Você está na página 1de 11

Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Contents lists available at ScienceDirect

Autonomic Neuroscience: Basic and Clinical


journal homepage: www.elsevier.com/locate/autneu

Review

Cardiac neuroanatomy - Imaging nerves to define functional control T


a a,b a a a
Peter Hanna , Pradeep S. Rajendran , Olujimi A. Ajijola , Marmar Vaseghi , J. Andrew Armour ,
Jefrrey L. Ardella,b, Kalyanam Shivkumara,b,⁎
a
University of California Los Angeles (UCLA) Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, David Geffen School of Medicine, UCLA,
Los Angeles, CA, USA
b
Molecular, Cellular & Integrative Physiology Program, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA

A R T I C L E I N F O A B S T R A C T

Keywords: The autonomic nervous system regulates normal cardiovascular function and plays a critical role in the pa-
Neurocardiology thophysiology of cardiovascular disease. Further understanding of the interplay between the autonomic nervous
Intracardiac nervous system system and cardiovascular system holds promise for the development of neuroscience-based cardiovascular
Myocardial innervation therapeutics. To this end, techniques to image myocardial innervation will help provide a basis for under-
Autonomic nervous system
standing the fundamental underpinnings of cardiac neural control. In this review, we detail the evolution of
gross and microscopic anatomical studies for functional mapping of cardiac neuroanatomy.

1. Introduction of cardiac nerves depicted in anatomical texts since the late 1500s are
reproduced in books and anatomical studies without histological con-
The autonomic nervous system (ANS) enables integrative control of firmation of whether structures labeled as ‘nerves’ are actually neural.
the viscera to ensure survival of the organism (Jänig, 2008). Specifi- Indeed, when neural structures are surgically targeted for therapeutics
cally, the cardiac ANS plays a crucial role to maintain normal rhythm in clinical care (e.g. bilateral cardiac sympathetic decentralization),
and sustain the circulation of blood. The cardiac ANS intricately reg- intra-operative histological confirmation is sought to confirm that
ulates all the critical physiological functions of the heart (chronotropy, neural tissue was indeed removed (Bourke et al., 2010). The problem at
dromotropy, inotropy, and lusitropy). The interplay of the ANS and the the level of the heart is compounded by the lack of technology for high-
heart is readily apparent in the pathophysiology of most cardiovascular resolution microscopic studies until very recently. Furthermore,
diseases including hypertension, heart failure, myocardial infarction, methods to handle the very large image datasets and the bioinformatics
and arrhythmias (Barron and Lesh, 1996; Nolan et al., 1998; La Rovere tools for neuronal tracing in images and three-dimensional re-
et al., 1998; Floras, 2003; Shen and Zipes, 2014). The study of such construction have only recently been developed. Placing these images
interactions has led to the development of neuromodulatory therapies in their functional context, using state-of-the-art physiological ap-
that treat cardiac disease, ranging from vagus nerve stimulation to bi- proaches from micro-stimulation to neural recordings, are required for
lateral cardiac sympathetic decentralization for heart failure and ven- interpretation of normal physiology. Finally, integrating anatomical
tricular arrhythmias (Beaumont et al., 2015; Shen and Zipes, 2015; and functional data would provide the framework needed to precisely
Schwartz, 2014). A more thorough characterization of cardiac in- define the changes in innervation due to disease (neural remodeling)
nervation will promote further study of neural control of cardiac and assure correct utilization and monitoring of new or pre-existing
function and allow for the development of targeted cardiovascular methods for neuromodulation.
therapeutics (Shivkumar et al., 2016). In this review, we describe
available and burgeoning techniques in mapping the structure and 3. Gross anatomy of neurotransmission to and from the heart
function of cardiac neuroanatomy.
The gross anatomy of cardiac innervation has been the subject of
2. Functional organization of the cardiac nervous system investigation for a number of years. Dissections of mammals and human
cadavers have illustrated that myocardial innervation is conserved to a
Anatomically, cardiac nerves are complex, and detailed atlases of significant degree among species (Kuntz and Morehouse, 1930;
cardiac innervation are few in number (Figs. 1-3). Macroscopic images Saccomanno, 1943; Mizeres, 1963; Randall et al., 1972; Hopkins and


Corresponding author at: UCLA Cardiac Arrhythmia Center, 100 UCLA Medical Plaza, Suite 660, Los Angeles, CA 90095, United States.
E-mail address: kshivkumar@mednet.ucla.edu (K. Shivkumar).

http://dx.doi.org/10.1016/j.autneu.2017.07.008
Received 3 February 2017; Received in revised form 22 July 2017; Accepted 28 July 2017
1566-0702/ © 2017 Elsevier B.V. All rights reserved.

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Fig. 1. Neural control of the heart. Autonomic control of the heart is


comprised of a series of nested feedback loops, from the intracardiac
neurons (level 1) to the intrathoracic extracardiac neurons (levels 2) and
the central nervous system (level 3). SG, sympathetic ganglion; DRG,
dorsal root ganglion; ICNS, intracardiac nervous system. Adapted from
Jänig (2016).

Level 3

Level 2

Level 1

Armour, 1984; Janes et al., 1986; Kawashima, 2005). The ANS in- intermediolateral cell column of the spinal cord and project via C7-T6
nervating the heart has been categorized into: i) central; ii) in- rami to postganglionic sympathetic neurons in the superior cervical,
trathoracic extracardiac; and iii) intrinsic cardiac components (Figs. 1- middle cervical, cervicothoracic (stellate; also known as the sympa-
3). The intrathoracic extrinsic cardiac nervous system connects the in- thetic chain), and mediastinal ganglia (Hopkins and Armour, 1984;
trinsic cardiac nervous system within the heart to the central nervous Kawashima, 2005). These postganglionic neurons project axons via
system. It is composed of parasympathetic and sympathetic motor multiple cardiopulmonary nerves to atrial and ventricular myocardium
components that exert opposing effects on cardiac electrical and me- and limited populations of intrinsic cardiac adrenergic neurons. Ka-
chanical indices. washima published a detailed study in 2005 that analyzed the origin
and course of the autonomic nerves in human cadavers using micro-
scopy, highlighting the conserved nature of the superior and middle
3.1. Parasympathetic motor neurons
cervical, mediastinal, and stellate ganglia across species (Kawashima,
2005).
Preganglionic neurons of the parasympathetic nervous system are
located in the nucleus ambiguus and dorsal motor nucleus of the me-
dulla oblongata as well as scattered regions in between these two 3.3. Sensory neurons
structures (Standish et al., 1994; Standish et al., 1995). These project
axons via the vagus nerve and its multiple intrathoracic cardio- Cardiac afferents provide beat-to-beat sensory information of car-
pulmonary branches to efferent postganglionic parasympathetic neu- diac function and microenvironment to the neuraxis, and additional
rons in the numerous intrinsic cardiac ganglia (Hopkins et al., 1996). information is conveyed by arterial mechano- and chemoreceptors
Postganglionic efferent parasympathetic neurons located in individual (Fig. 2). Afferent nerves travel within the vagal trunk and ‘sympathetic’
cardiac ganglia receive preganglionic inputs from both the right and left fibers to the nodose and dorsal root ganglia, respectively. The proces-
vagal trunks (Ardell et al., 2015). Anatomic dissections in canines have sing of afferent information at multiple levels, including the intrinsic
demonstrated that postganglionic vagal neurons to the sinoatrial (SA) cardiac nervous system, extracardiac intrathoracic ganglia, spinal cord,
node are found predominantly in a ganglionated plexus next to the right brain stem, and higher centers, provides an elegant mechanism of in-
pulmonary vein-atrial junction, while the postganglionic vagal neurons teracting feedback loops that modulate efferent cardiomotor (sympa-
that influence the atrioventricular (AV) node are located predominantly thetic and parasympathetic) signals for maintaining normal rhythm and
in the region adjacent to the inferior vena cava-inferior left atrium life-sustaining circulation.
junction (Randall and Ardell, 1985).
3.4. The intrinsic cardiac nervous system
3.2. Sympathetic motor neurons
In contrast to the extrinsic components of the ANS, intrinsic cardiac
Sympathetic efferent preganglionic neurons originate in the neurons are found in intramural ganglia and in epicardial fat pads that

49

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Fig. 2. Neurohumoral control and functional organization of cardiac innervation. Aff, afferent; β1, β-adrenergic receptor; C, cervical; DRG, dorsal root ganglion; Gi, inhibitory G-protein;
Gs, stimulatory G-protein; L, lumbar; LCN, local circuit neuron; M2, muscarinic receptor; T, thoracic. Adapted from Shivkumar et al. (2016).

obscure direct visualization (Fig. 4) (Yuan et al., 1994; Armour et al., marker has also proved helpful in delineating locations of afferent
1997). While these neuronal somata were identified early in the last neurons in nodose and dorsal root ganglia that transduce the mechan-
century, the location and connections of these neurons have remained ical and/or chemical milieu of various regions of the heart or great
poorly understood (McFarland and Anders, 1913). Subsequent study vessels to the nervous system (Oldfield and McLachlan, 1978; Kalia and
using light and electron microscopy confirmed these structures in the Mesulam, 1980; Hopkins and Armour, 1989). Xrhodamine-5-(and 6)-
human heart (Figs. 4C-E) (Armour et al., 1997). It is now known that isothiocyanate (XRITC) is another marker that serves as a retrograde
the intrinsic cardiac ganglionated plexuses contain complex networks of fluorescent tracer for preganglionic vagal neurons (Mendelowitz and
neurons and interconnecting nerves that include the following sub- Kunze, 1991; Wang et al., 2014). As mentioned above, visualization of
types: i) afferent neurons; ii) motor (parasympathetic and sympathetic) the intrinsic cardiac nervous system has proved challenging, and
neurons; and iii) interconnecting local circuit neurons (Armour, 2008). fluorescent dyes, Fast Blue and bisbenzimide, have been used to de-
Taken together, this neural network at the level of the heart is thought monstrate retrograde transport that confirm the presence of inter-
to play an important role in modulating cardio-cardiac reflexes. neuronal connections (Kuypers et al., 1979; Bentivoglio et al., 1980;
Tomney et al., 1985). Antibodies against cholera toxin subunit B have
also been used in immunohistochemical studies of the intracardiac
4. Histochemical and immunohistochemical techniques ganglia (Grkovic et al., 2005).
Specific identification of neuron types in the two limbs of the ANS is
Histochemical techniques have characterized the richness of myo- made possible by targeting select neurotransmitters associated with
cardial innervation. Cresyl violet staining has been used to identify the sympathetic or parasympathetic neuronal somata located in the per-
cytoplasmic Nissl substance in autonomic neurons (Janes et al., 1986; ipheral cardiac ANS. Postganglionic parasympathetic neurons utilize
Yuan et al., 1994; Pickel et al., 1975). The retrograde transport of the neurotransmitter acetylcholine, degraded by the enzyme acet-
horseradish peroxidase and vital dyes applied to various cardiac tissues ylcholinesterase selectively associated with parasympathetic motor
had been used to identify the locations of cardiac vagal preganglionic neurons. As such, many studies use acetylcholine to mimic the effects of
neurons in the medulla (Todo et al., 1977; Mesulam, 1978). This vagal stimulation (DiFrancesco and Tromba, 1988; Petit-Jacques et al.,
technique has helped characterize the locations of sympathetic post- 1993). Karnovsky and Roots published a histochemical method in 1964
ganglionic neuronal somata and vagal preganglionic efferent neurons that utilized a thiocholine ester that is hydrolyzed by cholinesterase to
innervating feline and canine hearts (Hopkins and Armour, 1984). This

50

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Fig. 3. A, Detailed illustration of the course of the autonomic cardiac nerves. The innervation of the heart via the right thoracic cardiac nerve and the inferior cardiac nerve is shown by
the black stars. The arrowheads highlight the course of the left thoracic cardiac nerve. The sympathetic cardiac nerves, vagal cardiac branches, and the cardiac plexuses are colored in
orange, green and purple, respectively. AI, anterior interventricular branch; Ao, aorta; Az, azygos vein; CB, circumflex branch; CC, common carotid artery; CT, cervicothoracic (stellate)
ganglion; GV, great cardiac vein; IB, inferior (vagal) cardiac branch; IG, inferior cervical ganglion; IN, inferior cervical cardiac nerve; L, lung; LA, left atrium; LCA, left coronary artery;
MG, middle cervical ganglion; MN, middle cardiac nerve; P, pectoral nerve; Ph, phrenic nerve; PT, pulmonary trunk; RA, right atrium; RCA, right coronary artery; RL, recurrent laryngeal
nerve of vagus nerve; SB, superior (vagal) cardiac branch; Sbc, nerve to subclavian muscle; SG, superior cervical ganglion; SN, superior cardiac nerve; SS, suprascapular nerve; SVC,
superior vena cava; TB, thoracic (vagal) cardiac branch; TG, thoracic ganglia; TN, thoracic cardiac nerve; VG, vertebral ganglion; VN, vertebral nerve; X, vagus nerve; XI, accessory nerve;
XII, hypoglossal nerve. Adapted from Kawashima (2005). Course of the right (B) and left (C) sympathetic trunks, vagus nerve and their major branches in an embalmed cadaver. Adapted
from Janes et al. (1986) and Rajendran et al. (2017). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

form copper thiocholine, which appears brown when treated with 2010; Pauza et al., 2002; Kawano et al., 2003). For example, Pauza
ammonium sulfide (Karnovsky and Roots, 1964). This method and re- et al. showed that the number of intrinsic cholinergic neurons in the
finements of this method including, but not limited to, the use of heart was directly proportional to ganglia area in guinea pig, rat, dog,
acetylthiocholine and the use of a marker of the peroxidase activity of and human hearts using such methods (Pauza et al., 2002; Pauza et al.,
the reaction product have been used to study the distribution of cho- 2014; Pauziene et al., 2017).
linergic neurons in the heart (Fig. 5) (Tago et al., 1986; Roberts et al., While this histologic method identifies cholinesterase activity as-
1989; Wharton et al., 1990; Batulevicius et al., 2005; Ulphani et al., sociated with select populations of neuronal somata, it is not specific for

51

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

A B

C D E

Fig. 4. A-B, Distribution of the intrinsic cardiac ganglionated plexuses (GP) in the heart. A, posterior; B, superior. Adapted from Rajendran et al. (2017). C-E, Light photomicrographs of
human intrinsic cardiac nerves, ganglia and neurons. C, Network of ganglia and nerves stained with methylene blue and dissected from the posteromedial left atrial ganglionated plexus.
The ganglia appear as expansions along a nerve (box). D, Enlargement of boxed area in A illustrating a ganglion composed of approximately 150–200 nerve cell bodies. E, High-
magnification micrograph of a multipolar neuron. Note the accumulation of lipofuscin granules (arrowheads). Scale bars 52.5 mm in C, 250 μm in D, 25 μm in E. Adapted from Armour
et al. (1997). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

acetylcholinesterase activity in vagal neurons. As such, im- and protein gene product 9.5 that are found almost exclusively in
munohistochemcial methods have been used to identify para- neurons (Fig. 6) (Wharton et al., 1990; Pauza et al., 2014; Mabe et al.,
sympathetic neurons using antibodies against choline acetyltransferase 2006; Crick et al., 1999; Khan and Ludueña, 1996; Mahmoud et al.,
(ChAT, Fig. 5A-C). This methodology was initially utilized in the 2015; White et al., 2015; Hoard et al., 2008). Presynaptic nerves are
characterization of guinea pig myenteric plexus neurons associated targeted using antibodies against synapsin I, which is a member of a
with the gastrointestinal tract (Schemann et al., 1993). It was thereafter family of neuron-specific proteins that interact with synaptic vesicles
applied to study of guinea pig cardiac ganglia and is now in widespread (White et al., 2015; Gitler et al., 2004).
use (Mawe et al., 1996; Fedorov et al., 2006). Additional markers such While the majority of these intracardiac neurons are either para-
as choline transporter and vesicular acetylcholine transporter, which sympathetic or sympathetic in nature, intracardiac nerves are not ex-
are more specific for the identification cholinergic neurons, have since clusively parasympathetic or sympathetic. Thus, the use of additional
been developed (Hoover et al., 2004; Mabe et al., 2006). markers has helped elucidate the mixed cholinergic and adrenergic
Similarly, sympathetic neurons can be identified using im- phenotype of intrinsic cardiac neurons and nerves in the heart. For
munohistochemcial markers that target an enzyme specific to these example, Hoard et al. demonstrated the presence of cholinergic neurons
neurons. With respect to adrenergic neurons, tyrosine hydroxylase (TH) in intrinsic cardiac ganglia that are associated with tyrosine hydro-
is the enzyme involved in the rate-limiting step in norepinephrine xylase, dopamine β-hydroxylase, and norepinephrine transporter, yet
synthesis and, as such, is specific for norepinephrine-producing neurons do not contain vesicular monoamine transporter type 2 (VMAT2) and
(Fig. 5D-F). Pickel et al. developed antibodies against TH that per- thereby precludes the ability to store norepinephrine (Hoard et al.,
formed well for localization of neurons involved in biosynthesis of ca- 2008). Rysevaite et al. showed that 83% of intrinsic cardiac neurons
techolamines (Pickel et al., 1975). Immunohistochemistry targeting of were ChAT immunoreactive, 4% were TH immunoreactive, and 14%
TH has been used regularly to characterize the sympathetic innervation were immunoreactive for both ChAT and TH (Rysevaite et al., 2011).
of the heart across different animal models (Wharton et al., 1990; This study also demonstrated immunoreactivity to calcitonin gene-re-
Kawano et al., 2003; Mawe et al., 1996; Crick et al., 1999; Kreipke and lated peptide and substance P, neuropeptides found in nociceptive
Birren, 2015). Pan-neuronal markers are used to identify cardiac neurons, to identify a population of afferent neurons in the intrinsic
nerves, and examples include the neurofilament markers β-tubulin III cardiac nervous system (Rysevaite et al., 2011).

52

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Fig. 5. Confocal images of human intrinsic cardiac neurons expressing the cholinergic and adrenergic phenotypes. A–C, Confocal images of a ganglion that was double labeled to show
CHT (A) and ChAT (B). CHT immunoreactivity (A, red) was prominent in varicose nerve fibers around intrinsic cardiac neurons and faint or absent in the neuronal cell bodies. In contrast,
staining for ChAT (B, green) was notable in neuronal cell bodies and generally less intense in surrounding nerve processes. C, Colocalization of CHT and ChAT (yellow) was noted in some
cell bodies and nerve processes in the overlay image (CHT + ChAT). Inserts at lower left show boxed areas at higher magnification. All panels contain maximum projection images
compiled from confocal scans that spanned 8 μm. Scale bar = 100 μm in A–C. D-F, Confocal images of a section that was double labeled to show TH (D, green) and VMAT2 (E, red). D,
Few neurons and nerve fibers stained for TH. E, Prominent staining for VMAT2 occurred in most neurons and many nerves fibers. F, Overlap (OVL) of TH and VMAT2 images shows that a
significant amount of the TH colocalizes (yellow) with VMAT2. All panels contain maximum projection images compiled from confocal scans that spanned 10 μm. Scale bar = 150 μm.
Adapted from Hoover et al. (2009). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

Antibody-based approaches have inherent limitations with respect inhomogeneity has hindered imaging larger volumes of tissue. Tissue
to fidelity of results. In that regard, the use of transgenic mice has al- clearing approaches, initially developed to better visualize neural net-
lowed for a more detailed structural analysis, as in the morphogenesis works in the central nervous system, attempt to minimize light scatter
of the cardiac nervous system. In mouse studies, wingless type in- by decreasing tissue inhomogeneity and allowing for deeper imaging in
tegration site family, member 1 (Wnt1)-Cre recombinase (Cre) mice thick sections or intact tissues while preserving the three-dimensional
have been used to identify tissues derived from neural crest cells, as molecular and cellular architecture. Two main subgroups of techniques
Wnt1 is only expressed during embryonic development of the central include solvent-based methods, such as BABB (Miller et al., 2005),
nervous system (Echelard et al., 1994). White et al. demonstrated that tetrahydrofuran and dibenzylether (Becker et al., 2012), 3DISCO
crossing these mice with mice expressing the tdTomato reporter results (Ertürk et al., 2012) and iDISCO, or aqueous-based methods, including
in neural crest-derived cells that are labeled with red fluorescence and Scale, SeeDB, CUBIC, ClearT, ClearT2, CLARITY, and PACT, are used to
imaged using epifluorescent stereomicroscopy to image the sub- remove the lipid membranes of cells, which are the major source of
epicardial neural network in exquisite detail (White et al., 2015). tissue heterogeneity and, hence, light diffraction, to generate optically
Mapping the fine neural network across large distances in organs transparent tissue (Becker et al., 2012; Ertürk et al., 2012; Hama et al.,
has typically been performed using histological sections that are then 2011; Renier et al., 2014; Ke et al., 2013; Tainaka et al., 2014;
reconstructed, a time-consuming process that also suffers from loss in Kuwajima et al., 2013; Chung et al., 2013; Yang et al., 2014). While
fidelity due to tissue distortion. Light scatter due to tissue various challenges face these differing techniques including time

53

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

in decreased chronotropy and decreased AV conduction, while Loeb


and DeTarnowsky showed how sympathetic nerve stimulation resulted
RAA
in increased AV conduction through the use of intracardiac electro-
grams in in vivo experiments in dog (Randall et al., 1985; Loeb and de
Tarnowsky, 1988). More recently, a model of isolated Langendorff
rabbit heart with intact dual autonomic innervation has allowed for in
vitro preparations that are devoid of confounding neurohormonal in-
fluences of in vivo models (Ng et al., 2001).
Intrinsic cardiac
ganglia 6. Optogenetic techniques

In contrast to optical mapping, which is helpful at the organ level,


LAA
CS optogenetics provide functional information on myocardial innervation
at the cellular level. Optogenetic methods utilize light-activated ion
channels called opsins to activate or inhibit specific cell types such as
neural populations in vivo, providing spatiotemporal resolution in un-
RV precedented detail and allowing the study of structure and function.
The application of optogenetics in neurons was developed over a
decade ago and has since been translated to the study in neuro-
LV cardiology (Zemelman et al., 2002; Nagel et al., 2003; Boyden et al.,
2005). Channelrhodopsin 2 (ChR2) is an opsin, or light-gated ion
channel, identified in unicellular green alga Chlamydomonas reinhardtii
(Nagel et al., 2003). When ChR2 is expressed in a neuron, inward
currents are evoked within 50 ms of a flash of blue light, and this
technology has be used for fast neuronal photostimulation in mammals
(Boyden et al., 2005). Improvements of this tool include ChR2 variants
that increase the magnitude of photocurrents induced (Berndt et al.,
2011; Kleinlogel et al., 2011; Lin et al., 2009). In the brain, multidiode
probe arrays have been used in studies of local brain tissues to allow for
multisite neuronal light-activation (Stark et al., 2012). Alternatively,
1 mm various ways of patterned illumination have been developed for dif-
PGP9.5
ferential light activation in a sample (Packer et al., 2013).
Fig. 6. Confocal image of dorsal surface of mouse heart stained with pan-neuronal marker
Gene delivery systems, such as viruses are currently been used to
protein gene product 9.5 (PGP9.5) and demonstrating the rich innervation of the heart. deliver opsins to select neuronal populations. Examples of such vectors
CS, coronary sinus; LAA, left atrial appendage; LV, left ventricle; RAA, right atrial ap- include herpes, rabies and adeno- and lenti-associated viruses, with the
pendage; RV, right ventricle. latter two being the most commonly used (Yizhar et al., 2011;
Deverman et al., 2016; Chan et al., 2017). For example, Boyden et al.
intensity, degree of tissue clearing, preservation of fluorescence of re- transfected cultured neurons with lentiviruses bearing the ChR2 protein
porters, and tissue expansion, the rapidly evolving field holds the tagged with a yellow fluorescent protein (Boyden et al., 2005). Adeno-
promise of improved three-dimensional imaging at depth. In conjunc- associated viruses are small, nonpathogenic and replication-defective
tion with these developments, advances in microscopy in the form of parvoviruses with single-stranded DNA genome. Multiple subtypes
laser-scanning confocal, two-photon, and light-sheet microscopy permit exist; for instance, AAV2 in particular has been shown to have high
volumetric imaging of cleared tissues. The premise of these techniques transduction efficiency in neurons while AAV9 has been used to deliver
is to image discrete planes within a volume and minimize out-of-focus the cop4 gene, which encodes the ChR2 protein, into rat ventricular
light to yield improved imaging at depth. cardiomyocytes in vivo (Packer et al., 2013; Yizhar et al., 2011;
Nussinovitch and Gepstein, 2015). Additional delivery systems include
the use of transgenes and the Cre/loxP recombination systems, as
5. Functional mapping mentioned above, to target photoactivation of specific neural popula-
tions. In this system, loxP cassettes flank the cop4 gene with a reporter
Given the complex structure of parasympathetic, sympathetic motor gene encoding fluorescent protein and the Cre driver expressing Cre
and other intrinsic cardiac neurons within the heart, mapping studies recombinase under the control of a cell-specific promoter such as tyr-
have been performed to elucidate the functional architecture of the osine hydroxylase or choline acetyltransferase (Wang et al., 2014;
cardiac nervous system. Studies have evaluated how autonomic nerve Wengrowski et al., 2015; Kalmbach et al., 2012).
stimulation affects mechanical and electrocardiographic parameters of The initial application of optogenetics to cardiovascular research
the heart (Fig. 7) (Ito and Zipes, 1994; Chiou et al., 1997; Ajijola et al., was in 2010 to locate and control cardiac pacemaker cells in zebrafish
2017; Yamakawa et al., 2015). Norris et al. placed strain gauges on the and in embryonic stem cell-derived cardiomyocytes in mice, and sev-
epicardium and electrically stimulated sympathetic nerves in canines to eral studies have since been published regarding optogenetic control of
measure regional inotropy to determine gross functional innervation cardiomyocytes (Arrenberg et al., 2010; Bruegmann et al., 2010; Jia
patterns (Norris et al., 1974; Norris et al., 1977). The positive and ne- et al., 2011; Boyle et al., 2013; Beiert et al., 2014; Bingen et al., 2014;
gative chronotropic effect of sympathetic and parasympathetic simu- Park et al., 2014). Wang et al. have been able to demonstrate that
lation on SA node activity, respectively, were originally described in optogenetic stimulation of noradrenergic neurons in the locus cereleus
studies employing nerve stimulation while monitoring heart rate. These of the brain inhibits parasympathetic cardiac vagal neuron outflow that
studies determine that the right vagus nerve primarily affecting the SA results in tachycardia (Wang et al., 2014). Their group have further
node while the left vagus nerve exerted predominant influence on the evaluated the neural axis in the murine heart by identifying and sti-
AV node in the canine heart (Cohn, 1912; Levy and Zieske, 1969). mulating a subpopulation of cardiomyocytes that bear the phenyletha-
Randall et al. demonstrated that parasympathetic stimulation resulted nolamine n-methyltransferase gene, which encodes the enzyme that

54

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Vagal nerve stimulation in normal porcine heart

Stellate ganglion stimulation in normal porcine heart

Fig. 7. A, Activation recovery interval (ARI) maps at baseline and during right (RVNS) and left (LVNS) vagal nerve stimulation in a normal porcine heart. No significant regional
differences in responses were found. Adapted from Yamakawa et al. (Yamakawa et al., 2015) B, ARI maps in control porcine hearts at baseline (BL) and during right (RSG), left (LSG) and
bilateral (BSG) stellate ganglion stimulation. Myocardial regions are displayed in the BL control map. Adapted from Ajijola et al. (2013).

converts norepinephrine to epinephrine, to exert control on heart degradation in the sympathetic nerve terminal and outlined the dy-
rhythm through the use of optogenetics (Wang et al., 2017). Wen- namic regeneration process of sympathetic nerve function in the
growski et al. have shown photoactivation of cardiac sympathetic transplanted heart (Bravo et al., 2015). In studies of mIBG, the radio-
nerves affects cardiac electrical properties to increase heart rate along tracer is injected and the mean counts per pixel in the heart are com-
with cardiac contractile force (Fig. 8) (Wengrowski et al., 2015). To pared to those of the mediastinum to generate a heart/mediastinal (H/
study the effects of vagal tone on exercise capacity, Machhada et al. M) uptake ratio. Quantification of the H/M ratio has resulted in the
utilized a viral vector system to target neurons in the rat brainstem identification of cutoffs that have been associated with disease pro-
dorsal ventral motor nucleus, the main vagal nucleus in the central gression and response to therapy (Jacobson et al., 2010; Gerson et al.,
nervous system, with channelrhodopsin ChIEF (Machhada et al., 2017). 2002; Toyama et al., 1999; Kasama et al., 2005; Somsen et al., 1996;
Activation of the dorsal ventral motor nucleus neurons resulted in in- Kasama et al., 2002; Nagahara et al., 2008; Tomoda et al., 1994). In
creased cardiac contractility and exercise capacity. In sum, modern short, cardiac imaging employing radiotracers of sympathetic output to
viral-based as well as Cre transgenic approaches, have been used to map the heart has diagnostic and prognostic implications in heart failure and
the structural and functional properties of the autonomic nervous arrhythmic and ischemic heart disease that warrants further study.
system controlling the heart.
8. Summary and future directions
7. Clinical neuroimaging
The functional anatomy of the cardiac nervous system is complex.
Imaging techniques of myocardial innervation, although limited, do Early studies focused on describing the anatomy of the cardiac nervous
allow for the study of innervation in animal studies and living humans. system to help elucidate the role of the ANS in regional cardiac control.
[14C]2-deoxyglucose has been used to radiolabel neurons in culture and Concepts regarding cardiac neural control have been revised in recent
in vivo in sympathetic ganglia in canine with neural uptake induced by years with new physiological data and have created a new and exciting
stimulation and imaged using autoradiography (Saji and Obata, 1981; framework for understanding regulatory control of the mammalian
Kostreva et al., 1985). C-11 hydroxyephedrine or radioiodinated me- heart. Direct single neural and network recordings from intrinsic car-
taiodobenzylguanidine (mIBG), which are catecholamine analogues diac and extracardiac ganglia provide the methods to study organ level
that are taken up by sympathetic nerve terminals, can be used to vi- physiology and a proper framework for interpretation (Armour, 2008;
sualize sympathetic innervation to the heart in vivo in dogs and humans Armour, 2004; Beaumont et al., 2013). The field is now poised from a
using a gamma camera and positron emission tomography, respectively clinical and instrumentation front to take full advantage of the cellular
(Kline et al., 1981; Wieland et al., 1981; Sisson et al., 1988; Dae et al., and molecular work that has been done in the past to probe autonomic
1989; Schwaiger et al., 1990; Calkins et al., 1993). Bravo et al. used circuits.
three different C-11 labeled catecholamines to identify neuro- More recently, technological advances in optical mapping and op-
transmitter metabolites involved in vesicular storage, transport and togenetics hold the promise of functionally mapping the specificities of

55

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Fig. 8. Effects of blue light photostimulation on heart rate and contractile force in Langendorff-perfused mouse hearts. A, Stability of force and heart rate shown over 15 min (top) and
expressed as percent change (bottom, n = 8). B, Percent change from control phase after stimulating with blue light (grey, n = 10), administering isoproterenol (black, n = 5), or
stimulating with blue light after administering propranolol (white, n = 5). C, Changes in force and heart rate after two rounds of blue light photostimulation (*) and addition of
isoproterenol (+). D, Changes in force and heart rate after several rounds of blue light photostimulation (*) and response to photostimulation after administration of propranolol (+).
**P < 0.05 statistically different form baseline. Adapted from Wengrowski et al. (2015). (For interpretation of the references to colour in this figure legend, the reader is referred to the
web version of this article.)

cardiac neural innervation to understand the role that the autonomic Ardell, J.L., Rajendran, P.S., Nier, H.A., KenKnight, B.H., Armour, J.A., 2015. Central-
peripheral neural network interactions evoked by vagus nerve stimulation: functional
nervous system plays in the pathophysiology of cardiovascular disease, consequences on control of cardiac function. Am. J. Physiol. Heart Circ. Physiol. 309,
with particular regard to therapeutically targeting select neural ele- H1740–1752.
ments therein. As such, further discoveries in this arena hold the pro- Armour, J.A., 2004. Cardiac neuronal hierarchy in health and disease. Am. J. Phys. Regul.
Integr. Comp. Phys. 287, R262–271.
mise of developing neuroscience-based cardiovascular therapeutics in Armour, J.A., 2008. Potential clinical relevance of the ‘little brain’ on the mammalian
the future. The recent National Institutes of Health (NIH) initiative for heart. Exp. Physiol. 93, 165–176.
Stimulating Peripheral Activity to Relieve Conditions (SPARC) has Armour, J.A., Murphy, D.A., Yuan, B.X., Macdonald, S., Hopkins, D.A., 1997. Gross and
microscopic anatomy of the human intrinsic cardiac nervous system. Anat. Rec. 247,
provided new impetus for the characterization of organ innervation
289–298.
including the heart (https://commonfund.nih.gov/sparc). Arrenberg, A.B., Stainier, D.Y.R., Baier, H., Huisken, J., 2010. Optogenetic control of
cardiac function. Science 330, 971–974.
Barron, H.V., Lesh, M.D., 1996. Autonomic nervous system and sudden cardiac death. J.
Acknowledgements
Am. Coll. Cardiol. 27, 1053–1060.
Batulevicius, D., Pauziene, N., Pauza, D.H., 2005. Architecture and age-related analysis of
None. the neuronal number of the guinea pig intrinsic cardiac nerve plexus. Ann. Anat.
Anat. Anz. Off. Organ Anat. Ges. 187, 225–243.
Beaumont, E., et al., 2013. Network interactions within the canine intrinsic cardiac
Funding nervous system: implications for reflex control of regional cardiac function. J.
Physiol. 591, 4515–4533.
This work is supported by NIH/National Heart, Lung, and Blood Beaumont, E., et al., 2015. Vagus nerve stimulation mitigates intrinsic cardiac neuronal
and adverse myocyte remodeling postmyocardial infarction. Am. J. Physiol. Heart
Institute (NHLBI) Grant F31HL127974 to P.S.R; NIH/NHLBI HL125730- Circ. Physiol. 309, H1198–1206.
03 to O.A.A.; the UCLA Specialty Training and Advanced Research Becker, K., Jährling, N., Saghafi, S., Weiler, R., Dodt, H.-U., 2012. Chemical clearing and
Program (STAR) fellowship to P.H.; NIH Stimulating Peripheral Activity dehydration of GFP expressing mouse brains. PLoS One 7, e33916.
Beiert, T., Bruegmann, T., Sasse, P., 2014. Optogenetic activation of Gq signalling mod-
to Relieve Conditions (SPARC) Grant 5U18EB021799 to J.L.A.; and ulates pacemaker activity of cardiomyocytes. Cardiovasc. Res. 102, 507–516.
NIH/NHLBI HL084261 & NIH SPARC Grant OT2OD023848 to K.S. Bentivoglio, M., Kuypers, H.G., Catsman-Berrevoets, C.E., Loewe, H., Dann, O., 1980. Two
new fluorescent retrograde neuronal tracers which are transported over long dis-
tances. Neurosci. Lett. 18, 25–30.
References Berndt, A., et al., 2011. High-efficiency channelrhodopsins for fast neuronal stimulation
at low light levels. Proc. Natl. Acad. Sci. U. S. A. 108, 7595–7600.
Ajijola, O.A., et al., 2013. Focal myocardial infarction induces global remodeling of Bingen, B.O., et al., 2014. Light-induced termination of spiral wave arrhythmias by op-
cardiac sympathetic innervation: neural remodeling in a spatial context. Am. J. togenetic engineering of atrial cardiomyocytes. Cardiovasc. Res. 104, 194–205.
Physiol. Heart Circ. Physiol. 305, H1031–1040. Bourke, T., et al., 2010. Neuraxial modulation for refractory ventricular arrhythmias:
Ajijola, O.A., et al., 2017. Sympathetic modulation of electrical activation in normal and value of thoracic epidural anesthesia and surgical left cardiac sympathetic denerva-
infarcted myocardium: implications for arrhythmogenesis. Am. J. Physiol. Heart Circ. tion. Circulation 121, 2255–2262.
Physiol. 00575, 2016. http://dx.doi.org/10.1152/ajpheart.00575.2016. Boyden, E.S., Zhang, F., Bamberg, E., Nagel, G., Deisseroth, K., 2005. Millisecond-

56

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

timescale, genetically targeted optical control of neural activity. Nat. Neurosci. 8, Kalmbach, A., Hedrick, T., Waters, J., 2012. Selective optogenetic stimulation of choli-
1263–1268. nergic axons in neocortex. J. Neurophysiol. 107, 2008–2019.
Boyle, P.M., Williams, J.C., Ambrosi, C.M., Entcheva, E., Trayanova, N.A., 2013. A Karnovsky, M.J., Roots, L., 1964. A ‘direct-coloring’ THIOCHOLINE method for
comprehensive multiscale framework for simulating optogenetics in the heart. Nat. CHOLINESTERASES. J. Histochem. Cytochem. 12, 219–221.
Commun. 4, 2370. Kasama, S., et al., 2002. Spironolactone improves cardiac sympathetic nerve activity and
Bravo, P.E., et al., 2015. Mechanistic insights into sympathetic neuronal regeneration: symptoms in patients with congestive heart failure. J. Nucl. Med. Off. Publ. Soc. Nucl.
multitracer molecular imaging of catecholamine handling after cardiac transplanta- Med. 43, 1279–1285.
tion. Circ. Cardiovasc. Imaging 8, e003507. Kasama, S., et al., 2005. Effects of candesartan on cardiac sympathetic nerve activity in
Bruegmann, T., et al., 2010. Optogenetic control of heart muscle in vitro and in vivo. Nat. patients with congestive heart failure and preserved left ventricular ejection fraction.
Methods 7, 897–900. J. Am. Coll. Cardiol. 45, 661–667.
Calkins, H., Lehmann, M.H., Allman, K., Wieland, D., Schwaiger, M., 1993. Scintigraphic Kawano, H., Okada, R., Yano, K., 2003. Histological study on the distribution of auto-
pattern of regional cardiac sympathetic innervation in patients with familial long QT nomic nerves in the human heart. Heart Vessel. 18, 32–39.
syndrome using positron emission tomography. Circulation 87, 1616–1621. Kawashima, T., 2005. The autonomic nervous system of the human heart with special
Chan, K.Y., et al., 2017. Engineered AAVs for efficient noninvasive gene delivery to the reference to its origin, course, and peripheral distribution. Anat. Embryol. 209,
central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179. 425–438.
Chiou, C.W., Eble, J.N., Zipes, D.P., 1997. Efferent vagal innervation of the canine atria Ke, M.-T., Fujimoto, S., Imai, T., 2013. SeeDB: a simple and morphology-preserving op-
and sinus and atrioventricular nodes. The third fat pad. Circulation 95, 2573–2584. tical clearing agent for neuronal circuit reconstruction. Nat. Neurosci. 16,
Chung, K., et al., 2013. Structural and molecular interrogation of intact biological sys- 1154–1161.
tems. Nature 497, 332–337. Khan, I.A., Ludueña, R.F., 1996. Phosphorylation of beta III-tubulin. Biochemistry (Mosc)
Cohn, A.E., 1912. On the differences in the effects of stimulation of the two vagus nerves 35, 3704–3711.
on rate and conduction of the dog's heart. J. Exp. Med. 16, 732–757. Kleinlogel, S., et al., 2011. Ultra light-sensitive and fast neuronal activation with the
Crick, S.J., Anderson, R.H., Ho, S.Y., Sheppard, M.N., 1999. Localisation and quantitation Ca2 +-permeable channelrhodopsin CatCh. Nat. Neurosci. 14, 513–518.
of autonomic innervation in the porcine heart II: endocardium, myocardium and Kline, R.C., et al., 1981. Myocardial imaging in man with I-123 meta-iodobenzylguani-
epicardium. J. Anat. 195 (Pt 3), 359–373. dine. J. Nucl. Med. 22, 129–132.
Dae, M.W., et al., 1989. Scintigraphic assessment of regional cardiac adrenergic in- Kostreva, D.R., Armour, J.A., Bosnjak, Z.J., 1985. Metabolic mapping of a cardiac reflex
nervation. Circulation 79, 634–644. mediated by sympathetic ganglia in dogs. Am. J. Phys. 249, R317–322.
Deverman, B.E., et al., 2016. Cre-dependent selection yields AAV variants for widespread Kreipke, R.E., Birren, S.J., 2015. Innervating sympathetic neurons regulate heart size and
gene transfer to the adult brain. Nat. Biotechnol. 34, 204–209. the timing of cardiomyocyte cell cycle withdrawal. J. Physiol. 593, 5057–5073.
DiFrancesco, D., Tromba, C., 1988. Inhibition of the hyperpolarization-activated current Kuntz, A., Morehouse, A., 1930. Thoracic sympathetic cardiac nerves in man: their re-
(if) induced by acetylcholine in rabbit sino-atrial node myocytes. J. Physiol. 405, lation to cervical sympathetic ganglionectomy. Arch. Surg. 20, 607–613.
477–491. Kuwajima, T., et al., 2013. ClearT: a detergent- and solvent-free clearing method for
Echelard, Y., Vassileva, G., McMahon, A.P., 1994. Cis‑acting regulatory sequences gov- neuronal and non-neuronal tissue. Development 140, 1364–1368.
erning Wnt-1 expression in the developing mouse CNS. Development 120, Kuypers, H.G., Bentivoglio, M., van der Kooy, D., Catsman-Berrevoets, C.E., 1979.
2213–2224. Retrograde transport of bisbenzimide and propidium iodide through axons to their
Ertürk, A., et al., 2012. Three-dimensional imaging of solvent-cleared organs using parent cell bodies. Neurosci. Lett. 12, 1–7.
3DISCO. Nat. Protoc. 7, 1983–1995. La Rovere, M.T., Bigger, J.T., Marcus, F.I., Mortara, A., Schwartz, P.J., 1998. Baroreflex
Fedorov, V.V., Hucker, W.J., Dobrzynski, H., Rosenshtraukh, L.V., Efimov, I.R., 2006. sensitivity and heart-rate variability in prediction of total cardiac mortality after
Postganglionic nerve stimulation induces temporal inhibition of excitability in rabbit myocardial infarction. ATRAMI (autonomic tone and reflexes after myocardial in-
sinoatrial node. Am. J. Physiol. Heart Circ. Physiol. 291, H612–623. farction) investigators. Lancet Lond. Engl. 351, 478–484.
Floras, J.S., 2003. Sympathetic activation in human heart failure: diverse mechanisms, Levy, M.N., Zieske, H., 1969. Autonomic control of cardiac pacemaker activity and at-
therapeutic opportunities. Acta Physiol. Scand. 177, 391–398. rioventricular transmission. J. Appl. Physiol. 27, 465–470.
Gerson, M.C., et al., 2002. Carvedilol improves left ventricular function in heart failure Lin, J.Y., Lin, M.Z., Steinbach, P., Tsien, R.Y., 2009. Characterization of engineered
patients with idiopathic dilated cardiomyopathy and a wide range of sympathetic channelrhodopsin variants with improved properties and kinetics. Biophys. J. 96,
nervous system function as measured by iodine 123 metaiodobenzylguanidine. J. 1803–1814.
Nucl. Cardiol. 9, 608–615. Loeb, J.M., de Tarnowsky, J.M., 1988. Integration of heart rate and sympathetic neural
Gitler, D., et al., 2004. Molecular determinants of synapsin targeting to presynaptic effects on AV conduction. Am. J. Phys. 254, H651–657.
terminals. J. Neurosci. 24, 3711–3720. Mabe, A.M., Hoard, J.L., Duffourc, M.M., Hoover, D.B., 2006. Localization of cholinergic
Grkovic, I., Fernandez, K., McAllen, R.M., Anderson, C.R., 2005. Misidentification of innervation and neurturin receptors in adult mouse heart and expression of the
cardiac vagal pre-ganglionic neurons after injections of retrograde tracer into the neurturin gene. Cell Tissue Res. 326, 57–67.
pericardial space in the rat. Cell Tissue Res. 321, 335–340. Machhada, A., et al., 2017. Vagal determinants of exercise capacity. Nat. Commun. 8,
Hama, H., et al., 2011. Scale: a chemical approach for fluorescence imaging and re- 15097.
construction of transparent mouse brain. Nat. Neurosci. 14, 1481–1488. Mahmoud, A.I., et al., 2015. Nerves regulate Cardiomyocyte proliferation and heart re-
Hoard, J.L., et al., 2008. Cholinergic neurons of mouse intrinsic cardiac ganglia contain generation. Dev. Cell 34, 387–399.
noradrenergic enzymes, norepinephrine transporters, and the neurotrophin receptors Mawe, G.M., Talmage, E.K., Lee, K.P., Parsons, R.L., 1996. Expression of choline acetyl-
tropomyosin-related kinase A and p75. Neuroscience 156, 129–142. transferase immunoreactivity in guinea pig cardiac ganglia. Cell Tissue Res. 285,
Hoover, D.B., Ganote, C.E., Ferguson, S.M., Blakely, R.D., Parsons, R.L., 2004. 281–286.
Localization of cholinergic innervation in guinea pig heart by immunohistochemistry McFarland, J., Anders, A., 1913. The morbid histology of the cardiac nervous ganglia. J.
for high-affinity choline transporters. Cardiovasc. Res. 62, 112–121. Med. Res. 27, 425–435.
Hoover, D.B., et al., 2009. Localization of multiple neurotransmitters in surgically derived Mendelowitz, D., Kunze, D.L., 1991. Identification and dissociation of cardiovascular
specimens of human atrial ganglia. Neuroscience 164, 1170–1179. neurons from the medulla for patch clamp analysis. Neurosci. Lett. 132, 217–221.
Hopkins, D.A., Armour, J.A., 1984. Localization of sympathetic postganglionic and Mesulam, M.M., 1978. Tetramethyl benzidine for horseradish peroxidase neurohis-
parasympathetic preganglionic neurons which innervate different regions of the dog tochemistry: a non-carcinogenic blue reaction product with superior sensitivity for
heart. J. Comp. Neurol. 229, 186–198. visualizing neural afferents and efferents. J. Histochem. Cytochem. 26, 106–117.
Hopkins, D.A., Armour, J.A., 1989. Ganglionic distribution of afferent neurons in- Miller, C.E., et al., 2005. Confocal imaging of the embryonic heart: how deep? Microsc.
nervating the canine heart and cardiopulmonary nerves. J. Auton. Nerv. Syst. 26, Microanal. Off. J. Microsc. Soc. Am. Microbeam Anal. Soc. Microsc. Soc. Can. 11,
213–222. 216–223.
Hopkins, D.A., Bieger, D., de Vente, J., Steinbusch, W.M., 1996. Vagal efferent projec- Mizeres, N.J., 1963. The cardiac plexus in man. Am. J. Anat. 112, 141–151.
tions: viscerotopy, neurochemistry and effects of vagotomy. Prog. Brain Res. 107, Nagahara, D., et al., 2008. Predicting the need for an implantable cardioverter defi-
79–96. brillator using cardiac metaiodobenzylguanidine activity together with plasma na-
Ito, M., Zipes, D.P., 1994. Efferent sympathetic and vagal innervation of the canine right triuretic peptide concentration or left ventricular function. J. Nucl. Med. Off. Publ.
ventricle. Circulation 90, 1459–1468. Soc. Nucl. Med. 49, 225–233.
Jacobson, A.F., et al., 2010. Myocardial iodine-123 meta-iodobenzylguanidine imaging Nagel, G., et al., 2003. Channelrhodopsin-2, a directly light-gated cation-selective
and cardiac events in heart failure. Results of the prospective ADMIRE-HF (AdreView membrane channel. Proc. Natl. Acad. Sci. U. S. A. 100, 13940–13945.
Myocardial Imaging for Risk Evaluation in Heart Failure) study. J. Am. Coll. Cardiol. Ng, G.A., Brack, K.E., Coote, J.H., 2001. Effects of direct sympathetic and vagus nerve
55, 2212–2221. stimulation on the physiology of the whole heart—a novel model of isolated
Janes, R.D., et al., 1986. Anatomy of human extrinsic cardiac nerves and ganglia. Am. J. Langendorff perfused rabbit heart with intact dual autonomic innervation. Exp.
Cardiol. 57, 299–309. Physiol. 86, 319–329.
Jänig, W., 2008. Integrative Action of the Autonomic Nervous System: Neurobiology of Nolan, J., et al., 1998. Prospective study of heart rate variability and mortality in chronic
Homeostasis. Cambridge University Press. heart failure: results of the United Kingdom heart failure evaluation and assessment
Jänig, W., 2016. Neurocardiology: a neurobiologist's perspective. J. Physiol. 594, of risk trial (UK-heart). Circulation 98, 1510–1516.
3955–3962. Norris, J.E., Foreman, R.D., Wurster, R.K., 1974. Responses of the canine heart to sti-
Jia, Z., et al., 2011. Stimulating cardiac muscle by light: cardiac optogenetics by cell mulation of the first five ventral thoracic roots. Am. J. Phys. 227, 9–12.
delivery. Circ. Arrhythm. Electrophysiol. 4, 753–760. Norris, J.E., Lippincott, D., Wurster, R.D., 1977. Responses of canine endocardium to
Kalia, M., Mesulam, M.M., 1980. Brain stem projections of sensory and motor components stimulation of the upper thoracic roots. Am. J. Phys. 233, H655–659.
of the vagus complex in the cat: II. Laryngeal, tracheobronchial, pulmonary, cardiac, Nussinovitch, U., Gepstein, L., 2015. Optogenetics for in vivo cardiac pacing and re-
and gastrointestinal branches. J. Comp. Neurol. 193, 467–508. synchronization therapies. Nat. Biotechnol. 33, 750–754.

57

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
P. Hanna et al. Autonomic Neuroscience: Basic and Clinical 207 (2017) 48–58

Oldfield, B.J., McLachlan, E.M., 1978. Localization of sensory neurons traversing the Somsen, G.A., et al., 1996. Increased myocardial [123I]-metaiodobenzylguanidine uptake
stellate ganglion of the cat. J. Comp. Neurol. 182, 915–922. after enalapril treatment in patients with chronic heart failure. Heart Br. Card. Soc.
Packer, A.M., Roska, B., Häusser, M., 2013. Targeting neurons and photons for optoge- 76, 218–222.
netics. Nat. Neurosci. 16, 805–815. Standish, A., Enquist, L.W., Schwaber, J.S., 1994. Innervation of the heart and its central
Park, S.A., Lee, S.-R., Tung, L., Yue, D.T., 2014. Optical mapping of optogenetically medullary origin defined by viral tracing. Science 263, 232–234.
shaped cardiac action potentials. Sci Rep 4, 6125. Standish, A., Enquist, L.W., Escardo, J.A., Schwaber, J.S., 1995. Central neuronal circuit
Pauza, D.H., Pauziene, N., Pakeltyte, G., Stropus, R., 2002. Comparative quantitative innervating the rat heart defined by transneuronal transport of pseudorabies virus. J.
study of the intrinsic cardiac ganglia and neurons in the rat, guinea pig, dog and Neurosci. 15, 1998–2012.
human as revealed by histochemical staining for acetylcholinesterase. Ann. Anat. Stark, E., Koos, T., Buzsáki, G., 2012. Diode probes for spatiotemporal optical control of
Anat. Anz. Off. Organ Anat. Ges. 184, 125–136. multiple neurons in freely moving animals. J. Neurophysiol. 108, 349–363.
Pauza, D.H., et al., 2014. A combined acetylcholinesterase and immunohistochemical Tago, H., Kimura, H., Maeda, T., 1986. Visualization of detailed acetylcholinesterase fiber
method for precise anatomical analysis of intrinsic cardiac neural structures. Ann. and neuron staining in rat brain by a sensitive histochemical procedure. J.
Anat. Anat. Anz. Off. Organ Anat. Ges. 196, 430–440. Histochem. Cytochem. 34, 1431–1438.
Pauziene, N., et al., 2017. Neuroanatomy of the pig cardiac ventricles. A stereomicro- Tainaka, K., et al., 2014. Whole-body imaging with single-cell resolution by tissue de-
scopic, confocal and electron microscope study. Anat. Rec. 2007http://dx.doi.org/10. colorization. Cell 159, 911–924.
1002/ar.23619. (Hoboken NJ). Todo, K., et al., 1977. Origins of vagal preganglionic fibers to the sino-atrial and atrio-
Petit-Jacques, J., Bois, P., Bescond, J., Lenfant, J., 1993. Mechanism of muscarinic control ventricular node regions in the cat heart as studied by the horseradish peroxidase
of the high-threshold calcium current in rabbit sino-atrial node myocytes. Pflugers method. Brain Res. 130, 545–550.
Arch. 423, 21–27. Tomney, P.A., Hopkins, D.A., Armour, J.A., 1985. Axonal branching of canine sympa-
Pickel, V.M., Joh, T.H., Field, P.M., Becker, C.G., Reis, D.J., 1975. Cellular localization of thetic postganglionic cardiopulmonary neurons. A retrograde fluorescent labeling
tyrosine hydroxylase by immunohistochemistry. J. Histochem. Cytochem. 23, 1–12. study. Brain Res. Bull. 14, 443–452.
Rajendran, P.S., et al., 2017. Neural control of cardiac function in health and disease. In: Tomoda, H., et al., 1994. Regional sympathetic denervation detected by iodine 123
Dilsizian, V., Narula, J. (Eds.), Atlas of Cardiac Innervation, 1st ed. Springer metaiodobenzylguanidine in non-Q-wave myocardial infarction and unstable angina.
International Publishing, Switzerland, pp. 13–35. Am. Heart J. 128, 452–458.
Randall, W.C., Ardell, J.L., 1985. Selective parasympathectomy of automatic and con- Toyama, T., et al., 1999. Cardiac sympathetic activity estimated by 123I-MIBG myo-
ductile tissues of the canine heart. Am. J. Phys. 248, H61–68. cardial imaging in patients with dilated cardiomyopathy after beta-blocker or an-
Randall, W.C., Armour, J.A., Geis, W.P., Lippincott, D.B., 1972. Regional cardiac dis- giotensin-converting enzyme inhibitor therapy. J. Nucl. Med. Off. Publ. Soc. Nucl.
tribution of the sympathetic nerves. Fed. Proc. 31, 1199–1208. Med. 40, 217–223.
Randall, W.C., Ardell, J.L., Becker, D.M., 1985. Differential responses accompanying se- Ulphani, J.S., et al., 2010. Quantitative analysis of parasympathetic innervation of the
quential stimulation and ablation of vagal branches to dog heart. Am. J. Phys. 249, porcine heart. Heart Rhythm. 7, 1113–1119.
H133–140. Wang, X., Piñol, R.A., Byrne, P., Mendelowitz, D., 2014. Optogenetic stimulation of locus
Renier, N., et al., 2014. iDISCO: a simple, rapid method to immunolabel large tissue ceruleus neurons augments inhibitory transmission to parasympathetic cardiac vagal
samples for volume imaging. Cell 159, 896–910. neurons via activation of brainstem α1 and β1 receptors. J. Neurosci. 34, 6182–6189.
Roberts, L.A., Slocum, G.R., Riley, D.A., 1989. Morphological study of the innervation Wang, Y., et al., 2017. Optogenetic control of heart rhythm by selective stimulation of
pattern of the rabbit sinoatrial node. Am. J. Anat. 185, 74–88. Cardiomyocytes derived from Pnmt(+) cells in murine heart. Sci Rep 7, 40687.
Rysevaite, K., et al., 2011. Immunohistochemical characterization of the intrinsic cardiac Wengrowski, A.M., et al., 2015. Optogenetic release of norepinephrine from cardiac
neural plexus in whole-mount mouse heart preparations. Heart Rhythm. 8, 731–738. sympathetic neurons alters mechanical and electrical function. Cardiovasc. Res. 105,
Saccomanno, G., 1943. The components of the upper thoracic sympathetic nerves. J. 143–150.
Comp. Neurol. 79, 355–378. Wharton, J., et al., 1990. Immunohistochemical demonstration of human cardiac in-
Saji, M., Obata, K., 1981. Stimulus-dependent labeling of cultured ganglionic cell with nervation before and after transplantation. Circ. Res. 66, 900–912.
[14C]2-deoxyglucose. Brain Res. 212, 435–446. White, I.A., Gordon, J., Balkan, W., Hare, J.M., 2015. Sympathetic reinnervation is re-
Schemann, M., Sann, H., Schaaf, C., Mäder, M., 1993. Identification of cholinergic neu- quired for mammalian cardiac regeneration. Circ. Res. 117, 990–994.
rons in enteric nervous system by antibodies against choline acetyltransferase. Am. J. Wieland, D.M., et al., 1981. Myocardial imaging with a radioiodinated norepinephrine
Phys. 265, G1005–1009. storage analog. J. Nucl. Med. 22, 22–31.
Schwaiger, M., et al., 1990. Noninvasive evaluation of sympathetic nervous system in Yamakawa, K., et al., 2015. Vagal nerve stimulation activates vagal afferent fibers that
human heart by positron emission tomography. Circulation 82, 457–464. reduce cardiac efferent parasympathetic effects. Am. J. Physiol. Heart Circ. Physiol.
Schwartz, P.J., 2014. Cardiac sympathetic denervation to prevent life-threatening ar- 309, H1579–1590.
rhythmias. Nat. Rev. Cardiol. 11, 346–353. Yang, B., et al., 2014. Single-cell phenotyping within transparent intact tissue through
Shen, M.J., Zipes, D.P., 2014. Role of the autonomic nervous system in modulating car- whole-body clearing. Cell 158, 945–958.
diac arrhythmias. Circ. Res. 114, 1004–1021. Yizhar, O., Fenno, L.E., Davidson, T.J., Mogri, M., Deisseroth, K., 2011. Optogenetics in
Shen, M.J., Zipes, D.P., 2015. Interventional and device-based autonomic modulation in neural systems. Neuron 71, 9–34.
heart failure. Heart Fail. Clin. 11, 337–348. Yuan, B.X., Ardell, J.L., Hopkins, D.A., Losier, A.M., Armour, J.A., 1994. Gross and mi-
Shivkumar, K., et al., 2016. Clinical neurocardiology defining the value of neuroscience- croscopic anatomy of the canine intrinsic cardiac nervous system. Anat. Rec. 239,
based cardiovascular therapeutics. J. Physiol. 594, 3911–3954. 75–87.
Sisson, J.C., et al., 1988. Scintigraphic detection of regional disruption of adrenergic Zemelman, B.V., Lee, G.A., Ng, M., Miesenböck, G., 2002. Selective photostimulation of
neurons in the heart. Am. Heart J. 116, 67–76. genetically charged neurons. Neuron 33, 15–22.

58

Descargado para Anonymous User (n/a) en Universidad Libre de Colom de ClinicalKey.es por Elsevier en mayo 23, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.

Você também pode gostar