You are on page 1of 29

Pharmacology & Therapeutics 139 (2013) 260288

Contents lists available at SciVerse ScienceDirect

Pharmacology & Therapeutics


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

Associate Editor: P. Molenaar

Structure, function and clinical relevance of the cardiac conduction


system, including the atrioventricular ring and outow tract tissues
Halina Dobrzynski a,, Robert H. Anderson a, Andrew Atkinson a, Zoltan Borbas a, Alicia D'Souza a,
John F. Fraser b, c, Shin Inada d, Sunil J.R.J. Logantha a, Oliver Monfredi a, Gwilym M. Morris a,
Anton F.M. Moorman e, Thodora Nikolaidou a, Heiko Schneider a, Viktoria Szuts f, Ian P. Temple a,
Joseph Yanni a, Mark R. Boyett a,
a
University of Manchester, UK
b
Critical Care Research Group, University of Queensland, Australia
c
Prince Charles Hospital, Australia
d
National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
e
University of Amsterdam, Netherlands
f
University of Szeged, Hungary

a r t i c l e i n f o a b s t r a c t

Keywords: It is now over 100 years since the discovery of the cardiac conduction system, consisting of three main parts, the
Cardiac conduction system sinus node, the atrioventricular node and the HisPurkinje system. The system is vital for the initiation and co-
Atrioventricular ring tissue ordination of the heartbeat. Over the last decade, immense strides have been made in our understanding of
Right ventricular outow tract the cardiac conduction system and these recent developments are reviewed here. It has been shown that the sys-
Arrhythmias tem has a unique embryological origin, distinct from that of the working myocardium, and is more extensive
Biopacemaking
than originally thought with additional structures: atrioventricular rings, a third node (so called retroaortic
Channelopathies
node) and pulmonary and aortic sleeves. It has been shown that the expression of ion channels, intracellular
Ca2+-handling proteins and gap junction channels in the system is specialised (different from that in the ordinary
working myocardium), but appropriate to explain the functioning of the system, although there is continued de-
bate concerning the ionic basis of pacemaking. We are beginning to understand the mechanisms (brosis and
remodelling of ion channels and related proteins) responsible for dysfunction of the system (bradycardia, heart
block and bundle branch block) associated with atrial brillation and heart failure and even athletic training.
Equally, we are beginning to appreciate how naturally occurring mutations in ion channels cause congenital car-
diac conduction system dysfunction. Finally, current therapies, the status of a new therapeutic strategy (use of a
specic heart rate lowering drug) and a potential new therapeutic strategy (biopacemaking) are reviewed.
2013 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
2. Embryonic development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
3. Cardiac conduction system: anatomy, function and clinical relevance . . . . . . . . . . . . . . . . . . 261
4. Atrioventricular ring tissue: anatomy, function and clinical relevance . . . . . . . . . . . . . . . . . . 261
277
5. Right ventricular outow tract: anatomy, function and clinical relevance . . . . . . . . . . . . . . . . . 279
261
6. Channelopathies in humans responsible for dysfunction of the cardiac conduction system . . . . . . . . . 280
261
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 282
261
Conict of Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 282
262
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263
282

Corresponding authors at: Institute of Cardiovascular Sciences, CTF Building, 46 Grafton Street, University of Manchester, Manchester M13 9NT, UK. Tel.: +44 161 2751182,
+44 161 2751192; fax: +44 161 2751183.
E-mail addresses: halina.dobrzynski@manchester.ac.uk (H. Dobrzynski), mark.boyett@manchester.ac.uk (M.R. Boyett).

0163-7258/$ see front matter 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.pharmthera.2013.04.010
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 261

1. Introduction tracts. Parts of this primary myocardium also retain their initial embry-
onic phenotype so as to form the cardiac nodes and AV conduction axis
The cardiac conduction system consists of the sinus (or sinuatrial or (Fig. 2C; Davis et al., 2001; Rentschler et al., 2001; Hoogaars et al.,
sinoatrial) node, the atrioventricular (AV) conduction axis (including 2004), with some of the ventricular trabeculations being incorporated
the AV node), its right and left bundle branches, and the terminal to form the rapidly conducting Purkinje network. These changes are
Purkinje network. All of these structures, made up of specialised cardiac achieved by so-called patterning, or regional expression of develop-
myocytes, have unique anatomical, molecular and functional properties mental gene programmes (Christoffels et al., 2004), with the other
that permit them to work collectively as the electrical system of the parts of the initial primary myocardium becoming transformed into
heart. The system has been the subject of extensive studies since its chamber myocardium. Development of the cardiac conduction system
elucidation in the rst decade of the 20th century. Recent advances in is controlled by transcription factors and for an excellent review of this
technologies have aided our further understanding of this specialised fast developing eld see Christoffels et al. (2010).
system of the heart. In particular, other myocytes with comparable
properties, such as the AV ring tissue and myocytes in the ventricular 2.2. Atrioventricular ring tissue
outow tracts, especially the right ventricular outow tract, have
attracted notice because of their arrhythmogenic properties. A common As described above, only a small part of the primary myocardium
feature of the different tissues of the cardiac conduction system is the eventually develops into nodal tissues, retaining the characteristics of
ability to show pacemaking and Fig. 1 shows the perhaps surprising automaticity, and failing to be converted into working myocardium. It
widespread distribution of tissues with pacemaking potentiality in the is now well established that the expression of the transcriptional inhibi-
heart. Our aim in this review is to describe our current understanding tor Tbx3 is responsible for this fundamental process (Hoogaars et al.,
of the expanded concept of the cardiac conduction system, in terms of 2007). Ectopic expression of Tbx3 in the atrial chambers has been
anatomy, function and clinical relevance. So-called channelopathies shown to result in the formation of functional ectopic nodes exhibiting
related to the cardiac conduction system are also discussed. pacemaker activity. The expression of Tbx3, however, marks not only
the normal cardiac conduction system, but also the so-called AV ring
2. Embryonic development tissues (Anderson et al., 1974a, 1974b; Yanni et al., 2009a). The areas
of the primary heart tube such as the AV canal, the interventricular
2.1. Cardiac conduction system in relation to working myocardium ring and the ventricular outow tract can be considered as supportive
of this concept, albeit that the rings do not appear as new entities,
The heart initially forms as a midline tube ventral to the foregut at the but rather are part of the initial primary heart tube, becoming evident
stage of embryonic folding. Its cells are derived from visceral mesoderm, as rings only subsequent to the formation of the chamber myocardium.
with the cells forming a linear primary heart tube containing the primor- In particular, part of the embryonic AV canal persists in the denitive
dium for little more than the left ventricle, or even less (Cai et al., 2003; heart within the atrial vestibules as the specialised AV rings that
Aanhaanen et al., 2009). Ongoing development depends on the addition surround the orices of the tricuspid and mitral valves, with the ring
of cells to the primary tube from the heart-forming areas at both the tissues originating from the dorsal aspect of the AV node, and crossing
venous and arterial poles (Fig. 2A). At this early undifferentiated stage, over the bundle of His to join so as to form a ventral retroaortic node
the walls of the heart tube are made up of so-called primary myocardi- (Yanni et al., 2009a).
um (Moorman & Christoffels, 2003). The myocytes of this myocardium
have a phenotype that includes slow growth, slow contraction, slow 2.3. Right ventricular outow tract
conduction and the ability to depolarize spontaneously (Moorman &
Christoffels, 2003). As the new material is added at the venous and The initial common outow tract is exclusively composed of the
arterial poles (Kelly & Buckingham, 2002), the tube itself undergoes primary myocardium of the linear heart tube, and is entirely supported
looping. Subsequent to looping, the primordiums of the eventual atrial by the right ventricle. The distal portion of this common outow tract is
and ventricular chambers balloon from the cavity of the primary tube, remodelled by further addition of cells from the visceral mesenchyme
with the right and left atrial appendages ballooning in parallel from the to form the intrapericardial arterial trunks. At this stage, the more
atrial component of the tube, and the apical ventricular components proximal parts retain their muscular walls, with the pulmonary and
ballooning in series from the inlet and outlet parts of the ventricular aortic valves, along with their sinuses, developing within a muscular
loop (Fig. 2B). The myocytes making up these new parts together make sleeve. The most proximal part of the common outow tract will form
up the chamber myocardium (Moorman & Christoffels, 2003). Chamber the right ventricular outow tract and the aortic vestibule, with the
myocardium is distinct from primary myocardium in a number of ways, proximal cushions themselves fusing and muscularising to form the
including the possession of gap junctions made up of connexin40 (Cx40) dorsal part of the infundibulum, this part also fusing with the crest of
and connexin43 (Cx43), which permit rapid conduction, along with the muscular ventricular septum so as to commit the aorta to the left
rapid rates of cell division. There is also formation of pectinate muscles ventricle. As development progresses, the proximal right ventricular
and trabeculations in the newly forming chamber components of the outow tract largely differentiates into working right ventricular
atria and the ventricles, respectively. Already at this early stage, however, myocardium (Rana et al., 2007), with the more distal parts that initially
while it is still possible to distinguish the areas of primary and chamber surrounded the developing arterial valves disappearing by apoptosis
myocardium, and while there is muscular continuity throughout the (Barbosky et al, 2006). It has been suggested that not all myocytes in
developing heart, it is possible to record a relatively normal electrocar- the right ventricular outow tract differentiate into working ventricular
diogram (ECG). The tracing incorporates a period of AV delay, even myocytes and some myocytes retain their initial nodal-like phenotype
though at this stage it is not possible to recognise any of the components (Yanni et al., 2009a; Boukens et al., 2009; Monfredi et al., 2010a, 2010b).
of the denitive cardiac conduction system. The areas of the tube made
up of primary as opposed to chamber myocardium are also different in 3. Cardiac conduction system:
that blocks of cushion mesenchyme are formed within their lumens anatomy, function and clinical relevance
that will ultimately form the valves of the heart, and set the scene for
separation of the systemic and pulmonary pathways of the circulation. 3.1. Sinus node
It is also within these components of the primary tube that the
remodelling takes place to allow correct alignment of the separating In health, the sinus node is the pacemaker of the mammalian
atrial and ventricular chambers and the systemic and pulmonary outow heart. Even more than 100 years after its rst description by Keith &
262 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

Fig. 1. Pacemaker potentiality in the heart. The schematic diagram of the heart shows different components of the cardiac conduction system. Orange, sinus node; red, AV ring tissue and AV
node; purple, HisPurkinje network. Typical pacemaker action potentials from the sinus node centre (32 C; Tellez et al., 2006), sinus node periphery (32 C; Tellez et al., 2006), subsidiary
atrial pacemaker site (36 C; Rozanski & Lipsius, 1985), Eustachian ridge (warm temperature; Rubenstein et al., 1987), coronary sinus (36 C; Wit & Craneeld, 1977; Wit et al., 1981), right
ring tissue (37 C; S.J.R.J. Logantha, unpublished data), tricuspid valve leaet (37 C; Rozanski & Jalife, 1986; Rozanski, 1987), Purkinje bre (free running; 37 C; S.J.R.J. Logantha, unpublished
data), AV node (3537 C; Cheng et al., 2011), right ventricular outow tract (37 C; S.J.R.J. Logantha, unpublished data), mitral valve (36 C; Wit et al., 1979), left ring tissue (37 C; S.J.R.J.
Logantha, unpublished data), pulmonary vein (37 C; Wongcharoen et al., 2006) and superior vena cava (37 C; Chen et al., 2002) of different species are shown around the schematic
diagram. All records were obtained in tissue preparations (either in isolated preparations or in tissues with sinus node activity supressed) with the exception of the AV node and superior
vena cava recordings, which were obtained from isolated myocytes. In each trace, the vertical scale bar represents 60 mV and its upper limit indicates the zero potential. Unless otherwise
stated, the horizontal scale bar denotes 200 ms.

Flack in, 1907, the molecular mechanisms underlying its pacemaker and blood vessels. For a review of the possibility of exit sites see Fedorov
potential are hotly disputed (e.g., Monfredi et al., 2010a, 2010b). et al. (2012). Although it is well established that there is a block zone be-
tween the sinus node and the interatrial septum in the human and other
3.1.1. Anatomy species (Fedorov et al., 2010; Boyett et al., 2000), we have observed no
In most human hearts, the sinus node is a banana-shaped structure evidence of an anatomical block zone between the sinus node and the
located in the subepicardial intercaval region of the heart at the junction terminal crest in the human and other species (Sanchez-Quintana et
of the superior caval vein and the right atrium, with a tail extending al., 2005; Chandler et al., 2011). We suggest that there are no discrete
along the terminal crest (Dobrzynski et al., 2007; Wiese et al., 2009; exit sites; the preferential supero-inferior action potential propagation
Chandler et al., 2011). The nodal myocytes are embedded in a network within the sinus node as a result of the preferential supero-inferior
of connective tissue and surround the sinus node artery (Fig. 3A). The myocyte orientation (Boyett et al., 2000; Chandler et al., 2011) may
action potential originates in the centre of the sinus node and here give the appearance of discrete superior and inferior exit sites. In the
the myocytes are small when compared to working atrial myocytes human, a further histologically specialised area has now been noted
(Fig. 4). In the centre of the sinus node, the nodal myocytes also have at the periphery of the sinus node (Fig. 3A; Chandler et al., 2009).
few mitochondria and sparse and poorly organised myolaments This paranodal area is distinct from both the sinus node and atrial
(Boyett et al., 2000; Christoffels et al., 2010). From the centre to the pe- muscle for example, the myocytes of the paranodal area are loose-
riphery of the sinus node, there is a gradual transition from true (central) ly packed in fatty tissue, whereas atrial myocytes are densely packed
nodal myocytes to peripheral nodal myocytes, which share some charac- and the myocytes of the sinus node are packed in connective tissue
teristics with the surrounding atrial myocytes (Boyett et al., 2000). For (Fig. 4A; Chandler et al., 2009). The paranodal area has a myocardial
example, peripheral nodal myocytes are larger with more and better phenotype in between that of the working myocardium and the sinus
organised myolaments (Boyett et al., 2000). Throughout the periphery node, for example in terms of myocyte size (Fig. 4; Chandler et al.,
of the sinus node, the sinus node can be seen to interdigitate with the 2009) and connexin expression (Fig. 4BD; Chandler et al., 2009). It
surrounding atrial muscle at many sites (Sanchez-Quintana et al., 2005; also contains a mixture of atrial and nodal myocytes (Chandler et al.,
Liu et al., 2007). The interdigitations have been suggested to assist the 2009). The paranodal area of the human is similar to the periphery of
exit of the action potential from the sinus node into the atrial muscle the sinus node in the rabbit compare Chandler et al. (2009) and
(Boyett et al., 2006). Perhaps in contrast, Bromberg et al. (1995) and Dobrzynski et al. (2005). In terms of location, the paranodal area is ex-
Fedorov et al. (2009, 2010) have argued that exit of the action potential tensive, and runs parallel to the sinus node along the terminal crest
from the sinus node occurs at specic superior and inferior exit sites and (Fig. 5A; Chandler et al., 2009). Fig. 5B shows the position of the leading
exit of the action potential at all other points is blocked by brous tissue pacemaker site in the right atrium of the human heart and Fig. 5C shows
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 263

Fig. 2. Cartoons showing the changes involved in forming the denitive conduction tissues. A, lateral view of the developing heart tube subsequent to the beginning of addition of new ma-
terial from the heart-forming areas at both the arterial and venous poles. The central component, the initial linear heart tube, eventually forms only part of the left ventricle. The
intrapericardial components, shown in grey, are all formed of primary myocardium at this stage. The extrapericardial parts are shown in yellow. B, cartoon showing how the chamber com-
ponents balloon from the primary tube, with the atrial appendages (AA) ballooning from the newly formed atrial component of the heart tube, and the left and right ventricles (LV and RV)
ballooning from the inlet and outlet parts of the ventricular loop. Note that, during this intermediate stage, the persisting primary myocardium forms the atrioventricular canal, and an atrial
channel labelled in the cartoon as dorsal since it eventually forms the dorsal atrial wall, albeit being developed from the initially ventral wall of the primary heart tube. Note also that the
developing outow tract is formed at this stage by primary myocardium. C, cartoon showing that the primary myocardium persists only as the sinus node and the atrioventricular conduction
axis in the fully formed heart, albeit that further remnants can be identied, e.g. as the AV ring tissues (not shown).
From R.H. Anderson and A.F. Moorman.

the position of the sinus node (and the neighbouring paranodal area) in 3.1.2. Function
the human heart; the two correspond. The function of the paranodal area The electrical activity of the sinus node differs from that of the
is not known, but it is possible that it may be responsible for the cristal working myocardium (Irisawa et al., 1993; Boyett et al., 2000;
tachycardias observed by Kalman et al. (1998) or can take over as subsid- Dobrzynski et al., 2007; Mangoni & Nargeot, 2008). The sinus node
iary pacemaker. action potential has a slow upstroke (phase 0; Fig. 6B), because the

A
Terminal crest
Pectinate
RA Paranodal area
muscle

Sinus node

Aatrioventricular conduction axis


B
Compact
node
Penetrating
INE bundle

Fig. 3. Low magnication images of tissue sections through human cardiac conduction system stained with Masson's trichrome. A, histology of the sinus node, paranodal area and atrial
muscle. B, histology of the AV junction (the inferior nodal extension, compact node and penetrating bundle). CFB, central brous body; INE, inferior nodal extension; TA transitional area.
From Chandler et al. (2009) and Greener et al. (2011).
264 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

Fig. 4. High magnication images of human sinus node, paranodal area and right atrial muscle stained with Masson's trichrome (A) and immunolabelled for Cx43 (B, C) and Cx40
and caveolin3 (D). The combination of the markers (Cx40, Cx43 and caveolin3) distinguished the three tissues. Cx40 and Cx43 are absent in the sinus node, but present in the atrial
muscle. In the atrial muscle, Cx40 and Cx43 are present at gap junctions. In the paranodal region, there is a mixture of Cx40 and Cx43 positive and negative myocytes (white arrows
highlight myocytes that express Cx40 and Cx43; red arrows highlight myocytes that do not express Cx40 and Cx43). Caveolin3 is present in the cell membrane of all myocytes and
shows that atrial myocytes are large, nodal myocytes are small and paranodal myocytes are intermediate in size.
From Chandler et al. (2011).

upstroke is not generated by the Na+ current, INa, as in the working to be responsible for diastolic depolarization and they are the subject of
myocardium instead it is largely generated by smaller and slower T- Section 3.1.2.1. The different ionic currents etc. involved in the sinus
and L-type Ca2+ currents, ICa,T and ICa,L (Irisawa et al., 1993; Mangoni & node action potential are shown in Fig. 6A. In parallel with the histolog-
Nargeot, 2008). The sinus node action potential usually lacks an early ical transition from the centre to the periphery of the sinus node
phase of repolarization (phase 1), but it has a prominent plateau (Section 3.1.1), there are changes in the electrical properties of the
(phase 2; Fig. 6B) and, as a result, the sinus node action potential is sinus node: the maximum diastolic potential becomes more negative,
longer than the action potential of the surrounding atrial muscle the action potential upstroke becomes faster, the action potential
(Boyett et al., 1999). The downward gradient in action potential duration becomes shorter, and paradoxically the intrinsic pacemaker activity
from the sinus node has been argued to prevent sinus node reentry becomes faster (Boyett et al., 1999). In the rabbit, we have suggested
(Boyett et al., 1999). Repolarization of the sinus node, as in the rest of that these changes are in part at least the result of an increase in the
the heart, is the result of the inactivation of ICa,L and the activation of a va- density of If, INa and IK,r (Boyett et al., 2000). The specialisations in electri-
riety of K+ currents: transient outward K+ current (Ito) and ultra-rapid cal activity in the periphery of the sinus node are likely to be further
(possibly), rapid and slow delayed rectier K+ currents (IK,ur, IK,r and IK, adaptations to enable the sinus node to drive the surrounding right atrial
s, respectively). During diastole (i.e. phase 4), the sinus node lacks the muscle (Honjo et al., 1996; Boyett et al., 2009). It is possible that the func-
stable resting potential characteristic of the working myocardium tion of the paranodal area in the human is similar computer modelling
(Fig. 6B). This is because the sinus node has little or no background suggests that the paranodal area aids propagation of the action potential
inward rectier K+ current, IK,1. In the sinus node, the maximum diastol- from the sinus node to the atrial muscle (Aslanidi et al., 2011).
ic potential (peak negative potential immediately following the action Electrical coupling between myocytes is well known to be poor in
potential) is less negative than the resting potential in the working myo- the centre of the sinus node as compared to the surrounding atrial
cardium and, furthermore, throughout diastole there is a slow diastolic muscle (Boyett et al., 2000, 2006). This is one reason why the conduc-
depolarization or pacemaker potential (Fig. 6B). On reaching threshold, tion velocity of the action potential is lower in the centre of the sinus
the diastolic depolarization triggers the next action potential (Fig. 6B). node (several cm/s in the rabbit) than in the atrial muscle (~70 cm/s
A variety of ionic currents (such as the hyperpolarization-activated or in the rabbit; Sano & Yamagishi, 1965; Boyett et al., 2000). The poor
funny current, If) and mechanisms (such as the Ca2+ clock) are known electrical coupling in the centre of the sinus node is thought to insulate
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 265

Fig. 5. Three-dimensional reconstruction of the human sinus node and paranodal area and comparison of the activation sequence of the sinus node in a patient and in the
three-dimensional anatomical model. A, three-dimensional reconstruction. Green, atrial muscle; red, sinus node; yellow, paranodal area; blue, adipose tissue. Most of the sinus
node tissue (red) is located on the epicardial side and is a banana-shaped structure. The paranodal area is located on the endocardial side. From Chandler et al. (2011). B, dorsal
view of the atria of a patient during open heart surgery showing the leading pacemaker site (red) and the activation sequence of the atria; the activation sequence is shown as
a colour scale and by the isochrones in ms. From Boineau et al. (1988). C, model of the sinus node (red) and paranodal area (yellow) superimposed on a schematic diagram of
the heart (dorsal view). The activation sequence is shown by the isochrones in ms; the leading pacemaker site is shown by the white spot. Simulated action potentials from the
sinus node, paranodal area and atrial muscle are shown. From Chandler et al. (2009). Ao, aorta; IVC, inferior vena cava; LAA, left atrial appendage; PA, pulmonary artery; PV,
pulmonary vein; RA, right atrium; RAA, right atrial appendage; RV, right ventricle; SVC, superior vena cava.

the centre from the hyperpolarizing inuence of the surrounding atrial action potential, the net ionic current through these channels switches
muscle, which acts to slow pacemaker activity (Joyner & van Capelle, from outward to inward to drive diastolic depolarization: there is a
1986; Boyett et al., 2006). It has been postulated that there is a gradual voltage-dependent deactivation of outward K+ currents (IK,r and IK,s),
increase of electrical coupling from the centre to the periphery of the as well as a voltage-dependent activation of inward currents: If,
sinus node as yet another adaptation to enable the sinus node to drive hyperpolarization-activated Cl current, sustained inward current (Ist),
the surrounding right atrial muscle (Joyner & van Capelle, 1986; ICa,T and ICa,L (Fig. 6A; Mangoni & Nargeot, 2008; Huang et al., 2009;
Boyett et al., 2006). In the periphery of the sinus node, the conduction Chen et al., 2010; Park & Fishman, 2011). The importance of If in pace-
velocity is intermediate between that in the centre of the sinus node making has been shown by conditional knockout of HCN4 in the adult
and atrial muscle (Sano & Yamagishi, 1965) and this is consistent with mouse it causes deep bradycardia (Baruscotti et al., 2011). The impor-
a gradient in electrical coupling from the centre to the periphery of tance of If in pacemaking in the human has been highlighted by pharma-
the sinus node. cological intervention (such as ivabradine) targeting If (Section 3.1.3.2)
and also reports describing a slower heart rate in response to mutations
3.1.2.1. Clock mechanisms in sinus node pacemaking. Pacemaker auto- in the If channel, HCN4 (Section 6). Activation of inward INa is also impor-
maticity is governed by spontaneous diastolic depolarization during tant: in the rabbit, although INa is absent from the centre of the sinus
phase 4. Although the events that underlie diastolic depolarization node (Section 3.1.2), INa is present and plays a role in pacemaking in
are not fully understood, diastolic depolarization and sinus node the periphery of the sinus node (Kodama et al., 1997a, 1997b); INa has
pacemaking are currently thought to be the result of the synergistic been recorded in human sinus node myocytes (Verkerk et al., 2009);
interaction between two oscillators: the membrane voltage clock and, nally, in the human, mutations in the Na+ channel, Nav1.5, cause
and the Ca 2+ clock (Fig. 6A) (Lakatta & DiFrancesco, 2009; Joung bradycardia (Section 6). In the mouse, INa may be particularly important
et al., 2009). The membrane voltage clock comprises an ensemble (Lei et al., 2004); knock-out of the Na+ channel, Nav1.5, causes bradycar-
of voltage-dependent ion channels (in the surface membrane or dia (Lei et al., 2005). In addition to the inward currents above, a
sarcolemma) and the corresponding ionic currents. Following the background inward Na+ current (Ib,Na) owing throughout diastole is
266 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

Fig. 6. Sinus node pacemaking mechanisms. A, the multiple ion channels, ionic currents and Ca2+-handling proteins comprising the membrane and Ca2+ clocks involved in pacemaking.
During the pacemaker potential, there is a voltagedependent decay of outward currents, IK,r and IK,s (carried by ERG and KvLQT1), and a voltagedependent activation of inward currents:
If (carried by HCN1 and HCN4), Cl current (carried by ClC-2), ICa,L (carried by Cav1.2 and Cav1.3) and ICa,T (carried by Cav3.1 and Cav3.2). The Ca2+ clock is also involved in pacemaking:
during the nal phase of the pacemaker potential, there is an activation of inward INaCa (generated by the electrogenic Na+Ca2+ exchanger, NCX1) in response to a spontaneous release of
Ca2+ from the SR via the ryanodine receptor (RYR2). Ca2+ release from the SR also occurs as a result of Ca2+induced Ca2+ release in response to Ca2+ entry into the cell via ICa,L and ICa,T.
The SR is replenished with Ca2+ by reuptake of Ca2+ via SERCA2 (Sarco/Endoplasmic Reticulum Ca2+ ATPase). Storeoperated Ca2+ channels at the cell surface membrane also help to
replenish the sarcoplasmic reticulum with Ca2+. Modied from Monfredi et al. (2010a, 2010b). B, summary of the expression of the main ion channels contributing to the different phases
of the nodal action potential. Rabbit sinus node myocyte action potentials shown.
From O. Monfredi (unpublished data).

often invoked in models of the sinus node action potential (e.g., Zhang et activate the electrogenic operation of the sarcolemmal Na +Ca2+ ex-
al., 2000). The Ca2+ clock contributes to sinus node diastolic depolariza- changer (NCX1). This generates an inward current (INaCa) that imparts
tion through localised Ca2+ release from the sarcoplasmic reticulum (SR) a steep, exponential increase to the late phase of diastolic depolarization
via the type 2 ryanodine receptor (RYR2; Huser et al., 2000; Bogdanov et (Fig. 6; Maltsev & Lakatta, 2010; Lakatta et al., 2010). Li+-induced INaCa
al., 2001). Whether the Ca2+ release is spontaneous (Vinogradova et al., blockade has been demonstrated to abolish sinus node pacemaking
2005) or occurs in response to Ca2+-induced Ca2+ release activated by (Bogdanov et al., 2001). Mutations in genes encoding components
ICa,T (Huser et al., 2000) is controversial. Oscillatory spikes in intracellular of the Ca2+ clock have a detrimental effect on sinus node function
Ca2+ occur in voltage clamped and chemically skinned sinus node (Section 6). While recent years have seen controversy over the relative
myocytes and this suggests that the Ca2+ release is spontaneous importance of the membrane voltage and Ca2+ clocks, mutual entrain-
(Vinogradova et al., 2005). The localised Ca2+ releases are thought to ment of the clock mechanisms is likely to underlie normal sinus node
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 267

Table 1
Ion channel expression in the human at the mRNA level in the sinus node, paranodal area, transitional area, inferior nodal extension, compact node, penetrating bundle, Purkinje
bres and ventricular muscle as compared to that in the atrial muscle. *, signicantly upregulated; *, signicantly downregulated; , trend to upregulation; , trend to
downregulation; =, no difference. Based on Chandler et al. (2009), Greener et al. (2011) and Gaborit et al. (2007).

function (Lakatta & DiFrancesco, 2009; Maltsev & Lakatta, 2009; Maltsev Na+ channel, Nav1.5, is poorly expressed or not expressed in the sinus
and Lakatta, 2010; Himeno et al., 2011; DiFrancesco & Noble, 2012). node, whereas it is highly expressed in the working myocardium in
the human and other species (Table 1; Tellez et al., 2006; Hoogaars et
3.1.2.2. Ion channels. There is ample evidence to show that the special- al., 2007; Chandler et al., 2009; Greener et al., 2011). This is why INa is
isations of electrical activity in the centre and periphery of the sinus small or absent in the centre of the sinus node. However, another Na+
node are the result of specialisations in the expression of ion channels channel, Nav1.1, although much less abundant than Nav1.5, is preferen-
(responsible for ionic currents), gap junction proteins (responsible tially expressed in the sinus node (Tellez et al., 2006) and is responsible
for electrical coupling between myocytes) and Ca2+-handling proteins for part of INa (at least in the mouse; Lei et al., 2004). Specialised Ca 2+
(involved in the Ca2+ clock mechanism of pacemaking). Table 1 channels, Cav1.3 and Cav3.1, more appropriate for pacemaking, are
shows the expression levels of a few key players (at the mRNA level) more highly expressed in the sinus node than in the atrial muscle
in different regions of the human heart including the sinus node and (Table 1; e.g. Chandler et al., 2009). In the human, some K + channels
paranodal area. Kir2.1 is primarily responsible for IK,1 and this is more are more poorly expressed (e.g. ERG responsible for IK,r; Table 1;
poorly expressed in the sinus node than in the atrial muscle and this Chandler et al., 2009) and this may result in the action potential being
explains why the sinus node lacks a stable resting potential (Table 1; longer in the sinus node than in the atrial muscle (Boyett et al., 1999).
Irisawa et al., 1993; Chandler et al., 2009). HCN4 is primarily responsible It is curious that, despite the importance of the Ca2+ clock in sinus
for If, an important pacemaker current as described above, and it is high- node pacemaking, two key components of the Ca2+ clock (SERCA2
ly expressed in the sinus node as expected, but not in the working myo- and RYR2) are downregulated in the sinus node (Table 1; Chandler et
cardium (Table 1; Dobrzynski et al., 2007; Mangoni & Nargeot, 2008; al., 2009). Electrical coupling between cardiac myocytes is provided
Yanni et al., 2009a, 2009b). HCN1 also plays an important role in If by gap junctions connecting neighbouring myocytes. Gap junction
and it too is highly expressed in the sinus node as expected, but not in channels are comprised of connexins: in the heart, Cx40 forms large
the working myocardium (Table 1; e.g. Chandler et al., 2009). The conductance (200 pS) channels, Cx43 forms intermediate conductance
inward hyperpolarization-activated Cl current is biophysically similar (60100 pS) channels and Cx45 forms small conductance (2040 pS)
to If and the underlying channel, ClC-2, is preferentially expressed in the channels (Boyett et al., 2006). The two higher conductance connexins,
sinus node (Huang et al., 2009; Yanni et al., 2011a, 2011b). The cardiac Cx40 and Cx43, are more poorly expressed in the sinus node than in
268 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

the atrial muscle (Fig. 4C and D; Table 1; e.g. Coppen et al., 1999a, Faggiano et al., 2001). It is well known that there is a resting bradycardia
1999b; Chandler et al., 2009) and this explains why electrical coupling in athletes the heart rate of race t Tour de France cyclists can be as
is poor in the centre of the sinus node. In the centre of the sinus node, low as 30 beats/min. Although this is assumed to be a physiological
Cx45 is expressed (Table 1; e.g. Coppen et al., 1999a, 1999b) and this adaptation in athletes, there are indications that it can become
is a low conductance connexin (expected to provide poor electrical cou- pathological there is a higher incidence of the implantation of elec-
pling). In the periphery of the sinus node, including the paranodal area tronic pacemakers in endurance athletes (Baldesberger et al., 2008).
in the human, expression of ion channels etc. can be transitional. For ex- Histopathological approaches to studying the underlying aetiology of
ample, in the rabbit and human, there is evidence of little expression of sinus node dysfunction suggest a central role for degenerative brosis
Nav1.5, Kir2.1 and Cx43 in the centre of the sinus node, intermediate ex- in the abnormal sinus node automaticity and conduction (DeMelo et
pression in the periphery of the sinus node/paranodal area and high ex- al., 2002; Adan & Crown, 2003). Recently, in a mouse model of heart
pression in the atrial muscle (Coppen et al., 1999a, 1999b; Tellez et al., failure triggered by angiotensin II infusion, Swaminathan et al. (2011)
2006; Chandler et al., 2009). We suggest that a gradient in ion channel observed sinus node dysfunction together with sinus node myocyte
expression is an important adaptation to enable to the sinus node to apoptosis caused by activation of NADPH oxidase and increased levels
drive the surrounding atrial muscle (Boyett et al., 2000). of oxidised calmodulin kinase II. In addition, in a rat model of pulmo-
It is clear from the above discussion that there is not just one nary hypertension with sinus node dysfunction, Yanni et al. (2009b)
pacemaker mechanism there is a multiplicity of mechanisms in- observed upregulation of brosis genes in the sinus node (although
volved in pacemaking: Ib,Na, If, hyperpolarization-activated Cl cur- no overt brosis). However, brosis as well as an upregulation of
rent, INa, ICa,L, ICa,T, INaCa and the Ca 2+ clock (Fig. 6). There are also brosis genes has been observed in the sinus node of the ageing
possible new mechanisms emerging: a TRP channel, TRPM4, is expressed mouse (Hao et al., 2011). Nevertheless, the concept that sinus node
in mouse sinus node myocytes and is possibly responsible for a dysfunction is the result of degenerative brosis has been challenged
Ca2+-activated non-specic current (Demion et al., 2007). Its role is no evidence has been seen in the ageing human, cat and rat (Ailings et
not known, but evidence suggests that at least in Purkinje bres it is al., 1995; Yanni et al., 2010). Instead of degenerative brosis, remodelling
functionally important (Section 3.3.2). Two-pore-domain K+ channels of ion channel gene expression could be responsible. Downregulation of
are responsible for K+ leak or background current. ORK1 is a two- Nav1.5, Cav1.2 and other Ca2+ channel subunits, HCN1, HCN2, various
pore-domain K+ channel in Drosophila and has been shown to control K+ channels, RYR2 and Cx43 has been documented in the sinus node
the rate of diastolic depolarization and, therefore, the heart rate in in the ageing guinea-pig, rat and mouse (Jones et al., 2004, 2007; Yanni
Drosophila: downregulation of ORK1 increases the heart rate and et al., 2010; Hao et al., 2011). Atrial brillation has recently been associ-
upregulation decreases it (Lalevee et al., 2006). Various two-pore- ated with downregulation of HCN2, HCN4 and minK (and corresponding
domain K+ channels are known to be expressed in the mammalian reductions in If and IK,s) and Ca2+ clock malfunction in the sinus node of a
sinus node (TASK1, TREK1, TWIK1 and TWIK2; Tellez et al., 2011). dog atrial brillation model (Yeh et al., 2009; Joung et al., 2010).
There is some evidence that one of these channels, TREK1, is functionally Downregulation of HCN2 and HCN4 has been reported in the sinus
important in mouse sinus node pacemaking (Froese et al., 2012). node of the failing dog heart (ventricular tachypacing-induced con-
gestive heart failure; Zicha et al., 2005). A widespread remodelling
3.1.3. Clinical relevance of ion channels (e.g. a downregulation of HCN4) has been observed
in the sinus node of a rabbit heart failure model (volume and pres-
3.1.3.1. Sinus node dysfunction. Sinus node dysfunction, sometimes used sure overload; Yanni et al., unpublished data). In the same rabbit
interchangeably with the term sick sinus syndrome, is a congenital or model, a downregulation of If and IK,s has been observed in the
acquired malfunction of the sinus node. Clinically, tachy-brady syn- sinus node (Verkerk et al., 2003). A widespread remodelling of ion
drome is used to refer to sinus node dysfunction in the setting of atrial channels has been observed in the sinus node of rat models of myo-
brillation (see below) and the term will be avoided here. Sinus node cardial infarction (Yanni et al., 2011a, 2011b) and pulmonary hyper-
dysfunction presents as inappropriate sinus bradycardia, sinus arrest/ tension (Yamanushi et al., 2010). In athletically trained rats, a
sinus pause and sinus node exit block (Rubenstein et al., 1972). There widespread remodelling of ion channels has been observed in the
can be a decrease of the intrinsic heart rate (heart rate in the absence sinus node (Monfredi et al., 2011).
of autonomic inuences) and increases of the sinus node recovery time Fibrosis and ion channel remodelling may not be mutually exclusive
(time taken for pacemaking to resume after rapid pacing) and sinus mechanisms there is evidence that ion channels and brosis are
node conduction time (time taken for the action potential once initiat- linked (van Veen et al., 2005; Hao et al., 2011). In the mouse,
ed in the sinus node to conduct into the surrounding atrial muscle; knock-out of one copy of the SCN5A gene (responsible for Nav1.5 and
Benditt et al., 1995; Lau et al., 2011). Sinus node dysfunction can be INa) results in an age-dependent brosis in the sinus node (Hao et al.,
congenital (hereditary) and in some families it has been linked to 2011) and elsewhere (van Veen et al., 2005). Indeed, Hao et al. (2011)
mutations in ion channels (Section 6). However, sinus node dysfunc- showed that block of INa in cultured human cardiac myocytes and
tion is primarily a disease of ageing and its incidence increases in an broblasts results in an upregulation of the brosis gene, TGF1. Anoth-
exponential-like manner with age (Benditt et al., 1995). In the er brosis gene, NF-B, has been shown to bind to the promoter region
human and all other mammals studied, there is a decline in the intrin- of SCN5A and regulate transcription of the gene (Shang et al., 2008).
sic heart rate with age (e.g., Benditt et al., 1995). Sick sinus syndrome is
the second commonest indication for bradyarrhythmia pacing world- 3.1.3.2. If blockade in clinical practice. The BEAUTIFUL trial tested a
wide (after atrioventricular block) and pacemakers are primarily novel selective blocker of If, ivabradine, in patients with stable coronary
implanted in the elderly (Mond & Proclemer, 2011). artery disease and left-ventricular systolic dysfunction. The trial showed
In the human and/or animal models, sinus node dysfunction is ivabradine to reduce the heart rate and thereby the incidence of
also associated with heart disease: with atrial brillation (see earli- coronary artery disease outcomes (in patients with heart rates of
er comment; e.g., Elvan et al., 1996; Yeh et al., 2009), heart failure 70 beats/min) (Fox et al., 2008). Ivabradine is now approved for
(e.g., Opthof et al., 2000; Sanders et al., 2004; Zicha et al., 2005), treating angina and heart failure in Europe. Perhaps more interestingly,
myocardial infarction (Yanni et al., 2011a, 2011b), pulmonary hyper- the use of ivabradine for heart rate control in patients with left ventric-
tension (Yanni et al., 2009b), diabetes (Howarth et al., 2005, 2006) ular systolic dysfunction has been shown to signicantly improve
and possibly obesity (Yanni et al., 2010). Bradyarrhythmias account mortality in comparison to standard medical therapy (Swedberg et al.,
for a signicant number of deaths of heart failure patients, particularly 2010). The mechanism of this effect is debated; clinical data have sug-
of patients with advanced heart failure (Stevenson et al., 1993; gested that the degree of heart rate reduction correlates to the
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 269

reduction in mortality implicating a direct adverse physiological effect A newer and novel approach is to use upstream transcription factors
of elevated heart rate per se in heart failure patients (Bohm et al., to reprogramme working myocardium as pacemaker tissue. Tbx3 is one
2010). A more intriguing possibility is that electrical remodelling of of the most important transcription factors directing the development of
the myocardium in the syndrome of heart failure leads to upregulation the cardiac conduction system as discussed above in Section 2. Transfec-
of If outside of the cardiac conduction system, which then acts as an tion of Tbx3 into neonatal cardiomyocytes increases their automaticity
arrhythmogenic substrate (Song et al., 2011). and pacing rate in vitro (Boink et al., 2008). Tbx3, however, has not
It is well established that electrical remodelling of ventricular been used in animals to create a biopacemaker. Lineage analysis and
myocardium occurs in hypertension, heart failure and following Tbx3 knockout suggests that Tbx18 is necessary for the development of
myocardial infarction; furthermore it has been demonstrated that If the sinus node head (Wiese et al., 2009). Transfection of Tbx18 to neona-
is upregulated as part of this process (Cerbai et al., 1994, 1996, tal cardiomyocytes induced a pacemaker phenotype: the myocytes
2001; Cerbai & Mugelli, 2006; Stillitano et al., 2008; Xia et al., shrank in size, exhibited sparse myobrils, ceased to express Cx43 and
2010). Prevention of electrical remodelling following myocardial atrial natriuretic peptide and exhibited increased levels of cAMP and
infarction by established therapies such as ACE inhibitors may HCN4 (Kapoor et al., 2011a). Furthermore, adenoviral transduction of
include reduction of the upregulation of If, providing a mechanism Tbx18 to guinea-pig left ventricle in vivo created a biopacemaker with
for suppression of arrhythmia by pharmacological therapy that is a slow ventricular rhythm under conditions of heart block (Kapoor et
not primarily antiarrhythmic (Song et al., 2011). Blocking If directly al., 2011b). The authors elegantly demonstrated reprogramming of the
with ivabradine both reduces ventricular remodelling and increases ventricular myocytes to a pacemaker phenotype: diminished cell size,
ventricular brillation threshold following myocardial infarction myobrillar disorganisation and suppression of Cx43 and atrial natri-
(Ceconi et al., 2011; Vaillant et al., 2011) and also reduces uretic peptide expression. It has been shown that DNA histone DNA
remodelling in heart failure (Tardif et al., 2011). However, reduction methylation of the HCN4 gene locus is inactive in the ventricle normally
of death in the SHIFT study was primarily driven by reduction in and activated after Tbx18 expression. If successful, this approach has the
heart failure death rather than arrhythmia (Swedberg et al., 2010). advantage of changing the phenotype of the working myocardium rather
than just expressing a single additional channel and, therefore, the
3.1.3.3. Biopacemaking: the status quo. Regardless of the underlying biopacemaker is more likely to function like the native sinus node.
aetiology, sinus node dysfunction and resultant bradycardia account The mode of delivery of the biopacemaker product represents a
for a substantial proportion of electronic pacemaker implantations. separate challenge. Adeno-associated, lentiviral, and stem cell based
Modern electronic pacing technology is sophisticated and in many strategies have been developed to temper the shortcomings of tradi-
instances provides sufcient chronotropic modulation in response tional adenoviral gene transfer methods such as short term expression,
to physiological demands. Shortcomings, such as the risk of infec- in vivo immune responses, and lack of genome integration. Lentiviral
tion, inadequate sensitivity to adrenergic regulation, suboptimal methods may allow longer term gene expression, but pose the possibil-
cardiac activation pathways, limited battery life and magnetic inter- ity of carcinogenicity, because viral DNA is integrated into the host
ference (Den Haan et al., 2011), nonetheless, have underscored an in- genome (Wu & Burgess, 2004). An alternative approach is the use of
terest in a biological pacemaker that can offer the all elusive cure stem cells. These can be forced along a cardiogenic lineage to produce
for sinus node dysfunction. The earliest proof-of-principle report of a subset of cells expressing pacemaker characteristics, including diastol-
biopacemaking described the overexpression of 2 adrenoceptors in ic depolarization attributable to If and electromechanical integration in
mouse hearts, thereby temporarily increasing rhythm but presenting vitro (Kehat et al., 2004; Xue et al., 2005). Stem cells have also been
the possibility for arrythmogenicity (Edelberg et al., 1998). Since then, used as a vector to deliver an HCN transgene on implantation into the
the basic intent, and hence design, of the ideal biopacemaker has been dog ventricle under conditions of complete heart block (Plotnikov et
the successful mimicry of sinus node function at an ectopic site in the al., 2007). Using this approach, stable spontaneous rhythms were
working myocardium, based on gene and cell therapies. Given this de- recorded, and persisted up to three months (Kehat et al., 2004). In
scription, several strategies based on ion channel manipulation have another innovative approach, Potapova et al. (2004) described that
been advanced, although identifying the optimal construct remains a HCN2-overexpressing human mesenchymal stem cells (hMSCs) are
challenge (Rosen et al., 2009). Initial ion channel-based approaches in- able to spread depolarizing current to adjacent dog ventricular
cluded dominant-negative inhibition of Kir2.1-encoded inward rectier myocytes in co-culture, while generating a higher spontaneous beating
K+ channels (responsible for IK,1) by viral gene transfer, and thus sup- rate than co-cultures with control hMSCs overexpressing enhanced
pression of the repolarizing inuence of IK,1 in guinea-pig left ventricle, green uorescent protein alone. Taken together, currently applied cell
resulting in the production of spontaneous ventricular rhythms from and viral vector based strategies show promise in that they appear to
the implant site in the intact heart (Miake et al., 2002). A major shortcom- sustain cardiac rhythm under changing physiological demands over
ing of this approach was the proarrhythmic prolongation of action poten- limited periods of time. Critical concerns, such as the eventual fate of
tial duration resulting from the inhibition of IK,1 (Miake et al., 2002). implanted cells, the potential for carcinogenicity, and the possibility of
To overcome this limitation, more recent efforts have focussed on immuno-rejection, nonetheless, need to be thoroughly addressed
the use of the pacemaker current, If, by the adenovirus-mediated before bedside transition can become a reality.
overexpression of HCN channels in the myocardium (Tse et al., 2006).
Expression of HCN2 in the canine left atrial appendage resulted in spon- 3.2. Atrioventricular node
taneous activity during initial vagal stimulation (sinus node suppres-
sion; Qu et al., 2003). Higher levels of vagal stimulation suppressed 3.2.1. Anatomy
pacemaking in the engineered pacemaker, indicating the possibility of The AV node is part of the AV conduction axis, which is the electrical
vagal modulation. To date, pacemaking from biopacemakers in working conduit from the atria to the ventricles. It is located at the base of the
myocardium is relatively bradycardic. This is because If displays context atrial septum, at the apex of the area known as the triangle of Koch
dependence; the rate of diastolic depolarization is slower when the cells (Fig. 7; Ko et al., 2004; Li et al., 2008; Anderson et al., 2009). The triangle
are hyperpolarized as is the case in the atrial and ventricular myocardi- is bounded by the ostium of the coronary sinus, the tendon of Todaro
um (Zhang et al., 2011). Efforts to overcome this have included and the septal leaet of the tricuspid valve (Fig. 7). The three-
enhancement of HCN2 kinetics using custom designed channel genes dimensional description of the different components of the AV con-
(Kashiwakura et al., 2006), mutant or chimeric HCN channels (Bucchi duction axis is complicated by different orientations used in clinical
et al., 2006), and other HCN constructs such as HCN1/HCN4 (Rosen et electrophysiology, as well as differences amongst species. In 1999, the
al., 2009). Working Group on Arrhythmias of the European Society of Cardiology
270 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

to
ventricles
fast
pathway

FO
Right
CN
ventricle
Right
atrium
Transit-
ional
zone
Tricuspid
valve
IVC CS
Crista
terminalis

slow
pathway

Fig. 7. Schematic diagrams of the rabbit heart and AV junction. Left, schematic diagram of the rabbit heart showing the location of the AV node. From Li et al. (2006). Right,
schematic diagram of the AV junction within the triangle of Koch. Arrows show the fast and slow pathways into the AV junction. Modied from Greener et al. (2009). Ao, aorta;
CN, compact node; CS, coronary sinus; FO, fosa ovalis; IVC, inferior vena cava; LAA, left atrial appendage; PA, pulmonary artery; PV, pulmonary vein; RA, right atrium; RAA, right
atrial appendage; RV, right ventricle; SVC, superior vena cava.

and the North American Society of Pacing and Electrophysiology highlight- the AV conduction axis becomes embedded within the central brous
ed the importance of using anatomically-correct nomenclature (Cosio body, with the site of insulation being promoted by Tawara as the
et al, 1999). Sunao Tawara, a Japanese scientist working in the laborato- landmark for distinction between the compact node and the penetrat-
ry of Ludwig Aschoff in Germany, pointed out that the overall ar- ing bundle (Figs. 3B and 7). The bundle runs a short penetrating course,
rangement of the AV conduction axis can be compared to a tree, emerging on the crest of the ventricular septum as the branching AV
with roots commencing in the atrial transitional myocytes (Fig. 7), (His) bundle, albeit that in some people there can be a short non-
the trunk made of the bundle of His, and branches ending in the branching component. The branching bundle divides on the crest of
Purkinje network (discussed in more detail in Section 3.3; Tawara, the muscular ventricular septum to form the left and right bundle
2000). The reader can demonstrate this by inverting Tawara's dia- branches (see Section 3.3).
gram of the human left ventricle Purkinje system in his monograph
(Tawara, 2000) the AV conduction axis resembles a weeping wil- 3.2.2. Function
low. Studies since have testied to the accuracy of Tawara's The principal function of the AV node is to conduct the action poten-
description. tial from the atria to the ventricles. It must conduct the action potential
Spach et al. (1971) showed that, from the sinus node, the action slowly in order to introduce a delay between atrial and ventricular
potential follows two routes to the AV node: the faster route is via the systole. The delay allows atrial systole to take place, providing complete
interatrial septum and the slower route is via the terminal crest. These ventricular lling prior to initiation of ventricular systole. The AV node
connect to two inputs (or pathways) into the AV node (the AV node is has other roles. The relatively long refractory period of the AV node
said to show dual pathway electrophysiology; Moe et al., 1956; protects the ventricles from atrial tachycardias, such as atrial brillation
Meijler & Janse, 1988): the route via the interatrial septum connects and focal atrial tachycardia, by reducing the frequency of action poten-
to the fast pathway and the route via the terminal crest connects to tials transmitted to the ventricles (there is no longer 1:1 conduction).
the slow pathway (Fig. 7). The fast and slow AV node pathways are Finally, when the sinus node fails as a pacemaker, the AV node can
so-called because they are the fastest and slowest pathways for the take over as the pacemaker, because it too shows pacemaker activity,
action potential through the AV node. In summary, the fastest (and, although it is slower than that of the sinus node (Dobrzynski et al.,
therefore, normal) route for the action potential from the sinus node 2003).
through the AV node is via the interatrial septum and the fast pathway Within the AV node of the rabbit, Billette (1987) described the
of the AV node. Atrial transitional myocytes provide the fast pathway presence of six types of myocytes based on their action potential charac-
into the AV node (Fig. 7). The transitional myocytes make contact teristics. Three types of myocytes have been more extensively studied:
with the compact node, a half-oval of specialised myocytes set against nodal (N), atrio-nodal (AN) and nodo-His (NH; e.g. deCarvalho et al.,
the central brous body (Figs. 3B and 7), the brous area being the 1959; Anderson et al., 1974a, 1974b; N myocytes exhibit diastolic depo-
strongest part of the so-called brous skeleton (Anderson & Ho, larization and are capable of pacemaking. They have a Ca2+-dependent
2003). Inferiorly, the compact node has an extension (Figs. 3B and 7), action potential with a slow upstroke and occupy the inferior nodal
which passes through the septal isthmus between the coronary sinus extensions and compact node (Greener et al., 2009). AN and NH cells
and the hinge of the tricuspid leaet (Inoue & Becker, 1998; Hucker et are intermediate in nature, and are thought to be present in the transi-
al., 2008; Li et al., 2008). This inferior nodal extension constitutes the tional zone and penetrating bundle, respectively (Greener et al., 2009).
slow pathway into the AV node (Inoue & Becker, 1998; Hucker et al., Differences in action potential morphology in different areas within the
2008; Li et al., 2008); it is also continuous with the right AV ring around AV node are thought to be related to differences in ionic currents and
the tricuspid valve (Section 4.1; Yanni et al., 2009a). Traced superiorly, ion channel expression (Table 1; Hancox et al., 2003; Greener et al.,
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 271

2009, 2011). N myocytes exhibit a more positive diastolic membrane po- node. Pollack (1976) injected uorescein intracellularly by micro-
tential compared not only to atrial and ventricular myocytes, but also AN iontophoresis into the rabbit AV node and tracked its diffusion and
and NH myocytes (Inada et al., 2009a). This is due to the absence or a low showed that the rate of passage of dye between N myocytes is at least
density of IK,1 (Hancox et al., 2003). mRNA for Kir2.1 (one of the principal three orders of magnitude lower than between myocytes of the other
channels responsible for IK,1) is reduced in the AV conduction axis tissues studied; this result is consistent with published reports indicating
compared to working myocardium in both human and rabbit (Table 1; few gap junctions between AV node myocytes (Pollack, 1976). The poor
Greener et al., 2009, 2011). The poor expression of Kir2.1 in the AV coupling in the AV node can be explained by the make-up of gap
node will facilitate pacemaking caused by other currents. As in the junctions in the AV node as well as the paucity of gap junctions in the
sinus node, various ionic currents are likely to be involved in pacemaking AV node. Whereas the intermediate conductance Cx43 is the main
at the AV node. Some but not all nodal myocytes exhibit the pacemaker isoform expressed in the working myocardium, Cx43 (mRNA and
current, If (Noma et al., 1980a, 1980b; Hancox et al., 2003). Munk et al. protein) is poorly or not expressed in the inferior nodal extension and
(1996) studied rod-shaped myocytes with an AN action potential prole compact node in the human, rabbit and rat (Table 1; Yoo et al., 2006;
and oval-shaped myocytes with a N or NH action potential prole isolat- Greener et al., 2009, 2011). In contrast, the small conductance Cx45 is
ed from the rabbit AV node. Munk et al. (1996) reported that ~100% of the main connexin expressed in these regions (Table 1; Coppen et al.,
ovoid myocytes exhibit If, whereas only ~10% of rod-shaped myocytes 1999a,b; Severs et al., 2001; Coppen & Severs, 2002; Severs et al.,
exhibit the current. In the AV node, HCN4 is likely to be the most impor- 2004). Expression of Cx43 (mRNA and protein) in the transitional zone
tant isoform responsible for If (Greener et al., 2011). In the human, rabbit is also reduced in the human, rabbit and rat (Yoo et al., 2006; Greener
and rat, HCN4 (both mRNA and protein) is abundant in all areas of the AV et al., 2009, 2011). Cx43 is expressed in the penetrating bundle in the
conduction axis, particularly in the inferior nodal extension and compact human and rabbit (but not the rat; Yoo et al., Greener et al., 2009,
node (Table 1; Yoo et al., 2006; Greener et al., 2009, 2011). Block of If by 2 2011). The large conductance Cx40 can be expressed in the atrial
mM Cs+ slows pacemaking (increases the cycle length by 55%) at the muscle. In the human and rat, although, Cx40 (mRNA and protein)
rabbit AV node (Dobrzynski et al., 2003). The Ca2+ clock is also impor- is not expressed in the proximal part of the atrioventricular conduc-
tant in AV node pacemaking (in the rabbit, pacemaking is slowed by tion axis (e.g. inferior nodal extension), it is expressed in the distal
ryanodine and inhibitors of NCX1 and SERCA2; Nikmaram et al., 2008; parts (e.g. the compact node and penetrating bundle; Yoo et al.,
Cheng et al., 2011). 2006; Greener et al., 2011). Interestingly, the penetrating bundle is
N myocytes have a slow action potential upstroke: the maximum divided into upper and lower bundles with different connexin expres-
upstroke velocity of the action potential (dV/dtmax) is high in the work- sion proles: the lower bundle expresses Cx40 or Cx43, whereas the
ing myocardium, lower in the transitional AN and NH myocytes and upper bundle does not (Yoo et al., 2006; Hucker et al., 2008). This
lowest in the N myocytes. dV/dtmax is high in the working myocardium, may explain the presence of two conduction pathways within the His
because INa is responsible for the action potential upstroke in the work- bundle (Zhang et al., 2001). Interestingly, Kreuzberg et al. (2006)
ing myocardium. Munk et al. (1996) reported that 100% of rod-shaped showed that Cx30.2 slows atrioventricular conduction in mouse heart.
myocytes (with AN-type action potentials) from the rabbit AV node See Temple et al., 2013 for further discussion.
exhibit INa, whereas only ~30% of oval-shaped myocytes (with N- or What are the consequences of the pattern of expression of ion chan-
NH-type action potentials) exhibit INa (Munk et al., 1996). Whereas nels within the atrioventricular conduction axis on electrical activity?
atrial and ventricular myocytes are large, AV nodal myocytes are small As with a previous study from our laboratory on the human sinus
(Li et al., 2008). Ren et al. (2006) isolated myocytes from the rabbit node (Chandler et al., 2009), for the human AV conduction axis, we
AV node and observed that the smallest (spider-like) myocytes exhibit have explored this question by assuming that the whole-cell conduc-
an N-type action potential, intermediate-sized (spindle-shaped) tance for a particular ionic current is roughly proportional to the abun-
myocytes exhibit a transitional AN- or NH-like action potential and dance of the mRNAs responsible for the relevant ion channel; Na+-Ca2+
the largest (rod-shaped) myocytes exhibit an atrial-like action poten- exchange, SR Ca2+ release and SR Ca 2+ uptake were scaled in an anal-
tial. Ren et al. (2006) showed that both dV/dtmax and the density of ogous way (Inada et al., 2009a, 2009b). Control values for human atrial
INa correlate with the cell capacitance (a measure of cell size). In keep- muscle were taken from the mathematical model of Courtemanche et
ing with the electrophysiological ndings, Nav1.5 (primarily responsi- al. (1998) of the human atrial action potential, which is based on exper-
ble for INa) at both mRNA and protein levels has been shown to be imental voltage clamp data from human atrial myocytes. Fig. 8A shows
absent or poorly expressed in the inferior nodal extension and compact computed action potentials calculated in this way for different parts of
node in the human, rabbit and rat (Table 1; Yoo et al., 2006; Greener et the AV conduction axis in the human. The predicted action potential
al., 2009, 2011). In the human, the level of Nav1.5 (mRNA) has been waveforms are feasible for example, the inferior nodal extension
shown to be intermediate in the transitional zone (presumed to be and compact node are predicted to show pacemaking, whereas dV/
made up of AN myocytes; Greener et al., 2011). In the mouse, knock- dtmax is high in the working myocardium and least in the inferior
out of Nav1.5 slows AV nodal conduction (Papadatos et al., 2002) and nodal extension and compact node (Fig. 8A). Can the pattern of gene
mutations in SCN5A affects AV nodal conduction in the human expression explain the slow conduction of the action potential along
(Section 6). the AV conduction axis? The conduction time of the action potential
In the AV node, ICa,L rather than INa is responsible for the action poten- along a string of 300 electrically coupled myocytes was calculated
tial upstroke (e.g. Hancox & Levi, 1994). Cav1.2 is the L-type Ca2+ (Fig. 8B). In the case of atrial myocytes, the coupling conductance
channel responsible for ICa,L in much of the heart. However, in the rabbit, (1000 nS) was chosen to give an appropriate conduction velocity
there is evidence of an isoform switch from Cav1.2 to Cav1.3 in the AV (53 cm/s). In the rst simulation (white bars in Fig. 8C), atrial myocytes
node (Greener et al., 2009). In the human, Cav1.3 is upregulated in the were assumed to occupy all regions, and the coupling conductance was
AV node (Table 1; Greener et al., 2011). Cav1.3 has a more negative scaled based on the expression of connexin mRNAs in the different
activation threshold than Cav1.2 and this may be more appropriate for regions: the predicted conduction velocity was lower in the inferior
a Ca2+-dependent action potential. Another Ca2+ channel with a more nodal extension as a result of the low expression of connexins in this
negative activation threshold is Cav3.1 responsible for ICa,T. Cav3.1 region. In the second simulation (grey bars in Fig. 8C), the myocyte
(mRNA and protein) is more highly expressed in the AV node (especially type was appropriate for each region, but the coupling conductance
in the compact node) than in the working myocardium in the human was assumed to be the same in all regions (1000 nS): the predicted
(Greener et al., 2011). Knock-out of the Cav3.1 channel slows AV node conduction velocity was lower in the inferior nodal extension, this
conduction in the mouse (Mangoni et al., 2006). Since the pioneering time as a result of the low dV/dtmax in this tissue (a consequence of
work of Pollack (Pollack, 1976), coupling is known to be poor in the AV the low expression of Nav1.5 in this tissue). In the third simulation
272 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

Fig. 8. Conduction through the AV node. A, predicted action potential waveforms for the atrial muscle (AM), transitional area (TA), inferior nodal extension (INE), compact node
(CN), penetrating bundle (PB) and ventricular muscle (VM) in the human. The action potentials were based on the model of the human atrial action potential from Courtemanche
et al. (1998) and ion channel expression in the different tissues (relative to that in the atrial muscle). See Inada et al. (2009b) for details. Membrane potential is shown in the rst
row, the rate of change of membrane potential in the second row and the intracellular Ca2+ concentration in the third row. All action potentials triggered by stimuli except those in
inferior nodal extension and compact node, which are spontaneous (spontaneous cycle length, CL, shown). In simulations, the conductance for Ito was decreased by 30% in all tissues
(including the atrial muscle). B, calculation of the conduction velocity. One end of a string of 300 electrically-coupled myocytes (atrial muscle, transitional area, inferior nodal ex-
tension, compact node, penetrating bundle or ventricular muscle type) was stimulated. Pairs of action potentials (recorded at the stimulation site and the distal end of the string)
are shown. Conduction velocity was calculated from the interval between the two action potentials (assuming cells are 100 m in length and joined end to end). In examples shown,
coupling conductance was scaled according to connexin mRNA expression and the appropriate action potential model was used for each region. C, calculated conduction velocity
(D) and dV/dtmax (E) in the atrial muscle, transitional area, inferior nodal extension, compact node, penetrating bundle and ventricular muscle. In the rst set of simulations (Cx40/
Cx43/Cx45; white bars), the atrial muscle action potential model was used for all regions and the coupling conductance was scaled according to connexin mRNA expression. In the
second set of simulations (Nav1.5; grey bars), the appropriate action potential model was used for each region, but the coupling conductance not varied. In the nal set of simu-
lations (Cx40/Cx43/Cx45/Nav1.5; red bars), the coupling conductance was scaled according to connexin mRNA expression and the appropriate action potential model was used for
each region. From S. Inada (unpublished data).

(red bars in Fig. 8C), the coupling conductance was scaled based on the extension (Figs. 3B and 7; Li et al., 2008). There are differences in
expression of connexin mRNAs and the myocyte type was appropriate the refractory period of the two pathways: the refractory period of
for each region: the predicted conduction velocity was low in the infe- the fast pathway is longer than that of the slow pathway (Inada et
rior nodal extension, lower than in the two previous cases. In the inferi- al., 2009a, 2009b). Consequently, in response to a premature stimulus
or nodal extension, the predicted conduction velocity was ~4 cm/s, there can be unidirectional block (of the fast pathway with the longer
similar to that recorded experimentally in the rabbit (Inada et al., refractory period) and this sets the scene for slow-fast AVNRT
2009b). Fig. 8C suggests that the slow conduction along the human (Nikolski et al., 2003). Slow-fast AVNRT in the rabbit AV conduction
atrioventricular conduction axis is the result of the low expression of axis has been simulated using a biophysically-detailed model of the
both Cx43 and Nav1.5. In the third simulation, the predicted conduction dual pathway electrophysiology (Inada et al., 2009a).
velocity was, as expected, highest in the penetrating bundle (Fig. 8C). In the acute setting, adenosine is highly effective in terminating
AVNRT (Wilbur & Marchlinski, 1997). In nodal tissue, the effects of
3.2.3. Clinical relevance adenosine are predominantly mediated via activation of the ACh-
The AV node can act as a substrate for the generation of tachyar- activated K+ current (IK,ACh), which hyperpolarizes the cell and
rhythmias transmitted to the ventricles. The dual pathways of the shortens the action potential (Wilbur & Marchlinski, 1997). This leads
AV node form the substrate for AV nodal reentrant tachycardia to a transient conduction block within the AV node, mainly in the
(AVNRT). In AVNRT, a reentrant circuit is established, with the action slow pathway, which is sufcient to terminate the reentry circuit and
potential travelling down one pathway (usually the slow) and up the restore normal sinus rhythm (Wilbur & Marchlinski, 1997). In the
other (usually the fast) causing incessant tachycardia. Use of mapping chronic setting, many different pharmacological agents have been
strategies in the human and rabbit heart has helped elucidate these used for the treatment of AVNRT including -blockers, Ca2+ channel
pathways (McGuire et al., 1993; Inoue & Becker, 1998; Li et al., blockers, ecainide, propafenone, sotalol, dofetilide and amiodarone
2008; Kurian et al., 2010). With the use of optical mapping tech- (Blomstrom-Lundqvist et al., 2003). Because the maintenance of the
niques, Emov and his colleagues have clearly shown the rotation of reentrant circuit in AVNRT is dependent on the balance of conduction
the action potential around the two pathways (Nikolski et al., velocity and the refractory period of both the fast and slow pathways
2003). Optical mapping followed by histological examination has of the AV node, any agent that alters either of these parameters in either
suggested that the fast pathway corresponds to the transitional pathway may disrupt the AVNRT circuit. Ca2+ channel blocking drugs
zone, whereas the slow pathway corresponds to the inferior nodal bind to the L-type Ca 2+ channel directly to inhibit ICa,L, whilst
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 273

-blockers act indirectly by antagonising the increase in ICa,L due to networks are complex three-dimensional structures, with a combi-
-stimulation (Abernethy & Schwartz, 1999). The reduction in ICa,L nation of subendocardial and free running bres (Fig. 10). Typically,
leads to a reduction of conduction velocity within the AV node the left bundle branch is a fan-like structure (Fig. 10A,Ci,Cii), where-
(Van De Ven et al., 1996; Zicha et al., 2006). The VaughanWilliams as the right bundle branch is cord-like (Fig. 10B; Atkinson et al.,
class 1 agents, ecainide and propafenone, bind to the Na + channel 2011). The left bundle branch extends to the mid portion of the septum
reducing INa and slowing conduction in the working atrial myocardi- before it detaches from the underlying endocardium and forms
um and the fast pathway (Hoff et al., 1988). The VaughanWilliams free-running false tendons that traverse the ventricular chamber
class III agents, amiodarone and dofetilide, act by increasing the (Fig. 10A). The free-running strands reattach at the free wall myocardi-
refractory period of both the fast and the slow pathway, predomi- um, forming a complex sub-endocardial network that covers a large
nantly by block of the outward K + currents IK,r and IK,s (Kodama et portion of the free wall, but projecting predominantly towards the pap-
al., 1997a, 1997b). Despite the use of multiple classes of drugs, some- illary muscles (Fig. 10A,Civ).
times in combination, there is a high risk of recurrence of AVNRT in The cord-like right bundle branch emerges beneath the membranous
addition to the medications' potential side effects and adverse septum and runs towards the apex of the heart on the septal endocardi-
events. This has led to the adoption of catheter ablation of the slow um, with free running strands extending from the bundle branch
pathway as rst line therapy for AVNRT with success rates estimated towards the apical trabeculated portion of the ventricle (Fig. 10B). The
to be around 95% (Blomstrom-Lundqvist et al., 2003). ventricular cavity is also bridged via connections through the moderator
Conduction through the AV node can be pathologically slowed band (Fig. 10Cvi). The bundle branches are insulated from the underlying
resulting in AV or heart block (Fig. 9). First-degree heart block is a myocardium by connective tissue sheaths (Ansari et al., 1999), enabling
delay in conduction of the action potential through the AV node, the transfer of action potentials to the apex of the heart without
resulting in prolongation of the PR interval on the ECG. In second- activating the ventricular myocardium at the base of the heart rst,
degree heart block, only some atrial action potentials are transmitted ensuring the controlled activation of the ventricular myocardium. The
to the ventricles, which results in some P waves without an accompany- sub-endocardial Purkinje network covers large portions of the ventricu-
ing QRS complex in the ECG. Third-degree heart block is the absence of lar myocardium and forms the terminal portion of the ventricular
conduction from the atria to the ventricles. An escape ventricular conduction system. At specic sites the insulating sheath is lost, allowing
rhythm is usually present with third degree heart block. The higher action potentials to propagate to the ventricular working myocardium
the degree of block, the more likely mechanical permanent pacemaker (Tranum-Jensen et al., 1991; Ryu et al., 2009). Transitional myocytes
implantation will be required to restore AV synchrony. Heart block is are present at the junction of the sinus node with the atrial muscle
commonly idiopathic. However, it may be congenital (Section 6). The (Section 3.1.1) and the junction of the atrial muscle with the AV node
incidence of heart block increases with ageing (Piccini & Calkins, (Section 3.2.1) and it is interesting that there also exists transitional
2005). Heart block may be secondary to ischaemic heart disease, cardio- myocytes at the junction of the Purkinje myocytes with the ventricular
myopathy, heart failure (Crisel et al., 2011) or increased vagal tone muscle (Severs, 2000). Presumably the transitional myocytes are present
(Imaizumi et al., 1990). Fig. 9 shows standard 12 lead ECG recordings to minimise source-sink mismatch.
from a patient with a normal ECG (Fig. 9A) and two heart failure
patients (Fig. 9B, C). The second of the heart failure patients (Fig. 9C) 3.3.2. Function
has rst-degree heart block (the PR interval in Fig. 9C is >200 ms and The principle function of the HisPurkinje system is to rapidly
approximately double that in Fig. 9A) as well as other conduction disor- conduct the action potential throughout the ventricles to ensure the
ders: mild sinus bradycardia (the heart rate in Fig. 9C is ~60 beats/min ventricular muscle contracts in the correct sequence. A second func-
compared to ~92 beats/min in Fig. 9A) and an intermittent left bundle tion is to act as a ventricular pacemaker in the event of heart block.
branch pattern (intermittent broad QRS complexes in Fig. 9C compared The HisPurkinje system is highly specialised for the rapid conduction
to narrow complexes in Fig. 9A). Heart failure patients with a prolonged of the action potential with a conduction velocity of 2.3 m/s as com-
PR interval have a poor outcome in terms of death, urgent transplanta- pared to 0.75 m/s in the ventricular muscle (Desplantez et al., 2007).
tion or cardiovascular hospitalisation (Gervais et al., 2009). Fascinating- One reason for the high conduction velocity is the expression prole
ly, the incidence of heart block is increased in athletes (Maron & of connexins: unlike the sinus node and the AV node where the small
Pelliccia, 2006). Finally, heart block can occur as a side-effect of medical conductance Cx45 is predominantly expressed, the HisPurkinje sys-
therapy or ablation procedures. In an analogous way as gene therapy is tem shows expression of large and medium conductance connexins,
being used to generate a biopacemaker (Section 3.1.3.3), gene therapy Cx40 (Gaborit et al., 2007; Atkinson et al., 2011) and Cx43 (Gourdie et
has been used to control the AV node (Bauer et al., 2004). al., 1993). In the rat, Cx40 is predominantly expressed in the proximal
His-bundle and bundle branches, whereas both Cx40 and Cx43 co-
3.3. His bundle, bundle branches and Purkinje network localise in the distal Purkinje bre network (Gourdie et al., 1993).
In genetically modied mice with Cx40 deciency, action potential
3.3.1. Anatomy conduction in the left bundle branch is impaired and a lack of ex-
The ventricular portion of the cardiac conduction system from the pression of Cx40 in the proximal bundles correlates with right
AV node to the ventricular working myocardium is vital. The structure bundle-branch block (van Rijen et al., 2001), demonstrating the
of the system ensures the synchronous activation of the ventricular importance of connexin expression for effective conduction in the
myocardium to achieve the most efcient pumping activity. The His HisPurkinje system.
bundle, or penetrating bundle, is the insulated component of the AV A second reason for the fast conduction velocity of the action
conduction axis, becoming recognisable anatomically when the potential in Purkinje bres concerns the nature of the action poten-
axis becomes insulated from the surrounding atrial myocardium by tial in the Purkinje bres. The Purkinje bre action potential is not
collagenous tissue (Fig. 3B). The bundle thus formed provides, in nodal-like. As compared to the ventricular action potential, the
the normal heart, the only AV conduction pathway. The asymmetric action potential in Purkinje bres has a faster upstroke velocity
left bundle branch and right bundle branch are formed by the bifur- (dV/dtmax), higher amplitude, more prominent early phase of rapid
cation of the His bundle at the crest of the ventricular septum repolarization (phase 1) and a more negative plateau potential and,
(Miquerol et al., 2004; Atkinson et al., 2011). The Purkinje networks most importantly, it is longer in duration (Fig. 11; mouse, Anumonwo
formed at the terminations of the bundle branches are comparable et al., 2001; rabbit, Persson et al., 2007; dog, Balti et al., 2000; Koncz
between species, as shown initially by Tawara (2000) and now con- et al., 2011). The fast upstroke and large amplitude of the action poten-
rmed by others (Massing & James, 1976; Miquerol et al., 2004). The tial is the second reason for the fast conduction velocity of the action
274 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 275

Fig. 10. HisPurkinje network in the left and right ventricles of the rabbit. A, macroscopic image of immunoenzyme-labelling (dark brown signal) of middle neurolament on the
endocardial surface of the left ventricle. B, macroscopic image of immunolabelling of middle neurolament on the endocardial surface of the right ventricle. C, high magnication
images. Labels in A and B indicate the location of the high magnication images in C. Ci, origin of left bundle branch (LBB) in the membranous septum near the root of the aorta. Cii,
mid-septal left bundle branch. Ciii, division of the left bundle branch prior to forming free-running Purkinje bres. Civ, left bundle branch and free-running Purkinje bres. Connec-
tion to anterior papillary muscle (PM) is visible. Cv, free-running Purkinje bres in the left ventricle. Cvi, right bundle branch (RBB) and moderator band. Cvii, right bundle branch
running around the base of the papillary muscle on the right ventricular septum with free-running Purkinje bres branching from it. The right bundle branch runs endocardially on
the septal surface crossing the ventricular chamber via the moderator band. Cviii, peripheral Purkinje bre network on the right ventricular free wall.
From Atkinson et al. (2011).

potential in Purkinje bres. Fig. 8 shows the importance of both strong have suggested that the action potential differences between Purkinje
electrical coupling provided by connexins and a fast action potential in bres and ventricular muscle in the rabbit are the result of ICa,T in
determining a high conduction velocity in the penetrating bundle at the Purkinje bres but not ventricular muscle and increases in the densi-
start of the HisPurkinje system. The long duration of the Purkinje bre ty of INa and the late Na + current (INa,L) and decreases in the density
action potential makes the Purkinje bres prone to certain drug- of ICa,L, IK,r, IK,s and IK,1 in the Purkinje bres (Fig. 11; Aslanidi et al.,
induced early after depolarizations (EADs; Roden & Hoffman, 1985; 2010). These differences are consistent with the ion channel expres-
Nattel & Quantz, 1988; Kaseda et al., 1989; Boyden et al., 2000; sion prole in Purkinje bres in the human (Table 1; Gaborit et al.,
Cordeiro et al., 2001a, 2001b; Persson et al., 2007) can lead to torsades 2007) and rabbit (Table 1; Atkinson et al., 2011). There is higher ex-
des pointes, a life-threatening cardiac arrhythmia. Gender differences pression of Nav1.5 mRNA in rabbit Purkinje bres at least explaining
have been observed in dog Purkinje bres: action potentials are longer the higher density of INa (and possibly INa,L) and, therefore, the faster
in female dogs in spite of there being no difference in the resting poten- action potential upstroke (Fig. 11; Atkinson et al., 2011). There is a
tial, dV/dtmax or action potential amplitude (Abi-Gerges et al., 2004, lower expression of Cav1.2, ERG, KvLQT1 and Kir2.1 mRNAs in rabbit
2006). Such differences are thought to underlie the increased suscepti- Purkinje bres at least explaining the lower densities of ICa,L, IK,r, IK,s
bility of women to torsades de pointes (Makkar et al., 1993; Abi-Gerges and IK,1, respectively, and, therefore, helping to explain the longer
et al., 2006). Computer simulation studies based on patch clamp data action potential (Fig. 11; Atkinson et al., 2011).

Fig. 9. Standard 12 lead ECG recordings from normal and heart failure patients. A, ECG from normal patient in sinus rhythm. The PR interval is normal (b200 ms) and the QRS com-
plex narrow (b120 ms). B, ECG from heart failure (HF) patient with left bundle branch block (as indicated by broad QRS complexes; >120 ms), left axis deviation and atrial bril-
lation (as indicated by the absence of P waves and an irregularly irregular ventricular rhythm). C, ECG from heart failure patient with a slow heart rate (prolonged RR interval),
rst-degree heart block (prolonged PR interval; >200 ms) and intermittent left bundle branch pattern (broad QRS complexes; >120 ms).
From Dr. J. Fraser (unpublished data).
276 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

al., 1987). Several ionic currents are thought to contribute to the


diastolic depolarization in Purkinje bres. A low density of IK,1 and
poor expression of Kir2.1 in Purkinje bres is one reason why Purkinje
bres can show pacemaking (Fig. 11; Gaborit et al., 2007; Aslanidi et
al., 2010; Atkinson et al., 2011). If (DiFrancesco, 1981a, 1981b;
Callewaert et al., 1984; Yu et al., 1995; Schram et al., 2002) and the
HCN ion channel family (Shi et al., 1999; Gaborit et al., 2007; Atkinson
et al., 2011) responsible for If are present in Purkinje bres of a number
of species including human. Whole cell patch-clamp studies have
revealed that steady-state activation of If occurs at physiological poten-
tials in dog Purkinje cells (80 to 130 mV). In contrast, If activation in
dog ventricular myocytes occurs at more negative potentials (120 to
170 mV; Yu et al., 1995). Koncz et al. (2011) have shown that
inhibiting If with ivabradine blocks pacemaking in dog Purkinje bres,
suggesting an important role for this current. However, an earlier
study had reported that lidoazine, a drug that decreases If in
sheep Purkinje bres (Hart & Dukes, 1984), had no effect on the
pacemaker activity in dog Purkinje bres (Dangman & Miura,
1987). Another potential contributor to the diastolic depolarization,
ICa,T, is substantially larger in Purkinje myocytes, but its inhibition
with mibefradil does not affect pacemaking (Pinto et al., 1999).
Several other currents including a voltage- and time-dependent K + cur-
rent (IK,dd) (Vassalle et al., 1995) and a voltage- and time-dependent
inward Na + current (INa3) have been reported to contribute to pace-
making in Purkinje bres (Rota & Vassalle, 2003; Vassalle, 2007). The
pacemaker activity of Purkinje bres is usually suppressed by the
phenomenon of overdrive suppression. In normal hearts during
regular sinus rhythm, the Purkinje bres are excited at frequencies
higher than their intrinsic rate, leading to increased inux of Na+ and
a rise of the intracellular Na+ concentration (Boyett et al., 1987)
followed by enhanced electrogenic Na +/K+ pump activity that results
in hyperpolarization of the resting membrane and a suppression of
diastolic depolarization (Vassalle, 1970; Boyett & Fedida, 1984).
Purkinje bres are susceptible to another type of pacemaker activity.
However, this pacemaker activity is abnormal and occurs when the
Purkinje bres are Ca2+-overloaded and has all of the characteristics of
Fig. 11. Purkinje bres relating mRNA expression to function. A, current densities in the Ca2+ clock (Tsien et al., 1979). Following an action potential, there
Purkinje bres comparison of the best estimates from voltage clamp experiments can be a spontaneous release of Ca2+ from the SR leading to a Ca2+
with the predictions from ion channel mRNA expression levels. Black bars, current wave (the Ca2+ waves are sensitive to treatment with ryanodine and
density in rabbit left ventricular Purkinje cells (as a fraction of current density in rabbit
thapsigargin conrming a role for Ca2+ release from the SR; Boyden et
left ventricular endocardial myocytes; based on Aslanidi et al., 2010). The peak INa den-
sity was measured during a voltage clamp pulse to 25 mV from a holding potential of al., 2000, 2004). Some of the released Ca2+ is extruded out of the cell
75 mV. The peak ICa,L density was measured during a pulse to +10 mV from a hold- by the Na+Ca2+ exchanger resulting in a transient inward current
ing potential 40 mV. The IK,r tail current density was measured at the holding poten- (ITI) that elicits a depolarization: a delayed afterdepolarization (DAD;
tial of 50 mV after a 300 ms pulse to +20 mV. The IK,s tail current density was
Boyden et al., 2000). If the DAD reaches threshold an ectopic action
measured at the holding potential of 50 mV after a 1 s pulse to +20 mV. The IK,1 den-
sity was measured as the positive peak value at 70 mV. Grey bars, mRNA expression
potential is initiated. Purkinje bres are susceptible to DADs, which can
level of the ion channel or ion channels responsible for each of the ionic currents in be triggered by rapid stimulation and drug-induced (Ferrier et al., 1973;
rabbit left ventricular Purkinje bres (as a fraction of the expression level in rabbit Lederer & Tsien, 1976). Despite this, expression of Ca2+-handling pro-
left ventricular muscle). B, computed action potentials. Black trace VM, action poten- teins such as RYR2, RYR3, SERCA2a and NCX1 in the Purkinje bres is
tial computed for a rabbit ventricular endocardial myocyte using the model of Aslanidi
low (Table 1; Gaborit et al., 2007; Atkinson et al., 2011). The Purkinje -
et al. (2010). Grey trace PF (mRNA), action potential computed using the rabbit ven-
tricular endocardial myocyte model modied by changing ionic current conductances bres share this in common with the two nodes (Table 1). Perhaps this is
based on the expression of the corresponding ion channel mRNAs. The computed ac- consistent with the limited contractile function of the Purkinje bres
tion potential resembles that of a Purkinje bre. Black dashed trace PF, action poten- and the two nodes.
tial computed from the rabbit Purkinje bre model of Aslanidi et al. (2010). It has already been mentioned that Purkinje bres have a lower den-
From Atkinson et al. (2011).
sity of IK,1 (Cordeiro et al., 1998) and lower expression of Kir2.1
(Atkinson et al., 2011). It is perhaps for this reason that the steady-
Purkinje bres can show pacemaking, although it is slower and state currentvoltage relationship of dog Purkinje bres is shallow
less robust than pacemaking in the two nodes. Pacemaker activity and N-shaped and crosses the current axis twice, i.e. there are two
with diastolic depolarization during phase 4 of the action potential potentials at which the net current is zero (Gadsby & Craneeld, 1977;
has been observed in isolated (non-driven) multicellular Purkinje Boyden et al., 1989). As a result, dog Purkinje bres have two levels of
bre preparations of the mouse (Anumonwo et al., 2001), rabbit stable resting potential, one near 90 mV and one near 50 mV (corre-
(S.J.R.J. Logantha, unpublished observation) and dog (Davis & sponding to the two zero current potentials) and the Purkinje bre can
Temte, 1969; Kus & Sasyniuk, 1975; Balti et al., 2000; Koncz et al., be ipped from one resting potential to the other by a small perturba-
2011). In single myocyte studies, Purkinje myocytes with less nega- tion (Wiggins & Craneeld, 1976; Gadsby & Craneeld, 1977). This is
tive diastolic potentials show pacemaker activity (Callewaert et al., functionally important, because at the high resting potential the action
1984; Robinson et al., 1987), but well polarised (85 mV) myocytes potential has a fast INa-dependent upstroke and is fast conducting
remain quiescent even after catecholamine treatment (Robinson et (Callewaert et al., 1984), whereas at the low resting potential the action
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 277

potential is slow conducting and sensitive to treatment with Ca2+ antag- cause left ventricular dysfunction with abnormal lling times and
onists (Molyvdas & Sperelakis, 1983; Amerini et al., 1985) and perhaps ejection fractions (Grines et al., 1989). Just like sinus node dysfunction
has a ICa,L-dependent slow upstroke (i.e., nodal-like). Experimentally, and heart block, the incidence of bundle branch block (left and right) in-
the slowly conducting action potential can lead to re-entry around a creases during ageing; the prevalence of bundle-branch block increases
network of Purkinje bres (Wit et al., 1972). from 1% at age 50 years to 17% at age 80 years (Eriksson et al., 1998). Bun-
dle branch block is associated with coronary artery disease, congestive
3.3.3. Clinical relevance heart failure and progression to complete heart block (Maddali, 2010).
The HisPurkinje system plays an important role in arrhythmogenesis For example, 26% of heart failure patients are reported to have left bundle
and is linked with a number of different arrhythmias (Boyden et al., 2003; branch block (Padeletti et al., 2010). It is for this reason that some heart
Scheinman, 2009). Purkinje bres have been shown to be prone to EADs failure patients require resynchronisation therapy. It is likely that bundle
due to their long action potential, which can lead to the torsade de branch block in the setting of heart failure at least is the result of
pointes arrhythmia in patients with long QT syndrome (Schram et al., remodelling of ion channels etc. in the HisPurkinje network. Maguy et
2002; Ben et al., 2008; Dun & Boyden, 2008). Abnormal automaticity al. (2009) have demonstrated a downregulation of Kir2.1, Nav1.5, Kv3.4,
(e.g. as a result of DADs) in the HisPurkinje system is another potential Kv4.3, Cx40 and Cx43 in the Purkinje bres of a dog model of heart failure
cause of arrhythmias. Arnar et al. (1997) speculated that such abnormal (ventricular tachypacing model). There was a decrease in dV/dtmax
automaticity may be responsible for the episodes of spontaneous ven- (a likely consequence of the downregulation of Nav1.5) as well as the
tricular tachycardia (VT) of focal Purkinje origin (60% of total) occurring conduction velocity (a likely consequence of the decrease in dV/dtmax
in the rst 30 min after coronary artery occlusion in a dog model. A and downregulation of Cx43) and this could explain the dyssynchronous
slowing of conduction in the HisPurkinje system is an important risk ventricular activation (Maguy et al., 2009). Consistent with these nd-
factor for reentrant arrhythmia formation and can lead to bundle branch ings, we have shown a widespread remodelling of ion channels etc. in a
reentry VT and fascicular VT. Bundle branch reentry VT typically occurs in rabbit model of heart failure (Yanni et al. unpublished data).
patients with impaired ventricular function, particularly in patients with
dilated cardiomyopathy. There are a number of different bundle branch 4. Atrioventricular ring tissue:
reentrant circuits causing ventricular tachycardia. The commonest form anatomy, function and clinical relevance
involves retrograde activation of the left bundle branch and antegrade ac-
tivation of the right bundle branch (Balasundaram et al., 2008). Common- 4.1. Anatomy
ly, ablation of the right bundle branch is performed to abolish these
re-entrant circuits (Balasundaram et al., 2008). Ablation of the left bundle In the normal postnatal heart, the atrial and ventricular muscle
branch is possible to perform, but due to the broad structure of the left masses are separated at the AV junctions by connective tissues, with
bundle branch it is more difcult to perform (Balasundaram et al., the plane of insulation thus formed crossed only by the AV conduction
2008; Crijns et al., 1995). Reentrant HisPurkinje system circuits can axis, thus allowing normal conduction of the action potential generated
also be established in the opposite direction or involving only the two fas- by the sinus node to the ventricles. In addition to the histologically-
cicles of the left bundle branch. The HisPurkinje system can also be in- specialised tissues forming the AV conduction axis, we now know that
volved in ventricular arrhythmias in structurally normal hearts. In additional histologically specialised rings encircle the orices of the
fascicular VT a reentrant circuit involving both fast conduction and slow tricuspid and mitral valves, these rings taking their origin from inferior
conduction within then Purkinje network, most commonly the left poste- extensions of the AV node (Fig. 12A; Yanni et al., 2009a). The rightward
rior Purkinje network, causes VT. During ablation procedures the His inferior extension from the AV node extends around the vestibule of the
Purkinje system is mapped to identify the region of slow conduction tricuspid valve, whereas the leftward inferior extension encircles the
within the Purkinje system and then radio frequency ablation is used to orice of the mitral valve (Fig. 12A). On reaching the atrial septum,
abolish the Purkinje network at this site. Ectopic beats from the Purkinje having passed round the right AV orice, the tricuspid ring crosses
network have also been shown to be the initiating trigger for arrhythmia over the AV conduction axis, separated from it by the central brous
in a high proportion of survivors of VF without structural heart disease. body, and reunites with the mitral ring to produce a superiorly located
Ablation at the site of these triggers has been shown to be effective in re- retroaortic node. The retroaortic node is located within the atrial walls
ducing the recurrence of VF (Hassaguerre et al., 2002; Wright et al., immediately dorsal to the aortic root. Taken overall, therefore, the AV
2009). The Purkinje-muscle junctions have also been shown to be impor- rings form a gure-of-eight structure around the mitral and tricuspid
tant sites for the generation of triggered activity involved in arrhythmias junctions, effectively creating a solitary ring of specialised tissue that
(Berenfeld & Jalife, 1988; Gilmour & Watanabe, 1994; Li et al., 1994; encircles the right and left AV junctions (Fig. 12A; Yanni et al., 2009a).
Mazur et al., 2005; Nogami, 2011). The HisPurkinje system may be in- The histologically-specialised cardiomyocytes forming the rings are
volved in catecholaminergic polymorphic ventricular tachycardia less extensive around the mitral orice than the tricuspid orice. In
(CPVT) and this is dealt with in Section 6. The HisPurkinje system is sen- terms of their histology, the myocytes making up the rings satisfy two
sitive to a number of drugs that increase action potential duration of the three criteria established by Aschoff and Monckeberg in 1910
and trigger arrhythmias (Gupta et al., 2007; Champeroux et al., (Anderson et al., 2000) to justify recognition as being specialised, in
2011). Drugs that reduce action potential duration or limit automa- that the myocytes are histologically discrete, and can be traced from
ticity (e.g. verapamil and lidocaine) can be used to treat arrhythmias section to section in serially prepared material. The rings, however,
linked to the HisPurkinje system (Hirata et al., 1979; Dersham & are not insulated from the atrial myocardium, albeit being separated
Han, 1980; Kuo & Reddy, 1981). Magnesium has been used as a treat- by the plane of AV insulation from the ventricular myocardium.
ment for torsade de pointes arrhythmia originating in the His
Purkinje system in long QT syndrome and has been shown to behave 4.2. Function
in a similar fashion to TTX (which blocks INa; Kaseda et al., 1989;
Gupta et al., 2007). There are only a few studies of the electrophysiology of the AV ring
Another problem with the HisPurkinje system is bundle branch tissue. Studies of pig, dog and rabbit multicellular preparations have
block. An example of left bundle branch block in the setting of heart revealed nodal-type action potentials in the AV ring region (deCarvalho
failure is shown in Fig. 9B and is characterised by a broad QRS complex et al., 1959; Horibe, 1961; McGuire et al., 1994). In pig and dog hearts,
(compare to the normal narrow QRS complex in Fig. 9A) as a result of such nodal-type action potentials are present within 12 mm of the
slowed ventricular activation. Right bundle branch block is thought to annulus, surrounding the entire tricuspid valve. A circumferential zone
be asymptomatic, but left bundle branch block has been shown to (1 cm wide) of myocytes with transitional action potentials separates
278 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

A B SN

RA

LR
LV
RR AVN
RV VS 1 mm

C D
RR

LR

Total RA 73% Total LA 27%

Fig. 12. AV ring tissue. A, schematic diagram of AV ring tissue and its association with the established AV conduction axis. From Yanni et al. (2009a). B, section through a rat heart
double labelled for HCN4 (green signal) and Cx43 (red signal). HCN4 labelling identies the sinus node, AV node and right and left rings, whereas Cx43 labelling identies the work-
ing myocardium of the right atrium, right ventricle, ventricular septum and left ventricle. From H. Dobrzynski et al. (unpublished image). C, location of sites (black circles) in the
atria of patients ablated to terminate atrial tachcardia. Sites around the tricuspid and mitral valves are shown. From Kistler et al. (2006). D, pacemaker action potentials recorded
from the sinus node and right AV ring in the rabbit heart. From Rozanski and Jalife (1986). AVN, atrioventricular node; LR, left ring; LV, left ventricle; MV, mitral valve; RA, right
atrium; RR, right ring; RV, right ventricle; SN, sinus node; TV, tricuspid valve; VS, ventricular septum.

the myocytes with nodal-type action potentials from atrial myocytes level (Table 2). In the rabbit and the rat, it has been shown that the
(McGuire et al., 1994, 1996). When action potentials were recorded left and/or right AV rings express HCN4 and less Nav1.5 and Cx43
from myocytes progressively closer to the tricuspid valve, from any at the protein level (Fig. 12B; Yoo et al., 2006; Yamamoto et al.,
point around the AV ring, the maximum diastolic potential became 2006; Yanni et al., 2009a). This expression prole is consistent with
less negative and there was a gradual diminution of dV/dtmax of the the known electrophysiology of AV ring tissue.
action potential, disappearance of the spiked reversal and, nally,
appearance of slowly rising, rounded nodal-type action potentials 4.3. Clinical relevance
(deCarvalho et al., 1959; Horibe, 1961; McGuire et al., 1994). The
gradual change in action potential characteristics within the AV A number of clinical investigations have identied tachycardia foci
ring tissue is accompanied by a decrease in conduction velocity around the tricuspid valve annulus as well as the mitral annulus
(deCarvalho et al., 1959). Recent work in our laboratory has shown (Fig. 12C; Morton et al., 2001; Kistler et al., 2003, 2006; Tada et al.,
that the AV ring tissue in the rat shows pacemaking involving both 2007). It is likely that the tachycardia is arising in the AV ring tissue.
the membrane and Ca2+ clock mechanisms: both block of If (by Cs+) The AV rings in the human heart were erroneously identied by
and RYR2 (by ryanodine) slows pacemaking (S.J.R.J. Logantha, Kent (1913) as providing multiple pathways for AV conduction in
unpublished observations). In the hearts of a number of species, there the normal heart. The WolffParkinsonWhite syndrome is a heart
are sparsely arranged myocytes in the AV valve leaets (Wit et al., condition in which there is pre-excitation (i.e. premature excitation)
1973; Basset et al., 1976; Wit & Craneeld, 1976; Rozanski & Jalife, of the ventricles (Anderson et al., 1996). The WolffParkinsonWhite
1986). The myocytes in the AV valve leaets exhibit pacemaking syndrome is caused by the presence of an abnormal accessory electrical
(Fig. 12D) that is sensitive to block of If (with Cs+) and ICa,L (with Ni+ conduction pathway, which bypasses the AV node, between the atria
or verapamil; Rozanski & Jalife, 1986; Rozanski, 1987; Anumonwo et and the ventricles. While the majority of cases are the result of an ab-
al., 1990). If has been recorded from myocytes from the AV valve leaets normal tract of working myocardium connecting the atria and ventricles,
(Anumonwo et al., 1990). Pacemaking in the valve leaets is potentiat- the Mahaim variant of the syndrome is the result of decremental con-
ed by treatment with adrenaline and inhibited by ACh, suggesting that duction along an abnormal tract of nodal-like myocytes (a Kent bundle)
intrinsic autonomic stimulation might regulate ectopic activity in this connecting the atria and ventricles (Anderson et al., 1996). In the case of
tissue (Wit et al., 1973; Basset et al., 1976; Wit & Craneeld, 1976; the Mahaim variant, the tracts of nodal-like myocytes arise in the nodes
Wit et al., 1979). of Kent, i.e. the AV rings (Anderson et al., 1996). The accessory pathway
The AV rings have a distinct prole of expression of major ion may form part of a macroreentrant circuit that also involves the atri-
channels similar to that of the sinus and AV nodes (Table 2). In the um, AV node and ventricle termed atrioventricular reentrant tachy-
rat, like the sinus and AV nodes, the right AV ring and the retroaortic cardia (AVRT) (Blomstrom-Lundqvist et al., 2003). There is also a
node express HCN1 and HCN4, but they tend to express less (as com- risk of sudden cardiac death associated with accessory pathways
pared to atrial myocardium) Kir2.1, Nav1.5 and Cx43 at the mRNA in patients with atrial brillation (pre-excited atrial brillation)
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 279

Table 2 are separated by a large muscular ridge, the supraventricular crest


Ion channel expression in the rat at the mRNA level in the sinus node, AV node, right AV
(Anderson et al., 1977). It is the basal part of the crest that forms the
ring, retroaortic node and ventricular muscle as compared to that in the atrial muscle.
AM, atrial muscle. *, signicantly upregulated; *, signicantly downregulated; , free-standing support for the leaets of the pulmonary valve.
trend to upregulation; , trend to downregulation; =, no difference. From Atkinson et
al. (unpublished data). 5.2. Function and clinical relevance
Rat
Action Region (expression relative to atrial muscle) Over the last quarter of a century, arrhythmias arising in the
Ionic potential
Ion
channel current Phase Sinus Atrial Right AV Retroaortic Ventricular
right ventricular outow tract have acquired a degree of notoriety,
AV node
4 0 1 2 3
node muscle Ring node muscle because of their propensity to afict young patients, potentially
HCN1 /f leading to sudden cardiac death. In some patients thus aficted,
HCN4 /f the arrhythmias have a clear macroscopic anatomical basis, such
Kir2.1 /K1 as those associated with arrhythmogenic right ventricular dyspla-
/Na =
Nav1.5 sia or tetralogy of Fallot. In other patients, the arrhythmias do not
Cav3.1 /CaT = = = =
appear to be associated with overt macroscopic anatomical abnor-
Cav1.2 /CaL = = = =
Cav1.3
malities, such as those seen in Brugada syndrome, or right ventric-
/CaL

Kv1.4 /to = = = = =
ular outow tract-VT. Brugada syndrome is a genetic arrhythmia
Kv4.2 /to = = = syndrome linked to abnormalities in the cardiac Na + channel,
Kv1.5 /Kur = Nav1.5 (discussed in more detail in Section 6). It is a recognised
ERG /Kr = = = = cause of sudden cardiac death in patients with structurally normal
KvLQT1 /Ks = = = = = hearts. Patients affected may have a characteristic resting ECG pat-
Kir3.1 /KACh tern, although this may only become apparent with provocation,
Cx40 = = = =
and so estimating its true prevalence is difcult (Antzelevitch,
Cx43 =
= =
2006). Death usually occurs due to polymorphic VT or ventricular
Cx45 = =
NCX1 = = =
brillation (Brugada et al., 2002). The right ventricular outow
SERCA2 = = = tract is often implicated in the occurrence of these terminal ar-
RYR2 = = = = rhythmias through a number of proposed mechanisms, including
impaired or heterogeneous repolarization (Yan & Antzelevitch,
1999; Capulzini et al., 2010), conduction delay (Meregalli et al.,
2005) or even abnormalities of embryogenesis leading to erroneous
migration of neural crest cells and consequent non-homogenous
expression of connexins (Elizari et al., 2007). Right ventricular outow
as the rapid atrial rates in atrial brillation may be conducted to the tract-VT is generally a more benign condition. It belongs to the group
ventricles much more rapidly than would be permitted by the AV of disorders known as idiopathic VTs, usually presenting between the
node (Blomstrom-Lundqvist et al., 2003). Medical therapy for AVRT is ages of 20 and 50 with palpitations or presyncope. Idiopathic VTs are
similar to that for AVNRT (Section 3.2.3) and includes -blockers, Ca2+ so-called because, when investigated fully, there are no denable ab-
channel blockers, ecainide, propafenonoe, sotalol, dofetilide and normalities on clinical examination, the resting ECG, echocardiography
amiodarone (Blomstrom-Lundqvist et al., 2003). Given that both the or coronary angiography. The advent of high resolution magnetic reso-
AV node and the accessory pathways are part of the reentry circuit, ei- nance imaging (MRI) has uncovered a proportion of such cases that
ther element can be targeted. -blockers and Ca2+ channel blockers tar- would previously have been dened as idiopathic VT, which have a
get the AV node via ICa,L, ecainide and propafenone slow conduction clearly dened pathological process, albeit often at an early stage,
via blockade of INa and amiodarone and dofetilide prolong the refractory including early or limited arrhythmogenic right ventricular dysplasia.
period predominantly by block of IK,r and IK,s (Abernethy & Schwartz, Still more patients have focal abnormalities on MRI, including wall
1999; Kodama et al., 1997a, 1997b; Zicha et al., 2006). Just as medica- thinning or abnormal wall motion, though whether these are clinically
tion often fails in AVNRT, it is also often unsuccessful in AVRT and, par- relevant is unclear (O'Donnell et al., 2003; Capulzini et al., 2010).
ticularly given the risk of sudden death associated with AF and an There then remains a signicant population, which experiences VT
accessory pathways, catheter ablation of the accessory pathway is the despite having a completely structurally normal heart, meaning that
usual rst line therapy (Blomstrom-Lundqvist et al., 2003). the classication of idiopathic VT has remained clinically relevant. This
group of disorders accounts for as many as 10% of all newly presenting
5. Right ventricular outow tract: cases of VT (Brooks & Burgess, 1988; Bunch & Day, 2006; Arya et al.,
anatomy, function and clinical relevance 2007), with outow tract VTs representing the largest subtype of
idiopathic VTs (Joshi & Wilber, 2005; Miller et al., 2006), and right
The right ventricular outow tract is an intriguing region of the ventricular outow tract-VT in turn making up 8090% of this particular
heart, with an embryological development and anatomy that is unique subgroup (Miles, 2001; Arya et al., 2007). Unlike most forms of VT, right
(see Section 2), and subsequently an electrophysiological phenotype ventricular outow tract-VT can be terminated with adenosine (Lerman
that contributes disproportionately to arrhythmias in the adult heart. et al., 2002). This has led to the view that the mechanism underlying
right ventricular outow tract-VT is a triggered activity due to
5.1. Anatomy cAMP-mediated intracellular Ca2+ overload and DADs (Lerman et al.,
2002). Long term therapy includes Ca2+ channel blockers which act
The right ventricular outow tract connects the working myocardi- directly on the L type Ca2+ channel to reduce ICa,L and -blockers
um of the right ventricle with the pulmonary trunk via the pulmonary which act indirectly by antagonising the increase in ICa,L due to
valve. It is grossly distinct from the left ventricular outow tract in a -adrenergic stimulation (Lerman et al., 2002).
number of ways. On the right side, the leaets of the pulmonary valve There remains debate surrounding the group of patients who expe-
are completely supported by an encircling muscular infundibulum, rience malignant right ventricular outow tract-VT (i.e. that which
whilst on the left side, two of the leaets of the aortic valve are in brous deteriorates into ventricular brillation and ultimately death), and
continuity with the aortic leaet of the mitral valve (Anderson & risk factors for sudden cardiac death in these patients have been identi-
Weinberg, 2005). This means that the pulmonary and tricuspid valves ed (Viskin et al., 2005) in patients at risk, catheter ablation is carried
280 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

out. Mapping studies performed during catheter ablation procedures the working myocardium, they lack expression of HCN4, like nodal
for right ventricular outow tract-VT have identied that the site of tissues, they are embedded in connective tissue and lack expression of
origin in most cases of this condition lies in the region immediately Nav1.5, Kir2.1 and Cx43 (Fig. 13; Monfredi et al., unpublished data).
proximal to the leaets of the pulmonary valve, especially on the Whilst the lack of expression of Nav1.5 and Cx43 will result in slow
anteromedial aspect of the right ventricular outow tract (Kamakura conduction, the lack of expression of Kir2.1 will promote ectopic
et al., 1988). pacemaker activity. In the rat, we have recorded nodal-like pacemaker
It remains unclear why the right ventricular outow tract should be activity from this region (S. Logantha and H. Schneider, unpublished
so arrhythmogenic, but the answer is likely to lie with its complex observations). Could it be that these myocytes hold the key to unlocking
development and late transformation from primary to chamber the arrhythmogenicity of the right ventricular outow tract?
myocardium, leading to substantial electrophysiological heterogeneity
between itself and the adjacent working right ventricular myocardium 6. Channelopathies in humans responsible
(Boukens et al., 2009). It has also been hypothesised that the presence for dysfunction of the cardiac conduction system
of nodal remnants in the right ventricular outow tract could be
responsible for the genesis of arrhythmias (Yanni et al., 2009a). We In humans, naturally occurring mutations (inherited or acquired) in
have demonstrated a previously undescribed population of cells that ion channels or associated proteins are the cause of a variety of cardiac
exist within the right ventricular outow tract of guinea-pigs and rats arrhythmias, including arrhythmias arising from dysfunction of the
that have a histological and immunohistochemical phenotype interme- cardiac conduction system. Channel (or channel-related) genes, muta-
diate between nodal myocytes and myocytes from the working right tions in which are associated with dysfunction of the cardiac conduc-
ventricular myocardium (Fig. 13; Yanni et al., 2009a). Although, like tion system, are listed in the rst part of Table 3. Mutations in other

Fig. 13. Nodal myocytes in unusual places. A, cartoon of heart showing region of interest (dashed box). B, sagittal section (from boxed region in A) through the roots of the aorta and
pulmonary artery of the guinea-pig immunolabelled for Cx43 (green signal) and NCX1 (Na+Ca2+ exchanger; red signal). Nodal-like tissues are Cx43-negative and NCX1-positive
and appear red (B). C, enlargement of the boxed region in B to show the region of the right ventricular outow tract. Modied from Yanni et al. (2009a). D, high magnication image
of Masson's trichrome stained section (corresponding to the boxed region in C) from the rat heart to show the morphology of the right ventricular outow tract infundibulum, pul-
monary valve and pulmonary artery. There is a sleeve of cardiac myocytes reaching up into the pulmonary artery corresponding to the area of Cx43-negative and NCX1-positive
cells in C. Smooth muscle cells are seen in the pulmonary artery vessel wall. From H. Schneider, O. Monfredi and H. Dobrzynski (unpublished data). Ao-lumen, lumen of the
aorta; PA-lumen, lumen of the pulmonary artery; RA, right atrium, RV-infundibulum: right ventricular infundibulum.
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 281

Table 3
Channelopathies responsible for cardiac conduction disease in the human.

Gene Protein Electrogenic process affected Disorder Associated disorders Reference

Gene mutations
KCNJ2 Kir2.1 IK,1 Sinus bradycardia Andersen syndrome Andelnger et al. (2002)
First-degree heart block Ventricular arrhythmias
Left bundle branch block
HCN4 HCN4 If Asymptomatic to severe sinus Intermittent atrial Schulze-Bahr et al. (2003)
bradycardia brillation Ueda et al. (2004)
Syncope QT prolongation Milanesi et al. (2006)
Chronotropic incompetence Polymorphic VT Nof et al. (2007)
SCN5A Nav1.5 INa Sick sinus syndrome or dysfunction Cardiomyopathy Kyndt et al. (2001)
Sinus bradycardia Brugada syndrome Grant et al. (2002)
Sinus bradyarrhythmia Long QT syndrome Benson et al. (2003)
Sinus arrest Ventricular arrhythmias Bezzina et al. (2003)
Sinus pauses Veldkamp et al. (2003)
Chronotropic incompetence Makiyama et al. (2005)
Heart block Probst et al. (2006)
Progressive cardiac conduction system Hu et al. (2010)
disease (Lengre-Lev disease) Neu et al. (2010)
Prolonged QRS interval
Prolonged His-ventricle conduction time
Right and left bundle branch block (various forms)
SCN1B Nav1 INa Cardiac conduction disease Brugada syndrome Watanabe et al. (2008)
Heart block
Left and right bundle branch block
KCNH2 ERG IK,r Sinus bradycardia Torsade de points Hoorntje et al. (1999)
Heart block QT prolongation Piipo et al. (2000)
Long QT syndrome Johnson et al. (2003)
Neonatal long QT Lupoglazoff et al. (2004)
syndrome Chevalier et al. (2007)
Oka et al. (2010)
KCNQ1 KVLQT1 IK,s Sinus bradycardia Torsade de points Lupoglazoff et al. (2004)
Heart block QT prolongation Oka et al. (2010)
Neonatal long QT
syndrome
TRPM4 TRPM4 Ca2+-activated non-specic Progressive familial heart block Kruse et al. (2009)
cation current type I Liu et al. (2010)
Heart block Stallmeyer et al. (2012)
Right bundle branch block
RYR2 Type 2 Ca2+ clock Sinus node dysfunction CPVT Bhuiyan et al. (2007)
ryanodine receptor Sinus arrest Atrial brillation Postma et al. (2005)
Sinus node exit block
Chronotropic incompetence
Heart block

Gene mutations
CASQ2 Type 2 calsequestrin Ca2+ clock Sinus bradycardia CPVT Postma et al. (2002)
ANK2 Ankyrin-B ICa,L Sinus node dysfunction Atrial brillation Le Scouarnec et al. (2008)
INaCa Sinus bradycardia Abnormal ventricular
repolarization
Supraventricular and
ventricular
arrhythmias

Other
CACNA1C Cav1.2 ICa,L/ICa,T Sinus bradycardia Boutjdir et al. (1998)
Heart block Qu et al. (2001)
Hu et al. (2004)

genes (non-channel related) giving rise to cardiac conduction system IK,1 (Section 3.3.2; Table 1). Various mutations in the HCN4 gene,
dysfunction are listed elsewhere (Park & Fishman, 2011). Unsurpris- expected to lead to a decrease in the pacemaker current If, have been
ingly, the channel (or channel-related) genes, mutations in which are shown to be responsible for sinus bradycardia (Milanesi et al., 2006;
responsible for cardiac conduction system dysfunction, are the major Nof et al., 2007; Schulze-Bahr et al., 2003; Ueda et al., 2004). Many mu-
ones involved in the electrical activity of the cardiac conduction sys- tations in the SCN5A gene encoding Nav1.5 have been discovered: these
tem. Mutation in KCNJ2 responsible for Kir2.1 and expected to lead to mutations lead to cardiac conduction system dysfunction: sinus brady-
a decrease in IK,1 is responsible for Andersen syndrome, but has been as- cardia, sinus node dysfunction, heart block and bundle branch block as
sociated with sinus bradycardia, heart block and left bundle branch well as other arrhythmias (e.g. Brugada syndrome and long QT
block in some individuals (Andelnger et al., 2002). The effects on the syndrome; Table 3). The mutations in SCN5A causing cardiac conduc-
two nodes is surprising, because Kir2.1 and IK,1 are responsible for the tion system dysfunction are generally loss-of-function and result in a
resting potential in the working myocardium and are largely absent decrease in peak INa. The loss-of-function in INa is presumably re-
from the nodes (Tables 1 and 2). However, the effects could be second- sponsible for the Brugada syndrome, which can be associated with
ary or the result of the expected depolarization and loss of excitability in cardiac conduction system dysfunction. However, the mutations
the working myocardium. The effect on the bundle branches is not sur- causing cardiac conduction system dysfunction linked with long QT
prising, because Purkinje bres are known to express Kir2.1 and possess syndrome are also gain-of-function in that they increase the late current
282 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

through Nav1.5 (Grant et al., 2002; Veldkamp et al., 2003). A loss of determining its embryological development to the ion channels
function mutation in a Na + channel auxiliary subunit, Nav1, also and related proteins underlying its electrical activity. Clinically, a
causes heart block and bundle branch block (Watanabe et al., 2008). picture is emerging of dysfunction of the cardiac conduction system
In new born infants, neonatal long QT syndrome can be associated in various disease states, such as heart failure and atrial brillation,
with sinus bradycardia and/or second degree heart block and these and we are uncovering the remodelling of the cardiac conduction
cases have been attributed to mutations in KCNH2 (ERG) and KCNQ1 system in the disease states that is responsible for the dysfunction.
(KvLQT1), responsible for IK,r and IK,s, respectively (Hoorntje et al., This raises the possibility of designing new treatments for sinus
1999; Piipo et al., 2000; Johnson et al., 2003; Lupoglazoff et al., 2004). bradycardia, heart block and bundle branch block etc. in the future.
In the case of IK,r at least, one of the ERG mutations is expected to
cause a decrease in IK,r, one has been shown to cause a decrease in IK,r Conict of Interest
and another has been shown to alter the kinetics of IK,r (Hoorntje et
al., 1999; Piipo et al., 2000; Johnson et al., 2003). Mutations in KCNH2 The authors declare that there are no conicts of interests.
(ERG) and KCNQ1 (KvLQT1) demonstrated to result in decreases in IK,r
and IK,s have also been shown to be responsible for heart block associat- References
ed with long QT syndrome in adults (Chevalier et al., 2007; Oka et al.,
2010). In the two nodes (in the absence of IK,1) IK,r and IK,s are responsi- Aanhaanen, W. T., Brons, J. F., Domnguez, J. N., Rana, M. S., Norden, J., Airik, R., et al. (2009).
The Tbx2+ primary myocardium of the atrioventricular canal forms the atrioventricular
ble for the maximum diastolic potential (Fig. 6) and a decrease in either node and the base of the left ventricle. Circ Res 104, 12671274.
current is expected to result in depolarization and inexcitability Abernethy, D. R., & Schwartz, J. B. (1999). Calcium-antagonist drugs. N Engl J Med 341,
(Kodama et al., 1999). Mutations in TRPM4, responsible for a 14471457.
Abi-Gerges, N., Small, B. G., Lawrence, C. L., Hammond, T. G., Valentin, J. P., & Pollard, C. E.
Ca 2+-activated non-specic cation current, have been linked to (2004). Evidence for gender differences in electrophysiological properties of canine
heart block and right bundle branch block (Kruse et al., 2009; Liu Purkinje bres. Br J Pharmacol 142, 12551264.
et al., 2010; Stallmeyer et al., 2012). A number of the mutations Abi-Gerges, N., Small, B. G., Lawrence, C. L., Hammond, T. G., Valentin, J. P., & Pollard, C. E.
(2006). Gender differences in the slow delayed (IKs) but not in inward (IK1) rectier
have been shown to be gain-of-function and increase the current K+ currents of canine Purkinje bre cardiac action potential: key roles for IKs,
(Kruse et al., 2009; Liu et al., 2010). How TRPM4 acts is not known. beta-adrenoceptor stimulation, pacing rate and gender. Br J Pharmacol 147, 653660.
It is no surprise that mutations in components of the Ca 2+ clock also Adan, V., & Crown, L. A. (2003). Diagnosis and treatment of sick sinus syndrome. Am
Fam Physician 67, 17251732.
have functional consequences for the cardiac conduction system. Muta-
Ailings, A. M., Abbas, R. F., & Bouman, L. N. (1995). Age-related changes in structure and
tions in RYR2 (SR Ca2+ release channel) and CASQ2 (calsequestrin SR relative collagen content of the human and feline sinoatrial node. A comparative
Ca2+ storage protein) cause catecholaminergic polymorphic ventricular study. Eur Heart J 16, 16551667.
Amerini, S., Giotti, A., & Mugelli, A. (1985). Effect of verapamil and diltiazem on
tachycardia (CPVT) and this can be associated with sinus bradycardia
calcium-dependent electrical activity in cardiac Purkinje bres. Br J Pharmacol 85,
and heart block (Table 3; Postma et al., 2002, 2005; Bhuiyan et al., 8996.
2007); RYR2 and CASQ2 mutations are likely to affect the Ca 2+ clock Andelnger, G., Tapper, A. R., Welch, R. C., Vanoye, C. G., George, A. L., Jr., & Benson, D. W.
and, therefore, it is understandable that the mutations should cause (2002). KCNJ2 mutation results in Andersen syndrome with sex-specic cardiac and
skeletal muscle phenotypes. Am J Hum Genet 71, 663668.
sinus bradycardia. However, why it should cause heart block is un- Anderson, R. H., Becker, A. E., & Van Mierop, L. H. (1977). What should we call the crista? Br
known. Interestingly in a mouse model of CPVT, there is evidence that Heart J 39, 856859.
intracellular Ca2+ handling in Purkinje myocytes is more profoundly Anderson, R. H., Davies, M. J., & Becker, A. E. (1974a). Atrioventricular ring specialized
tissue in the normal heart. Eur J Cardiol 2, 219230.
affected than in ventricular myocytes and ventricular arrhythmias Anderson, R. H., Janse, M. J., van Capelle, F. J., Billette, J., Becker, A. E., & Durrer, D. (1974b).
arise in the Purkinje myocytes (Kang et al., 2010). Mutation in ANK2 A combined morphological and electrophysiological study of the atrioventricular
has been shown to cause severe bradycardia (Table 3; Le Scouarnec et node of the rabbit heart. Circ Res 35, 909922.
Anderson, R. H., & Ho, S. Y. (2003). The morphology of the cardiac conduction system.
al., 2008). ANK2 is responsible for ankyrin-B. Ankyrins are adaptor pro- Novartis Found Symp 250, 617.
teins responsible for targeting channels and transporters in various cell Anderson, R. H., Ho, S. Y., & Becker, A. E. (2000). Anatomy of the human atrioventricular
types. Ankyrin-B is enriched in the sinus node and heterozygous knock- junctions revisited. Anat Rec 260, 8191.
Anderson, R. H., Ho, S. Y., Gillette, P. C., & Becker, A. E. (1996). Mahaim, Kent and abnormal
out of ankyrin-B in the mouse: disrupts cell membrane targetting of
atrioventricular conduction. Cardiovasc Res 31, 480491.
Cav1.3 (in part responsible for ICa,L in the sinus node), NCX1 (responsi- Anderson, R. H., & Weinberg, P. M. (2005). The clinical anatomy of tetralogy of fallot.
ble for the Na+Ca2+ exchanger), the Na+K + pump and the IP3 recep- Cardiol Young 1, 3847 (Supplement).
Anderson, R. H., Yanni, J., Boyett, M. R., Chandler, N. J., & Dobrzynski, H. (2009). The
tor; reduces ICa,L and INaCa; disrupts intracellular Ca2+ homeostasis; and
anatomy of the cardiac conduction system. Clin Anat 22, 99113.
causes bradycardia (Table 3; Le Scouarnec et al., 2008; Hund & Mohler, Ansari, A., Ho, S. Y., & Anderson, R. H. (1999). Distribution of the Purkinje bres in the
2008). sheep heart. Anat Rec (Hoboken) 254, 9297.
Boutjdir and colleagues (Boutjdir et al., 1998; Qu et al., 2001; Hu et Antzelevitch, C. (2006). Brugada syndrome. Pacing Clin Electrophysiol 9, 11301159.
Anumonwo, J. M., Delmar, M., & Jalife, J. (1990). Electrophysiology of single heart cells
al., 2004) have obtained evidence that the mothers of children born from the rabbit tricuspid valve. J Physiol 425, 145167.
with congenital heart block (presumably, one type of congenital Anumonwo, J. M., Tallini, Y. N., Vetter, F. J., & Jalife, J. (2001). Action potential characteristics
heart block, at least, because there are other causes of congenital and arrhythmogenic properties of the cardiac conduction system of the murine heart.
Circ Res 89, 329335.
heart block as shown in Table 3) produce IgG autoantibodies which Arnar, D. O., Bullinga, J. R., & Martins, J. B. (1997). Role of the Purkinje system in spontaneous
enter the foetal circulation and reduce ICa,T and ICa,L. This can be ventricular tachycardia during acute ischemia in a canine model. Circulation 96,
described as a channelopathy, although it is not the normal kind and 24212429.
Arya, A., Piorkowski, C., Sommer, P., Gerds-Li, J. -H., Kottkamp, H., & Hindricks, G. (2007).
it is listed separately in Table 3. A reduction in ICa,T at least is expected Idiopathic outow tract tachycardias: Current perspectives. Herz 32, 218225.
to result in heart block: knock-out of Cav3.1 in the mouse causes a Aslanidi, O. V., Colman, M. A., Stott, J., Dobrzynski, H., Boyett, M. R., Holden, A. V., et al.
slowing of AV conduction (Mangoni et al., 2006). Knock-out of both (2011). 3D virtual human atria: A computational platform for studying clinical
atrial brillation. Prog Biophys Mol Biol 107, 156168.
Cav1.3 and Cav3.1 in the mouse also causes sinus bradycardia
Aslanidi, O. V., Sleiman, R. N., Boyett, M. R., Hancox, J. C., & Zhang, H. (2010). Ionic mecha-
(Mangoni et al., 2003, 2006). nisms for electrical heterogeneity between rabbit Purkinje ber and ventricular cells.
Biophys J 98, 24202431.
Atkinson, A., Inada, S., Li, J., Tellez, J. O., Yanni, J., Sleiman, R., et al. (2011). Anatomical
7. Conclusions
and molecular mapping of the left and right ventricular HisPurkinje conduction
networks. J Mol Cell Cardiol 51, 689701.
Over the last few decades, as well as developing a greater under- Balasundaram, R., Rao, H. B., Kalavakolanu, S., & Narasimhan, C. (2008). Catheter ablation
standing of the embryonic development and anatomy of the cardiac of bundle branch reentrant ventricular tachycardia. Hear Rhythm 5, S68S72.
Balti, B., Lost, N., Simon, J., Varr, A., & Papp, J. G. (2000). Analysis of the electrophysio-
conduction system, we have learnt much about the cardiac conduc- logical effects of ambasilide, a new antiarrhythmic agent, in canine isolated ventric-
tion system at the molecular level, from the transcription factors ular muscle and purkinje bers. Gen Pharmacol 34, 8593.
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 283

Baldesberger, S., Bauersfeld, U., Candinas, R., Seifert, B., Zuber, M., Ritter, M., et al. Boyett, M. R., Hart, G., Levi, A. J., & Roberts, A. (1987). Effects of repetitive activity on
(2008). Sinus node disease and arrhythmias in the long-term follow-up of former developed force and intracellular sodium in isolated sheep and dog Purkinje bres.
professional cyclists. Eur Heart J 29, 7178. J Physiol 388, 295322.
Barbosky, L., Lawrence, D. K., Karunamuni, G., Wikenheiser, J. C., Doughman, Y. Q., Boyett, M. R., & Fedida, D. (1984). Changes in the electrical activity of dog cardiac
Visconti, R. P., et al. (2006). Apoptosis in the developing mouse heart. Dev Dyn Purkinje bres at high heart rates. J Physiol 350, 361391.
235, 25922602. Bromberg, B. I., Hand, D. E., Schuessler, R. B., & Boineau, J. P. (1995). Primary negativity does
Baruscotti, M., Bucchi, A., Viscomi, C., Mandelli, G., Consalez, G., Gnecchi-Rusconi, T., et al. not predict dominant pacemaker location: implications for sinoatrial conduction. Am J
(2011). Deep bradycardia and heart block caused by inducible cardiac-specic knock- Physiol 269, H877H887.
out of the pacemaker channel gene Hcn4. Proc Natl Acad Sci U S A 108, 17051710. Brooks, R., & Burgess, J. H. (1988). Idiopathic ventricular tachycardia. Medicine 67, 71294.
Basset, A. L., Fenoglio, J. J., Jr., Wit, A. L., Myerburg, R. J., & Gelband, H. (1976). Brugada, J., Brugada, R., Antzelevitch, C., Towbin, J., Nademanee, K., & Brugada, P.
Eelctrophysiologic and ultrastructural characteristics of the canine tricuspid valve. (2002). Long-term follow-up of individuals with the electrocardiographic pattern
Am J Physiol 230, 13661373. of right bundle-branch block and st-segment elevation in precordial leads v1 to
Bauer, A., McDonald, A. D., Nasir, K., Peller, L., Rade, J. J., Miller, J. M., et al. (2004). Inhibitory v3. Circulation 105, 7378.
G protein overexpression provides physiologically relevant heart rate control in persis- Bucchi, A., Tognati, A., Milanesi, R., Baruscotti, M., & DiFrancesco, D. (2006). Properties
tent atrial brillation. Circulation 110, 31153120. of ivabradine-induced block of HCN1 and HCN4 pacemaker channels. J Physiol 572,
Ben, Caref E., Boutjdir, M., Himel, H. D., & El-Sherif, N. (2008). Role of subendocardial 335346.
Purkinje network in triggering torsade de pointes arrhythmia in experimental Bunch, T. J., & Day, J. D. (2006). Right meets left: A common mechanism underlying
long QT syndrome. Europace 10, 12181223. right and left ventricular outow tract tachycardias. J Cardiovasc Electrophysiol
Benditt, D. G., Sakaguchi, S., Goldstein, M. A., Lurie, K. G., Gornick, C. C., & Adler, S. W. 17, 10591061.
(1995). Sinus node dysfunction: pathophysiology, clinical features, evaluation, Cai, C. L., Liang, X., Shi, Y., Chu, P. H., Pfaff, S. L., Chen, J., et al. (2003). Isl1 identies a cardiac
and treatment. In D. P. Zipes, & J. Jalife (Eds.), Cardiac electrophysiology: from cell progenitor population that proliferates prior to differentiation and contributes a major-
to bedside (pp. 12151247). Philadelphia, Pa: WB Saunders Company. ity of cells to the heart. Dev Cell 5, 877889.
Benson, D. W., Wang, D. W., Dyment, M., Knilans, T. K., Fish, F. A., Strieper, M. J., et al. Callewaert, G., Carmeliet, E., & Vereecke, J. (1984). Single cardiac Purkinje cells: general
(2003). Congenital sick sinus syndrome caused by recessive mutations in the cardiac electrophysiology and voltage-clamp analysis of the pace-maker current. J Physiol
sodium channel gene (SCN5A). J Clin Invest 112, 10191028. 349, 643661.
Berenfeld, O., & Jalife, J. (1988). Purkinje-muscle reentry as a mechanism of polymorphic Champeroux, P., Ouill, A., Martel, E., Fowler, J. S., Maurin, A., Richard, S., et al. (2011). A
ventricular arrhythmias in a 3-dimensional model of the ventricles. Circ Res 82, step towards characterisation of electrophysiological prole of torsadogenic drugs.
10631077. J Pharmacol Toxicol Methods 63, 269278.
Bezzina, C. R., Rook, M. B., Groenewegen, W. A., Herfst, L. J., van der Wal, A. C., Lam, J., Chandler, N., Aslanidi, O., Buckley, D., Inada, S., Birchall, S., Atkinson, A., et al. (2011). A
et al. (2003). Compound heterozygosity for mutations (W156X and R225W) in new twist in the origin of the heartbeat. Anat Rec (Hoboken) 294, 970979.
SCN5A associated with severe cardiac conduction disturbances and degenerative Chandler, N. J., Greener, I. D., Tellez, J. O., Inada, S., Musa, H., Molenaar, P., et al. (2009).
changes in the conduction system. Circ Res 92, 159168. Molecular architecture of the human sinus node: insights into the function of the
Bhuiyan, Z. A., van den Berg, M. P., van Tintelen, J. P., Bink-Boelkens, M. T., Wiesfeld, A. cardiac pacemaker. Circulation 119, 15621575.
C., Alders, M., et al. (2007). Expanding spectrum of human RYR2-related disease: Capulzini, L., Brugada, P., Brugada, J., & Brugada, R. (2010). Arrhythmia and right heart dis-
new electrocardiographic, structural, and genetic features. Circulation 116, ease: From genetic basis to clinical practice. Rev Esp Cardiol 63, 963983.
15691576. Ceconi, C., Comini, L., Suffredini, S., Stillitano, F., Bouly, M., Cerbai, E., et al. (2011). Heart rate
Billette, J. (1987). Atrioventricular nodal activation during periodic premature stimula- reduction with ivabradine prevents the global phenotype of left ventricular remodeling.
tion of the atrium. Am J Physiol 252, H163H177. Am J Physiol 300, H366H373.
Blomstrom-Lundqvist, C., Scheinman, M. M., Aliot, E. M., Alpert, J. S., Calkins, H., Camm, Cheng, H., Smith, G. L., Hancox, J. C., & Orchard, C. H. (2011). Inhibition of spontaneous
A. J., et al. (2003). ACC/AHA/ESC guidelines for the management of patients with activity of rabbit atrioventricular node cells by KB-R7943 and inhibitors of sarcoplasmic
supraventricular arrhythmias-executive summary: a report of the American Col- reticulum Ca(2+) ATPase. Cell Calcium 49, 5665.
lege of Cardiology/American Heart Association Task Force on Practice Guidelines Chen, Y. -J., Chen, Y. C., Yeh, H. I., Lin, C. I., & Chen, S. A. (2002). Electrophysiology and
and the European Society of Cardiology Committee for Practice Guidelines (Writ- arrhythmogenic activity of single cardiomyocytes from canine superior vena cava.
ing Committee to Develop Guidelines for the Management of Patients With Supra- Circulation 105, 26792685.
ventricular Arrhythmias). Circulation 108, 18711909. Cerbai, E., Barbieri, M., & Mugelli, A. (1994). Characterization of the hyperpolarization-
Bogdanov, K. Y., Vinogradova, T. M., & Lakatta, E. G. (2001). Sinoatrial nodal cell ryanodine activated current, If, in ventricular myocytes isolated from hypertensive rats. J Physiol
receptor and Na+-Ca2+ exchanger: molecular partners in pacemaker regulation. Circ 481, 585591.
Res 88, 12541258. Cerbai, E., Barbieri, M., & Mugelli, A. (1996). Occurrence and properties of the
Bohm, M., Swedberg, K., Komajda, M., Borer, J. S., Ford, I., Dubost-Brama, A., et al. (2010). hyperpolarization-activated current If in ventricular myocytes from normotensive
Heart rate as a risk factor in chronic heart failure (SHIFT): the association between and hypertensive rats during aging. Circulation 94, 16741681.
heart rate and outcomes in a randomised placebo-controlled trial. Lancet 376, Cerbai, E., & Mugelli, A. (2006). If in non-pacemaker cells: Role and pharmacological
886894. implications. Pharmacol Res 53, 416423.
Boineau, J. P., Canavan, T. E., Schuessler, R. B., Cain, M. E., Corr, P. B., & Cox, J. L. (1988). Cerbai, E., Sartiani, L., DePaoli, P., Pino, R., Maccherini, M., Bizzarri, F., et al. (2001). The
Demonstration of a widely distributed atrial pacemaker complex in the human properties of the pacemaker current If in human ventricular myocytes are modulated
heart. Circulation 77, 12211237. by cardiac disease. J Mol Cell Cardiol 33, 441448.
Boink, G. J., Bakker, M. L., Verkerk, A. O., Bakker, D., de Bakker, J. M., Seppen, J., et al. (2008). Chevalier, P., Bellocq, C., Millat, G., Piqueras, E., Potet, F., Schott, J. J., et al. (2007). Torsades
Inducible TBX3 overexpression as a tool for biopacemaker engineering. Circulation 118, de pointes complicating atrioventricular block: Evidence for a genetic predisposition.
S340. Hear Rhythm 4, 170174.
Boukens, B. J., Christoffels, V. M., Coronel, R., & Moorman, A. F. (2009). Developmental basis Kapoor, N., Marbn, E., & Cho, H. C. (2011a). Tbx18-reprogrammed cardiomyocytes exhibit
for electrophysiological heterogeneity in the ventricular and outow tract myocardium pacemaker phenotype with upregulated calcium and voltage-sependent clock mecha-
as a substrate for life-threatening ventricular arrhythmias. Circ Res 104, 1931. nisms. Circulation 124, A14024.
Boutjdir, M., Chen, L., Zhang, Z. H., Tseng, C. E., El-Sherif, N., & Buyon, J. P. (1998). Serum Kapoor, N., Marbn, E., & Cho, H. C. (2011b). Biological pacemaker induced in vivo by focal
and antibody IgG from the mother of a child with congenital heart block induce Tbx18 gene transfer in the guinea-pig left ventricle. Circulation 124, A15845 (2011).
conduction abnormalites and inhibit L-type Ca channels in a rat heart model. Chen, P. S., Joung, B., Shinohara, T., Das, M., Chen, Z., & Lin, S. F. (2010). The initiation of
Pediatr Res 44, 1119. the heart beat. Circulation 74, 221225.
Boyden, P. A., Dun, W., Barbhaiya, C., & Ter Keurs, H. E. (2004). 2APB- and Coppen, S. R., Kodama, I., Boyett, M. R., Dobrzynski, H., Takagishi, Y., Honjo, H., et al. (1999a).
JTV519(K201)-sensitive micro Ca2+ waves in arrhythmogenic Purkinje cells that Connexin45, a major connexin of the rabbit sinoatrial node, is co-expressed with
survive in infarcted canine heart. Hear Rhythm 1, 218226. connexin43 in a restricted zone at the nodal-crista terminalis border. J Histochem
Boyden, P. A., Pu, J., Pinto, J., & ter Keurs, H. E. (2000). Ca2+ transients and Ca2+ waves Cytochem 47, 907918.
in purkinje cells: role in action potential initiation. Circ Res 86, 448455. Coppen, S. R., Severs, N. J., & Gourdie, R. G. (1999b). Connexin45 (alpha 6) expression
Boyden, P. A., Barbhaiya, C., Lee, T., & ter Keurs, H. E. (2003). Nonuniform Ca2+ transients delineates an extended conduction system in the embryonic and mature rodent
in arrhythmogenic Purkinje cells that survive in the infarcted canine heart. heart. J Gene Dev 24, 8290.
Cardiovasc Res 57, 681693. Coppen, S. R., & Severs, N. J. (2002). Diversity of connexin expression patterns in the
Boyden, P. A., Albala, A., & Dresdner, K. P., Jr. (1989). Electrophysiology and ultrastructure atrioventricular node: vestigial consequence or functional specialization? J
of canine subendocardial Purkinje cells isolated from control and 24-hour infarcted Cardiovasc Electrophysiol 13, 625626.
hearts. Circ Res 65, 955970. Cordeiro, J. M., Bridge, J. H., & Spitzer, K. W. (2001a). Early and delayed afterdepolarizations
Boyett, M. R., Honjo, H., & Kodama, I. (2000). The sinoatrial node, a heterogeneous in rabbit heart Purkinje cells viewed by confocal microscopy. Cell Calcium 29, 289297.
pacemaker structure. Cardiovasc Res 47, 658687. Cordeiro, J. M., Spitzer, K. W., & Giles, W. R. (1998). Repolarizing K+ currents in rabbit
Boyett, M. R., Honjo, H., Yamamoto, M., Nikmaram, M. R., Niwa, R., & Kodama, I. (1999). heart Purkinje cells. J Physiol 508, 811823.
Downward gradient in action potential duration along the conduction path in and Cordeiro, J. M., Spitzer, K. W., Giles, W. R., Ershler, P. E., Cannell, M. B., & Bridge, J. H.
around the sinoatrial node. Am J Physiol 276, H686H698. (2001b). Location of the initiation site of calcium transients and sparks in rabbit
Boyett, M. R., Inada, S., Yoo, S., Li, J., Liu, J., Tellez, J., et al. (2006). Connexins in the sinoatrial heart Purkinje cells. J Physiol 531, 301314.
and atrioventricular nodes. Adv Cardiol 42, 175197. Cosio, F. G., Anderson, R. H., Becker, A., Borggrefe, M., Campbell, R. W., Gaita, F., et al.
Boyett, M. R., Tellez, J. O., & Dobrzynski, H. (2009). The sinoatrial node its complex (1999). Living anatomy of the atrioventricular junctions. A guide to electrophysiolog-
structure and unique ion channel gene programme. In D. P. Zipes, & J. Jalife ical mapping. A Consensus Statement from the Cardiac Nomenclature Study Group,
(Eds.), Cardiac electrophysiology: from cell to bedside (5th ed.). (pp.x-x). Working Group of Arrythmias, European Society of Cardiology, and the Task Force
284 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

on Cardiac Nomenclature from NASPE. North American Society of Pacing and Electro- Gaborit, N., Le Bouter, S., Szuts, V., Varro, A., Escande, D., Nattel, S., et al. (2007). Regional
physiology. Eur Heart J 20, 10681075. and tissue specic transcript signatures of ion channel genes in the non-diseased
Courtemanche, M., Ramirez, R. J., & Nattel, S. (1998). Ionic mechanisms underlying human heart. J Physiol 582, 675693.
human atrial action potential properties: insights from a mathematical model. Gadsby, D. C., & Craneeld, P. F. (1977). Two levels of resting potential in cardiac Purkinje
Am J Physiol 275, H301H321. bers. J Gen Physiol 70, 725746.
Crijns, H. J., Smeets, J. L., Rodriguez, L. M., Meijer, A., & Wellens, H. J. (1995). Cure of Gervais, R., Leclercq, C., Shankar, A., Jacobs, S., Eiskjaer, H., Johannessen, A., et al. (2009). Sur-
interfascicular reentrant ventricular tachycardia by ablation of the anterior fascicle face electrocardiogram to predict outcome in candidates for cardiac resynchronization
of the left bundle branch. J Cardiovasc Electrophysiol 6, 486492. therapy: a sub-analysis of the CARE-HF trial. Eur J Heart Fail 11, 699705.
Christoffels, V. M., Burch, J. B., & Moorman, A. F. (2004). Architectural plan for the heart: Gilmour, R. F., Jr., & Watanabe, M. (1994). Dynamics of circus movement re-entry
Early patterning and delineation of the chambers and the nodes. Trends Cardiovasc across canine Purkinje bre-muscle junctions. J Physiol 476, 473485.
Med 14, 301307. Gourdie, R. G., Severs, N. J., Green, C. R., Rothery, S., Germroth, P., & Thompson, R. P.
Christoffels, V. M., Smits, G. J., Kispert, A., & Moorman, A. F. M. (2010). Development of (1993). The spatial distribution and relative abundance of gap-junctional
the pacemaker tissues of the heart. Circ Res 106, 240254. connexin40 and connexin43 correlate to functional properties of components of
Crisel, R. K., Farazneh-Far, R., Na, B., & Whooley, M. A. (2011). First-degree atrioventricular the cardiac atrioventricular conduction system. J Cell Sci 105, 985991.
block is associated with heart failure and death in persons with stable coronary artery Grant, A. O., Carboni, M. P., Neplioueva, V., Starmer, C. F., Memmi, M., Napolitano, C., et al.
disease: data from the Heart and Soul Study. Eur Heart J 32, 18751880. (2002). Long QT syndrome, Brugada syndrome, and conduction system disease
Dangman, K. H., & Miura, D. S. (1987). Does if control normal automatic rate in canine linked to a single sodium channel mutation. J Clin Invest 110, 12011209.
cardiac Purkinje bers? Studies on the negative chronotropic effects of lidoazine. Greener, I. D., Monfredi, O., Inada, S., Chandler, N. J., Tellez, J. O., Atkinson, A., et al. (2011).
J Cardiovasc Pharmacol 10, 332340. Molecular architecture of the human specialised atrioventricular conduction axis.
Davis, D. L., Edwards, A. V., Juraszek, A. L., Phelps, A., Wessels, A., & Burch, J. B. (2001). A gata-6 J Mol Cell Cardiol 50, 642651.
gene heart-region-specic enhancer provides a novel means to mark and probe a dis- Greener, I. D., Tellez, J. O., Dobrzynski, H., Yamamoto, M., Graham, G. M., Billeter, R., et al.
crete component of the mouse cardiac conduction system. Mech Dev 108, 105119. (2009). Ion channel transcript expression at the rabbit atrioventricular conduction
Davis, L. D., & Temte, J. V. (1969). Electrophysiological actions of lidocaine on canine axis. Circ Arrhythm Electrophysiol 2, 305315.
ventricular muscle and Purkinje bers. Circ Res 24, 639655. Grines, C. L., Bashore, T. M., Boudoulas, H., Olson, S., Shafer, P., & Wooley, C. F. (1989). Func-
deCarvalho, A. P., De Mello, W. C., & Hoffman, B. F. (1959). Electrophysiological evi- tional abnormalities in isolated left bundle branch block. The effect of interventricular
dence for specialized ber types in rabbit atrium. Am J Physiol 196, 483488. asynchrony. Circulation 79, 845853.
Demion, M., Bois, P., Launay, P., & Guinamard, R. (2007). TRPM4, a Ca2+-activated Gupta, A., Lawrence, A. T., Krishnan, K., Kavinsky, C. J., & Trohman, R. G. (2007). Current
nonselective cation channel in mouse sino-atrial node cells. Cardiovasc Res 73, concepts in the mechanisms and management of drug-induced QT prolongation
531538. and torsade de pointes. Am Heart J 153, 891899.
DeMelo, S. R., DeSouza, R. R., & Mandarim-de-Lacerda, C. A. (2002). Stereologic study of Hassaguerre, M., Shah, D. C., Jas, P., Shoda, M., Kautzner, J., Arentz, T., et al. (2002).
the sinoatrial node of rats age related changes. Biogerontology 3, 383390. Role of Purkinje conducting system in triggering of idiopathic ventricular brilla-
Den Haan, D., Verkerk, A. O., & Tan, H. L. (2011). Creation of a biopacemaker: lessons tion. Lancet 359, 677688.
from the sinoatrial node. In M. K. Das (Ed.), Modern pacemakers-present and future Hancox, J. C., & Levi, A. J. (1994). L-type calcium current in rod- and spindle-shaped
(pp. 495524). : Intech. myocytes isolated from rabbit atrioventricular node. Am J Physiol 267, H1670H1680.
Dersham, G. H., & Han, J. (1980). Effects of verapamil on action potentials of Purkinje Hancox, J. C., Yuill, K. H., Mitcheson, J. S., & Convery, M. K. (2003). Progress and gaps in un-
bers. J Electrocardiol 13, 6772. derstanding the electrophysiological properties of morphologically normal cells from
Desplantez, T., Dupont, E., Severs, N. J., & Weingart, R. (2007). Gap junction channels the cardiac atrioventricular node. Int J Bifurcation Chaos 13, 36753691.
and cardiac impulse propagation. J Membr Biol 18, 1328. Hao, X. Z. Y., Zhang, X., Nirmalan, M., Davies, L., Dobrzynski, H., Wang, X., et al. (2011).
DiFrancesco, D. (1981a). A new interpretation of the pace-maker current in calf TGF-1 mediated brosis and ion channel remodeling are key mechanisms producing
Purkinje bres. J Physiol 314, 359376. sinus node dysfunction associated with SCN5A deciency and aging. Circ Arrhythm
DiFrancesco, D. (1981b). A study of the ionic nature of the pace-maker current in calf Electrophysiol 2011(4), 397406.
Purkinje bres. J Physiol 314, 377393. Hart, G., & Dukes, I. D. (1984). An analysis of the rate-dependent action of
DiFrancesco, D., & Noble, D. (2012). Rebuttal: The funny current in the context of the lidoazine in mammalian sino-atrial node and Purkinje bres. J Mol Cell Cardiol
coupled clock pacemaker cell system. Hear Rhythm 9, 457458. 16, 3342.
Dobrzynski, H., Boyett, M. R., & Anderson, R. H. (2007). New insights into pacemaker ac- Himeno, Y., Toyoda, F., Satoh, H., Amano, A., Cha, C. Y., Matsuura, H., et al. (2011). Minor
tivity: promoting understanding of sick sinus syndrome. Circulation 115, 19211932. contribution of cytosolic Ca2+ transients to the pacemaker rhythm in guinea pig
Dobrzynski, H., Li, J., Tellez, J., Greener, I. D., Nikolski, V. P., Wright, S. E., et al. (2005). sinoatrial node cells. Am J Physiol Heart Circ Physiol 300, H251H261.
Computer three-dimensional reconstruction of the sinoatrial node. Circulation Hirata, Y., Kodama, I., Iwamura, N., Shimizu, T., Toyama, J., & Yamada, K. (1979). Effects of
111, 846854. verapamil on canine Purkinje bres and ventricular muscle bres with particular
Dobrzynski, H., Nikolski, V. P., Sambelashvili, A. T., Greener, I. D., Yamamoto, M., Boyett, M. reference to the alternation of action potential duration after a sudden increase in
R., et al. (2003). Site of origin and molecular substrate of atrioventricular junctional driving rate. Cardiovasc Res 13, 18.
rhythm in the rabbit heart. Circ Res 93, 11021110. Hoff, P. I., Tronstad, A., Oie, B., & Ohm, O. J. (1988). Electrophysiologic and clinical effects
Dun, W., & Boyden, P. A. (2008). The Purkinje cell; 2008 style. J Mol Cell Cardiol 45, of ecainide for recurrent paroxysmal supraventricular tachycardia. Am J Cardiol 62,
617624. 585589.
Edelberg, J. M., Aird, W. C., & Rosenberg, R. D. (1998). Enhancement of murine cardiac Honjo, H., Boyett, M. R., Kodama, I., & Toyama, J. (1996). Correlation between electrical
chronotropy by the molecular transfer of the human beta2 adrenergic receptor activity and the size of rabbit sinoatrial node cells. J Physiol 496, 795808.
cDNA. J Clin Invest 101, 337343. Hoogaars, W. M., Engel, A., Brons, J. F., Verkerk, A. O., de Lange, F. J., Wong, L. Y., et al.
Elizari, M. V., Levi, R., Acunzo, R. S., Chiale, P. A., Civetta, M. M., Ferreiro, M., et al. (2007). Tbx3 Controls the Sinoatrial Node Gene Program and Imposes Pacemaker
(2007). Abnormal expression of cardiac neural crest cells in heart development: Function on the Atria. Genes Dev 21, 10981112.
A different hypothesis for the etiopathogenesis of brugada syndrome. Hear Hoogaars, W. M., Tessari, A., Moorman, A. F., de Boer, P. A., Hagoort, J., Soufan, A. T., et al.
Rhythm 4, 359365. (2004). The Transcriptional Repressor Tbx3 Delineates the Developing Central
Elvan, A., Wylie, K., & Zipes, D. P. (1996). Pacing-induced chronic atrial brillation impairs Conduction System of the Heart. Cardiovasc Res 62, 489499.
sinus node function in dogs. Electrophysiological remodeling. Circulation 94, 29532960. Hoorntje, T., Alders, M., van Tintelen, P., van der Lip, K., Sreeram, N., van der Wal, A.,
Eriksson, P., Hansson, P. O., Eriksson, H., & Dellborg, M. (1998). Bundle-branch block in et al. (1999). Homozygous premature truncation of the HERG protein: the
a general male population: the study of men born 1913. Circulation 98, 24942500. human HERG knockout. Circulation 100, 12641267.
Faggiano, P., d'Aloia, A., Gualeni, A., Gardini, A., & Giordano, A. (2001). Mechanisms and Horibe, H. (1961). Studies on the spread of the right atrial activation by means of intracel-
immediate outcome of in-hospital cardiac arrest in patients with advanced heart lular microelectrode. Jpn Circ J 25, 583593.
failure secondary to ischemic or idiopathic dilated cardiomyopathy. Am J Cardiol 87, Howarth, F. C., Jacobson, M., Shaullah, M., & Adeghate, E. (2005). Long-term effects of
655657. streptozotocin-induced diabetes on the electrocardiogram, physical activity and
Fedorov, V. V., Glukhov, A. V., Chang, R., Kostecki, G., Aferol, H. A., Hucker, W. J., et al. body temperature in rats. Exp Physiol 90, 827835.
(2010). Optical mapping of the isolated coronary-perfused human sinus node. J Howarth, F. C., Jacobson, M., Shaullah, M., & Adeghate, E. (2006). Effects of insulin treat-
Am Coll Cardiol 56, 13861394. ment on heart rhythm, body temperature and physical activity in streptozotocin-
Fedorov, V. V., Glukhov, A. V., & Chang, R. (2012). Conduction barriers and pathways of the induced diabetic rats. Clin Exp Pharmacol Physiol 33, 327331.
sino-atrial pacemaker complex: their role in normal rhythm and atrial arrhythmias. Am Hu, D., Barajas-Martinez, H., Nesterenko, V. V., Pfeiffer, R., Guerchicoff, A., Cordeiro, M.
J Physiol 302, H1773H1783. J., et al. (2010). Dual variation in SCN5A and CACNB2b underlies the development of
Fedorov, V. V., Schuessler, R. B., Hemphill, M., Ambrosi, C. M., Chang, R., Voloshina, A. S., cardiac conduction disease without Brugada syndrome. PACE 33, 274285.
et al. (2009). Structural and functional evidence for discrete exit pathways that Hu, K., Qu, Y., & Boutjdir, M. (2004). Functional basis of sinus bradycardia in congenital
connect the canine sinoatrial node and atria. Circ Res 104, 915923. heart block. Circ Res 94, 3238.
Ferrier, G. R., Saunders, J. H., & Mendez, C. (1973). A cellular mechanism for the generation Huang, Z. M., Prasad, C., Britton, F. C., Ye, L. L., Hatton, W. J., & Duan, D. (2009). Functional
of ventricular arrhythmias by acetylstrophanthidin. Circ Res 32, 600609. role of CLC-2 chloride inward rectier channels in cardiac sinoatrial nodal pacemaker
Fox, K., Ford, I., Steg, P. G., Tendera, M., & Ferrari, R. (2008). Ivabradine for patients with cells. J Mol Cell Cardiol 47, 121132.
stable coronary artery disease and left-ventricular systolic dysfunction (BEAUTIFUL): Hucker, W. J., McCain, M. L., Laughner, J. I., Iaizzo, P. A., & Emov, I. R. (2008). Connexin
a randomised, double-blind, placebo-controlled trial. Lancet 372, 807816. 43 expression delineates two discrete pathways in the human atrioventricular
Froese, A., Breher, S. S., Waldeyer, C., Schindler, R. F., Nikolaev, V. O., Rinn, S., et al. (2012). junction. Anat Rec (Hoboken) 291, 204215.
Popeye domain containing proteins are essential for stress-mediated modulation of Hund, T. J., & Mohler, P. J. (2008). Ankyrin-based targeting pathway regulates human
cardiac pacemaking in mice. J Clin Invest 122, 11191130. sinoatrial node automaticity. Channels (Austin) 2, 404406.
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 285

Huser, J., Blatter, L. A., & Lipsius, S. L. (2000). Intracellular Ca2+ release contributes to Kurian, T., Ambrosi, C., Hucker, W., Fedorov, V. V., & Emov, I. R. (2010). Anatomy
automaticity in cat atrial pacemaker cells. J Physiol 524, 415422. and electrophysiology of the human AV node. Pacing Clin Electrophysiol 33,
Imaizumi, S., Mazgalev, T., Dreifus, L. S., Michelson, E. L., Miyagawa, A., Bharati, S., et al. 754762.
(1990). Morphological and electrophysiological correlates of atrioventricular nodal Kus, T., & Sasyniuk, B. I. (1975). Electrophysiological actions of disopyramide phosphate
response to increased vagal activity. Circulation 82, 951964. on canine ventricular muscle and purkinje bers. Circ Res 37, 844854.
Inada, S., Boyett, M. R., & Dobrzynski, H. (2009a). Mathematical models of human sinus Kyndt, F., Probst, V., Potet, F., Demolombe, S., Chevallier, J. C., Baro, I., et al. (2001).
and atrioventricular node action potential. Comput Cardiol 36, 7780. Novel SCN5A mutation leading either to isolated cardiac conduction defect or
Inada, S., Hancox, J. C., Zhang, H., & Boyett, M. R. (2009b). One-dimensional mathematical Brugada syndrome in a large French family. Circulation 104, 30813086.
model of the atrioventricular node including atrio-nodal, nodal, and nodal-his cells. Lakatta, E. G., & DiFrancesco, D. (2009). What keeps us ticking: a funny current, a calcium
Biophys J 97, 21172127. clock, or both? J Mol Cell Cardiol 47, 157170.
Inoue, S., & Becker, A. E. (1998). Posterior extensions of the human compact atrioventricular Lakatta, E. G., & Maltsev, V. (2009). A new functional pacemaker for the heart's paradigm:
node: a neglected anatomic feature of potential clinical signicance. Circulation 97, Mutual entrainment of intracellular calcium clocks and surface membrane ion
188193. channel Clocks. In D. P. Zipes, & J. Jalife (Eds.), Cardiac Electrophysiology: From Cell
Irisawa, H., Brown, H. F., & Giles, W. (1993). Cardiac pacemaking in the sino-atrial node. to bedside (pp. 235245) (5th ed.). Philadelphia, Pa: WB Saunders Company.
Physiol Rev 73, 197227. Lakatta, E. G., Maltsev, V. A., & Vinogradova, T. M. (2010). A coupled SYSTEM of intracellular
Johnson, W. H., Jr., Yang, P., Yang, T., Lau, Y. R., Mostella, B. A., Wolff, D. J., et al. (2003). calcium clocks and surface membrane voltage clocks controls the timekeepsing mech-
Clinical, genetic, and biophysical characterization of a homozygous HERG mutation anism of the heart's pacemaker. Circ Res 106, 659673.
causing severe neonatal long QT syndrome. Pediatr Res 53, 744748. Lalevee, N., Monier, B., Senatore, S., Perrin, L., & Semeriva, M. (2006). Control of cardiac
Jones, S. A., Boyett, M. R., & Lancaster, M. K. (2007). Declining into failure: the age- rhythm by ORK1, a Drosophila two-pore domain potassium channel. Curr Biol 16,
dependent loss of the L-type calcium channel within the sinoatrial node. Circulation 15021508.
115, 11831190. Lau, D. H., Roberts-Thomson, K. C., & Sanders, P. (2011). Sinus node revisited. Curr Opin
Jones, S. A., Lancaster, M. K., & Boyett, M. R. (2004). Ageing-related changes of connexins Cardiol 26, 5559.
and conduction within the sinoatrial node. J Physiol 560, 429437. Le Scouarnec, S., Le Scouarnec, S., Bhasin, N., Vieyres, C., Hund, T. J., Cunha, S. R., et al.
Joshi, S., & Wilber, D. J. (2005). Ablation of idiopathic right ventricular outow tract (2008). Dysfunction in ankyrin-B-dependent ion channel and transporter targeting
tachycardia: Current perspectives. J Cardiovasc Electrophysiol 16, S52S58. causes human sinus node disease. Proc Natl Acad Sci U S A 105, 1561715622.
Joung, B., Lin, S. F., Chen, Z., Antoun, P. S., Maruyama, M., Han, S., et al. (2010). Mechanisms Lederer, W. J., & Tsien, R. W. (1976). Transient inward current underlying arrhythmogenic
of sinoatrial node dysfunction in a canine model of pacing-induced atrial brillation. effects of cardiotonic steroids in Purkinje bres. J Physiol 263, 73100.
Hear Rhythm 7, 8895. Lei, M., Goddard, C., Liu, J., Loni, A. L., Royer, A., Fung, S. S., et al. (2005). Sinus node
Joung, B., Ogawa, M., Lin, S. F., & Chen, P. S. (2009). The calcium and voltage clocks in dysfunction following targeted disruption of the murine cardiac sodium channel
sinoatrial node automaticity. Korean Circ J 39, 217222. gene Scn5a. J Physiol 567, 387400.
Joyner, R. W., & Van Capelle, F. J. L. (1986). Propagation through electrically coupled Lei, M., Jones, S. A., Liu, J., Lancaster, M. K., Fung, S. S., Dobrzynski, H., et al. (2004). Re-
cells: how a small SA node drives a large atrium. Biophys J 50, 11571164. quirement of neuronal- and cardiac-type sodium channels for murine sinoatrial
Kalman, J. M., Olgin, J. E., & Karch, M. R. (1998). Cristal tachycardias: origin of right atrial node pacemaking. J Physiol 559, 835848.
tachycardias from the crista terminalis identied by intracardiac echocardiography. J Lerman, B. B., Stein, K. M., Markowitz, S. M., & Slotwiner, D. J. (2002). Right ventricular
Am Coll Cardiol 31, 451459. outow tract tachycardia: an update. Card Electrophysiol Rev 6, 6871.
Kamakura, S., Shimizu, W., Matsuo, K., Taguchi, A., Suyama, K., Kurita, T., et al. Li, J., Greener, I. D., Inada, S., Nikolski, V. P., Yamamoto, M., Hancox, J. C., et al. (2008). Com-
(1988). Localization of optimal ablation site of idiopathic ventricular tachycardia puter three-dimensional reconstruction of the atrioventricular node. Circ Res 102,
from right and left ventricular outow tract by body surface ecg. Circulation 98, 975985.
15251533. Li, Z. Y., Wang, Y. H., Maldonado, C., & Kupersmith, J. (1994). Role of junctional zone
Kang, G., Giovannone, S. F., Liu, N., Liu, F. Y., Zhang, J., Priori, S. G., et al. (2010). Purkinje cells between Purkinje bres and ventricular muscle in arrhythmogenesis.
cells from RyR2 mutant mice are highly arrhythmogenic but responsive to targeted Cardiovasc Res 28, 12771284.
therapy. Circ Res 107, 512519. Liu, J., Dobrzynski, H., Yanni, J., Boyett, M. R., & Lei, M. (2007). Organisation of the
Kaseda, S., Gilmour, R. F., Jr., & Zipes, D. P. (1989). Depressant effect of magnesium on mouse sinoatrial node: structure and expression of HCN channels. Cardiovasc
early afterdepolarizations and triggered activity induced by cesium, quinidine, and Res 73, 729738.
4-aminopyridine in canine cardiac Purkinje bers. Am Heart J 118, 458466. Liu, H. El, Zein, L., Kruse, M., Guinamard, R., Beckmann, A., Bozio, A., et al. (2010).
Kashiwakura, Y., Cho, H. C., Barth, A. S., Azene, E., & Marbn, E. (2006). Gene transfer of a Gain-of-function mutations in TRPM4 cause autosomal dominant isolated cardiac
synthetic pacemaker channel into the heart: a novel strategy for biological pacing. conduction disease. Circ Cardiovasc Gene 3, 374385.
Circulation 114, 16821686. Lupoglazoff, J. M., Denjoy, I., Villain, E., Fressart, V., Simon, F., Bozio, A., et al. (2004). Long
Kehat, I., Khimovich, L., Caspi, O., Gepstein, A., Shofti, R., Arbel, G., et al. (2004). Electrome- QT syndrome in neonates: conduction disorders associated with HERG mutations
chanical integration of cardiomyocytes derived from human embryonic stem cells. and sinus bradycardia with KCNQ1 mutations. J Am Coll Cardiol 43, 826830.
Nat Biotechnol 22, 12821289. Maddali, M. (2010). Cardiac pacing in left bundle/bifasciculr block patients. Ann Card
Keith, A., & Flack, M. (1907). The form and nature of the muscular connections between Anaesth 13, 715.
the primary divisions of the vertebrate heart. J Anat Physiol 41, 172189. Maguy, A., Le Bouter, S., Comtois, P., Chartier, D., Villeneuve, L., Wakili, R., et al. (2009).
Kelly, R. G., & Buckingham, M. E. (2002). The anterior heart-forming eld: Voyage to Ion channel subunit expression changes in cardiac Purkinje bers: a potential role
the arterial pole of the heart. Trends Genet 18, 210216. in conduction abnormalities associated with congestive heart failure. Circ Res 104,
Kent, A. F. S. (1913). The structure of the cardiac tissues at the auricularventricular 11131122.
junction. J Physiol 47, xviixviii. Makiyama, T., Akao, M., Tsuji, K., Doi, T., Ohno, S., Takenaka, K., et al. (2005). High risk
Kistler, P. M., Sanders, P., Hussin, A., Morton, J. B., Vohra, J. K., Sparks, P. B., et al. (2003). for brayarrhythmic complications in patients with Brugada syndrome caused by
Focal atrial tachycardia arising from the mitral annulus: electrocardiographic and SCN5A gene mutations. J Am Coll Cardiol 46, 21002106.
electrophysiologic characterization. J Am Coll Cardiol 41, 22122219. Makkar, R. R., Fromm, B. S., Steinman, R. T., Meissner, M. D., & Lehmann, M. H. (1993).
Kistler, P. M., Roberts-Thomson, K. C., Haqqani, H. M., Fynn, S. P., Suresh Singarayar, S., Female gender as a risk factor for torsades de pointes associated with cardiovascu-
Jitendra, K., et al. (2006). P-Wave morphology in focal atrial tachycardia develop- lar drugs. J Am Med Assoc 270, 25902597.
ment of an algorithm to predict the anatomic site of origin. J Am Coll Cardiol 48, Maltsev, V. A., & Lakatta, E. G. (2009). Synergism of coupled subsarcolemmal Ca2+
10101017. clocks and sarcolemmal voltage clocks confers robust and exible pacemaker func-
Ko, Y. S., Yeh, H. I., Ko, Y. L., Hsu, Y. C., Chen, C. F., Wu, S., et al. (2004). Three-dimensional tion in a novel pacemaker cell model. Am J Physiol 296, H594H615.
reconstruction of the rabbit atrioventricular conduction axis by combining histolog- Maltsev, V. A., & Lakatta, E. G. (2010). A novel quantitative explanation for the auto-
ical, desmin, and connexin mapping data. Circulation 109, 11721179. nomic modulation of cardiac pacemaker cell automaticity via a dynamic system
Kodama, I., Boyett, M. R., Nikmaram, M. R., Yamamoto, M., Honjo, H., & Niwa, R. (1999). of sarcolemmal and intracellular proteins. Am J Physiol 298, H2010H2023.
Regional differences in the effects of E-4031 within the sinoatrial node. Am J Physiol Mangoni, M. E., Couette, B., Bourinet, E., Platzer, J., Reimer, D., Striessnig, J., et al. (2003).
276, H793H802. Functional role of L-type Cav1.3 Ca2+ channels in cardiac pacemaker activity. Proc
Kodama, I., Kamiya, K., & Toyama, J. (1997a). Cellular electropharmacology of Natl Acad Sci U S A 100, 55435548.
amiodarone. Cardiovasc Res 35, 1329. Mangoni, M. E., & Nargeot, J. (2008). Genesis and regulation of the heart automaticity.
Kodama, I., Nikmaram, M. R., Boyett, M. R., Suzuki, R., Honjo, H., & Owen, J. M. (1997b). Physiol Rev 88, 919982.
Regional differences in the role of the Ca 2+ and Na + currents in pacemaker activ- Mangoni, M. E., Traboulsie, A., Leoni, A. L., Couette, B., Marger, L., Le Quang, K., et al.
ity in the sinoatrial node. Am J Physiol 272, H2793H2806. (2006). Bradycardia and slowing of the atrioventricular conduction in mice lacking
Koncz, I., Szl, T., Bitay, M., Cerbai, E., Jaeger, K., Flp, F., et al. (2011). Electrophysiological Cav 3.1 check T-type calcium channels. Circ Res 98, 14221430.
effects of ivabradine in dog and human cardiac preparations: potential antiarrhythmic Maron, B. J., & Pelliccia, A. (2006). The heart of trained athletes: cardiac remodeling and
actions. Eur J Pharmacol 668, 419426. the risks of sports, including sudden death. Circulation 114, 6331644.
Kreuzberg, M. M., Willecke, K., & Bukauskas, F. F. (2006). Connexin-mediated cardiac Massing, G. K., & James, T. N. (1976). Anatomical conguration of the His bundle and
impulse propagation: connexin 30.2 slows atrioventricular conduction in mouse bundle branches in the human heart. Circulation 53, 609621.
heart. Trends Cardiovasc Med 16, 266272. Mazur, A., Kusniec, J., & Strasberg, B. (2005). Bundle branch reentrant ventricular
Kruse, M., Schulze-Bahr, E., Coreld, V., Beckmann, A., Stallmeyer, B., Kurtbay, G., et al. tachycardia. Indian Pacing Electrophysiol J 5, 8695.
(2009). Impaired endocytosis of the ion channel TRPM4 is associated with human McGuire, M. A., Bourke, J. P., Robotin, M. C., Johnson, D. C., Meldrum-Hanna, W., Nunn,
progressive familial heart block type I. J Clin Invest 119, 27372744. G. R., et al. (1993). High resolution mapping of Koch's triangle using sixty elec-
Kuo, C. S., & Reddy, C. P. (1981). Effect of lidocaine on escape rate in patients with complete trodes in humans with atrioventricular junctional (AV nodal) reentrant tachycar-
atrioventricular block: B. Proximal His bundle block. Am J Cardiol 47, 13151320. dia. Circulation 88, 23152328.
286 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

McGuire, M. A., de Bakker, J. M., Vermeulen, J. T., Moorman, A. F., Loh, P., Thibault, B., Papadatos, G. A., Wallerstein, P. M., Head, C. E., Ratcliff, R., Brady, P. A., Benndorf, K., et al.
et al. (1996). Atrioventricular junctional tissue. Discrepancy between histological (2002). Slowed conduction and ventricular tachycardia after targeted disruption of
and electrophysiological characteristics. Circulation 94, 571577. the cardiac sodium channel gene Scn5a. Proc Natl Acad Sci U S A 99, 62106215.
McGuire, M. A., de Bakker, J. M., Vermeulen, J. T., Opthof, T., Becker, A. E., & Janse, M. J. Park, D. S., & Fishman, G. I. (2011). The cardiac conduction system. Circulation 123,
(1994). Origin and signicance of double potentials near the atrioventricular node. 904915.
Correlation of extracellular potentials, intracellular potentials, and histology. Circu- Persson, F., Andersson, B., Duker, G., Jacobson, I., & Carlsson, L. (2007). Functional effects
lation 89, 23512360. of the late sodium current inhibition by AZD7009 and lidocaine in rabbit isolated
Meijler, F. L., & Janse, M. J. (1988). Morphology and electrophysiology of the mamma- atrial and ventricular tissue and Purkinje bre. Eur J Pharmacol 558, 133143.
lian atrioventricular node. Physiol Rev 68, 608647. Piccini, J. P., & Calkins, H. (2005). Arrhythmia management in the elderly. Cardiovascular
Meregalli, P. G., Wilde, A. A., & Tan, H. L. (2005). Pathophysiological mechanisms of Disease in the Elderly (pp. 261300). Totowa, New Jersey: Humana Press.
brugada syndrome: Depolarization disorder, repolarization disorder, or more? Piipo, K., Laitinen, P., Swan, H., Toivonen, L., Viitasalo, M., Pasternack, M., et al. (2000).
Cardiovasc Res 67, 367378. Homozygosity for a HERG potassium channel mutation causes a severe form of
Miake, J., Maraban, E., & Nuss, B. H. (2002). Gene therapy: Biological pacemaker created long QT syndrome: identication of an apparent founder mutation in the Finns.
by gene transfer. Nature 419, 132133. J Am Coll Cardiol 35, 19191925.
Milanesi, R., Baruscotti, M., Gnecchi-Ruscone, T., & DiFrancesco, D. (2006). Familial Pinto, J. M., Sosunov, E. A., Gainullin, R. Z., Rosen, M. R., & Boyden, P. A. (1999). Effects of
sinus bradycardia associated with a mutation in the cardiac pacemaker channel. mibefradil, a T-type calcium current antagonist, on electrophysiology of Purkinje
N Engl J Med 354, 151157. bers that survived in the infarcted canine heart. J Cardiovasc Electrophysiol 10,
Miller, J. M., Pezeshkian, N. G., & Yadav, A. V. (2006). Catheter mapping and ablation of 12241235.
right ventricular outow tract ventricular tachycardia. J Cardiovasc Electrophysiol Plotnikov, A. N., Shlapakova, I., Szabolcs, M. J., Danilo, P., Jr., Lorell, B. H., Potapova, I. A.,
17, 800802. et al. (2007). Xenografted Adult Human Mesenchymal Stem Cells Provide a Platform
Miles, W. M. (2001). Idiopathic ventricular outow tract tachycardia: Where does it for Sustained Biological Pacemaker Function in Canine Heart. Circulation 116,
originate? J Cardiovasc Electrophysiol 2, 536537. 706713.
Miquerol, L., Meysen, S., Mangoni, M., Bois, P., van Rijen, H. V., Abran, P., et al. (2004). Pollack, G. H. (1976). Intercellular coupling in the atrioventricular node and other tissues
Architectural and functional asymmetry of the HisPurkinje system of the murine of the rabbit heart. J Physiol 255, 275298.
heart. Cardiovasc Res 63, 7786. Postma, A. V., Denjoy, I., Hoorntje, T. M., Lupoglazoff, J. M., Da Costa, A., Sebillon, P., et al.
Moe, G. K., Preston, J. B., & Burlington, H. (1956). Physiologic evidence for a dual A-V (2002). Absence of calsequestrin 2 causes severe forms of catecholaminergic polymor-
transmission system. Circ Res 4, 357375. phic ventricular tachycardia. Circ Res 91, e21e26.
Mond, H. G., & Proclemer, A. (2011). The 11th world survey of cardiac pacing and im- Postma, A. V., Denjoy, I., Kamblock, J., Alders, M., Lupoglazoff, J. M., Vaksmann, G., et al.
plantable cardioverter-debrillators: calendar year 2009-a World Society of (2005). Catecholaminergic polymorphic ventricular tachycardia: RYR2 mutations,
Arrhythmia's project. PACE 34, 10131027. bradycardia, and follow up of the patients. J Med Genet 42, 863887.
Monfredi, O., Dobrzynski, H., Mondal, T., Boyett, M. R., & Morris, G. M. (2010a). The Potapova, I., Plotnikov, A., Lu, Z., Danilo, P., Jr., Valiunas, V., Qu, J., et al. (2004). Human
anatomy and physiology of the sinoatrial nodea contemporary review. Pacing mesenchymal stem cells as a gene delivery system to create cardiac pacemakers.
Clin Electrophysiol 33, 13921406. Circ Res 94, 952959.
Monfredi, O. J., Johnsen, A. B., Dobrzynski, H., Lloyd, T., Yanni, J., Morris, G. M., et al. Probst, V., Allouis, M., Sacher, F., Pattier, S., Babuty, D., Mabo, P., et al. (2006). Progressive
(2011). Is athletic training-induced bradycardia caused by a downregulation cardiac conduction defect is the prevailing phenotype in carriers of a Brugada syn-
of the Ca 2+ clock pacemaker mechanism in the sinoatrial node? Eur Heart J drome SCN5A mutation5. J Cardiovasc Electrophysiol 17, 270275.
32, 99. Qu, J., Plotnikov, A. N., Danilo, P., Jr., Shlapakova, I., Cohen, I. S., Robinson, R. B., et al.
Monfredi, O. J., Anderson, R. H., Boyett, M. R., & Dobrzynski, H. (2010b). Nodal-like cells (2003). Expression and function of a biological pacemaker in canine heart. Circulation
exist in the right ventricular outow tract. Eur Heart J 311, 696. 107, 11061109.
Molyvdas, P. A., & Sperelakis, N. (1983). Comparison of the effects of several calcium Qu, Y., Xiao, G. -Q., Chen, L., & Boutjdir, M. (2001). Autoantibodies from mothers from
antagonistic drugs on the electrical activity of guinea pig Purkinje bers. Eur J children with congenital heart block down-regulate inhibit L-type Ca channels. J Mol
Pharmacol 88, 205214. Cell Cardiol 33, 11531163.
Moorman, A. F., & Christoffels, V. M. (2003). Cardiac chamber formation: Development, Rana, M. S., Horsten, N. C., Tesink-Taekema, S., Lamers, W. H., Moorman, A. F., & van den
genes, and evolution. Physiol Rev 83, 12231267. Hoff, M. J. (2007). Trabeculated right ventricular free wall in the chicken heart
Morton, J. B., Sanders, P., Das, A., Vohra, J. K., Sparks, P. B., & Kalman, J. M. (2001). Focal atrial forms by ventricularization of the myocardium initially forming the outow
tachycardia arising from the tricuspid annulus: electrophysiologic and electrocardio- tract. Circ Res 100, 10001007.
graphic characteristics. J Cardiovasc Electrophysiol 12, 653659. Ren, F. X., Niu, X. L., Ou, Y., Han, Z. H., Ling, F. D., Zhou, S. S., et al. (2006). Morphological
Munk, A. A., Adjemian, R. A., Zhao, J., Ogbaghebriel, A., & Shrier, A. (1996). Electrophys- and electrophysiological properties of single myocardial cells from Koch triangle of
iological properties of morphologically distinct cells isolated from the rabbit atrio- rabbit heart. Chin Med J (Engl) 119, 20752084.
ventricular node. J Physiol 493, 801813. Rentschler, S., Vaidya, D. M., Tamaddon, H., Degenhardt, K., Sassoon, D., Morley, G. E., et al.
Nattel, S., & Quantz, M. A. (1988). Pharmacological response of quinidine induced early (2001). Visualization and functional characterization of the developing murine cardiac
afterdepolarisations in canine cardiac Purkinje bres: insights into underlying conduction system. Development 128, 17851792.
ionic mechanisms. Cardiovasc Res 22, 808817. Robinson, R. B., Boyden, P. A., Hoffman, B. F., & Hewett, K. W. (1987). Electrical restitution
Neu, A., Eiselt, M., Paul, M., Sauter, K., Stallmeyer, B., Isbrandt, D., et al. (2010). A process in dispersed canine cardiac Purkinje and ventricular cells. Am J Physiol 253,
homozigous SCN5A mutation in a severe, recessive type in a cardiac conduction H1018H1025.
disease. Hum Mutat 31, E1609E1621. Roden, D. M., & Hoffman, B. F. (1985). Action potential prolongation and induction
Nikmaram, M. R., Liu, J., Abdelrahman, M., Dobrzynski, H., Boyett, M. R., & Lei, M. of abnormal automaticity by low quinidine concentrations in canine Purkinje
(2008). Characterization of the effects of ryanodine, TTX, E-4031 and 4-AP on the bers. Relationship to potassium and cycle length. Circ Res 56, 857867.
sinoatrial and atrioventricular nodes. Prog Biophys Mol Biol 96, 452464. Rosen, M. R., Brink, P. R., Cohen, I. S., & Robinson, R. B. (2009). Biologic pacing. In D. P.
Nikolski, V. P., Jones, S. A., Lancaster, M. K., Boyett, M. R., & Emov, I. R. (2003). Cx43 and Zipes, & J. Jalife (Eds.), Cardiac electrophysiology: from cell to bedside (pp. 223234)
dual-pathway electrophysiology of the atrioventricular node and atrioventricular (5th ed.). Philadelphia, Pa: WB Saunders Company.
nodal reentry. Circ Res 92, 469475. Rota, M., & Vassalle, M. (2003). Patch-clamp analysis in canine cardiac Purkinje
Nogami, A. (2011). Purkinje-Related Arrhythmias Part I: Monomorphic Ventricular cells of a novel sodium component in the pacemaker range. J Physiol 548,
Tachycardias. Pacing Clin Electrophysiol 34, 624650. 147165.
Nof, E., Luria, D., Brass, D., Marek, D., Lahat, H., Reznik-Wolf, H., et al. (2007). Point mutation Rozanski, G. J., & Jalife, J. (1986). Automaticity in atrioventricular valve leaets of rabbit
in the HCN4 cardiac ion channel pore affecting synthesis, trafcking, and functional heart. Am J Physiol 19, H397H406.
expression is associated with familial asymptomatic sinus bradycardia. Circulation Rozanski, G. J. (1987). Electrophysiological properties of automatic bers in rabbit
116, 463470. atrioventricular valves. Am J Physiol 253, H720H727.
Noma, A., Irisawa, H., Kokobun, S., Kotake, H., Nishimura, M., & Watanabe, Y. (1980a). Rozanski, G. J., & Lipsius, S. L. (1985). Electrophysiology of functional subsidiary pacemakers
Slow current systems in the A-V node of the rabbit heart. Nature 285, 228229. in canine right atrium. Am J Physiol 249, H594H603.
Noma, A., Kotake, H., & Irisawa, H. (1980b). Slow inward current and its role mediating Rubenstein, J., Schulman, C., Yurchak, P. M., & DeSanctis, R. W. (1972). Clinical Spectrum
the chronotropic effect of epinephrine in the rabbit sinoatrial node. Pugers Arch of the Sick Sinus Syndrome. Circulation 46, 513.
388, 19. Rubenstein, D. S., Fox, L. M., McNulty, J. A., & Lipsius, S. L. (1987). Electrophysiology and
O'Donnell, D., Cox, D., Bourke, J., Mitchell, L., & Furniss, S. (2003). Clinical and electrophysi- ultrastructure of Eustachian ridge from cat right atrium: a comparison with SA
ological differences between patients with arrhythmogenic right ventricular dysplasia node. J Mol Cell Cardiol 19, 965976.
and right ventricular outow tract tachycardia. Eur Heart J 24, 801810. Ryu, S., Yamamoto, S., Andersen, C. R., Nakazawa, K., Miyake, F., & James, T. N. (2009).
Oka, Y., Itoh, H., Ding, W. G., Shimizu, W., Makiyama, T., Ohno, S., et al. (2010). Atrioventric- Intramural Purkinje cell network of sheep ventricles as the terminal pathway of
ular block-induced Torsades de Pointes with clinical and molecular backgrounds similar conduction system. Anat Rec (Hoboken) 292, 1222.
to congenital long QT syndrome. Circulation 74, 25622571. Sano, T., & Yamagishi, S. (1965). Spread of excitation from the sinus node. Circ Res 16,
Opthof, T., Coronel, R., Rademaker, H. M., Vermeulen, J. T., Wilms-Schopman, F. J., & 423430.
Janse, M. J. (2000). Changes in sinus node function in a rabbit model of heart failure Sanchez-Quintana, D., Cabera, J. A., Farre, J., Climent, V., Anderson, R. H., & Ho, S. Y.
with ventricular arrhythmias and sudden death. Circulation 101, 29752980. (2005). Sinus node revisited in the era of electroanatomical mapping and catheter
Padeletti, L., Valleggi, A., Vergaro, G., Luc, F., Rao, C. M., Perrotta, L., et al. (2010). ablation. Heart 91, 189194.
Concordant versus discordant left bundle branch block in heart failure patients: Sanders, P., Kistler, P. M., Morton, J. B., Spence, S. J., & Kalman, J. M. (2004). Remodeling
novel clinical value of an old electrocardiographic diagnosis. J Card Fail 16, of sinus node function in patients with congestive heart failure: reduction in sinus
320326. node reserve. Circulation 110, 897903.
H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288 287

Schram, G., Pourrier, M., Melnyk, P., & Nattel, S. (2002). Differential distribution of cardiac Vassalle, M. (1970). Electrogenic suppression of automaticity in sheep and dog
ion channel expression as a basis for regional specialization in electrical function. Circ purkinje bers. Circ Res 27, 361377.
Res 90, 939950. Vassalle, M. (2007). The vicissitudes of the pacemaker current I (Kdd) of cardiac
Scheinman, M. M. (2009). Role of the His.Purkinje system in the genesis of cardiac arrhyth- purkinje bers. J Biomed Sci 14, 699716.
mia. Hear Rhythm 6, 10501058. Vassalle, M., Yu, H., & Cohen, I. S. (1995). The pacemaker current in cardiac Purkinje
Schulze-Bahr, E., Neu, A., Friederich, P., Kaupp, U. B., Breithardt, G., Pongs, O., et al. myocytes. J Gen Physiol 106, 559578.
(2003). Pacemaker channel dysfunction in a patient with sinus node disease. J Veldkamp, M. W., Wilders, R., Baartscheer, A., Zegers, J. G., Bezzina, C. R., & Wilde, A. A.
Clin Invest 111, 15371545. (2003). Contribution of sodium channel mutations to bradycardia and sinus node
Severs, N. J. (2000). The cardiac muscle cell. Bioessays 22, 188199. dysfunction in LQT3 families. Circ Res 92, 976983.
Severs, N. J., Dupont, E., Coppen, S. R., Halliday, D., Inett, E., Baylis, D., et al. (2004). Verkerk, A. O., Wilders, R., van Borren, M. M., & Tan, H. L. (2009). Is sodium current
Remodelling of gap junctions and connexin expression in heart disease. Biochim present in human sinoatrial node cells? Int J Biol Sci 5, 201204.
Biophys Acta Biomembr 1662, 138148. Verkerk, A. O., Wilders, R., Coronel, R., Ravesloot, J. H., & Verheijck, E. E. (2003).
Severs, N. J., Rothery, S., Dupont, E., Coppen, S. R., Yeh, H. I., Ko, Y. S., et al. (2001). Immuno- Ionic remodeling of sinoatrial node cells by heart failure. Circulation 108,
cytochemical analysis of connexin expression in the healthy and diseased cardiovascu- 760766.
lar system. Microsc Res Tech 52, 301322. Vinogradova, T. M., Maltsev, V. A., Bogdanov, K. Y., Lyashkov, A. E., & Lakatta, E. G.
Shang, L. L., Sanyal, S., Pfahnl, A. E., Jiao, Z., Allen, J., Liu, H., et al. (2008). NF- (2005). Rhythmic Ca2+ oscillations drive sinoatrial nodal cell pacemaker function
kappaB-dependent transcriptional regulation of the cardiac scn5a sodium channel by to make the heart tick. Ann N Y Acad Sci 1047, 138156.
angiotensin II. Am J Physiol 294, C372C379. Viskin, S., Rosso, R., Rogowski, O., & Belhassen, B. (2005). The short-coupled
Shi, W., Wymore, R., Yu, H., Wu, J., Wymore, R. T., Pan, Z., et al. (1999). Distribution and variant of right ventricular outow ventricular tachycardia: A not-so-benign
prevalence of hyperpolarization-activated cation channel (HCN) mRNA expression form of benign ventricular tachycardia? J Cardiovasc Electrophysiol 16,
in cardiac tissues. Circ Res 85, 16. 912916.
Song, T., Yang, J., Yao, Y., Li, H., Chen, Y., Zhang, J., et al. (2011). Spironolactone Watanabe, H., Koopmann, T. T., Le Scouarnec, S., Yang, T., Ingram, C. R., Schott, J. J., et al.
diminishes spontaneous ventricular premature beats by reducing HCN4 (2008). Sodium channel betal subunit mutations associated with Brugada syndrome
protein expression in rats with myocardial infarction. Mol Med Rep 4, 569573. and cardiac conduction disease in humans. J Clin Invest 118, 22602268.
Spach, M. S., Barr, R. C., Serwer, G. S., Johnson, E. A., & Kootsey, J. M. (1971). Collision of Wiese, C., Grieskamp, T., Airik, R., Mommersteeg, M. T., Gardiwal, A., de Gier-de, V. C., et al.
excitation waves in the dog Purkinje system. Extracellular identication. Circ Res (2009). Formation of the sinus node head and differentiation of sinus node myocardi-
29, 499511. um are independently regulated by tbx18 and tbx3. Circ Res 104, 388397.
Stallmeyer, B., Zumhagen, S., Denjoy, I., Duthoit, G., Hbert, J. L., Ferrer, X., et al. (2012). Mu- Wilbur, S. L., & Marchlinski, F. E. (1997). Adenosine as an antiarrhythmic agent. Am J
tational spectrum in the Ca2+-activated cation channel gene TRPM4 in patients with Cardiol 79, 3037.
cardiac conductance disturbances. Hum Mutat 33, 109117. Wit, A. L., Craneeld, P. F., & Gadsby, D. C. (1981). Electrogenic sodium extrusion can stop
Stevenson, W. G., Stevenson, L. W., Middlekauff, H. R., & Saxon, L. A. (1993). Sudden triggered activity in the canine coronary sinus. Circ Res 49, 10291042.
death prevention in patients with advanced ventricular dysfunction. Circulation Wit, A. L., Craneeld, P. F., & Hoffman, B. F. (1972). Slow conduction and reentry in the
88, 29532961. ventricular conducting system. II. Single and sustained circus movement in net-
Stillitano, F., Lonardo, G., Zicha, S., Varro, A., Cerbai, E., Mugelli, A., et al. (2008). Molecular works of canine and bovine Purkinje bers. Circ Res 30, 1122.
basis of funny current (If) in normal and failing human heart. J Cell Mol Cardiol 45, Wit, A. L., Fenoglio, J. J., Jr., Hordof, A. J., & Reemtsma, K. (1979). Ultrastructure and
289299. transmembrane potentials of cardiac muscle in the human anterior mitral valve
Swaminathan, P. D., Purohit, A., Soni, S., Voigt, N., Singh, M. V., Glukhov, A. V., et al. (2011). leaet. Circulation 59, 12841292.
Oxidized CaMKII causes cardiac sinus node dysfunction in mice. J Clin Invest 121, Wit, A. L., Fenoglio, J. J., Jr., & Wagner, B. M. (1973). Electrophysiological properties of
32773288. cardiac muscle in the anterior mitral valve leaet and the adjacent atrium in the
Swedberg, K., Komajda, M., Bohm, M., Borer, J. S., Ford, I., Dubost-Brama, A., et al. dog. Possible implications for the genesis of atrial dysrhythmias. Circ Res 32,
(2010). Ivabradine and outcomes in chronic heart failure (SHIFT): a randomised 731745.
placebo-controlled study. Lancet 376, 875885. Wit, A. L., & Craneeld, P. F. (1976). Triggered activity in cardiac muscle bers of the
Tada, H., Tadokoro, K., Ito, S., Naito, S., Hashimoto, T., Kaseno, K., et al. (2007). Idiopathic simian mitral valve. Circ Res 38, 8598.
ventricular arrhythmias originating from the tricuspid annulus: Prevalence, electro- Wit, A. L., & Craneeld, P. F. (1977). Triggered and automatic activity in the canine coronary
cardiographic characteristics, and results of radiofrequency catheter ablation. Hear sinus. Circ Res 41, 434445.
Rhythm 4, 716. Wiggins, J. R., & Craneeld, P. F. (1976). Two levels of resting potential in canine cardiac
Tardif, J. C., O'Meara, E., Komajda, M., Bhm, M., Borer, J. S., Ford, I., et al. (2011). Ef- Purkinje bers exposed to sodium-free solutions. Circ Res 39, 466474.
fects of selective heart rate reduction with ivabradine on left ventricular Wongcharoen, W., Chen, Y. C., Chen, Y. J., Chang, C. M., Yeh, H. I., Lin, C. L., et al. (2006).
remodelling and function: results from the SHIFT echocardiography substudy. Effects of a Na+/Ca2+ exchanger inhibitor on pulmonary vein electrical activity
Eur Heart J 32, 25072515. and ouabain-induced arrhythmogenicity. Cardiovasc Res 70, 497508.
Tawara, S. (2000). The conduction system of the mammalian heart. London: Imperial College Wright, M., Sacher, F., & Hassaguerre, M. (2009). Catheter ablation for patients with
Press. ventricular brillation. Curr Opin Cardiol 24, 5660.
Tellez, J. O., Dobrzynski, H., Greener, I. D., Graham, G. M., Laing, E., Honjo, H., et al. (2006). Wu, X., & Burgess, S. M. (2004). Integration target site selection for retroviruses and
Differential expression of ion channel transcripts in atrial muscle and sinoatrial node transposable elements. Cell Mol Life Sci 61, 25882596.
in rabbit. Circ Res 99, 13841393. Xia, S., Wang, Y., Zhang, Y., Deng, S. -B., Du, J. -L., Wang, X. -C., et al. (2010). Dynamic
Tellez, J. O., Mczewski, M., Yanni, J., Sutyagin, P., Mackiewicz, U., Atkinson, A., et al. changes in HCN2, HCN4, KCNE1, and KCNE2 expression in ventricular cells from
(2011). Ageing-dependent remodelling of ion channel and Ca2+ clock genes un- acute myocardial infarction rat hearts. Biochem Biophys Res Commun 395,
derlying sino-atrial node pacemaking. Exp Physiol 96, 11631178. 330335.
Temple, I. P., Inada, S., Dobrzynski, H., & Boyett, M. R. (2013). Connexins and the atrioven- Xue, T., Cho, H. C., Akar, F. G., Tsang, S. Y., Jones, S. P., Marbn, E., et al. (2005). Func-
tricular node. Hear Rhythm 10, 297304. tional integration of electrically active cardiac derivatives from genetically
Tse, H. F., Xue, T., Lau, C. P., Siu, C. W., Wang, K., Zhang, Q. Y., et al. (2006). Bioarticial engineered human embryonic stem cells with quiescent recipient ventricular
sinus node constructed via in vivo gene transfer of an engineered pacemaker HCN cardiomyocytes: insights into the development of cell-based pacemakers. Circulation
channel reduces the dependence on electronic pacemaker in a sick sinus syndrome 111, 1120.
model. Circulation 114, 16821686. Zhang, H., Holden, A. V., Kodama, I., Zhang, H., Holden, A. V., Kodama, I., et al. (2000).
Tsien, R. W., Kass, R. S., & Weingart, R. (1979). Cellular and subcellular mechanisms of Mathematical models of action potentials in the periphery and center of the rabbit
cardiac pacemaker oscillations. J Exp Biol 81, 205215. sinoatrial node. Am J Physiol 279, H397H421.
Tranum-Jensen, J., Wilde, A. A., Vermeulen, J. T., & Janse, M. J. (1991). Morphology of Zhang, Y., Bharati, S., Mowrey, K. A., Zhuang, S., Tchou, P., & Mazgalev, T. N. (2001). His elec-
electrophysiologically identied junctions between Purkinje bers and ventricular trogram alternans reveal dual-wavefront inputs into and longitudinal dissociation
muscle in rabbit and pig hearts. Circ Res 69, 429437. within the bundle of His. Circulation 104, 832838.
Ueda, K., Nakamura, K., Hayashi, T., Inagaki, N., Takahashi, M., Arimura, T., et al. (2004). Zhang, H., Lau, D. H., Shlapakova, I. N., Zhao, X., Danilo, P., Robinson, R. B., et al.
Functional characterization of a trafcking-defective HCN4 mutation, D553N, as- (2011, March 8) Implantation of sinoatrial node cells into canine right ventricle:
sociated with cardiac arrhythmia. J Biol Chem 279, 2719427198. biological pacing appears limited by the substrate. Cell Transplant 20, 1112.
Vaillant, F., Dehina, L., Mazzadi, A., Descotes, J., Chevalier, P., Tabib, A., et al. (2011). Heart Zicha, S., Fernandez-Velasco, M., Lonardo, G., L'Heureux, N., & Nattel, S. (2005). Sinus
rate reduction with ivabradine increases ischaemia-induced ventricular brillation node dysfunction and hyperpolarization-activated (HCN) channel subunit re-
threshold: Role of myocyte structure and myocardial perfusion. Resuscitation 82, modeling in a canine heart failure model. Cardiovasc Res 66, 472481.
10921099. Zicha, S., Tsuji, Y., Shiroshita-Takeshita, A., & Nattel, S. (2006). Beta-blockers as antiar-
Van De Ven, L. L. M., Crijns, H. J. G. M., De Muinck, E. D., Van Gelder, I. C., Van Wijk, L. M., & rhythmic agents. Handb Exp Pharmacol 171235.
Lie, K. I. (1996). Electrophysiologic and antiarrhythmic effects of intravenous Yamamoto, M., Dobrzynski, H., Tellez, J., Niwa, R., Billeter, R., Honjo, H., et al. (2006).
bisoprolol in atrioventricular nodal reentry tachycardia. Curr Ther Res 57, 950958. Extended atrial conduction system characterised by the expression of the HCN4
van Rijen, H. V., van Veen, T. A., van Kempen, M. J., Wilms-Schopman, F. J., Potse, M., channel and connexin45. Cardiovasc Res 72, 271281.
Krueger, O., et al. (2001). Impaired conduction in the bundle branches of mouse Yamanushi, T. T., Yanni, J., Dobrzynski, H., Kabuto, H., & Boyett, M. R. (2010). Changes
hearts lacking the gap junction protein connexin40. Circulation 103, 15911598. in ion channel expression in right-sided congestive heart failure. J Mol Cell
van Veen, T. A., Stein, M., Royer, A., Le Quang, K., Charpentier, F., Colledge, W. H., et al. Cardiol 48, S73.
(2005). Impaired impulse propagation in Scn5a-knockout mice: combined contri- Yan, G. X., & Antzelevitch, C. (1999). Cellular basis for the brugada syndrome and other
bution of excitability, connexin expression, and tissue architecture in relation to mechanisms of arrhythmogenesis associated with st-segment elevation. Circulation
aging. Circulation 112, 19271935. 100, 16601666.
288 H. Dobrzynski et al. / Pharmacology & Therapeutics 139 (2013) 260288

Yanni, J., Boyett, M. R., Anderson, R. H., & Dobrzynski, H. (2009a). The extent of the ERG (KCNH2): channel (ether-a-go-go related gene voltage-gated K+ channel) and
specialized atrioventricular ring tissues. Hear Rhythm 6, 672680. gene responsible for IK,r
Yanni, J., Tellez, J. O., Sutyagin, P. V., Boyett, M. R., & Dobrzynski, H. (2010). Structural HCN1, HCN2, HCN4: hyperpolarization-activated cyclic nucleotide gated channels responsi-
remodelling of the sinoatrial node in obese old rats. J Mol Cell Cardiol 48, ble for If
653662. hMSC: human mesenchymal stem cells
Yanni, J., Tellez, J. O., Maczewski, M., Mackiewicz, U., Beresewicz, A., Billeter, R., et al. Ib,Na: background Na+ current
(2011a). Changes in ion channel gene expression underlying heart failure-induced ICa,L: L-type Ca2+ current
sinoatrial node dysfunction. Circ Heart Fail 4, 496508. ICa,T: T type Ca2+ current
Yanni, J., Yamanushi, T. T., Dobrzynski, H., & Boyett, M. R. (2009b). Remodelling of the If: hyperpolarization-activated or funny current
extracellular matrix in the rat sinoatrial node in congestive heart failure. Proc IK,1: background inward rectier K+ current
Physiol Soc 15, C26C. IK,ACh: acetylcholine-activated K+ current
Yanni, J., Cai, X., Jones, C. B., Corno, A. F., Hutcheon, R. C., Monfredi, O., et al. (2011b). IK,r: rapid delayed rectier K+ current
Mechanisms of delayed intraventricular conduction in heart failure. Hear Rhythm IK,s: slow delayed rectier K+ current
8, 1819. INa: Na+ current
Yeh, Y. H., Burstein, B., Qi, X. Y., Sakabe, M., Chartier, D., Comtois, P., et al. (2009). Funny INaCa: Na+Ca2+ exchange current
current downregulation and sinus node dysfunction associated with atrial tachyar- INa,L: late Na+ current
rhythmia: a molecular basis for tachycardia-bradycardia syndrome. Circulation 119, IP3: inositol trisphosphate
15761585. Ist: sustained inward current
Yoo, S., Dobrzynski, H., Fedorov, V. V., Xu, S. Z., Yamanushi, T. T., Jones, S. A., et al. Ito: transient outward K+ current
(2006). Localisation of Na+ channel isoforms at the atrioventricular junction and IK,ur: ultra-rapid delayed rectier K+ current
atrioventricular node. Circulation 114, 13601371. Kir2.1 (KCNJ2): inward rectier K+ channel and gene responsible for IK,1
Yu, H., Chang, F., & Cohen, I. S. (1995). Pacemaker current If in adult canine cardiac ventric- Kir3.1: inward rectier K+ channel in part responsible for IK,ACh
ular myocytes. J Physiol 485, 469483. Kv1.4, Kv1.5, Kv3.4, Kv4.2, Kv4.3: voltage-gated K+ channels
KvLQT1 (KCNQ1): voltage-gated K+ channel and gene responsible for IK,s
minK: auxiliary subunit responsible for IK,s
mRNA: messenger ribonucleic acid
MRI: magnetic resonance imaging
Nav1 (SCN1B): Na+ channel (and gene) subunit
Glossary Nav1.1: voltage-gated Na+ channel
Nav1.5 (SCN5A): principle voltage-gated Na+ channel and gene responsible for INa in
A, N, NH: types of myocyte at the atrioventricular node the heart
ANK2: ankyrin 2 NF-B: transcription factor
AV: atrioventricular NCX1: Na+Ca2+ exchanger
AVNRT: atrioventricular nodal reentrant tachycardia ORK1: two-pore-domain K+ channel (Drosophila)
CASQ2: calsequestrin 2 RYR2: type 2 ryanodine receptor (sarcoplasmic reticulum Ca2+ release channel)
Cav1.2 (CACNA1C): principal L-type Ca2+ channel and gene SERCA2: sarco/endoplasmic reticulum Ca2+ ATPase (sarcoplasmic reticulum Ca2+ pump)
Cav1.3: auxiliary L-type Ca2+ channel SR: sarcoplasmic reticulum
Cav3.1/Cav3.2: T-type Ca2+ channels TASK1/TREK1/TWIK1/TWIK2: two-pore-domain K+ channels
ClC-2: hyperpolarization-activated Cl channel Tbx3/Tbx18: T-box transcription factors
CPVT: catecholaminergic polymorphic ventricular tachycardia TGF1: transforming growth factor beta 1
Cx30.2, Cx40, Cx43, Cx45: connexins 30.2, 40, 43 and 45 TRP: transient receptor potential cation channels (relatively non-selective cation channels)
DAD: delayed afterdepolarization TRPM4: transient receptor potential cation channel subfamily M member 4
DNA: deoxyribonucleic acid VT: ventricular tachycardia
dV/dtmax: maximum upstroke velocity of the action potential
EAD: early afterdepolarization
ECG: electrocardiogram

You might also like