You are on page 1of 11

Clinical Anatomy 22:36–46 (2009)

REVIEW

Autonomic Innervation of the Developing Heart:


Origins and Function
VICTORIA HILDRETH,* ROBERT H. ANDERSON, AND DEBORAH J. HENDERSON
Institute of Human Genetics, Newcastle University, Centre for Life, Central Parkway,
Newcastle upon Tyne, United Kingdom

Maintenance of homeostatic circulation in mammals and birds is reliant upon


autonomic innervation of the heart. Neural branches of mixed cellular origin
and function innervate the heart at the arterial and venous poles as it
matures, eventually coupling autonomic output to the cardiac components,
including the conduction system. The development of neural identity is con-
trolled by specific networks of genes and growth factors, whereas functional
properties are governed by the use of different neurotransmitters. In this
review, we summarize briefly the anatomic arrangement of the vertebrate au-
tonomic nervous system and describe, in detail, the innervation of the heart.
We discuss the timing of cardiac innervation in the chick and mouse, empha-
sizing the relationship of the cardiac neural networks to the anatomical struc-
tures within the heart. We also discuss the variable contribution of the neural
crest to vagal cardiac nerves, and summarize the main neurotransmitters
secreted by the developing sympathetic and parasympathetic autonomic
divisions. We provide an overview of the main growth factor and gene fami-
lies involved in neural development, discussing how these factors may
impact upon the development of cardiac abnormalities in congenital syn-
dromes associated with autonomic dysfunction. Clin. Anat. 22:36–46,
2009. V 2008 Wiley-Liss, Inc.
C

Key words: cardiac development; neurons; neural crest; cardiac plexus;


cardiac ganglia; neurotransmitters

ORGANIZATION OF THE VERTEBRATE Sympathetic efferent nerves containing pregan-


NERVOUS SYSTEM glionic neurons arise segmentally from the central
portion of the spine, and terminate in sympathetic
The vertebrate nervous system is divided into two ganglia, which are organized into two bilateral chains
major components: the central nervous system, con- that run parallel to the spinal cord. Here, they syn-
sisting of the spinal cord and brain, and the periph- apse with postganglionic sympathetic neurons that
eral nervous system, which can be subdivided into travel to the effector organ (Fig. 1). These neurons,
somatic and autonomic components. The autonomic in the case of the heart, serve to increase heart rate
division of the peripheral system exerts functional and contraction. The main neurotransmitter of the
control over internal organs by way of efferent nerve sympathetic nervous system is norepinephrine,
fibers originating in the central system. Sympathetic
and parasympathetic efferent nerves generate com-
plementary motor outputs in response to sensory *Correspondence to: Victoria Hildreth, Institute of Human Genet-
inputs directed to the brain from internal organs via ics, Newcastle University, Centre for Life, Central Parkway, New-
castle upon Tyne, United Kingdom.
mechanoreceptive and chemoreceptive afferent
E-mail: Victoria.Hildreth@ncl.ac.uk
neurons. Both divisions of the autonomic system use
preganglionic to postganglionic relays, although Received 27 March 2008; Accepted 2 July 2008
they differ in their organization and biochemical Published online 8 October 2008 in Wiley InterScience (www.
properties. interscience.wiley.com). DOI 10.1002/ca.20695

C 2008
V Wiley-Liss, Inc.
Autonomic Innervation of the Developing Heart 37

nomic output in the heart, and are thought to have


alternative functions. The majority of sensory
impulses are relayed to motor neurons within the
central system. Parasympathetic afferent pathways
transverse the nodose ganglion, connecting to effer-
ent pathways via interneurons located within the
brainstem. Sympathetic afferent nerves travel
through the stellate ganglia of the sympathetic
chain, and connect with motor neurons in the spinal
cord. Sympathetic afferent reflex responses can also
occur through direct interactions with motor nerves
in the stellate ganglia, while it is thought that some
sensory afferent pathways are intrinsic to the heart
and can direct reflex responses through sympathetic
and parasympathetic neurons and interneurons
located in the network of cardiac ganglia.

INNERVATION OF THE HEART


Parasympathetic activity in the heart is controlled
by the vagus nerve, which originates in the medulla
oblongata. In the human, it is the superior, inferior,
and thoracic branches of the vagus nerve that inner-
vate the heart (Kawashima, 2005). In the chick, car-
diac branches of the developing right and left para-
sympathetic vagal nerves pass along the arteries
within the 6th pharyngeal arches before entering
into the arterial pole of the heart between the aorta
and pulmonary trunk (Kuratani and Tanaka, 1990;
Verberne et al., 1998). Staining with an antibody
against HNK1, a carbohydrate moiety originally iden-
Fig. 1. Innervation of the heart. The heart is inner-
tified on human natural killer cells (Abo et al., 1982),
vated by sympathetic (dark grey) and parasympathetic
has shown that innervation of the outflow tract takes
(black) efferent nerves that originate from the spinal cord
place at stage 29 of Hamburger and Hamilton (HH).
and brain stem respectively. Afferent nerves (dashed
More caudally, positive HNK1 staining is first
lines) also utilize these pathways to relay sensory inputs
observed around the 24th stage in the dorsal meso-
from the heart to the brain. Pre- to post-ganglionic sym-
cardium, reaching the systemic venous sinus by the
pathetic transfer occurs in the sympathetic ganglion
26th stage, and forming the cardiac sinal branch of
chain adjacent to the spinal cord. Post-ganglionic sympa-
the vagus nerve (Kuratani and Tanaka, 1990; Ver-
thetic neurons travel to the cardiac plexus, where pre-
berne et al., 1998). Between the 28th and 29th
ganglionic parasympathetic neurons synapse to post-
stages, further branches from the vagus extend to-
ganglionic elements. From the cardiac plexus, autonomic
ward the arterial and venous pole, entering along
nerves can synapse with intrinsic cardiac neurons (light
the arteries coursing through the 3rd and 4th pha-
grey), located within the network of cardiac ganglia.
ryngeal arches, and the right and left cardinal veins,
respectively (Verberne et al., 1998). In the mouse,
nerve cells are first seen in the dorsal mesocardium
although acetylcholine is used by sympathetic post- at E10.5 (10.5 days after fertilization). This is
ganglionic neurons. approximately equivalent to HH20 in the chick. Well-
Postganglionic efferent nerves of the parasympa- developed nerve tracts can be identified using the
thetic division differ in that they arise from the brain neurofilament marker NF160D that extend through
stem and spinal cord and synapse with post- this region and reach the heart by E12.5 (HH24),
ganglionic neurons located near to, or within, the (Hildreth et al., 2008) (Fig. 2). As in the chick heart,
effector organ (Fig. 1). Acetylcholine is the main innervation of the outflow tract also occurs slightly
neurotransmitter used in the preganglionic and post- later in the mouse, with the first neural elements
ganglionic neurons of the parasympathetic system, first seen between the separating aorta and pulmo-
with parasympathetic activity slowing the heart rate nary trunk at E11.5 (Hildreth et al., 2008) (Fig. 3).
and reducing the force of contraction. Sympathetic innervation has been shown to occur
Sensory afferent pathways involve both sympa- at later stages compared with parasympathetic
thetic and parasympathetic nerves to relay informa- innervation in both avian and mammalian species
tion from the organs to the brain (Fig. 1). Cardiac (Kirby et al., 1980; Shoba and Tay, 2000). Postgan-
afferent pathways made up of parasympathetic glionic sympathetic neurons join the vagus nerve at
nerves are responsible for controlling sympathetic the nodose ganglion, and then use the established
and parasympathetic output, while afferents associ- vagal tracts to gain entry into the heart. Studies in
ated with sympathetic nerves do not influence auto- the chick using silver staining, histofluorescence, and
38 Hildreth et al.

nerve fibers in comparison to neuropeptide-Y immu-


noactivity in the rat (Shoba and Tay, 2000).

THE CARDIAC PLEXUS AND THE


INTRINSIC CARDIAC NEURAL SUPPLY
The nerves supplying the heart join in the cardiac
plexus, an accumulation of mixed neurons located

Fig. 2. Innervation of the venous pole of the heart in


the mouse. a,b: NF160D antibody staining reveals the
presence of neurofilaments in the dorsal mesocardium at
E10.5. c,d: By E12.5, neurofilaments are seen extending
into the heart through the dorsal mesocardium and pri-
mary atrial septum toward the atrioventricular cushions.
AVC, atrioventricular cushion; LA, left atrium; LV, left
ventricle; DM, dorsal mesocardium; PAS, primary atrial
septum; RA, right atrium; RV, right ventricle; SG, sym-
pathetic ganglion; V, vagus nerve branches.

immunohistochemistry with the sympathetic nerve


marker tyrosine hydroxylase have determined that
bilateral sympathetic nerves from the first thoracic
ganglia, along with branches from the superior, mid-
dle, and lower cervical ganglia, all contribute to car-
diac innervation (Kirby et al., 1980; Verberne et al.,
1999). In the human, superior, middle, and inferior
cardiac nerves provide the supply to the heart, col-
lectively arising from regions of the sympathetic Fig. 3. At E11.5, NF160D staining reveals the pres-
chain spanning the superior cervical to the inferior ence of the first neurofilaments innervating the outflow
cervical ganglia (Kawashima, 2005). Additional sym- tract of the heart between the developing aorta and pul-
pathetic innervation is provided by nerves originating monary trunk (a). These nerves display parasympa-
from the region of the thoracic sympathetic trunk thetic and sympathetic activity, as shown using antibod-
below the inferior cervical ganglia (Kawashima, ies raised against VAChT (c) and TH (e). Nerves stained
2005). In the mouse, staining against tyrosine by NF160D innervating the venous pole of the heart at
hydroxylase and the parasympathetic neuron marker E12.5 (b), also show parasympathetic and sympathetic
vasoactive cholinesterase transporter protein activity (d, f), although the cardiac plexus displays a
(VAChT) reveals the presence of sympathetic and notable reduction in TH staining (f) when compared
parasympathetic neurons within the main nerves with VAChT staining (d), indicating a smaller proportion
innervating the arterial and venous poles of the of sympathetic nerves in the cardiac plexus at this
heart at E11.5 and E12.5 (Hildreth et al., 2008) stage. Ao, aorta; CP, cardiac plexus; NF160D, neurofila-
(Fig. 3). Autonomic efferent innervation precedes ment160D; PT, pulmonary trunk; TH, tyrosine hydroxy-
sensory innervation, as shown by the later appear- lase; VAChT, vasoactive cholinesterase transporter pro-
ance of calcitonin gene-related peptide-positive tein; V, vagus nerve branches.
Autonomic Innervation of the Developing Heart 39

believed to develop in response to cardiac innerva-


tion from migratory neural precursor cells that travel
along the established nerve tracts.
Recent studies have shown the importance of the
intrinsic cardiac ganglia in modulating relay between
extrinsic autonomic nerves and the nodal tissues of
the cardiac conduction system. Specifically, auto-
nomic inputs from the left and right vagal branches
are regulated to varying degrees by different cardiac
ganglia, revealing complex interlinking pathways in
the control of heart rate. These pathways have been
Fig. 4. a: At E14.5, parasympathetic nerves are
revealed in studies of atrial fibrillation, which can be
abundant in the cardiac plexus, as shown by VAChT
induced through electrical stimulation of autonomic
antibody staining. b: TH staining shows that sympa-
nerves or cardiac ganglia. (Patterson et al., 2005;
thetic nerves are present but are much reduced in the
Scherlag et al., 2005a,b). A study in the dog (Hou
cardiac plexus at the same stage, reflecting similar find-
et al, 2007) revealed that the right and left vagal
ings at earlier embryonic stages. CP, cardiac plexus;
trunks exert sympathetic control over heart rate, but
TH, tyrosine hydroxylase; VAChT, vasoactive cholines-
that both inputs are modulated through specific,
terase transporter protein; V, vagus nerve branches. interlinking pathways, with these pathways differing
between each node (Fig. 6). There is, however, a

cranial and dorsal to the heart (Fig. 1). In the human


heart, left and right-sided cardiac plexuses surround
the brachiocephalic trunk and the aortic arch,
respectively, and form part of a larger cardiac plexus
that lies between the aorta and pulmonary trunk
(Kawashima, 2005). Immunohistochemical staining
against VAChT has shown that parasympathetic neu-
rons are abundant the murine cardiac plexus at
E12.5 and later stages, with a smaller number of
sympathetic neurons present, visualized with anti-
body staining to tyrosine hydroxylase (Hildreth
et al., 2008) (Figs. 3 and 4). The cardiac plexus also
contains sensory afferent elements associated with
these autonomic neural components (Crick et al.,
2000). From the cardiac plexus, the majority of au-
tonomic neurons travel between the aorta and pul-
monary trunk, extending along the coronary arteries
before branching to innervate the atria and ven-
tricles (Kawashima, 2005). Neurons can innervate
the heart directly, or synapse with intrinsic inter-
neurons located in cardiac ganglia that are distri-
buted within the neuronal network of the heart
(Fig. 5). Cardiac ganglia differ in number and distri-
bution between species, but are mostly found within
the subepicardial layer. In mammals, the cardiac
ganglia are predominantly located around the atria
at the roots of caval and pulmonary veins, found
particularly within the infolded atrial walls often
described inappropriately as the ‘‘septum secun-
dum,’’ this area being better described as the supe- Fig. 5. Schematic representation of the locations of
rior interatrial fold. From these ganglia, nerves cardiac ganglia in the human heart. a: Cross sectional
extend to reach the sinus and atrioventricular nodes view of the heart showing cardiac ganglia (white circles)
of the conduction system (Armour et al., 1997; Bap- distributed throughout the atria. Ganglia can be found
tista and Kirby, 1997). In the human, it is estimated near the nodal tissues of the conduction system and
that there are, on average, around 700–900 cardiac around the entry points of the pulmonary veins and
ganglia, contained within seven separate subplex- caval veins. b: Rostral view of the heart showing cardiac
uses located in different regions of the heart. The left ganglia clustered around the aorta, pulmonary trunk,
atrium contains three of the subplexuses. Around and pulmonary veins, as well as the superior caval vein.
half of all the cardiac ganglia are located on the dor- Ao, Aorta; AVN, atrioventricular node; ICV, inferior
sal and dorsolateral surface of this chamber (Pauza caval vein; LA, left atrium; LV, left ventricle; PT, pulmo-
et al., 2000). Morphologically distinct at stage 35 in nary trunk; PV, pulmonary veins; RA, right atrium; RV,
the chick (Verberne et al., 1998), the cardiac ganglia right ventricle; SCV, superior caval vein; SN, sinus
and their associated postganglionic fibers are node.
40 Hildreth et al.

of parasympathetic nerves from the neural crest


were first suggested in early ablation studies, when
disrupted cardiac innervation was observed in addi-
tion to defects within the developing outflow tracts
(Kirby and Stewart, 1983; Kirby et al., 1985). Using
chimeral and retroviral methods in the chick, it was
shown that the precursors gained entry to the heart
by migration along the arterial and sinal branches of
the vagus nerve (Kirby and Stewart, 1983; Poel-
mann et al., 1998; Verberne et al., 1998, 2000). The
advent of reliable systems for marking the neural
crest has since confirmed that the cardiac ganglia
also take their origin from the neural crest in mam-
mals (Waldo et al., 1999; Pietri et al., 2003).
Compensation from other parts of the neural crest
has been suggested for the limited loss of parasym-
pathetic activity in response to ablation of the car-
diac component of the crest in the chick (Kirby et al.,
1987). In embryos where the cardiac neural crest
has been ablated; however, chimeral marking has
shown that parasympathetic neurons persisting in
the heart originate from the nodose placode, empha-
sizing the ability of nonneural crest-derived precur-
sors to form functional cholinergic neurons within the
heart (Kirby, 1988). Wnt1-cre lineage tracing of neu-
ral crest cells, in combination with NF160D staining,
in the mouse has shown that the vagal nerves con-
Fig. 6. Schematic drawing showing variable modu- tain a large proportion of nonneural crest-derived
lation of sympathetic (vagal) control of sinus node and neurons in addition to neurons of neural crest cell or-
atrioventricular node rate by three cardiac ganglionated igin, supporting a role for neurons derived from
plexuses in the heart. Control of the sinus node (grey sources other than the neural crest in parasympa-
arrows) predominantly occurs through the anterior right thetic relay (Hildreth et al., 2008) (Fig. 8). In the
ganglionated plexus, which receives similar inputs from cranial region, sensory ganglia within the vagus
the right vagal branch and the left vagal branch directly, nerve are also of mixed origin, containing neurons
and via the superior left ganglionated plexus. Control of derived from the neural crest and the nodose pla-
the atrioventricular node (dashed arrows) predomi- code (D’Amico-Martel and Noden, 1983).
nantly occurs through the inferior right ganglionated Sympathetic cardiac innervation is thought to
plexus. The majority of input from the left and right originate from more caudal levels of the neural crest
vagal branches is directed through the superior left gan- than do the parasympathetic nerves (Kirby and
glionated plexus to the anterior right ganglionated Bockman, 1984; Kirby et al., 1987). The loss of sym-
plexus before modulation by the inferior right ganglion- pathetic innervation is accompanied by an expansion
ated plexus. ARGP, anterior right ganglionated plexus; of the parasympathetic population, suggesting a
AVN, atrioventricular node; IRGP, inferior right gangli- possible compensatory effect (Kirby et al., 1987). In
onated plexus; LV, left vagus nerve; RV, right vagus the Splotch2H mouse, development of the vagal
nerve; SLGP, superior left ganglionated plexus; SN, nerves, cardiac plexus, and innervation at the ve-
sinus node. Figure adapted from Hu et al. (2007). nous pole of the heart appears normal (Fig. 9), de-
spite dramatic reductions in the number of cardiac
neural crest cells reaching the outflow tract, support-
ing a possible alternative origin for sympathetic and
degree of variation in these pathways between dif- parasympathetic elements at the venous pole (Hil-
ferent individuals. Failure completely to attenuate dreth et al., 2008).
the autonomic responses following ablation of these
ganglia suggested that the modulation involves other
ganglia within the cardiac network (Hou et al.,
2007). NEUROTRANSMITTERS IN CARDIAC
INNERVATION
CARDIAC INNERVATION Regulation of cardiac function by the autonomic
AND THE NEURAL CREST nervous system plays crucial roles in the response of
the organism to external stimulation. The sympa-
The majority of the autonomic nervous system is thetic nervous system increases heart rate and the
thought to arise from the neural crest (Fig. 7), with force of contraction via effects on the function of the
additional contributions from other sources to the sarcoplasmic reticulum within the cardiomyocytes
glia and a proportion of the neurons of the sensory and on ion channel activity. The parasympathetic
ganglia (D’Amico-Martel and Noden, 1983). Origins system, in contrast, counteracts the action of the
Autonomic Innervation of the Developing Heart 41

Fig. 9. Neural crest cell derivatives at the venous


pole of the Splotch2H (Sp2H) mouse. a: Wnt1-cre neural
crest cell lineage tracing in the Sp2H heterozygote
mouse, which undergoes normal development, reveals
a large contribution of neural crest cells in the cardiac
plexus and in the nerves innervating the venous pole of
the heart at E13.5 (blue). b: In the Sp2H homozygote
mutant mouse, which is null for the Pax3 gene and
shows reduced numbers of neural crest cells at the arte-
rial pole of the heart in addition to cardiovascular
defects, neural crest cell contributions to the cardiac
plexus and cardiac innervation appear similar to that of
Fig. 7. Neural crest contributions to the heart. The the control at the venous pole, indicating that the neural
neural crest (dark blue) is divided into cranial, cardiac, crest origin of nerves in this region may differ from
vagal, trunk, and sacral segments. Cells from the cardiac those innervating the arterial pole. CP, cardiac plexus;
division of the neural crest, located between the otic vesi- V, vagus nerve branches.
cle and the 3rd somite, migrate to the heart via the 3rd,
4th, and 6th pharyngeal arches and are involved in out- sympathetic nerves, slowing the heart rate. Under
flow tract septation and arch artery remodeling. Neural normal conditions, the balance between these two
crest cells also contribute to the formation of major auto- opposing systems is well maintained.
nomic nerves as well as the cardiac plexus and the car- The local environment plays an important role in
diac ganglia. Vagus nerve formation and parasympa- determining the specific neurotransmitter used by
thetic activity is disturbed when the cardiac neural crest presumptive autonomic postganglionic neurons.
is ablated, and cardiac ganglia are reduced in size and Most sympathetic neurons express norepinephrine as
number, suggesting overlap between the vagal and car- a primary neurotransmitter, along with a range of
diac divisions. Cells from the trunk neural crest contrib- peptides that modulate signaling. Norepinephrine
ute to the formation of sympathetic nerves. binds to a range of adrenergic receptors, broadly
classified as a and b. Under normal circumstances, a
high affinity b-adrenoceptor binds to multiple G pro-
teins, activating adenylyl cyclase to produce cAMP.
This then improves excitation-contraction coupling
within the cardiac muscle, increasing heart rate and
force. Norepinephrine is produced from its precursor,

Fig. 8. Contributions to cardiac innervation from


the neural crest and the nodose placode. a: Cells from
the hindbrain region of the cranial neural crest, the car-
diac neural crest, and the vagal neural crest contribute
to the formation of parasympathetic nerves and the car-
diac ganglia, as well some sensory nerves. Precursors
from the trunk neural crest form sympathetic nerves
that innervate the heart. The ectodermal nodose pla-
code also gives rise to sensory neural elements, and
may also contribute to the formation of parasympa-
thetic cardiac nerves. b: Wnt1-cre neural crest cell line-
age tracing in the embryonic mouse reveals that the
vagus nerve contains a mixed population of cells. A pro-
portion of the vagus nerve is derived from the neural
crest (blue), but many neurofilaments (brown, NF160D
antibody) do not stain blue, indicating an alternative or-
igin. These elements may be placodally derived. NP,
nodose placode.
42 Hildreth et al.

L-tyrosine, by the action of three enzymes, tyrosine ventricular nodes (Potter, 1987). Neuropeptide Y has
hydroxylase, dopamine b-hydroxylase, and phenyle- been suggested to play a crucial role as a neuromo-
thanolamine N-methyl transferase, all found within dulator in the complex interactions that take place
the cell body of sympathetic neurons. Once pro- between different branches of the sensory and auto-
duced, the norepinephrine is transported along the nomic innervation of the heart (reviewed in Crick
axon to nerve terminals where it is stored. In all spe- et al., 2000).
cies studied to date, maturation of the sympathetic Acetylcholine is the main neurotransmitter
innervation and onset of function occurs late in the released by preganglionic and postgangionic vagal
fetal, or early in the neonatal, heart. Despite this, nerve terminals. Two types of cholinergic receptor
response to catecholamines, in the form of tachycar- have been described, namely muscarinic and nico-
dia, occurs well before birth, suggesting that b-adre- tinic. Muscarinic receptors are the main type found
nergic receptors must be present during the fetal on cardiac effector cells, whereas nicotinic receptors
period, perhaps as early as E9.5 in the mouse (Liu are found on postganglionic neurons. Acetylcholine is
et al., 1999). Indeed, although few tyrosine hydrox- mainly synthesized at the nerve endings where it is
ylase-positive neurons are found in the heart until stored until release (Loffelholz and Pappano, 1985).
late in gestation, myocardial cells expressing tyro- Acetycholine can generally be detected in the heart
sine hydroxylase, dopamine b-hydroxylase, and phe- at an earlier stage than can norepinephrine, reflect-
nylethanolamine N-methyl transferase are found ing the earlier onset of parasympathetic innervation
interspersed throughout the myocardium in the than sympathetic ingrowth (Pappano, 1977).
mouse early in its gestation. By mid-gestation, they Vasoactive intestinal peptide is a peptidergic
have localized to the regions of formation of the cotransmitter in cholinergic parasympathetic neu-
sinus and atrioventricular nodes (Ebert and Thomp- rons. Neurons expressing this peptide are localized
son, 2001). These data are supported by the finding primarily in relation to blood vessels in microvascular
that, following sympathectomy, tyrosine hydroxylase beds (Della et al., 1983), and to the sinus and atrio-
activity can still be detected in the chick heart ventricular nodes and coronary vessels (Weihe et al.,
(Stewart and Kirby, 1985). Although the roles of 1984). The peptide has direct effects on heart rate.
these myocardially produced catecholamines remain In the dog, it has been reported to be twice as
unclear, the fact that the heart can respond to these potent as norepinephrine in increasing heart rate
factors from early in gestation suggests that they (Rigel, 1988). It has also been suggested to modu-
may be playing an important role. late the activity of acetylcholine and norepinephrine
A wide range of cotransmitters are found within (Ferron et al., 1985).
the sympathetic nervous system, functioning along- Somatostatin immunoreactivity is associated with
side norepinephrine. Although the mechanisms postganglionic parasympathetic neurons, localizing
underlying the specification of these cotransmitters to nerve fibers in the myocardium, endocardium,
remain unclear, cell lineage plays an important role. and the conduction system. The actions of somato-
Of these cotransmitters, the adenosinergic system, statin, slowing the heart rate and decreasing cardiac
using adenosine, is the earliest functionally respon- output, closely resemble those caused by vagal stim-
sive system in the rodent heart, with activation of ulation. It exerts its suppressing cardiac effects by
the adenosine receptor slowing the heart rate from acting on calcium channels excited by b-adrenocep-
E8 in the rat embryo (Porter and Rivkees, 2001). tor activity (Diez et al., 1985) and/or by inducing the
Adenosine acts to inhibit norepinephrine release release of acetylcholine from intracardiac parasym-
from sympathetic nerve endings and is important pathetic neurons (Wiley et al., 1989), but may also
both as a vasodilator and for its anti-arrhythmic modulate sympathetic neurotransmission by acting
properties. Similar to catecholamines, cardiac adeno- at a postjunctional site.
sine receptors are functional well before sympathetic The neurotransmitters substance P and calcitonin
innervation occurs. It has been suggested that fetal gene-related peptide act on the heart via the sen-
plasma may be the source early in gestation (Sawa sory-motor pathway. Both transmitters are stored in
et al., 1991). Neuropeptide Y is the most abundant granules within the nerve terminal, are coreleased
peptide in the heart, being found in postganglionic on stimulation, and interact with parasympathetic
sympathetic neurons synapsing on postganglionic nerves (Armour et al., 1993). Substance P-contain-
parasympathetic neurons in the cardiac ganglia, en- ing nerve fibers are found surrounding blood vessels
docardium, and myocardium (Palmiter et al., 1998). within the myocardium, and within the sinus and
It has been shown to inhibit release of catechol- atrioventricular nodes (Crick et al., 1994). Substance
amines and regulate the vasoconstrictor action of P, a potent vasodilator, although it has no effect on
norepinephrine. Neuropeptide Y has also been shown heart rate, may control parasympathetic activity
to produce vasoconstriction of coronary vessels in (Corr, 1992). Calcitonin gene-related peptide is
many species, including human (Michel et al., 1989). scarce in the human heart (Crick et al., 1994). It is a
Neuropeptide Y is first seen late in development in potent vasodilator of peripheral blood vessels,
the region of the sinus and atrioventricular nodes including the coronary arteries. In the mouse, it is
and in the intrinsic cardiac ganglia. Levels increase in expressed in sensory nerves within the heart,
the immediate postnatal period, and then remain appearing around the time of birth. These, with other
steady thereafter. Some studies have suggested that data, suggest that the sensory innervation of the
the peptide can produce long-term inhibition of the heart occurs later than autonomic innervation
responses to vagal stimulation in the sinus and atrio- (Gordon et al., 1993).
Autonomic Innervation of the Developing Heart 43

The myocardium also secretes endocrine factors sympathetic neuron precursors is disturbed (Eno-
that regulate the cardiac nervous system. The two moto et al., 2001) Normal control of these precur-
most abundantly expressed proteins are the atrial sors may be controlled by the activity of the GDNF
and brain natriuretic peptides, which exert an inhibi- family member artemin, expressed outside of the
tory effect on sympathetic input to the heart. These nervous system (Enomoto et al., 2001).
factors are mainly expressed in the atria, although Another growth factor known to affect neural de-
they are also expressed in other regions of the heart. velopment is neuregulin-1 (NRG1), which signals
They have additional roles, such as diuretic and na- through heterodimers of ErbB tyrosine kinase recep-
triuretic homeostatic function in the kidney, and in- tors, principally ErbB2/ErbB3 or ErbB2/ErbB4 (Car-
hibitory effects on the renin-angiotensin-aldosterone raway and Cantley, 1994). Studies on the develop-
system (McGrath et al., 2005). ment of the sympathetic ganglia in NGR1 / ,
A number of other nonadrenergic noncholinergic ErbB2 / , and ErbB3 / mice also reveal a critical
transmitters have been localized within the heart of role for NGR1 in the migration of neural crest cells
various species, including opioids, neurotensin, and destined to develop a sympathetic neuronal pheno-
peptide histidine isoleucine, albeit that their actions type (Britsch et al., 1998).
remain poorly defined. They will not be discussed Basic helix-loop-helix (HLH) genes are important
further here. For a more detailed review of these in the early determination of neural cell fate and
transmitters, see Crick et al. (2000). neural identity. In the central and peripheral compo-
nents of the nervous system in mammals, the HLH
gene Mash1, and another HLH gene neurogenin-1
GENES AND GROWTH FACTORS (Ngr-1), are expressed in complementary fashion,
and promote the development of autonomic and sen-
REGULATING NEURAL DEVELOPMENT sory neurons, respectively (Ma et al., 1996, 1998;
Development of the peripheral nervous system is Lo et al., 1998, 1999). Mash1 is necessary for the
reliant upon the interactions of several genes and development of all sympathetic and parasympathetic
growth factor families. The neurotrophin family of neurons and, in vitro, is expressed in neural crest
growth factors are particularly important, influencing cells, which display neuronal commitment but not a
autonomic and sensory neurogenesis and function neuronal phenotype, suggesting that it promotes dif-
(Huang and Reichardt, 2001). Nerve growth factor ferentiation of neuronal precursors rather than multi-
(NGF), brain-derived neurotrophic factor (BDNF), potent uncommitted cells (Sommer et al., 1995).
and neutrophins 3 and 4 (NT3 and NT4) make up the Proneural genes such as Mash1 also appear to have
family, acting through the high-affinity Trk receptor their activities balanced by negative regulators dur-
tyrosine kinases A, B, and C, and lower-affinity neu- ing normal neurogenesis. An example of a negative
rotrophin receptor p75 (p75NTR). regulator is Hes1. Hes1 null mice exhibit acceleration
NGF primarily acts through TrkA, and promotes of neural development, which is associated with the
growth, survival, and maintenance of sympathetic upregulation of Mash1 and other proneuronal factors
nerves (Crowley et al., 1994; Ieda et al., 2006). It is (Ishibashi et al., 1995; Hatakeyama et al., 2004). It
expressed by cardiomyocytes, and its overexpres- inhibits the function of Mash1 at the protein level
sion in the hearts of mice results in hyperinnervation (Sasai et al., 1992; Chen et al., 1997).
(Hassankhani et al., 1995). Its function is independ- Early development of sensory lineages is con-
ent of its role in neuronal survival (Glebova and trolled by the neurogenin (Ngn) family (Ma et al.,
Ginty, 2004). BDNF also supports the survival of a 1999). Early lineage specification of multipotent neu-
subset of sensory neurons and is released by sympa- ral crest cells is influenced by genes outside of the
thetic nerves (Causing et al., 1997; Kohn et al., HLH family such as Sry-related HMG box (Sox)10. In
1999; Jahed and Kawaja, 2005). vitro, Sox10 can prevent Tgf-b-induced smooth mus-
Like NGF, NT3 is implicated in the development cle differentiation of neural crest progenitors in favor
and maintenance of sensory and sympathetic neural of neuronal differentiation (Cheng et al., 2000;
elements before birth, and for sympathetic innerva- Britsch et al., 2001; Kim et al., 2003).
tion after birth (Farinas et al., 1994; ElShamy and
Ernfors, 1996a,b; ElShamy et al., 1996). NT3 is also
thought to act as a target-derived growth factor in CONGENITAL DISORDERS RELATED
mature neurons (Zhou et al., 1997, Zhang and Rush, TO ABNORMALITIES IN
2001). The increase in sensory ganglion loss in CARDIAC INNERVATION
TrkC / mice in comparison to NT3 / mice sug-
gests that this growth factor signals through other Dysautonomias are a group of diseases that result
receptors during neural development (Tessarollo from impaired autonomic interactions. The most well
et al., 1997). known of these is familial dysautonomia (Riley-Day
The glial cell line-derived (GDNF) family of neuro- syndrome; OMIM 223900). A decrease in the synthe-
receptors are also implicated in neural development, sis of norepinephrine has been reported in patients
signaling through a complex of RET tyrosine kinase with familial dysautonomia, possibly due to reduced
and GDNF family (GFR)a receptors (Hiltunen et al., levels of dopamine-beta-hydroxylase, the enzyme
2000; Mabe et al., 2006). Both are required for nor- that converts dopamine to norepinephrine. Some
mal parasympathetic innervation of the organ (Hil- dysautonomic children had no plasma dopamine-
tunen et al., 2000). In Ret / mice, migration of beta-hydroxylase activity and their mothers had
44 Hildreth et al.

decreased activity (Goodall et al., 1971). In 2001, the mal numbers but are abnormal in function and morphology.
syndrome was shown to result from mutations in the J Clin Invest 70:193–197.
IKBKAP gene (Anderson et al., 2001; Slaugenhaupt et Anderson SL, Coli R, Daly IW, Kichula EA, Rork MJ, Volpi SA, Ekstein
J, Rubin BY. 2001. Familial dysautonomia is caused by mutations
al., 2001). Depletion has been associated with
of the IKAP gene. Am J Hum Genet 68:753–758.
reduced transcription of a number of genes and defec- Armour JA, Huang MH, Smith FM. 1993. Peptidergic modulation
tive cell motility (Hawkes et al., 2002; Close et al., of in situ canine intrinsic cardiac neurons. Peptides 14:191–
2006). Patients with familial dysautonomia have an 202.
increased risk of sudden death and have abnormal or- Armour JA, Murphy DA, Yuan BX, Macdonald S, Hopkins DA. 1997.
thostatic blood pressure, along with responses of Gross and microscopic anatomy of the human intrinsic cardiac
heart rate and cardiac tone. Sudden Infant Death nervous system. Anat Rec 247:289–298.
Syndrome (SIDS) has also been linked to abnormal Baptista CA, Kirby ML. 1997. The cardiac ganglia: Cellular and mo-
development of the autonomic nervous system. SIDS lecular aspects. Kaohsiung J Med Sci 13:42–54.
Britsch S, Li L, Kirchhoff S, Theuring F, Brinkmann V, Birchmeier C,
is a multifactorial condition thought to be caused by
Riethmacher D. 1998. The ErbB2 and ErbB3 receptors and their
abnormalities in the control of the heart, breathing, ligand, neuregulin-1, are essential for development of the sym-
autonomic nervous system, and patterns of arousal pathetic nervous system. Genes Dev 12:1825–1836.
during sleep. Physiological studies suggest that there Britsch S, Goerich DE, Riethmacher D, Peirano RI, Rossner M, Nave
may be a delay in the maturation of the autonomic KA, Birchmeier C, Wegner M. 2001. The transcription factor
nervous system, which results in a decreased propen- Sox10 is a key regulator of peripheral glial development. Genes
sity to arouse from sleep and a decreased sympatho- Dev 15:66–78.
vagal cardiac balance (Kahn et al., 2003). Carraway KL III, Cantley LC. 1994. A neu acquaintance for erbB3
and erbB4: A role for receptor heterodimerization in growth sig-
naling. Cell 78:5–8.
Causing CG, Gloster A, Aloyz R, Bamji SX, Chang E, Fawcett J,
CLINICAL SIGNIFICANCE OF Kuchel G, Miller FD. 1997. Synaptic innervation density is regu-
CARDIAC INNERVATION lated by neuron-derived BDNF. Neuron 18:257–267.
Chen H, Thiagalingam A, Chopra H, Borges MW, Feder JN, Nelkin
From our review, it is clear that much more is BD, Baylin SB, Ball DW. 1997. Conservation of the Drosophila
known about the development of the cardiac nerves, lateral inhibition pathway in human lung cancer: A hairy-related
and their related growth factors and gene products, protein (HES-1) directly represses achaete-scute homolog-1
than is known about the specific distribution of the expression. Proc Natl Acad Sci USA 94:5355–5360.
autonomic nerves in the human heart. It is also Cheng Y, Cheung M, Abu-Elmagd MM, Orme A, Scotting PJ. 2000.
clear, nonetheless, that these autonomonic nerves Chick sox10, a transcription factor expressed in both early neu-
ral crest cells and central nervous system. Brain Res Dev Brain
play a crucial role in multiple diseases, such as the
Res 121:233–241.
congenital malformations discussed earlier, and Close P, Hawkes N, Cornez I, Creppe C, Lambert CA, Rogister B,
other anomalies such as atrial fibrillation, also dis- Siebenlist U, Merville MP, Slaugenhaupt SA, Bours V, Svejstrup
cussed previously. The role of disordered cardiac JQ, Chariot A. 2006. Transcription impairment and cell migration
innervation in syndromes such as cardiac failure, defects in elongator-depleted cells: Implication for familial dys-
however, remains to be elucidated. There is much to autonomia. Mol Cell 22:521–531.
be done, therefore, in matching the knowledge of Corr L. 1992. Neuropeptides and the conduction system of the
the distribution of the various elements of the auto- heart. Int J Cardiol 35:1–12.
nomic nervous system in clinical disease states to Crick SJ, Wharton J, Sheppard MN, Royston D, Yacoub MH, Ander-
son RH, Polak JM. 1994. Innervation of the human cardiac con-
that now existing for development of these nerves in
duction system. A quantitative immunohistochemical and histo-
experimental animals. chemical study. Circulation 89:1697–1708.
Crick SJ, Sheppard MN, Anderson RH. 2000. The nervous system
and the heart. Ter Horst GT, editor. Totowa, NJ: Humana Press.
SUMMARY Crowley C, Spencer SD, Nishimura MC, Chen KS, Pitts-Meek S,
Armanini MP, Ling LH, McMahon SB, Shelton DL, Levinson AD,
We have provided an account of autonomic and Philips HS. 1994. Mice lacking nerve growth factor display peri-
sensory innervation in the vertebrate heart, empha- natal loss of sensory and sympathetic neurons yet develop basal
sizing the importance of the intrinsic cardiac nerves forebrain cholinergic neurons. Cell 76:1001–1011.
in the control of cardiac conduction. We have D’Amico-Martel A, Noden DM. 1983. Contributions of placodal and
neural crest cells to avian cranial peripheral ganglia. Am J Anat
emphasized the contribution of the neural crest to
166:445–468.
the development of the autonomic nerves, along Della NG, Papka RE, Furness JB, Costa M. 1983. Vasoactive intesti-
with the roles of specifying genes, growth factors, nal peptide-like immunoreactivity in nerves associated with
and neurotransmitters in the establishment of car- the cardiovascular system of guinea-pigs. Neuroscience 9:605–
diac innervation. Future research into these factors, 619.
and the interactions between them, is imperative if Diez J, Tamargo J, Valenzuela C. 1985. Negative inotropic effect of so-
we are to elucidate the etiology of clinical disorders matostatin in guinea-pig atrial fibres. Br J Pharmacol 86:547–555.
associated with autonomic aberrations, and the Ebert SN, Thompson RP. 2001. Embryonic epinephrine synthesis in
potential significance of abnormal aoutonomic inner- the rat heart before innervation: Association with pacemaking
and conduction tissue development. Circ Res 88:117–124.
vation in clinical cardiac disease.
ElShamy WM, Ernfors P. 1996a. A local action of neurotrophin-3
prevents the death of proliferating sensory neuron precursor
REFERENCES cells. Neuron 16:963–972.
Elshamy WM, Ernfors P. 1996b. Requirement of neurotrophin-3 for
Abo T, Roder JC, Abo W, Cooper MD, Balch CM. 1982. Natural killer the survival of proliferating trigeminal ganglion progenitor cells.
(HNK-1+) cells in Chediak-Higashi patients are present in nor- Development 122:2405–2414.
Autonomic Innervation of the Developing Heart 45

ElShamy WM, Linnarsson S, Lee KF, Jaenisch R, Ernfors P. 1996. Kim J, Lo L, Dormand E, Anderson DJ. 2003. SOX10 maintains mul-
Prenatal and postnatal requirements of NT-3 for sympathetic tipotency and inhibits neuronal differentiation of neural crest
neuroblast survival and innervation of specific targets. Develop- stem cells. Neuron 38:17–31.
ment 122:491–500. Kirby ML. 1988. Nodose placode contributes autonomic neurons to
Enomoto H, Crawford PA, Gorodinsky A, Heuckeroth RO, Johnson the heart in the absence of cardiac neural crest. J Neurosci
EM Jr, Millbrandt J. 2001. RET signaling is essential for migra- 8:1089–1095.
tion, axonal growth and axon guidance of developing sympa- Kirby ML, Bockman DE. 1984. Neural crest and normal develop-
thetic neurons. Development 128:3963–3974. ment: A new perspective. Anat Rec 209:1–6.
Farinas I, Jones KR, Backus C, Wang XY, Reichardt LF. 1994. Severe Kirby ML, Stewart DE. 1983. Neural crest origin of cardiac ganglion
sensory and sympathetic deficits in mice lacking neurotrophin-3. cells in the chick embryo: Identification and extirpation. Dev Biol
Nature 369:658–661. 97:433–443.
Ferron A, Siggins GR, Bloom FE. 1985. Vasoactive intestinal poly- Kirby ML, McKenzie JW, Weidman TA. 1980. Developing innervation
peptide acts synergistically with norepinephrine to depress spon- of the chick heart: A histofluorescence and light microscopic
taneous discharge rate in cerebral cortical neurons. Proc Natl study of sympthetic innervation. Anat Rec 196:333–340.
Acad Sci USA 82:8810–8812. Kirby ML, Aronstam RS, Buccafusco JJ. 1985. Changes in cholinergic
Glebova NO, Ginty DD. 2004. Heterogeneous requirement of NGF for parameters associated with failure of conotruncal septation in
sympathetic target innervation in vivo. J Neurosci 24:743–751. embryonic chick hearts after neural crest ablation. Circ Res
Goodall MC, Gitlow SE, Alton H. 1971. Decreased noradrenaline 56:392–401.
(norepinephrine) synthesis in familial dysautonomia. J Clin Kirby ML, Conrad DC, Stewart DE. 1987. Increase in the cholinergic
Invest 50:2734–2740. cardiac plexus in sympathetically aneural chick hearts. Cell Tis-
Gordon L, Polak JM, Moscoso GJ, Smith A, Kuhn DM, Wharton J. sue Res 247:489–496.
1993. Development of the peptidergic innervation of human Kohn J, Aloyz RS, Toma JG, Haak-Frendscho M, Miller FD. 1999.
heart. J Anat 183 (Part 1):131–140. Functionally antagonistic interactions between the TrkA and p75
Hassankhani A, Steinhelper ME, Soonpaa MH, Katz EB, Taylor DA, neurotrophin receptors regulate sympathetic neuron growth and
Andrade-Rozental A, Factor SM, Steinberg JJ, Field LJ, Federoff target innervation. J Neurosci 19:5393–5408.
HJ. 1995. Overexpression of NGF within the heart of transgenic Kuratani S, Tanaka S. 1990. Peripheral development of the avian
mice causes hyperinnervation, cardiac enlargement, and hyper- vagus nerve with special reference to the morphological innerva-
plasia of ectopic cells. Dev Biol 169:309–321. tion of heart and lung. Anat Embryol (Berl) 182:435–445.
Hatakeyama J, Bessho Y, Katoh K, Ookawara S, Fujioka M, Guille- Liu W, Yasui K, Arai A, Kamiya K, Cheng J, Kodama I, Toyama J.
mot F, Kageyama R. 2004. Hes genes regulate size, shape 1999. Beta-adrenergic modulation of L-type Ca2+-channel cur-
and histogenesis of the nervous system by control of the tim- rents in early-stage embryonic mouse heart. Am J Physiol
ing of neural stem cell differentiation. Development 131: 276:H608–H613.
5539–5550. Lo L, Tiveron MC, Anderson DJ. 1998. MASH1 activates expression
Hawkes NA, Otero G, Winkler GS, Marshall N, Dahmus ME, Krapp- of the paired homeodomain transcription factor Phox2a, and
mann D, Scheidereit C, Thomas CL, Schiavo G, Erdjument- couples pan-neuronal and subtype-specific components of auto-
Bromage H, Tempst P, Svejstrup JQ. 2002. Purification and nomic neuronal identity. Development 125:609–620.
characterization of the human elongator complex. J Biol Chem Lo L, Morin X, Brunet JF, Anderson DJ. 1999. Specification of neuro-
277:3047–3052. transmitter identity by Phox2 proteins in neural crest stem cells.
Hildreth V, Webb S, Bradshaw L, Brown NA, Anderson RH, Henderson Neuron 22:693–705.
DJ. 2008. Cells migrating from the neural crest contribute to the Loffelholz K, Pappano AJ. 1985. The parasympathetic neuroeffector
innervation of the venous pole of the heart. J Anat 212:1–11. junction of the heart. Pharmacol Rev 37:1–24.
Hiltunen JO, Laurikainen A, Airaksinen MS, Saarma M. 2000. GDNF Ma Q, Kintner C, Anderson DJ. 1996. Identification of neurogenin, a
family receptors in the embryonic and postnatal rat heart and vertebrate neuronal determination gene. Cell 87:43–52.
reduced cholinergic innervation in mice hearts lacking ret or Ma Q, Chen Z, del Barco Barrantes I, de la Pompa JL, Anderson DJ. 1998.
GFRalpha2. Dev Dyn 219:28–39. Neurogenin1 is essential for the determination of neuronal precursors
Hou Y, Scherlag BJ, Lin J, Zhang Y, Lu Z, Truong K, Patterson E, for proximal cranial sensory ganglia. Neuron 20:469–482.
Lazzara R, Jackman WM, Po SS. 2007. Ganglionated plexi modu- Ma Q, Fode C, Guillemot F, Anderson DJ. 1999. Neurogenin1 and
late extrinsic cardiac autonomic nerve input: Effects on sinus neurogenin2 control two distinct waves of neurogenesis in devel-
rate, atrioventricular conduction, refractoriness, and inducibility oping dorsal root ganglia. Genes Dev 13:1717–1728.
of atrial fibrillation. J Am Coll Cardiol 50:61–68. Mabe AM, Hoard JL, Duffourc MM, Hoover DB. 2006. Localization of
Huang EJ, Reichardt LF. 2001. Neurotrophins: Roles in neuronal de- cholinergic innervation and neurturin receptors in adult mouse
velopment and function. Annu Rev Neurosci 24:677–736. heart and expression of the neurturin gene. Cell Tissue Res
Ieda M, Kanazawa H, Ieda Y, Kimura K, Matsumura K, Tomita Y, 326:57–67.
Yagi T, Onizuka T, Shimoji K, Ogawa S, Makino S, Sano M, McGrath MF, de Bold ML, de Bold AJ. 2005. The endocrine function
Fukuda K. 2006. Nerve growth factor is critical for cardiac sen- of the heart. Trends Endocrinol Metab 16:469–477.
sory innervation and rescues neuropathy in diabetic hearts. Cir- Michel MC, Wirth SC, Zerkowski HR, Maisel AS, Motulsky HJ. 1989.
culation 114:2351–2363. Lack of inotropic effects of neuropeptide Y in human myocar-
Ishibashi M, Ang SL, Shiota K, Nakanishi S, Kageyama R, Guillemot dium. J Cardiovasc Pharmacol 14:919–922.
F. 1995. Targeted disruption of mammalian hairy and Enhancer Palmiter RD, Erickson JC, Hollopeter G, Baraban SC, Schwartz MW.
of split homolog-1 (HES-1) leads to up-regulation of neural 1998. Life without neuropeptide Y. Recent Prog Horm Res
helix-loop-helix factors, premature neurogenesis, and severe 53:163–199.
neural tube defects. Genes Dev 9:3136–3148. Pappano AJ. 1977. Ontogenetic development of autonomic neuroef-
Jahed A, Kawaja MD. 2005. The influences of p75 neurotrophin re- fector transmission and transmitter reactivity in embryonic and
ceptor and brain-derived neurotrophic factor in the sympathetic fetal hearts. Pharmacol Rev 29:3–33.
innervation of target tissues during murine postnatal develop- Patterson E, Po SS, Scherlag BJ, Lazzara R. 2005. Triggered firing in
ment. Auton Neurosci 118:32–42. pulmonary veins initiated by in vitro autonomic nerve stimula-
Kahn A, Groswasser J, Franco P, Scaillet S, Sawaguchi T, Kelman- tion. Heart Rhythm 2:624–631.
son I, Dan B. 2003. Sudden infant deaths: Stress, arousal and Pauza DH, Skripka V, Pauziene N, Stropus R. 2000. Morphology,
SIDS. Early Hum Dev 75 (Suppl):S147–S166. distribution, and variability of the epicardiac neural ganglionated
Kawashima T. 2005. The autonomic nervous system of the human subplexuses in the human heart. Anat Rec 259:353–382.
heart with special reference to its origin, course, and peripheral Pietri T, Eder O, Blanche M, Thiery JP, Dufour S. 2003. The human
distribution. Anat Embryol (Berl) 209:425–438. tissue plasminogen activator-Cre mouse: A new tool for target-
46 Hildreth et al.

ing specifically neural crest cells and their derivatives in vivo. Sommer L, Shah N, Rao M, Anderson DJ. 1995. The cellular func-
Dev Biol 259:176–187. tion of MASH1 in autonomic neurogenesis. Neuron 15:1245–
Poelmann RE, Mikawa T, Gittenberger-de Groot AC. 1998. Neu- 1258.
ral crest cells in outflow tract septation of the embryonic Stewart DE, Kirby ML. 1985. Endogenous tyrosine hydroxylase ac-
chicken heart: Differentiation and apoptosis. Dev Dyn 212: tivity in the developing chick heart: A possible source of extra-
373–384. neuronal catecholamines. J Mol Cell Cardiol 17:389–398.
Porter GA Jr, Rivkees SA. 2001. Ontogeny of humoral heart rate Tessarollo L, Tsoulfas P, Donovan MJ, Palko ME, Blair-Flynn J,
regulation in the embryonic mouse. Am J Physiol Regul Integr Hempstead BL, Parada LF. 1997. Targeted deletion of all iso-
Comp Physiol 281:R401–R407. forms of the trkC gene suggests the use of alternate receptors
Potter E. 1987. Presynaptic inhibition of cardiac vagal postganglionic by its ligand neurotrophin-3 in neuronal development and impli-
nerves by neuropeptide Y. Neurosci Lett 83:101–106. cates trkC in normal cardiogenesis. Proc Natl Acad Sci USA
Rigel DF. 1988. Effects of neuropeptides on heart rate in dogs: 94:14776–14781.
Verberne ME, Gittenberger-de Groot AC, Poelmann RE. 1998. Line-
Comparison of VIP. PHI, NPY, CGRP, and NT. Am J Physiol
age and development of the parasympathetic nervous system of
255:H311–H317.
the embryonic chick heart. Anat Embryol (Berl) 198:171–184.
Sasai Y, Kageyama R, Tagawa Y, Shigemoto R, Nakanishi S. 1992.
Verberne ME, Gittenberger-De Groot AC, Van Iperen L, Poelmann
Two mammalian helix-loop-helix factors structurally related to
RE. 1999. Contribution of the cervical sympathetic ganglia to the
Drosophila hairy and Enhancer of split. Genes Dev 6:2620–
innervation of the pharyngeal arch arteries and the heart in the
2634.
chick embryo. Anat Rec 255:407–419.
Sawa R, Asakura H, Power GG. 1991. Changes in plasma adenosine
Verberne ME, Gittenberger-de Groot AC, van Iperen L, Poelmann
during simulated birth of fetal sheep. J Appl Physiol 70:1524–
RE. 2000. Distribution of different regions of cardiac neural crest
1528.
in the extrinsic and the intrinsic cardiac nervous system. Dev
Scherlag BJ, Nakagawa H, Jackman WM, Yamanashi WS, Patterson Dyn 217:191–204.
E, Po S, Lazzara R. 2005a. Electrical stimulation to identify neu- Waldo KL, Lo CW, Kirby ML. 1999. Connexin 43 expression reflects
ral elements on the heart: Their role in atrial fibrillation. J Interv neural crest patterns during cardiovascular development. Dev
Card Electrophysiol 13 (Suppl) 1:37–42. Biol 208:307–323.
Scherlag BJ, Yamanashi W, Patel U, Lazzara R, Jackman WM. Weihe E, Reinecke M, Forssmann WG. 1984. Distribution of vasoac-
2005b. Autonomically induced conversion of pulmonary vein tive intestinal polypeptide-like immunoreactivity in the mamma-
focal firing into atrial fibrillation. J Am Coll Cardiol 45:1878– lian heart. Interrelation with neurotensin- and substance P-like
1886. immunoreactive nerves. Cell Tissue Res 236:527–540.
Shoba T, Tay SS. 2000. Nitrergic and peptidergic innervation in the Wiley JW, Uccioli L, Owyang C, Yamada T. 1989. Somatostatin stim-
developing rat heart. Anat Embryol (Berl) 201:491–500. ulates acetylcholine release in the canine heart. Am J Physiol
Slaugenhaupt SA, Blumenfeld A, Gill SP, Leyne M, Mull J, Cuajungco 257:H483–487.
MP, Liebert CB, Chadwick B, Idelson M, Reznik L, Robbins C, Zhang SH, Rush RA. 2001. Neurotrophin 3 is increased in the spon-
Makalowska I, Brownstein M, Krappmann D, Scheidereit C, taneously hypertensive rat. J Hypertens 19:2251–2256.
Maayan C, Axelrod FB, Gusella JF. 2001. Tissue-specific expres- Zhou XF, Chie ET, Deng YS, Rush RA. 1997. Rat mature sympa-
sion of a splicing mutation in the IKBKAP gene causes familial thetic neurones derive neurotrophin 3 from peripheral effector
dysautonomia. Am J Hum Genet 68:598–605. tissues. Eur J Neurosci 9:2753–2764.

You might also like