortic wall andcarotid sinus that transduce pressure fluctuations tothe cardiovascular neural reflex pathways responsiblefor heart rate and blood pressure. There is a contin-uous strip of mechanosensory neurites along the in-ner aortic arch (21). They are believed to transducecombined mechanosensory information that mea-sures distortion from pulsatile waves in the aorta inspace and time. The arterial chemoreceptors are inthe carotid arteries, aortic bodies, and medulla, andrespond to hypoxemia and hypercapnia, respectively.The aortic arch mechanoreceptors can be found in thenodose ganglion, whereas some of the carotid sinusafferents are found in the petrosal ganglia. These re-ceptors then transmit information to the nucleus ofthe tractus solitarius, which modulates sympathetic
Page 4: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

FIGURE 1 Autonomic Neural Control of the Heart

Autonomic neural control of the heart. Modified with permission from Shivkumar et al. (4). DRG¼ dorsal root ganglion; ICNS¼ intrinsic nervous system; SG¼ stellate ganglion.

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1192

Page 5: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

FIGURE 2 Functional Organization of Cardiac Neural Control

Medulla

NodoseAff, soma

Spinal cord

Aortic arterialbaroreceptors

Aorticchemoreceptors

Carotid bodychemoreceptors

Carotid sinusbaroreceptors

DRGAff, soma

Afferentsoma

LCN

Kidneys

Muscle

T6-L2

C1-L5

SympathEfferent soma

SympathEfferent soma

ParasymEfferent soma

Petrosalganglia

C1-C2

T1-T4

LCN

Cortex

Heart

Neurite

Afferent soma

Afferent

Neurite NeuriteNeurite M2

Gs Gi

β1

Brainstem

Extracardiacintrathoracicganglia (stellate,middle cervical)

Intrinsic cardiacnervous system

EfferentPreganglionic

‘Parasympathetic’‘Sympathetic’

Postganglionic

Higher centers

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.

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1193

and parasympathetic output to the cardiovascularsystem.

Finally, the regulation of the cardiac ANS is alsounder central nervous system control. The balance ofactivation and inhibitory neurons is finely tuned andis designed to provide dynamic control of the heartunder a range of physiological conditions rangingfrom rest to severe exertion. This exquisite multilevelneural network is profoundly altered in the presenceof cardiac dysfunction and leads to progression ofheart disease (22).

AUTONOMIC DYSFUNCTION AND ITS ROLE IN

CARDIOVASCULAR DISEASE. Autonomic dysfunc-tion may arise from 2 mechanisms—intrinsic orextrinsic. Intrinsic autonomic dysfunction arises from

diseases that directly affect the autonomic nerves,such as diabetes mellitus and the various syndromesof primary autonomic failure. Extrinsic autonomicdysfunction reflects the changes in autonomic func-tion that are secondarily induced by cardiac or otherdisease. Cardiac diseases, such as myocardial infarc-tion (MI), can also primarily disrupt the autonomicnerves.

PRIMARY AUTONOMIC DYSFUNCTION. Pr imaryautonomic dysfunct ion resu l t ing from diabet i cneuropathy . Diabetes mellitus is associated withthe development of both peripheral and autonomicneuropathy. Because of its widespread prevalence,diabetes is the leading cause of primary autonomicdysfunction. Pathological studies have noted loss of

Page 6: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1194

and/or damage to myelinated vagus nerve axons (23–25). Although findings in the sympathetic nerves andganglia have been disputed (26), they include giantnerve cells, vacuolization, and neuroaxonal dystrophy(24,26,27). Appenzeller and Richardson (27) noted ab-normalities in 4 of 5 patients with diabetes with neu-ropathy and in 0 of 4 patients without neuropathy.These studies showed significant pathological changesin the autonomic nerves of patients with diabetes whopredominantly had advanced disease and peripheralneuropathy. The pathological basis of early diabeticautonomic neuropathy has not been elucidated.Numerous biochemical mechanisms for hyperglyce-mic injury have been implicated, including the pro-duction of advanced glycosylation end products,hyperglycemic activation of the polyol pathway, aswell as protein kinase C, immunological processes,and neurovascular insufficiency that leads to localischemia. The proposed mechanisms come largelyfrom research on somatic nerves in experimental dia-betic animal models. Although biochemically diverse,the various degenerative mechanisms have a commonpredisposition for distal axon terminals. The patho-genesis of diabetic cardiac autonomic neuropathyin vivo is likely to be heterogeneous and results fromthe interaction of multiple pathways (26,28–30).Because the vagus nerve is the longest autonomicnerve in the body, abnormalities in parasympatheticinnervation of the heart are typically the earliestmanifestation of cardiac autonomic neuropathy (31).

The prevalence of cardiac autonomic neuropathy inpatients with diabetes varies widely depending on thecohort studied and the tests and/or criteria used forassessment, but approximately 15% to 20% ofasymptomatic people with diabetes appear to haveabnormal cardiovascular autonomic function (28,32–34).Only later stages of the disease are associated withsymptoms. Autonomic abnormalities may even bedetected before the onset of diabetes (35–37). Earlyevidence of cardiac autonomic neuropathy can bedetected in children with type 1 diabetes (38). Manymethods are used to diagnose cardiac autonomicneuropathy. Because the heart rate is easily measuredand responds to autonomic stimuli (39), noninvasivestudies to assess cardiac autonomic neuropathy focusprimarily on heart rate or heart rate responses tophysiological manipulations. Cardiac autonomicneuropathy has been diagnosed based on abnormal-ities in heart rate variability (HRV), baroreflex sensi-tivity (BRS), Ewing’s tests (heart rate and/or bloodpressure responses to deep breathing, standing, Val-salva maneuver, and handgrip), and heart rate recov-ery (HRR) (40–44). These parameters are not highlycorrelated with each other (45,46), which presents a

unique challenge in the diagnostic approach to thisentity. Although cardiac autonomic reflex tests(Ewing’s tests) are generally recommended for diag-nosis (47), it is unclear which method, if any, ispreferred. It is also unclear how often any assessmentfor cardiac autonomic neuropathy is performed.

The presence of cardiac autonomic neuropathyconfers an adverse prognosis (48). In a meta-analysisof 15 studies among subjects with diabetes mellitus(49), the pooled relative risk for mortality related tocardiac autonomic neuropathy was 3.45 (95% confi-dence interval: 2.66 to 4.47; p < 0.001). The mecha-nism for the increased mortality has not beenclarified, but various possibilities have been pro-posed, including QT interval prolongation (50–54),renal disease (55–57), and asymptomatic myocardialischemia (28,56,58). Some of these potential mecha-nisms may arise directly from the primary distur-bance of cardiac autonomic function.Other primary disorders of autonomic dysfunction. Otherprimary disorders of autonomic dysfunction and/orfailure include a range of disorders—pure autonomicfailure, idiopathic orthostatic hypotension, Parkin-son’s disease with autonomic failure, and multiplesystem atrophy (59). These disorders are character-ized by orthostatic hypotension in combination withother manifestations. Postural orthostatic tachy-cardia syndrome is a complex syndrome with a majorautonomic component. An autoimmune etiology thattargets ganglionic receptors (nicotinic acetylcholinereceptors) may be the underlying etiology for some ofthese disorders (60). Extensive pathological studieshave not been reported, but in autonomic failure,Lewy bodies (abnormal protein aggregates) havebeen identified in the sympathetic ganglia (61,62).These disorders are discussed in further detail inother studies (63,64).

AUTONOMIC DYSFUNCTION SECONDARY TO MI,

CARDIOMYOPATHY, AND HEART FAILURE, AND ITS ROLE

IN THE GENESIS OF VENTRICULAR ARRHYTHMIAS. Incontrast to disorders that primarily affect the auto-nomic nerves, a variety of cardiac pathologies, suchas MI, heart failure, and cardiomyopathy result insecondary acute and chronic changes within the ANS(65). Direct ischemic damage to cardiac autonomicnerves may result from acute MI (66). In addition, thecardiac ANS responds acutely to preserve homeosta-sis. This may involve increased sympathetic anddecreased parasympathetic activity to maintain car-diac contractility and cardiac output in the setting ofa major insult. Hemodynamic changes that occur as aconsequence of these conditions cause a cascadeof changes in neurohumoral activity in the

Page 7: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1195

cardiovascular, peripheral vascular, and renalvascular systems. The problem arises when the im-mediate threat has abated, but the imbalance in theANS, driven by altered afferent signaling, persists andis further modulated by cytokines generated as aresponse to the disease state (65). This can lead to avicious cycle, with MI, cardiomyopathy, and heartfailure leading to an outpouring of catecholamines(and inflammatory cytokines), and the catechol-amines, in turn, lead to worsening of cardiomyopa-thy. This cycle of adrenergic activation and cardiacdysfunction has been well established in congestiveheart failure with reduced ejection fraction. Themarked benefit of beta-adrenergic blockers in this en-tity, which improve both ejection fraction and out-comes, is a testament to the critical role of thesecondary changes in the ANS in this disease state andreversibility of congestive heart failure with treatment.

Persistent maladaptations within the ANS as aresult of cardiac dysfunction have been linked toatrial and ventricular arrhythmias (4). These changesresult in a functional reorganization of the ANS andalterations in neural processing at each level of thecardiac neural hierarchy, which leads to a conflictbetween the central (central nervous system) andperipheral (intrathoracic cardiac ganglia) control.Cardiac dysfunction can cause short- and long-termchanges in the neural networks, which results inexaggerated reflex responses. Central sensitizationof neurons can occur secondarily to constantafferent signaling from the site of injury, such as theinfarct zone, which can elicit a cellular processthat changes the excitability of cell membranesand reduces the inhibitory mechanisms within thenervous system (67).

Morphological changes seen in the stellate gan-glion secondarily to ischemic and nonischemic car-diomyopathy include neuronal enlargement (68), aswell as alterations in neurochemical expression pat-terns in the stellate (69). Other changes such as nervesprouting around the border zones or periphery of anischemic territory also occur (70). Regional adrenergicoverexpression with functional denervation may beseen in the border zone after MI in animal models (71)and humans with ischemic cardiomyopathy (72).Clinically, it is well appreciated that excess adren-ergic activity is a negative prognostic indicator post-MI and in congestive cardiac failure. This forms thebasis for the success of beta-blocker therapy in theseconditions.

There is neuronal enlargement within the stellateganglion in both the right and left stellate ganglion,regardless of the territory of the MI (73). This is incontrast to the ICNS, where there are regional

changes as a result of MI (74). The afferent ICneuronal activity is attenuated in the territory of theMI, whereas those IC neurons in remote regions arepreserved. As such, this creates a heterogenousneural response within the myocardium (74). Thissuggests that the ICNS receives and coordinates theinformation that is transmitted to the intrathoracicand extrathoracic ganglia. In relation to otherganglionic changes seen as a result of MI, the dorsalroot ganglia appear not to change morphologically,but they do have a significant change in neurochem-ical properties of the neurons present (increasedneuronal nitric oxide synthase and calcitonin gene-related peptide) (69). Alterations in neurohumoralcontrol, circulating catecholamines, and the renin-angiotensin-aldosterone system also occur as aresult of cardiac injury.

Ventricular arrhythmias that occur in the setting ofischemic and nonischemic cardiomyopathy may beprecipitated as a result of sympathetic overactivity.These occur as a result of an excessive sympatho-neurohumoral response and a reduced para-sympathetic input that results in increased dispersionof electrical activation and repolarization in the ven-tricles from the endocardium to epicardium. In recentyears, neuromodulation, with the goal of increasingparasympathetic tone and suppressing sympathetictone, has become an emerging therapeutic strategyfor the treatment of ventricular arrhythmias.Emerging therapeutic approaches include left cardiacsympathetic denervation, cervical vagal stimulation,and spinal cord stimulation (75). Removal of sympa-thetic efferent input to the heart by surgical resectionor thoracic epidural anesthesia of the stellate gan-glion to T4 can significantly reduce ventricular ar-rhythmias in the setting of a severe life-threateningventricular electrical storm. Although animal studieshave clearly shown that vagal nerve stimulation cansuppress ventricular arrhythmias in heart failure,vagal stimulation has only been used in patients withheart failure in an attempt to improve cardiacremodeling and heart failure (76,75). Similarly, spinalcord stimulation has been shown to be improvesymptoms, functional status, and left ventricularfunction and remodeling in patients with severesymptomatic heart failure (77); however, spinal cordstimulation has not been systematically evaluatedfor its antiarrhythmic benefits in clinical studies.

Despite the potential success of neuromodulationin pre-clinical or clinical studies in preventing ven-tricular arrhythmias, it is important to remember thatcardiac dysfunction results in adverse adaptions ofthe afferent and efferent inputs at various levelsthroughout the cardiac neuroaxis. These adaptions

Page 8: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1196

may perpetuate cardiac dysfunction and are generallypro-arrhythmic. Further in-depth mechanistic un-derstanding is required to improve the sophisticationof these therapies.ROLE OF AUTONOMIC DYSFUNCTION IN CREATING A

VULNERABLE SUBSTRATE FOR ATRIAL FIBRILLATION.

Atrial fibrillation (AF) is the most common sustainedarrhythmia disturbance and is associated with sig-nificant morbidity and mortality. The morbidity andmortality associated with AF are especially increasedin the setting of congestive heart failure, with up toone-half of all patients with heart failure havingconcomitant AF (78). Several mechanisms contributeto the electrophysiological and structural substratefor AF, including fibrosis, stretch, oxidative stress,and altered calcium handling characteristics (79). TheANS has been hypothesized to have a likely role increation of a vulnerable AF substrate (80), with bothsympathetic and parasympathetic nerves believed tobe involved in the genesis of AF (81,82). Armour et al.(2) demonstrated an intricate pattern of autonomicinnervation in the heart with the atria being inner-vated by at least 5 major atrial fad pads or GPs (80).Direct nerve recordings from the stellate ganglia,vagus nerve, and the cardiac GPs also demonstrateda dynamic interaction between the sympatheticand parasympathetic nervous system in creating AF(83–86). Hou et al. (87,88) suggested the presence ofan intricate, interconnecting neural network in theleft atrium that could contribute to a substrate for AF.

Over the last several years, the pulmonary veins(PVs) and adjoining posterior left atrium have beenshown to play a significant role in the genesis of AF,with this region demonstrating unique structural,molecular, and electrophysiological characteristicsthat appear to contribute to the AF substrate.Anatomical and physiological studies of the auto-nomic innervation of the atria indicate that the pul-monary veins and posterior left atria have a uniqueautonomic profile (86,89–91). Chevalier et al. (92)described several gradients of innervation in andaround the PVs (92). In a canine study, Arora et al.(93) showed that the posterior left atrium was morerichly innervated than the rest of the left atrium, withnerve bundles containing both parasympathetic andsympathetic fibers, with parasympathetic fibers pre-dominating over sympathetic fibers. These caninestudies were in agreement with human studies thatdemonstrated co-localization of sympathetic andparasympathetic nerve fibers in the human leftatrium (80,94). Deneke et al. (95) also demonstratedco-localization of sympathetic and parasympatheticnerves, with patients in persistent AF that demon-strated a shift toward a lower density of cholinergic

nerves and a higher density of adrenergic nerves.There was evidence of sympathetic hyperinnervationin patients with persistent AF (96).

Using direct nerve recordings from the stellateganglia and the vagus nerve in a canine study, Ogawaet al. (86) showed increased sympathetic and vagalnerve discharge before the onset of atrial arrhythmiasin pacing-induced heart failure [these atrial arrhyth-mias were preventable by prophylactic ablation of thestellate ganglion and the T2 to T4 thoracic sympa-thetic ganglia (85)]. In a canine HF model, Ng et al.(78) demonstrated a profound increase in para-sympathetic—and to a lesser extent sympathetic—nerves in the left atrium (78), with nerve growth be-ing most pronounced in the PVs and posterior leftatrium. The increase in vagal innervation noted inthis model was believed to contribute to the AF sub-strate by affecting conduction characteristics of thePVs and posterior left atrium.

Taken together, the previous studies indicated animportant role for the ANS in the genesis of AF notonly in normal hearts, but also in structural heartdisease. These data underscore the potential impor-tance of the ANS as a suitable therapeutic target inAF. A variety of strategies targeting $1 GPs eithersurgically (97,98) or through a transvenous, endo-cardial approach (alone or together with PV isolation)have been used in patients with both paroxysmal andpersistent AF with variable success (99–101). Scana-vacca et al. (102) demonstrated the feasibility of se-lective atrial vagal denervation, as guided by evokedvagal reflexes, to treat patients with paroxysmal AF.Pokushalov et al. (103) reported that regional ablationat the anatomic sites of the left atrial GP could besafely performed and enabled maintenance of sinusrhythm in 71% of patients with paroxysmal AF. Kar-itsis et al (104) and others (105) demonstrated thatwhen GP ablation was combined with PV isolation, ityielded better results than PV isolation alone, withsuccess rates approaching 80% (104). Recent surgicalstudies also attempted to add GP ablation and/orexcision to PV isolation with varying efficacy(97,98,106,107). The efficacy and durability of thisapproach has not been established.

Renal denervation, which is known to lead to areduction of renal norepinephrine spillover (108) anda reduction in firing of single sympathetic vasocon-strictor fibers (a measure of central sympathetic nerveoutflow) (109), has been shown to reduce atrial sym-pathetic nerve sprouting, structural alterations,and AF complexity in goats with persistent AF,independent of changes in blood pressure (110).Early stage clinical data suggests that renal denerva-tion may improve the results of pulmonary vein

Page 9: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1197

isolation in patients with persistent AF and/or severeresistant hypertension (111). However, the SymplicityHTN-3 trial did not demonstrate efficacy of renaldenervation in reducing blood pressure in patientswith resistant hypertension (112). Its role as apotential therapeutic tool in AF has not beenestablished.

Recent approaches attempted to disrupt auto-nomic signaling by using novel pharmacological andbiological methods. Pokushalov et al. (113,114) re-ported that injection of botulinum toxin in the GPs atthe time of open-heart surgery led to a decrease in theincidence of post-operative AF, as well as a decreaseof AF burden at 1 year of follow-up. The clinicalusefulness of this approach was not established. Inanimal studies, Aistrup et al. (115) showed that para-sympathetic signaling in the atrium could be selec-tively disrupted by using G-protein inhibitorypeptides targeting the C-terminus of the Gai/o sub-units, with the peptides injected either directly (115)or as plasmid expression vectors to obtain constitu-tive administration of Gai2ctp and Gao1ctp (116). Inongoing preclinical studies, the same group of in-vestigators is actively exploring the efficacy andduration of expression of genes targeting the ANS incanine models of chronic AF.

ASSESSMENT OF CARDIAC

AUTONOMIC FUNCTION

Assessment of normal and abnormal autonomicfunction is challenging because of the proliferation oftechniques to assess cardiac autonomic function.Furthermore, most of the techniques currentlyavailable to assess autonomic function—whetherdirect nerve recordings or indirect assessments ofautonomic activity by HRV, heart rate turbulence(HRT), and so on—are difficult to use and reliablyinterpret in day-to-day clinical practice. Nonetheless,a brief review is presented to provide a historicalperspective on how autonomic testing in cardiovas-cular disease has evolved over the years. This reviewlargely focuses on the most widely used heartrate�based tests (HRV, HRT, BRS, HRR, and QT-RRslope), with a secondary emphasis on direct nerverecording techniques. Less widely used imaging-based techniques (sympathetic imaging with meta-iodobenzylguanidine, C-meta-hydroxyephedrine)will not be discussed here. There are many heartrate�based tests, but the core of these involve eval-uation of heart rate or heart rate changes. It is criticalto note that these heart rate evaluations largely relyon how autonomic input affects the sinus node.However, regional autonomic effects on the sinus

node, atrium, atrioventricular node, and ventriclesdiffer in normal subjects and may be even moreaccentuated in the presence of cardiac disease withregional abnormalities in innervation and ganglionicfunction. How closely abnormalities in sinus nodeautonomic function parallel abnormalities in otherareas of the heart may depend on regional differencesin innervation, interruption of feedback loops, andthe underlying disease state. This conundrum ex-tends to the prognostic significance of the heartrate�based measures. For example, although sym-pathoexcitation has been considered to be an impor-tant component in the pathogenesis of life-threatening ventricular arrhythmias, a meta-analysisof predictors of sudden cardiac death andarrhythmic events in patients with nonischemicdilated cardiomyopathy noted that HRV, BRS, or HRT,which are all tests that focus on autonomic effects onthe sinus node, were not significant predictors ofventricular arrhythmic events (117).

Heart rate can be evaluated under resting andsteady state, conditions in which parasympatheticeffects normally predominate. This manifests as res-piratory sinus arrhythmia, the “high-frequency” os-cillations of the heart rate that are noted at therespiratory frequency due to inspiratory suppressionof vagal nerve discharges. Next, the responsiveness ofthe sinus node to small perturbations from the steadystate and resting hemodynamic status can beassessed. Tests such as BRS and HRT evaluate theheart rate response to an acute increase in bloodpressure and premature ventricular beats, respec-tively. Finally, larger perturbations in autonomic ac-tivity can be provoked with exercise. Both theacceleration of heart rate with exercise and the HRRafter cessation of exercise have been studied. Thespectrum of evaluation from steady state to mildperturbations from steady state to the large changesnoted with exercise provides different assessments ofautonomic function, likely related to the differentmultilevel feedback loops that may be engaged ineach condition.HEART RATE VARIABILITY. HRV represents a mea-sure of the oscillation in the intervals betweenconsecutive heart beats. The most common methodsfor measuring the variation in heart rate can bebroadly categorized into either time- or frequency-domain analyses. Various nonlinear analyses havealso been proposed. Time-domain measures of HRV,such as the SD of normal RR intervals, the root meansquare of successive RR interval differences, and thepercentage of normal RR intervals that differ by>50 ms, are calculated based on statistical and/ormathematical operations on RR intervals (118).

Page 10: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

FIGURE 3 Diagram of the QT-RR Relationship

RR Interval (ms)

370

390

350

270

290

330

310

400 450 500 550 600 650 700 750

β-blockade

Doubleblockade

BaselineAtropine

QT In

terv

al (m

s)

The QT-RR relationship in the early post-exercise recovery period without autonomic

blockade (baseline [orange]), with beta-blockade (green), with parasympathetic

blockade (atropine [grey]), and double blockade with propranolol and atropine (blue).

Original data from Sundaram et al. (160).

TABLE 1 Common Ca

Conditions

Myocardial infarction

Congestive heart failur

Hypertension

Atrial fibrillation

Long QT syndrome

Neurocardiogenicsyncope

Postural orthostatictachycardiasyndrome

Diabetic cardiacautonomicneuropathy

AF ¼ atrial fibrillation.

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1198

Frequency-domain measures use spectral analysis ofa sequence of RR intervals and provide informationon how power (variance) is distributed as a functionof frequency (39), using either short- (2 to 5 min) orlong-term (24 h) recordings.

Respiratory sinus arrhythmia is a reflection ofthe oscillatory parasympathetic effects on thesinus node related to the respiratory cycle (119,120),which yields the high-frequency component of HRV.

rdiovascular Conditions Linked to the Autonomic Nervous System

LinkStrength ofEvidence

Imaging: sympathetic denervationBeta-blockers improve survival

þþþþ

e Imaging: diminished NE reuptakeElevated plasma catecholaminesDownregulation of beta-adrenergic receptorsBeta-blockers improve survival

þþþþ

Sympathetic activation þþþExperimental models: changes in innervation; induction of

AF by vagal stimulation, vagal stimulation plussympathetic activation

Clinical precipitants: vagal AF, sympathetic AF

þþþ

Beta-blockers prevent events þþþBezold-Jarisch reflex þþþþ

Sympathetic activation þ

Abnormalities in autonomic function tests involvingparasympathetic and sympathetic reflexes

þþþþ

The low-frequency and very–low-frequency compo-nents of HRV have a more complex physiology thatintegrates both sympathetic and parasympatheticactivities. More specifically, the absolute andnormalized low-frequency powers are influenced bysympathetic modulation of the heart rate (121). Up-right tilt-table testing increases sympathetic tone(39); the low frequency and the low-frequency/high-frequency ratio (122) increase with upright tilt, andbeta-blockade blunts these changes. Although thelow-frequency/high-frequency ratio is often consid-ered an index of sympathovagal balance (123,124), theRR interval may actually be a more precise index ofthe net effect of sympathetic and parasympatheticeffects on the sinus node (125) than the time- orfrequency-domain HRV parameters. Fluctuations inthe renin-angiotensin-aldosterone system and varia-tions in thermoregulatory mechanisms have beenspeculated to underlie the very–low-frequencycomponent. In long-term recordings, the physiolog-ical basis for ultralow frequency oscillations has notbeen well defined (126).

Nonlinear analysis techniques have been appliedto further characterize HRV (127,128). A variety ofanalysis techniques and parameters have been usedto measure nonlinear properties of HRV. Analysis of1/f characteristics (i.e., the inverse power–law slope),which describes the slope of the spectral powers inthe ultra-low and very–low-frequency areas has pro-vided prognostic information beyond the traditionalHRV measures (129). None of these techniques havebecome widely used. Importantly, autonomicblockade markedly attenuates all HRV, regardless ofthe measure, providing further evidence of thecomplexity of the various different approaches toassess autonomic effects on the sinus node.

Diminished HRV has been associated with bothsudden cardiac death and nonsudden death in MI andin chronic left ventricular dysfunction, independentof the left ventricular ejection fraction (130–132).Large epidemiological studies have also demon-strated the prognostic significance of diminishedHRV in the general population (133–135). At present,however, measures of HRV by themselves do notprovide adequate refinement of risk of sudden car-diac death due to ventricular tachyarrhythmias.Furthermore, the pathophysiological link betweenreduced HRV and increased mortality is unclear.Although HRV provides a measure of autonomicmodulation, it has not entered the realm of clinicalevaluation even after many decades of study.BAROREFLEX SENSITIVITY. BRS refers to the reflexbradycardia that accompanies a transient increase insystemic blood pressure, and, as such, reflects

Page 11: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

CENTRAL ILLUSTRATION 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.

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1199

intrinsic properties of arterial baroreceptors, but isalso attenuated by afferent cardiac sympatheticstimulation (136). BRS is typically measured after achange in blood pressure has been provoked. This isclassically done by acute administration of phenyl-ephrine (137). From the simultaneous record of RRintervals and blood pressure, BRS is calculated as theslope of the regression line (to assess the dependencyof RR intervals on systolic blood pressure values). As

a result, the greater the slope of the regression line,the stronger the baroreflex.

BRS can also be determined by using neckchamber devices that help activate and/or deactivatecarotid baroreceptors by applying positive pressureor suction to the neck (121). An increase in thepressure around the neck is perceived by the baro-receptors as a decrease in the arterial pressure,whereas neck suction stimulates an increase in

Page 12: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

TABLE 2 Noninvasive Tests of Cardiac Autonomic Function

Test Physiological Information Clinical Usefulness

Heart rate Net autonomic effect on the sinus node þþHeart rate variability Autonomic modulation of sinus node —

Heart rate recovery Parasympathetic reactivation aftercessation of exercise

Baroreflex sensitivity Sinus node response to baroreceptoractivation

Heart rate turbulence Sinus node response to hemodynamicperturbation by a PVC

Autonomic reflex testing(Ewing’s maneuvers)

Sinus node response to breathingmaneuvers, Valsalva, tilt, handgrip

Sympathetic nerve recordings Quantify regional sympathetic output —

Plasma/urinary catecholamines/turnover rates

Total body spillover to blood/urine þþþ(pheochromocytoma)

Cardiac sympathetic imaging Sympathetic nerve distribution andfunction

PVC ¼ premature ventricular complex.

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1200

blood pressure. Neck suction is typically bettertolerated and is therefore more commonly used.Using this method, BRS is typically quantified bythe maximum RR interval lengthening taken fromrepeated applications.

Spontaneous fluctuations in arterial pressure andRR interval may also be assessed to determine BRS.This is most often done by assessing the coherence ofthe power spectra of simultaneously recorded RR andblood pressure modulations. The coherence betweenthe power spectra is usually estimated in the low-frequency band and less often in the high-frequencyband (121).

Animal studies suggest that diminished BRS afterMI denotes an increased risk of ventricular fibrillation(138). Depressed BRS assessed by the phenylephrinemethod was found to be a significant predictor of5-year mortality in survivors of acute MI with pre-served ventricular function (139). However, data inhumans after MI are more conflicting (140,141). In theMarburg Cardiomyopathy Study, BRS did not appearto be helpful for arrhythmia risk stratification forpatients with idiopathic cardiomyopathy (142).

HEART RATE TURBULENCE. In 1999, Schmidt et al.described HRT that was manifested by short-termheart rate changes induced by a premature ventricu-lar beat (143). In healthy subjects, a ventricularpremature beat provokes an early acceleration fol-lowed by a late deceleration of heart rate, whereas insubjects with cardiac dysfunction, such a reaction isdiminished or even completely nonexistent. Basedon data from experimental and clinical studies,HRT is most likely mediated via the baroreceptorreflex; however, other mechanisms such as post-extrasystolic potentiation have been proposed (144–146). A premature ventricular ectopic beat results in

a transient drop in blood pressure that results inbaroreceptor activation and immediate vagal inhibi-tion, and therefore, an increase in heart rate.Augmented myocardial contractility followinga ventricular premature beat and the subsequentincrease in blood pressure lead to an opposite reac-tion with a subsequent decrease in sinus node activ-ity; thus, the biphasic HRT curve of acceleration anddeceleration is created (144).

Several large population studies have shown thatdecreased (or abnormal) HRT identifies patients athigh risk of mortality, including sudden death (147).HRT is represented by 2 numeric descriptors (144):turbulence onset, which reflects the initial accelera-tion of heart rate after a ventricular premature beat;and turbulence slope, which describes subsequentdeceleration of heart rate following a ventricularpremature beat. La Rovere et al. (148) studied therelationship between HRT and BRS in 157 heartfailure patients in whom Holter-derived HRT andphenylephrine-induced BRS were evaluated. Bothturbulence onset and turbulence slope significantlycorrelated with phenylephrine-derived BRS. Thesefindings strongly support the concept that HRT ismediated by the baroreflex response. HRT was eval-uated in 577 survivors of acute MI in the MulticenterPostinfarction Program and in 614 post-MI patientsrandomized to the placebo arm in the EMIAT (Euro-pean Myocardial Infarction Amiodarone Trial).Diminished HRT was a significant predictor of all-cause mortality (143). In contrast, HRT was not asso-ciated with a significant reduction in all-cause mor-tality in the MADIT-II (Multicenter AutomaticDefibrillator Implantation Trial II) (149).HEART RATE RECOVERY. During exercise, there is awell-described increase in sympathetic activity andwithdrawal of parasympathetic activity (150,151).During recovery from exercise, there is an initiallyrapid and then gradual return of heart rate to itsprevious resting level. The dynamic range of changesin sympathetic and parasympathetic activity relatedto exercise are greater than those assessed with HRV,BRS, or HRT. HRR after exercise reflects the sympa-thetic withdrawal and parasympathetic reactivationthat occurs. Although some earlier studies (152) sug-gested that sympathetic withdrawal is the majorautonomic limb contributing to HRR soon after peakexercise cessation (with parasympathetic activationcontributing later in recovery), more recent studiesindicated that parasympathetic reactivation was thekey factor in early HRR (153). Kannankeril et al. (154)studied heart rate and HRR in healthy individualsduring peak exercise and recovery under normalphysiological conditions, as well as during selective

Page 13: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1201

parasympathetic blockade with atropine. They notedthat even during peak exercise, parasympatheticwithdrawal was not complete. In recovery, para-sympathetic effects on heart rate appeared rapidlywithin the first minute, increased steadily until 4 mininto recovery, and then plateaued.

Another important factor in the analysis of HRR isthe parameter to use: heart rate or the RR interval.These variables are inversely related and cannot beused interchangeably. In an analysis of heart rateand the RR interval after submaximal exercise in33 healthy subjects at baseline conditions and duringselective beta-adrenergic blockade and/or para-sympathetic blockade (155), it was shown that theheart rate changes provided more physiological in-formation and should therefore be the preferredvariable.

Abnormal HRR is also associated with an adverseprognosis. Jouven et al. (156) reported a relative risk of2.2 for sudden cardiac death in those with1-min HRR <25 beats/min versus those with HRR>40 beats/min. Cole et al. (157) demonstrated a 4-foldrisk of death in those with abnormal HRR after peakexercise in a cohort of 2,428 subjects without a historyof heart failure, coronary revascularization, coronaryangiography, or exercise testing. After adjustment forage, sex, medications, perfusion defects on thalliumscintigraphy, standard cardiac risk factors, restingheart rate, change in heart rate with exercise, andworkload achieved, there was still a 2-fold risk ofdeath in those with an abnormal HRR.

QT-RR SLOPE. Ventricular repolarization can beassessed on the electrocardiogram (the QT interval)and is subject to autonomic effects. Because the QTinterval is also strongly influenced by heart rate,determining the independent autonomic effects onthe QT interval is challenging. A variety of approacheshave been proposed. Although rate correction for-mulas are widely used to adjust the QT interval forthe underlying heart rate, these formulas are inade-quate to assess autonomic effects on the QT interval.One approach, assessing the QT-RR slope, has beenshown to provide reliable and reproducible delinea-tion of cardiac repolarization (158,159). The auto-nomic effects on the QT-RR relationship in the first5 min of recovery has been defined (160) with selec-tive autonomic blockade. Figure 3 provides a diagramof the autonomic effects on the QT-RR relationship,showing a strong parasympathetic effect becauseatropine steepens the slope dramatically, and asmaller beta-adrenergic effect as propranolol bluntsthe slope. Thus, there are characteristic autonomiceffects on the QT-RR slope.

DIRECT NERVE RECORDINGS. Sympathet i cmicroneurography . Although heart rate�basedtests are simple noninvasive tools, they donot directly measure autonomic activity. Thedevelopment of microneurography, in which nerveactivity can be recorded directly from intraneuralmicroelectrodes inserted percutaneously in aperipheral nerve in awake patients, has provided awealth of information on the control of sympatheticoutflow to muscle and skin. Recordings of musclesympathetic nerve activity and skin sympatheticnerve activity in different disease states haveincreased our understanding of sympatheticfunction, both in physiological and pathologicalsettings. Several studies have suggested that musclesympathetic nerve activity is a reliable markerof sympathetic response in some internal organs(161). Compared with healthy individuals, musclesympathetic nerve activity is altered in the settingof orthostatic hypotension and syncope, inneurological disorders such as Parkinson’s disease,multiple system atrophy, familial dysautonomia,Guillain-Barre syndrome, and in cardiovasculardisease states such as hypertension and heart failure(162). Increased muscle sympathetic nerve activityin patients with heart failure is associated withreduced exercise capacity (163) and also helpspredict mortality in this patient population (164).Importantly, interventions, such as exercise training,appear to significantly decrease sympathetic activity,as assessed by muscle sympathetic nerve activity,as well as by noninvasive parameters such as HRVand HRR (165).

Direct recordings from cardiac autonomic nerves.Recently, direct recordings of autonomic nerve ac-tivity have been made from the stellate ganglia,vagus nerve, and from the cardiac GPs. These studies,in large animal models of AF, suggest that sympa-thetic and parasympathetic nerve dischargesfrequently precede the onset of atrial arrhythmias,both in the setting of heart failure and in a model ofrapid atrial pacing�induced AF (166). Recent datasuggest that it may be possible to record autonomicnerve activity by electrodes implanted in the subcu-taneous space of the left thorax, and that this sub-cutaneous nerve activity correlates with the stellageganglia nerve activity (167). Of great interest is thepossibility of making these types of recordings fromskin sympathetic activity in humans (168). Futurestudies are needed to assess the applicability of thesetechniques in humans, to better understand the roleof the ANS in the genesis of atrial and ventriculararrhythmias.

Page 14: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1202

SUMMARY

The link between ANS dysfunction and various car-diovascular disease states has been clearly estab-lished (Table 1). The role of exercise and otherconditions associated with sympathoexcitation as amajor precipitant for MI (169,170) and ventricular ar-rhythmias and/or sudden cardiac death (171,172), andthe proven usefulness of beta-adrenergic blockers toimprove survival after MI and in heart failure withreduced ejection fraction highlight the prominentrole autonomics play in clinical heart disease (CentralIllustration).

The appreciation of the importance of autonomicsled to the development of many noninvasive di-agnostics that interrogate the ANS, but have limitedto no clinical applicability at this time (Table 2). Thismay be due to the fact that the tests are focused onautonomic effects on the sinus node, whereas theadverse cardiac effects are generated by autonomiceffects on the ventricle. In addition, simple tests ofautonomic reflexes may not adequately interrogatethe complex interplay of a multilevel system withmultiple levels of feedback and excitatory and

inhibitory control. Because of the prominent role theANS plays in cardiac disease, other diagnostic ap-proaches are needed. Potentially useful approachesinclude direct nerve recordings and cardiac sympa-thetic imaging.

Therapeutically, the key intervention that hasdemonstrated benefit is beta-blocker therapy. Onlylimited data are available for other medical therapies,such as scopolamine, which can improve HRV andBRS (173) but may not affect survival (174). Other in-terventions that have been tested include autonomicnerve stimulation and autonomic nerve disruption,with mixed results. It is possible that further effortsto develop optimal approaches to interrogate thiscomplex system will enable targeted stimulation anddisruption interventions that can be more specificallytargeted to abnormalities that can be amelioratedwith these approaches.

ADDRESS FOR CORRESPONDENCE: Dr. Jeffrey J.Goldberger, University of Miami, Miller School ofMedicine, Cardiovascular Division, 1120 NW 14thStreet, Miami, Florida 33136. E-mail: [email protected]. Twitter: @DrJGoldberger, @UMiamiHealth.

RE F E RENCE S

1. Vaseghi M, Barwad P, Malavassi Corrales FJ,et al. Cardiac sympathetic denervation for re-fractory ventricular arrhythmias. J Am Coll Cardiol2017;69:3070–80.

2. Armour JA, Murphy DA, Yuan BX, Macdonald S,Hopkins DA. Gross and microscopic anatomy ofthe human intrinsic cardiac nervous system. AnatRec 1997;247:289–98.

3. Buckley U, Shivkumar K, Ardell JL. Autonomicregulation therapy in heart failure. Curr Heart FailRep 2015;12:284–93.

4. Shivkumar K, Ajijola OA, Anand I, et al. Clinicalneurocardiology defining the value ofneuroscience-based cardiovascular therapeutics.J Physiol 2016;594:3911–54.

5. Armour JA. Cardiac neuronal hierarchy in healthand disease. Am J Physiol Regul Integr CompPhysiol 2004;287:R262–71.

6. Hoover DB, Shepherd AV, Southerland EM,Armour JA, Ardell JL. Neurochemical diversity ofafferent neurons that transduce sensory signalsfrom dog ventricular myocardium. Auton Neurosci2008;141:38–45.

7. Randall DC, Brown DR, McGuirt AS,Thompson GW, Armour JA, Ardell JL. Interactionswithin the intrinsic cardiac nervous systemcontribute to chronotropic regulation. Am J PhysiolRegul Integr Comp Physiol 2003;285:R1066–75.

8. Csepe TA, Zhao J, Hansen BJ, et al. Humansinoatrial node structure: 3D microanatomy ofsinoatrial conduction pathways. Prog Biophys MolBiol 2016;120:164–78.

9. Irie T, Yamakawa K, Hamon D, Nakamura K,Shivkumar K, Vaseghi M. Cardiac sympatheticinnervation via middle cervical and stellate gangliaand antiarrhythmic mechanism of bilateral stel-lectomy. Am J Physiol Heart Circ Physiol 2017;312:H392–405.

10. Buckley U, Yamakawa K, Takamiya T, AndrewArmour J, Shivkumar K, Ardell JL. Targeted stel-late decentralization: implications for sympatheticcontrol of ventricular electrophysiology. HeartRhythm 2016;13:282–8.

11. Ellison JP, Williams TH. Sympathetic nervepathways to the human heart, and their variations.Am J Anat 1969;124:149–62.

12. Mizeres NJ. The cardiac plexus in man. Am JAnat 1963;112:141–51.

13. Ajijola OA, Vaseghi M, Zhou W, et al. Func-tional differences between junctional and extra-junctional adrenergic receptor activation inmammalian ventricle. Am J Physiol Heart CircPhysiol 2013;304:H579–88.

14. Wang HJ, Wang W, Cornish KG, Rozanski GJ,Zucker IH. Cardiac sympathetic afferent denerva-tion attenuates cardiac remodeling and improvescardiovascular dysfunction in rats with heart fail-ure. Hypertension 2014;64:745–55.

15. Vaseghi M, Zhou W, Shi J, et al. Sympatheticinnervation of the anterior left ventricular wall bythe right and left stellate ganglia. Heart Rhythm2012;9:1303–9.

16. Vaseghi M, Yamakawa K, Sinha A, et al. Mod-ulation of regional dispersion of repolarization andT-peak to T-end interval by the right and left

stellate ganglia. Am J Physiol Heart Circ Physiol2013;305:H1020–30.

17. Ulphani JS, Cain JH, Inderyas F, et al. Quanti-tative analysis of parasympathetic innervation ofthe porcine heart. Heart Rhythm 2010;7:1113–9.

18. Yamakawa K, Rajendran PS, Takamiya T, et al.Vagal nerve stimulation activates vagal afferentfibers that reduce cardiac efferent para-sympathetic effects. Am J Physiol Heart CircPhysiol 2015;309:H1579–90.

19. Yamakawa K, So EL, Rajendran PS, et al.Electrophysiological effects of right and left vagalnerve stimulation on the ventricular myocardium.Am J Physiol Heart Circ Physiol 2014;307:H722–31.

20. Ardell JL, Rajendran PS, Nier HA,KenKnight BH, Armour JA. Central-peripheralneural network interactions evoked by vagusnerve stimulation: functional consequences oncontrol of cardiac function. Am J Physiol Heart CircPhysiol 2015;309:H1740–52.

21. Kember GC, Armour JA, Zamir M. Mechanismof smart baroreception in the aortic arch. Phys RevE Stat Nonlin Soft Matter Phys 2006;74:031914.

22. Kember G, Ardell JL, Shivkumar K, Armour JA.Recurrent myocardial infarction: mechanisms offree-floating adaptation and autonomic derange-ment in networked cardiac neural control. PLoSOne 2017;12:e0180194.

23. Guo YP, McLeod JG, Baverstock J. Pathologicalchanges in the vagus nerve in diabetes and chronicalcoholism. J Neurol Neurosurg Psychiatry 1987;50:1449–53.

Page 15: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1203

24. Duchen LW, Anjorin A, Watkins PJ, Mackay JD.Pathology of autonomic neuropathy in diabetesmellitus. Ann Intern Med 1980;92:301–3.

25. Kristensson K, Nordborg C, Olsson Y,Sourander P. Changes in the vagus nerve in dia-betes mellitus. Acta Pathol Microbiol Scand A1971;79:684–5.

26. Schmidt RE. Neuropathology and pathogen-esis of diabetic autonomic neuropathy. Int RevNeurobiol 2002;50:257–92.

27. Appenzeller O, Richardson EP Jr. The sympa-thetic chain in patients with diabetic and alcoholicpolyneuropathy. Neurology 1966;16:1205–9.

28. Vinik AI, Maser RE, Mitchell BD, Freeman R.Diabetic autonomic neuropathy. Diabetes Care2003;26:1553–79.

29. Cameron NE, Cotter MA. Metabolic andvascular factors in the pathogenesis of diabeticneuropathy. Diabetes 1997;46 Suppl 2:S31–7.

30. Schonauer M, Thomas A, Morbach S,Niebauer J, Schonauer U, Thiele H. Cardiac auto-nomic diabetic neuropathy. Diab Vasc Dis Res2008;5:336–44.

31. Pop-Busui R. Cardiac autonomic neuropathy indiabetes: a clinical perspective. Diabetes Care2010;33:434–41.

32. O’Brien IA, O’Hare JP, Lewin IG, Corrall RJ. Theprevalence of autonomic neuropathy in insulin-dependent diabetes mellitus: a controlled studybased on heart rate variability. Q J Med 1986;61:957–67.

33. Neil HA, Thompson AV, John S, McCarthy ST,Mann JI. Diabetic autonomic neuropathy: theprevalence of impaired heart rate variability in ageographically defined population. Diabet Med1989;6:20–4.

34. Ziegler D, Gries FA, Spuler M, Lessmann F. Theepidemiology of diabetic neuropathy. DiabeticCardiovascular Autonomic Neuropathy MulticenterStudy Group. J Diab Comp 1992;6:49–57.

35. Carnethon MR, Prineas RJ, Temprosa M,Zhang ZM, Uwaifo G, Molitch ME. The associationamong autonomic nervous system function, inci-dent diabetes, and intervention arm in the Dia-betes Prevention Program. Diabetes Care 2006;29:914–9.

36. Carnethon MR, Jacobs DR Jr., Sidney S,Liu K. Influence of autonomic nervous systemdysfunction on the development of type 2diabetes: the CARDIA study. Diabetes Care2003;26:3035–41.

37. Carnethon MR, Golden SH, Folsom AR,Haskell W, Liao D. Prospective investigation ofautonomic nervous system function and thedevelopment of type 2 diabetes: the Atheroscle-rosis Risk In Communities study, 1987-1998. Cir-culation 2003;107:2190–5.

38. Lucini D, Zuccotti G, Malacarne M, et al. Earlyprogression of the autonomic dysfunctionobserved in pediatric type 1 diabetes mellitus.Hypertension 2009;54:987–94.

39. Lahiri MK, Kannankeril PJ, Goldberger JJ.Assessment of autonomic function in cardiovas-cular disease: physiological basis and prognosticimplications. J Am Coll Cardiol 2008;51:1725–33.

40. Vinik AI, Ziegler D. Diabetic cardiovascularautonomic neuropathy. Circulation 2007;115:387–97.

41. Spallone V, Menzinger G. Diagnosis of cardio-vascular autonomic neuropathy in diabetes. Dia-betes 1997;46 Suppl 2:S67–76.

42. Ng F, Wong S, Cruz AL, Hernandez MI,Gomis P, Passariello G. Heart rate recovery in thediagnosis of diabetic cardiovascular autonomicneuropathy. Comput Cardiol 2007:681–4.

43. Tesfaye S, Boulton AJ, Dyck PJ, et al. Diabeticneuropathies: update on definitions, diagnosticcriteria, estimation of severity, and treatments.Diabetes Care 2010;33:2285–93.

44. Ewing DJ, Martyn CN, Young RJ, Clarke BF.The value of cardiovascular autonomic functiontests: 10 years experience in diabetes. DiabetesCare 1985;8:491–8.

45. Mestivier D, Chau NP, Chanudet X,Bauduceau B, Larroque P. Relationship betweendiabetic autonomic dysfunction and heart ratevariability assessed by recurrence plot. Am JPhysiol 1997;272:H1094–9.

46. Khandoker AH, Jelinek HF, Palaniswami M.Identifying diabetic patients with cardiac auto-nomic neuropathy by heart rate complexity anal-ysis. Biomed Eng Online 2009;8:3.

47. American Diabetes Association. Standards ofmedical care in diabetes–2013. Diabetes Care2013;36 Suppl 1:S11–66.

48. Balcıo�glu AS, Müderriso�glu H. Diabetes andcardiac autonomic neuropathy: clinical manifesta-tions, cardiovascular consequences, diagnosis andtreatment. World J Diabetes 2015;6:80–91.

49. Maser RE, Mitchell BD, Vinik AI, Freeman R.The association between cardiovascular autonomicneuropathy and mortality in individuals with dia-betes: a meta-analysis. Diabetes Care 2003;26:1895–901.

50. Naas AA, Davidson NC, Thompson C, et al. QTand QTc dispersion are accurate predictors ofcardiac death in newly diagnosed non-insulindependent diabetes: cohort study. BMJ 1998;316:745–6.

51. Rossing P, Breum L, Major-Pedersen A, et al.Prolonged QTc interval predicts mortality in pa-tients with type 1 diabetes mellitus. Diabet Med2001;18:199–205.

52. Sawicki PT, Dahne R, Bender R, Berger M.Prolonged QT interval as a predictor of mortalityin diabetic nephropathy. Diabetologia 1996;39:77–81.

53. Sawicki PT, Kiwitt S, Bender R, Berger M. Thevalue of QT interval dispersion for identification oftotal mortality risk in non-insulin-dependent dia-betes mellitus. J Intern Med 1998;243:49–56.

54. Veglio M, Sivieri R, Chinaglia A, Scaglione L,Cavallo-Perin P. QT interval prolongation andmortality in type 1 diabetic patients: a 5-yearcohort prospective study. Neuropathy StudyGroup of the Italian Society of the Study of Dia-betes, Piemonte Affiliate. Diabetes Care 2000;23:1381–3.

55. Ewing DJ, Campbell IW, Clarke BF. Mortality indiabetic autonomic neuropathy. Lancet 1976;1:601–3.

56. Orchard TJ, Lloyd CE, Maser RE, Kuller LH.Why does diabetic autonomic neuropathy predictIDDM mortality? An analysis from the PittsburghEpidemiology of Diabetes Complications Study.Diabetes Res Clin Pract 1996;34 Suppl 1:S165–71.

57. Suarez GA, Clark VM, Norell JE, et al. Suddencardiac death in diabetes mellitus: risk factors inthe Rochester Diabetic Neuropathy Study.J Neurol Neurosurg Psychiatry 2005;76:240–5.

58. Wackers FJ, Young LH, Inzucchi SE, et al.Detection of silent myocardial ischemia inasymptomatic diabetic subjects: the DIAD study.Diabetes Care 2004;27:1954–61.

59. The Consensus Committee of the AmericanAutonomic Society, the American Academy ofNeurology. Consensus statement on the definitionof orthostatic hypotension, pure autonomic fail-ure, and multiple system atrophy. Neurology1996;46:1470.

60. Vernino S, Low PA, Fealey RD, Stewart JD,Farrugia G, Lennon VA. Autoantibodies to gangli-onic acetylcholine receptors in autoimmune auto-nomic neuropathies. N Engl J Med 2000;343:847–55.

61. Hague K, Lento P, Morgello S, Caro S,Kaufmann H. The distribution of Lewy bodies inpure autonomic failure: autopsy findings and re-view of the literature. Acta Neuropathol (Berl)1997;94:192–6.

62. Hishikawa N, Hashizume Y, Hirayama M, et al.Brainstem-type Lewy body disease presentingwith progressive autonomic failure and lethargy.Clin Auton Res 2000;10:139–43.

63. Freeman R, Abuzinadah AR, Gibbons C,Jones P, Miglis MG, Sinn DI. Orthostatic hypoten-sion: JACC state-of-the-art review. J Am CollCardiol 2018;72:1294–309.

64. Bryarly M, Phillips LT, Fu Q, Vernino S,Levine BD. Postural orthostatic tachycardia syn-drome: JACC focus seminar. J Am Coll Cardiol2019;73:1207–28.

65. Fukuda K, Kanazawa H, Aizawa Y, Ardell JL,Shivkumar K. Cardiac innervation and sudden car-diac death. Circ Res 2015;116:2005–19.

66. Inoue H, Zipes DP. Time course of denervationof efferent sympathetic and vagal nerves afterocclusion of the coronary artery in the canineheart. Circ Res 1988;62:1111–20.

67. Costigan M, Moss A, Latremoliere A, et al. T-cell infiltration and signaling in the adult dorsalspinal cord is a major contributor to neuropathicpain-like hypersensitivity. J Neurosci 2009;29:14415–22.

68. Ajijola OA, Wisco JJ, Lambert HW, et al.Extracardiac neural remodeling in humans withcardiomyopathy. Circ Arrhythm Electrophysiol2012;5:1010–116.

69. Nakamura K, Ajijola OA, Aliotta E, Armour JA,Ardell JL, Shivkumar K. Pathological effects ofchronic myocardial infarction on peripheral neu-rons mediating cardiac neurotransmission. AutonNeurosci 2016;197:34–40.

70. Li C-Y, Li Y-G. Cardiac sympathetic nervesprouting and susceptibility to ventricular ar-rhythmias after myocardial infarction. Cardiol ResPract 2015;2015:698368.

Page 16: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1204

71. Ajijola OA, Yagishita D, Patel KJ, et al. Focalmyocardial infarction induces global remodelingof cardiac sympathetic innervation: neuralremodeling in a spatial context. Am J PhysiolHeart Circ Physiol 2013;305:H1031–40.

72. Vaseghi M, Lux RL, Mahajan A, Shivkumar K.Sympathetic stimulation increases dispersion ofrepolarization in humans with myocardial infarc-tion. Am J Physiol Heart Circ Physiol 2012;302:H1838–46.

73. Ajijola OA, Yagishita D, Reddy NK, et al.Remodeling of stellate ganglion neurons afterspatially targeted myocardial infarction: neuro-peptide and morphologic changes. Heart Rhythm2015;12:1027–35.

74. Rajendran PS, Nakamura K, Ajijola OA, et al.Myocardial infarction induces structural andfunctional remodelling of the intrinsic cardiacnervous system. J Physiol 2016;594:321–41.

75. Hou Y, Zhou Q, Po SS. Neuromodulation forcardiac arrhythmia. Heart Rhythm 2016;13:584–92.

76. Premchand RK, Sharma K, Mittal S, et al.Autonomic regulation therapy via left or rightcervical vagus nerve stimulation in patients withchronic heart failure: results of the ANTHEM-HFtrial. J Card Fail 2014;20:808–16.

77. Tse HF, Turner S, Sanders P, et al. ThoracicSpinal Cord Stimulation for Heart Failure as aRestorative Treatment (SCS HEART study): first-in-man experience. Heart Rhythm 2015;12:588–95.

78. Ng J, Villuendas R, Cokic I, et al. Autonomicremodeling in the left atrium and pulmonary veinsin heart failure: creation of a dynamic substrate foratrial fibrillation. Circ Arrhythm Electrophysiol2011;4:388–96.

79. Iwasaki YK, Nishida K, Kato T, Nattel S. Atrialfibrillation pathophysiology: implications formanagement. Circulation 2011;124:2264–74.

80. Shen MJ, Choi EK, Tan AY, et al. Neuralmechanisms of atrial arrhythmias. Nat Rev Cardiol2011;9:30–9.

81. Amar D, Zhang H, Miodownik S, Kadish AH.Competing autonomic mechanisms precede theonset of postoperative atrial fibrillation. J Am CollCardiol 2003;42:1262–8.

82. Patterson E, Po SS, Scherlag BJ, Lazzara R.Triggered firing in pulmonary veins initiated byin vitro autonomic nerve stimulation.[seecomment]. Heart Rhythm 2005;2:624–31.

83. Tan AY, Zhou S, Ogawa M, et al. Neuralmechanisms of paroxysmal atrial fibrillation andparoxysmal atrial tachycardia in ambulatory ca-nines. Circulation 2008;118:916–25.

84. Choi EK, Shen MJ, Han S, et al. Intrinsic cardiacnerve activity and paroxysmal atrial tachyar-rhythmia in ambulatory dogs. Circulation 2010;121:2615–23.

85. Ogawa M, Tan AY, Song J, et al. Cryoablationof stellate ganglia and atrial arrhythmia in ambu-latory dogs with pacing-induced heart failure.Heart Rhythm 2009;6:1772–9.

86. Ogawa M, Zhou S, Tan AY, et al. Left stellateganglion and vagal nerve activity and cardiac ar-rhythmias in ambulatory dogs with pacing-induced

congestive heart failure. J Am Coll Cardiol 2007;50:335–43.

87. Hou Y, Scherlag BJ, Lin J, et al. Ganglionatedplexi modulate extrinsic cardiac autonomic nerveinput: effects on sinus rate, atrioventricular con-duction, refractoriness, and inducibility of atrialfibrillation. J Am Coll Cardiol 2007;50:61–8.

88. Hou Y, Scherlag BJ, Lin J, et al. Interactiveatrial neural network: determining the connectionsbetween ganglionated plexi. Heart Rhythm 2007;4:56–63.

89. Arora R, Ng J, Ulphani J, et al. Unique auto-nomic profile of the pulmonary veins and posteriorleft atrium. J Am Coll Cardiol 2007;49:1340–8.

90. Arora R, Ulphani JS, Villuendas R, et al. Neuralsubstrate for atrial fibrillation: implications fortargeted parasympathetic blockade in the poste-rior left atrium. Am J Physiol 2008;294:H134–44.

91. Arora R. Recent insights into the role of theautonomic nervous system in the creation ofsubstrate for atrial fibrillation: implications fortherapies targeting the atrial autonomic nervoussystem. Circ Arrhythm Electrophysiol 2012;5:850–9.

92. Chevalier P, Tabib A, Meyronnet D, et al.Quantitative study of nerves of the human leftatrium. Heart Rhythm 2005;2:518–22.

93. Arora R, Ulphani JS, Villuendas R, et al. Neuralsubstrate for atrial fibrillation: implications fortargeted parasympathetic blockade in the poste-rior left atrium. Am J Physiol Heart Circ Physiol2008;294:H134–44.

94. Tan AY, Li H, Wachsmann-Hogiu S, Chen LS,Chen PS, Fishbein MC. Autonomic innervation andsegmental muscular disconnections at the humanpulmonary vein-atrial junction: implications forcatheter ablation of atrial-pulmonary vein junc-tion. J Am Coll Cardiol 2006;48:132–43.

95. Deneke T, Chaar H, de Groot JR, et al. Shift inthe pattern of autonomic atrial innervation insubjects with persistent atrial fibrillation. HeartRhythm 2011;8:1357–63.

96. Gould PA, Yii M, McLean C, et al. Evidence forincreased atrial sympathetic innervation in persis-tent human atrial fibrillation. Pacing Clin Electro-physiol 2006;29:821–9.

97. Edgerton JR, Brinkman WT, Weaver T, et al.Pulmonary vein isolation and autonomic denerva-tion for the management of paroxysmal atrialfibrillation by a minimally invasive surgicalapproach. J Thorac Cardiovasc Surg 2010;140:823–8.

98. Edgerton JR, Edgerton ZJ, Weaver T, et al.Minimally invasive pulmonary vein isolation andpartial autonomic denervation for surgical treat-ment of atrial fibrillation. Ann Thorac Surg 2008;86:35–8; discussion 39.

99. Pokushalov E, Romanov A, Artyomenko S,Turov A, Shirokova N, Katritsis DG. Left atrialablation at the anatomic areas of ganglionatedplexi for paroxysmal atrial fibrillation. Pacing ClinElectrophysiol 2010;33:1231–8.

100. Pokushalov E, Romanov A, Artyomenko S,et al. Ganglionated plexi ablation for longstandingpersistent atrial fibrillation. Europace 2010;12:342–6.

101. Pokushalov E, Romanov A, Shugayev P, et al.Selective ganglionated plexi ablation for parox-ysmal atrial fibrillation. Heart Rhythm 2009;6:1257–64.

102. Scanavacca M, Pisani CF, Hachul D, et al.Selective atrial vagal denervation guided byevoked vagal reflex to treat patients with parox-ysmal atrial fibrillation. Circulation 2006;114:876–85.

103. Pokushalov E, Romanov A, Artyomenko S,et al. Ganglionated plexi ablation directed byhigh-frequency stimulation and complex frac-tionated atrial electrograms for paroxysmal atrialfibrillation. Pacing Clin Electrophysiol 2012;35:776–84.

104. Katritsis DG, Giazitzoglou E, Zografos T,Pokushalov E, Po SS, Camm AJ. Rapid pulmonaryvein isolation combined with autonomic gangliamodification: a randomized study. Heart Rhythm2011;8:672–8.

105. Zhou Q, Hou Y, Yang S. A meta-analysis ofthe comparative efficacy of ablation for atrialfibrillation with and without ablation of theganglionated plexi. Pacing Clin Electrophysiol2011;34:1687–94.

106. Nitta T, Ishii Y, Sakamoto S. Surgery for atrialfibrillation: recent progress and future perspec-tive. Gen Thorac Cardiovasc Surg 2012;60:13–20.

107. Bagge L, Blomstrom P, Nilsson L,Einarsson GM, Jideus L, Blomstrom-Lundqvist C.Epicardial off-pump pulmonary vein isolation andvagal denervation improve long-term outcomeand quality of life in patients with atrial fibrilla-tion. J Thorac Cardiovasc Surg 2009;137:1265–71.

108. Krum H, Schlaich M, Whitbourn R, et al.Catheter-based renal sympathetic denervation forresistant hypertension: a multicentre safety andproof-of-principle cohort study. Lancet 2009;373:1275–81.

109. Hering D, Lambert EA, Marusic P, et al.Substantial reduction in single sympathetic nervefiring after renal denervation in patients withresistant hypertension. Hypertension 2013;61:457–64.

110. Linz D, van Hunnik A, Hohl M, et al. Catheter-based renal denervation reduces atrial nervesprouting and complexity of atrial fibrillation ingoats. Circ Arrhythm Electrophysiol 2015;8:466–74.

111. Pokushalov E, Romanov A, Katritsis DG, et al.Renal denervation for improving outcomes ofcatheter ablation in patients with atrial fibrillationand hypertension: early experience. Heart Rhythm2014;11:1131–8.

112. Bhatt DL, Kandzari DE, O’Neill WW, et al.A controlled trial of renal denervation for resistanthypertension. N Engl J Med 2014;370:1393–401.

113. Pokushalov E, Kozlov B, Romanov A, et al.Long-term suppression of atrial fibrillation bybotulinum toxin injection into epicardial fat pads inpatients undergoing cardiac surgery: one-yearfollow-up of a randomized pilot study. CircArrhythm Electrophysiol 2015;8:1334–41.

114. Pokushalov E, Kozlov B, Romanov A, et al.Botulinum toxin injection in epicardial fat pads canprevent recurrences of atrial fibrillation after

Page 17: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9 Goldberger et al.M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6 Autonomic Nervous System Dysfunction

1205

cardiac surgery: results of a randomized pilotstudy. J Am Coll Cardiol 2014;64:628–9.

115. Aistrup GL, Villuendas R, Ng J, et al. TargetedG-protein inhibition as a novel approach todecrease vagal atrial fibrillation by selectiveparasympathetic attenuation. Cardiovasc Res2009;83:481–92.

116. Aistrup GL, Cokic I, Ng J, et al. Targeted non-viral gene-based inhibition of Gai/o-mediatedvagal signaling in the posterior left atrium de-creases vagal induced AF. Heart Rhythm 2011:1722–9.

117. Goldberger JJ, Subacius H, Patel T,Cunnane R, Kadish AH. Sudden cardiac death riskstratification in patients with nonischemic dilatedcardiomyopathy. J Am Coll Cardiol 2014;63:1879–89.

118. Kleiger RE, Stein PK, Bosner MS, Rottman JN.Time domain measurements of heart rate vari-ability. Cardiol Clin 1992;10:487–98.

119. Laude D, Weise F, Girard A, Elghozi JL.Spectral analysis of systolic blood pressure andheart rate oscillations related to respiration. ClinExp Pharmacol Physiol 1995;22:352–7.

120. Elghozi JL, Laude D, Girard A. Effects ofrespiration on blood pressure and heart rate vari-ability in humans. Clin Exp Pharmacol Physiol1991;18:735–42.

121. Malik MSG. Autonomic testing and cardiacrisk. In: Zipes DP, Jalife J, editors. Cardiac Elec-trophysiology: From Cell to Bedside. 6th ed.Philadelphia: Elsevier, 2014:649–56.

122. Vybiral T, Bryg RJ, Maddens ME, Boden WE.Effect of passive tilt on sympathetic and para-sympathetic components of heart rate variabilityin normal subjects. Am J Cardiol 1989;63:1117–20.

123. Pagani M, Lombardi F, Guzzetti S, et al. Powerspectral analysis of heart rate and arterial pressurevariabilities as a marker of sympatho-vagal inter-action in man and conscious dog. Circ Res 1986;59:178–93.

124. Chiou CW, Zipes DP. Selective vagal dener-vation of the atria eliminates heart rate variabilityand baroreflex sensitivity while preserving ven-tricular innervation. Circulation 1998;98:360–8.

125. Goldberger JJ. Sympathovagal balance: howshould we measure it? Am J Physiol 1999;276:H1273–80.

126. Fox K, Borer JS, Camm AJ, et al. Resting heartrate in cardiovascular disease. J Am Coll Cardiol2007;50:823–30.

127. Huikuri HV, Stein PK. Heart rate variability inrisk stratification of cardiac patients. Prog Car-diovasc Dis 2013;56:153–9.

128. Huikuri HV, Stein PK. Clinical application ofheart rate variability after acute myocardialinfarction. Front Physiol 2012;3:41.

129. Huikuri HV, Makikallio TH, Peng CK,Goldberger AL, Hintze U, Moller M. Fractal corre-lation properties of R-R interval dynamics andmortality in patients with depressed left ventric-ular function after an acute myocardial infarction.Circulation 2000;101:47–53.

130. Kleiger RE, Miller JP, Bigger JT Jr., Moss AJ.Decreased heart rate variability and its association

with increased mortality after acute myocardialinfarction. Am J Cardiol 1987;59:256–62.

131. La Rovere MT, Pinna GD, Maestri R, et al.Short-term heart rate variability strongly predictssudden cardiac death in chronic heart failure pa-tients. Circulation 2003;107:565–70.

132. Deyell MW, Krahn AD, Goldberger JJ. Suddencardiac death risk stratification. Circ Res 2015;116:1907–18.

133. Dekker JM, Crow RS, Folsom AR, et al. Lowheart rate variability in a 2-minute rhythm strippredicts risk of coronary heart disease and mor-tality from several causes: the ARIC Study.Atherosclerosis Risk In Communities. Circulation2000;102:1239–44.

134. de Bruyne MC, Kors JA, Hoes AW, et al. Bothdecreased and increased heart rate variability onthe standard 10-second electrocardiogram predictcardiac mortality in the elderly: the RotterdamStudy. Am J Epidemiol 1999;150:1282–8.

135. Tsuji H, Larson MG, Venditti FJ Jr., et al.Impact of reduced heart rate variability on risk forcardiac events. The Framingham Heart Study.Circulation 1996;94:2850–5.

136. Gnecchi Ruscone T, Lombardi F, Malfatto G,Malliani A. Attenuation of baroreceptive mecha-nisms by cardiovascular sympathetic afferent fi-bers. Am J Physiol 1987;253:H787–91.

137. Stein KM. Noninvasive risk stratification forsudden death: signal-averaged electrocardiog-raphy, nonsustained ventricular tachycardia, heartrate variability, baroreflex sensitivity, and QRSduration. Prog Cardiovasc Dis 2008;51:106–17.

138. Billman GE, Schwartz PJ, Stone HL. Barore-ceptor reflex control of heart rate: a predictor ofsudden cardiac death. Circulation 1982;66:874–80.

139. De Ferrari GM, Sanzo A, Bertoletti A,Specchia G, Vanoli E, Schwartz PJ. Baroreflexsensitivity predicts long-term cardiovascularmortality after myocardial infarction even in pa-tients with preserved left ventricular function.J Am Coll Cardiol 2007;50:2285–90.

140. Huikuri HV, Tapanainen JM, Lindgren K, et al.Prediction of sudden cardiac death after myocar-dial infarction in the beta-blocking era. J Am CollCardiol 2003;42:652–8.

141. La Rovere MT, Pinna GD, Hohnloser SH, et al.Baroreflex sensitivity and heart rate variability inthe identification of patients at risk for life-threatening arrhythmias: implications for clinicaltrials. Circulation 2001;103:2072–7.

142. Grimm W, Christ M, Bach J, Muller HH,Maisch B. Noninvasive arrhythmia risk stratifica-tion in idiopathic dilated cardiomyopathy: resultsof the Marburg Cardiomyopathy Study. Circulation2003;108:2883–91.

143. Schmidt G, Malik M, Barthel P, et al. Heart-rate turbulence after ventricular premature beatsas a predictor of mortality after acute myocardialinfarction. Lancet 1999;353:1390–6.

144. Cygankiewicz I. Heart rate turbulence. ProgCardiovasc Dis 2013;56:160–71.

145. Mrowka R, Persson PB, Theres H, Patzak A.Blunted arterial baroreflex causes “pathological”

heart rate turbulence. Am J Physiol Regul IntegrComp Physiol 2000;279:R1171–5.

146. Davies LC, Francis DP, Ponikowski P,Piepoli MF, Coats AJ. Relation of heart rate andblood pressure turbulence following prematureventricular complexes to baroreflex sensitivity inchronic congestive heart failure. Am J Cardiol2001;87:737–42.

147. Bauer A, Malik M, Schmidt G, et al. Heart rateturbulence: standards of measurement, physio-logical interpretation, and clinical use: Interna-tional Society for Holter and NoninvasiveElectrophysiology Consensus. J Am Coll Cardiol2008;52:1353–65.

148. La Rovere MT, Maestri R, Pinna GD, Sleight P,Febo O. Clinical and haemodynamic correlates ofheart rate turbulence as a non-invasive index ofbaroreflex sensitivity in chronic heart failure. ClinSci (Lond) 2011;121:279–84.

149. Berkowitsch A, Zareba W, Neumann T, et al.Risk stratification using heart rate turbulence andventricular arrhythmia in MADIT II: usefulness andlimitations of a 10-minute Holter recording. AnnNoninvasive Electrocardiol 2004;9:270–9.

150. Robinson BF, Epstein SE, Beiser GD,Braunwald E. Control of heart rate by the auto-nomic nervous system. Studies in man on theinterrelation between baroreceptor mechanismsand exercise. Circ Res 1966;19:400–11.

151. Ekblom B, Goldbarg AN, Kilbom A,Astrand PO. Effects of atropine and propranolol onthe oxygen transport system during exercise inman. Scand J Clin Lab Invest 1972;30:35–42.

152. Savin WM, Davidson DM, Haskell WL. Auto-nomic contribution to heart rate recovery fromexercise in humans. J Appl Physiol Respir EnvironExerc Physiol 1982;53:1572–5.

153. Imai K, Sato H, Hori M, et al. Vagally mediatedheart rate recovery after exercise is accelerated inathletes but blunted in patients with chronic heartfailure. J Am Coll Cardiol 1994;24:1529–35.

154. Kannankeril PJ, Le FK, Kadish AH,Goldberger JJ. Parasympathetic effects on heartrate recovery after exercise. J Investig Med 2004;52:394–401.

155. Goldberger JJ, Johnson NP, Subacius H, Ng J,Greenland P. Comparison of the physiologic andprognostic implications of the heart rate versusthe RR interval. Heart Rhythm 2014;11:1925–33.

156. Jouven X, Empana JP, Schwartz PJ,Desnos M, Courbon D, Ducimetiere P. Heart-rateprofile during exercise as a predictor of suddendeath. N Engl J Med 2005;352:1951–8.

157. Cole CR, Blackstone EH, Pashkow FJ,Snader CE, Lauer MS. Heart-rate recovery imme-diately after exercise as a predictor of mortality.N Engl J Med 1999;341:1351–7.

158. Batchvarov VN, Ghuran A, Smetana P, et al.QT-RR relationship in healthy subjects exhibitssubstantial intersubject variability and high intra-subject stability. Am J Physiol Heart Circ Physiol2002;282:H2356–63.

159. Fenichel RR, Malik M, Antzelevitch C, et al.Drug-induced torsades de pointes and implicationsfor drug development. J Cardiovasc Electrophysiol2004;15:475–95.

Page 18: ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY … · audio summary by Editor-in-Chief Dr. Valentin Fuster on JACC.org. JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 73, NO. 10,

Goldberger et al. J A C C V O L . 7 3 , N O . 1 0 , 2 0 1 9

Autonomic Nervous System Dysfunction M A R C H 1 9 , 2 0 1 9 : 1 1 8 9 – 2 0 6

1206

160. Sundaram S, Carnethon M, Polito K,Kadish AH, Goldberger JJ. Autonomic effectson QT-RR interval dynamics after exercise. AmJ Physiol Heart Circ Physiol 2008;294:H490–7.

161. Carter JR, Ray CA. Sympathetic neural adap-tations to exercise training in humans. AutonNeurosci 2015;188:36–43.

162. Macefield VG. Sympathetic micro-neurography. Handb Clin Neurol 2013;117:353–64.

163. Notarius CF, Millar PJ, Floras JS. Musclesympathetic activity in resting and exercisinghumans with and without heart failure. ApplPhysiol Nutr Metab 2015;40:1107–15.

164. Barretto AC, Santos AC, Munhoz R, et al.Increased muscle sympathetic nerve activity pre-dicts mortality in heart failure patients. Int J Car-diol 2009;135:302–7.

165. Pearson MJ, Smart NA. Exercise therapy andautonomic function in heart failure patients: asystematic review and meta-analysis. Heart FailRev 2018;23:91–108.

166. Chen PS, Chen LS, Fishbein MC, Lin SF,Nattel S. Role of the autonomic nervous system in

atrial fibrillation: pathophysiology and therapy.Circ Res 2014;114:1500–15.

167. Robinson EA, Rhee KS, Doytchinova A,et al. Estimating sympathetic tone by recordingsubcutaneous nerve activity in ambulatorydogs. J Cardiovasc Electrophysiol 2015;26:70–8.

168. Doytchinova A, Hassel JL, Yuan Y, et al.Simultaneous noninvasive recording of skin sym-pathetic nerve activity and electrocardiogram.Heart Rhythm 2017;14:25–33.

169. Mittleman MA, Maclure M, Tofler GH,Sherwood JB, Goldberg RJ, Muller JE. Trig-gering of acute myocardial infarction by heavyphysical exertion: protection against triggeringby regular exertion. N Engl J Med 1993;329:1677–83.

170. Mittleman MA, Maclure M, Sherwood JB,et al. Triggering of acute myocardial infarctiononset by episodes of anger. Determinants ofMyocardial Infarction Onset Study Investigators.Circulation 1995;92:1720–5.

171. Albert CM, Mittleman MA, Chae CU, Lee IM,Hennekens CH, Manson JE. Triggering of sudden

death from cardiac causes by vigorous exertion.N Engl J Med 2000;343:1355–61.

172. Lampert R, Joska T, Burg MM, Batsdord WP,McPherson CA, Jain D. Emotional and physicalprecipitants of ventricular arrhythmia. Circulation2002;106:1800–5.

173. De Ferrari GM, Mantica M, Vanoli E, Hull SS Jr.,Schwartz PJ. Scopolamine increases vagal tone andvagal reflexes in patients after myocardial infarc-tion. J Am Coll Cardiol 1993;22:1327–34.

174. Hull SS Jr., Vanoli E, Adamson PB, DeFerrari GM, Foreman RD, Schwartz PJ. Do in-creases in markers of vagal activity imply protec-tion from sudden death? The case of scopolamine.Circulation 1995;91:2516–9.

KEY WORDS arrhythmia, autonomic, heartfailure, myocardial infarction

Go to http://www.acc.org/jacc-journals-cme to takethe CME/MOC/ECME quizfor this article.


Recommended