ArticlePDF AvailableLiterature Review

Why Do We Have Purkinje Fibers Deep in Our Heart?

Authors:

Abstract and Figures

Purkinje fibers were the first discovered component of the cardiac conduction system. Originally described in sheep in 1839 as pale subendocardial cells, they were found to be present, although with different morphology, in all mammalian and avian hearts. Here we review differences in their appearance and extent in different species, summarize the current state of knowledge of their function, and provide an update on markers for these cells. Special emphasis is given to popular model species and human anatomy.
Content may be subject to copyright.
PHYSIOLOGICAL RESEARCH • ISSN 0862-8408 (print) • ISSN 1802-9973 (online)
© 2014 Institute of Physiology v.v.i., Academy of Sciences of the Czech Republic, Prague, Czech Republic
Fax +420 241 062 164, e-mail: physres@biomed.cas.cz, www.biomed.cas.cz/physiolres
Physiol. Res. 63 (Suppl. 1): S9-S18, 2014
REVIEW
Why Do We Have Purkinje Fibers Deep in Our Heart?
D. SEDMERA1,2, R. G. GOURDIE3,4,5
1Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic,
2Department of Cardiovascular Morphogenesis, Institute of Physiology Academy of Sciences of the
Czech Republic, Prague, Czech Republic, 3Virginia Tech Carilion Research Institute, Center for
Heart and Regenerative Medicine Research, Roanoke, Virginia, USA, 4Virginia Tech School of
Biomedical Engineering and Sciences, Roanoke, Virginia, USA, 5Department of Emergency
Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
Received October 21, 2013
Accepted October 29, 2013
Summary
Purkinje fibers were the first discovered component of the
cardiac conduction system. Originally described in sheep in 1839
as pale subendocardial cells, they were found to be present,
although with different morphology, in all mammalian and avian
hearts. Here we review differences in their appearance and
extent in different species, summarize the current state of
knowledge of their function, and provide an update on markers
for these cells. Special emphasis is given to popular model
species and human anatomy.
Key words
Cardiac conduction system Specialized tracts Gap junctions
Connexin
Corresponding author
D. Sedmera, Department of Cardiovascular Morphogenesis,
Institute of Physiology, Academy of Sciences of the Czech
Republic, Videnska 1083, 14220 Prague, Czech Republic. E-mail:
dsedmera@biomed.cas.cz
Introduction
The cardiac conduction system (CCS) is defined
as a network of specialized myocardial cells that
generates the cardiac rhythm and assures a coordinated
propagation of the electrical impulse for efficient
contraction of the heart. In the adult mammalian heart,
the CCS comprises the sinoatrial (SA) node, the
internodal tracts, the atrioventricular node, the
atrioventricular (His) bundle, its right and left branches,
and the network of Purkinje fibers. While the functional
equivalent of these components are present in some form
in all vertebrate hearts (Sedmera et al. 2003), all
morphologically distinct parts are present only in the
heart of mammals. While development of the CCS has
been subject to numerous reviews (Gourdie et al.
2003a,b, Christoffels et al. 2010, Burggren et al. 2013),
little was written on the comparative morphology of its
components in different species, with a few notable
exceptions (Davies 1930, Davies et al. 1952, 1994). The
goal of this overview is to put the Purkinje fibers into
context of other CCS components, briefly describe the
history of their discovery, provide functional insight into
equivalent structures in the lower vertebrates, and then to
focus in detail on their structure and function in the most
popular model species of mammals and birds.
Function of individual CCS components
The SA node is the principal pacemaker of the
heart. Under normal conditions it is the cardiac tissue that
autonomously sets the rhythm of the heart beat. It is also
subject to neurohumoral regulation, in particular by
autonomous nervous system. This allows the heart to
change its frequency in reaction to the functional status of
the organism. Morphologically, it is organized into a
three dimensionally complex compact structure
S10 Sedmera and Gourdie Vol. 63
(Mommersteeg et al. 2007, Fedorov et al. 2010), with
specific connection points to the working atrial
myocardium. In lower vertebrates, its function is
contained in a compartment termed the sinus venosus
(Koprla 1987, Jensen et al. 2012). New data on the
cellular origin of the SA node from outside the traditional
heart fields and role of Wnt signaling in recruitment of
mesodermal cells into pacemaker lineage were recently
reported by the Mikawa lab (Bressan et al. 2013).
Atrial special conduction pathways
The morphological distinction of specialized
intra-atrial and internodal conduction pathways is a
controversial topic. Some claim that there are tracts of
Purkinje-like cells connecting the sinoatrial and
atrioventricular node (James and Sherf 1971), while other
agree that the conduction through the atria is anisotropic,
see the main reason in holes caused by the entrance
vessels (Betts et al. 2002, Ho et al. 2002). Our view is
that these preferential conduction pathways, the best
example of which is the interatrial bundle of Bachmann
(Sedmera et al. 2006) can be explained by tissue
geometry (pectinate muscles), in agreement with previous
experimental data (Komuro et al. 1986), but are open to a
marker that would distinguish cells within those tracts
from the remaining atrial myocytes.
The AV node
The main function of the atrioventricular node is
generation of a delay between activation (and ensuing
contraction) of the atria and the ventricles. Due to its
prolonged refractory period, it also serves as a filter
against propagation of atrial tachyarrhythmias to the
ventricles. In mammals, it shows a distinct morphological
organization with specific cell phenotypes (Efimov et al.
1997, Aanhaanen et al. 2010). On the other hand, its
morphological localization in birds is still obscure
(Vicente-Steijn et al. 2011). In lower vertebrates, and
during embryonic development, the function of delay
generator is located in the atrioventricular canal region, a
slowly proliferating, conducting and contracting portion
of the cardiac tube that apparently retains its “primitive”
phenotype – not following the pathway of chamber
myocardium differentiation (Kirby 2007). The
transcriptional regulation of these events has been
recently uncovered, and Tbx2, BMP and Tbx3 are
implicated in maintaining this transcription programme
(Mommersteeg et al. 2007, Aanhaanen et al. 2009, 2010).
Electrophysiologically, this region shows a typical action
potential shape (Arguello et al. 1986) and has a level of
automaticity, which can manifest even in the embryo
when the sinoatrial node is perturbed (Raddatz 1997).
Interfaces between atria and node and node and His are
marked by distinct transitions in connexin expression
(Coppen et al. 1999, Gourdie and Sedmera 2008). Such
abrupt changes in cellular coupling can play a role in AV
delay generation in adult heart (Choi and Salama 1998).
The His bundle
Also known as the atrioventricular, or non-
branching bundle, it forms under normal conditions the
only conductive pathway between the atria and the
ventricles. It is a rapidly conducting tissue, with
propagation velocity an order of magnitude faster than
the working myocardium. As it traverses the
atrioventricular fibrous plane, the bundle is insulated
from the rest of the myocardium except of its proximal
connection with the AV node and distal bifurcation into
the left and right bundle branch.
The bundle branches
These form continuation of the His bundle,
sharing many of its characteristics – rapid conduction
speed, expression of gap junction protein connexin40
(without co-expression of connexin43) and fibrous
insulation from the working myocardium. This makes
these bundles well suited to act as electrical cables,
assuring rapid spread of the impulse through the
ventricles. There is a notable asymmetry between the left
and right bundle branch, the left being broad, in the
mouse composed of multiple parallel isolated strands,
while the right bundle being a narrow structure (Miquerol
et al. 2004). It is likely due to optimization of source:sink
ratio due to a marked asymmetry of the myocardial mass
between the left and right ventricle.
The Purkinje fibers
The Purkinje fibers are the terminal part of the
cardiac conduction system. They form a three-
dimensional subendocardial network originating from the
bundle branches and their main function is to distribute
the depolarization signal rapidly to the working
myocardium.
2014 Purkinje Fibers in Vertebrates S11
Discovery of CCS
The historical sequence, in which various CCS
components were discovered, is in reverse order to the
functional sequence of activation. In 1839 the Czech
scientist Jan Evangeliste Purkinje described a pale
network of cells in the sheep heart, and noted their
microscopic characteristics, including the presence of one
or two nuclei and cross striations, which made him, after
some discussion, consider them a special form muscular
tissue (Eliska 2006). It took more than fifty years before a
Swiss physiologist His found the elusive connection
between the atrial and ventricular myocardium, first by a
series of cuts in the beating heart, then by detailed
histological examination of the region, where such cuts
led to atrioventricular block (Suma 2001). Over a decade
later, Tawara (1906) connected this bundle by describing
the atrioventricular node proximally and the bundle
branches distally. The last component discovered was the
sinoatrial node, described by Keith and Flack (1907) only
a year later.
Original description by Purkinje
The original 1839 description by Purkinje was a
part of a larger volume dealing with innervation of various
organs and published in Polish. He followed up six years
later in 1845 with a more detailed account in German, then
the universal language of morphologists (Eliska 2006).
There are few interesting points in this description that
pertain to the present review. First, while the original
species was sheep, he noted similar structures in the cow,
pig, and horse. On the other hand, he was unable to see his
cells in the human, dog, rabbit, or hare heart. This is an
important reminder of the dangers of expecting that a
functionally similar structure will look the same in
different species (Robb 1965). Second, already by then he
stated with certainty that these structures are not nervous
fibers, especially significant in the context of general focus
of his 1839 text on nervous tissue. Third, although he did
seriously consider (due to their appearance) that these
fibers could be cartilage, the presence of striations made
him to decide that most likely these tissues were muscular.
Nevertheless, later studies using various neuronal markers
re-ignited the issue (Gorza et al. 1988, 1994), and the final
word on myogenic nature of the CCS was provided using
genetic lineage tracing techniques – showing its common
origin with working cardiomyocytes (Gourdie 1995,
Cheng et al. 1999).
Differentiation of Purkinje fibers during
development
It has been shown in experimental studies in the
chick heart, that ventricular Purkinje fibers share
common origin with the surrounding ventricular
myocytes and terminally differentiate during fetal period
(Gourdie et al. 1995). In the avian model, it was
convincingly demonstrated that the differentiation
towards the conduction phenotype is time-sensitive, and
locally produced endothelin-1 was demonstrated to be a
key signaling molecule (Gourdie et al. 1998, Hyer et al.
1999, Takebayashi-Suzuki et al. 2000). The clonal
relationship between the working ventricular myocytes
and Purkinje myocytes observed in birds was confirmed
recently in the mammalian (mouse) model (Meilhac et al.
2003, Miquerol et al. 2010). However, transcription or
any other regulation governing this process in mammals
is still elusive. While it was long speculated that
ventricular trabeculae, forming during chamber
differentiation as a means to increase myocardial mass
prior to presence of coronary perfusion, form the
precursors of the Purkinje network (de Jong et al. 1992,
Sedmera et al. 2004), it is clear that not all the trabeculae
will turn into Purkinje fibers – in fact, the majority will
form trabeculae carneae, or “meaty” trabeculations
composed largely of working myocardium, that we find
on the inside of the ventricles. As noted above, our
knowledge of mechanisms governing these decisions is
woefully incomplete.
Subendocardial and intramural network
The Purkinje network is composed of two
components: the subendocardial fibers, which have
connection to the bundle branches and assure the apex-to-
base activation of the ventricle, and a variably present
intramural component. While the first is invariably
reported, although with different morphology, from all
mammalian and avian hearts, the latter, presumably
accelerating transmural conduction, is only
morphologically distinguishable in some species –
in particular sheep (Ryu et al. 2009) (Fig. 1), cow
(Oosthoek et al. 1993), or pig (Fig. 2). In these animals,
connection between the subendocardial and intramural
network can be found at regular intervals (Fig. 6 in
Oosthoek et al. 1993), and the extent in cow is about
80 % of left ventricular wall thickness. On the other hand,
despite quite extensive searches, no intramural fibers
S12 Sedmera and Gourdie Vol. 63
have been located in the mouse or human heart (Fig. 3).
The presence of absence of the intramural component
does not seem to be related to heart size, as some small
animals – such as rats (Fazel et al. 1989, Thompson et al.
1990, Gourdie et al. 1992, 1993, Vuillemin et al. 1992)
or chicken (Gourdie et al. 1993) (Fig. 4) do have
intramural fibers – in the case of chicken clearly
associated with the coronary arteries.
Functionally, the Purkinje fibers are
characterized by a unique ion channel expression and
faster conductivity in comparison to the working
ventricular myocardium. This creates a local
heterogeneity in conduction velocity and coupling, which
can lead to re-entry. Their role as foci of arrhythmias was
recently and extensively reviewed in (Boyden et al.
2010).
Fig. 1. Purkinje fibers in the sheep
heart. A. Actinin staining shows that
both the subendocardially located
Purkinje fibers and the working
cardiomyocytes have well organized
contractile apparatus. B.
Hematoxylin and Eosin staining
shows clearly that they are larger
than the working ventricular
myocytes and their spatial and
fibrous isolation. C. Staining for
connexin43 shows very high density
of these gap junction proteins on
the entire cell membrane (compare
to neighboring working myocardium
where it is localized mostly at cell
ends). Wheat germ agglutinin
(WGA) staining highlights cell
boundaries and fibrous tissue. Scale
bars 25 µm.
2014 Purkinje Fibers in Vertebrates S13
Fig. 2. Purkinje fibers in the pig heart. Connexin40 staining labels specifically both subendocardial (A) and intramural (B) Purkinje
fibers. C and D: similar morphology with Connxin40 staining preferentially localized to cell ends and distribution of Purkinje cells is
found also in the right ventricle. E and F: in contrast, Connexin43 staining is distributed in the entire cell surface.
S14 Sedmera and Gourdie Vol. 63
Fig. 3. Purkinje fibers in the murine heart. Panels A and B show the transition of Connexin43-negative left bundle branches into
Purkinje fibers, which co-express both Connexin40 and 43. The tracts are spatially separated from the working myocardium and show a
thin fibrous sheath. Scale bars 10 µm. Panels C and D visualize the Purkinje network using Connexin40:GFP transgenic mouse. The
Purkinje network is formed by 1-3 cells thick strands of myocytes that are thinner but longer than the working myocytes. Panels
E and F compare the arrangement of the entire left ventricular network in mouse (from Miquerol
et al.
2004, withe permission) and
human (Tawara 1906). In neither species were described any intramural Purkinje fibers.
2014 Purkinje Fibers in Vertebrates S15
Fig. 4. Purkinje fibers in the
ED17 chick embryonic heart.
Similar to ungulates, there are
both subendocardial (A, B) as
well as intramural (C) Purkinje
cells; the later are located
periarterially. Coronary arteries
are labeled with asterisks.
Current state of the art future perspectives
As a closing paragraph, we would like to provide
some insight into some potentially fruitful areas of active
reseach of Purkinje fibers. First, new morphological
markers such as contactin (Pallante et al. 2010) allow us
better delineation of the entire network across species
(Ryu et al. 2009), and help to uncover signaling pathways
directing their differentiation from the working myocytes
in mammals such as notch signaling (Rentschler et al.
2012). This would be important to resolve the issue why
some closely related species do or do not possess the
intramural component (e.g. rat vs. mouse). Second little
explored question is the morphological and functional
imaging of contacts and conduction at the Purkinje-
myocyte junction, that should be enabled by coupling of
these markers with high-speed, high-resolution mapping
techniques. Is there a gradual transition of phenotype
from clear PF to working myocyte, or is the boundary
sharply defined? The third question worthy of attention is
the origin and differentiation of Purkinje myocytes. Is the
commitment to the conduction lineage irreversible, and if
yes, at which point of development? What is the default
program of ventricular myocytes – conduction, or
working phenotype? Resolution of this particular
question would be important piece of information
necessary to production of larger pieces of implantable
tissue-engineered myocardium.
Conflict of Interest
There is no conflict of interest.
Acknowledgements
Author’s primary research is supported by the Ministry of
Education of the Czech Republic PRVOUK-P35/LF1/5,
and institutional RVO: 67985823 from the Academy of
Sciences of the Czech Republic. Current support from the
Grant Agency of the Czech Republic P302/11/1308 and
13-12412S is also gratefully acknowledged. RGG is
supported by NIH RO1 HL56728.
References
AANHAANEN WT, BRONS JF, DOMINGUEZ JN, RANA MS, NORDEN J, AIRIK R, WAKKER V, DE GIER-DE
VRIES C, BROWN NA, KISPERT A, MOORMAN AF, CHRISTOFFELS VM: The Tbx2+ primary
myocardium of the atrioventricular canal forms the atrioventricular node and the base of the left ventricle. Circ
Res 104: 1267-1274, 2009.
AANHAANEN WT, MOMMERSTEEG MT, NORDEN J, WAKKER V, DE GIER-DE VRIES C, ANDERSON RH,
KISPERT A, MOORMAN AF, CHRISTOFFELS VM: Developmental origin, growth, and three-dimensional
architecture of the atrioventricular conduction axis of the mouse heart. Circ Res 107: 728-736, 2010.
ARGUELLO C, ALANIS J, PANTOJA O, VALENZUELA B: Electrophysiological and ultrastructural study of the
atrioventricular canal during the development of the chick embryo. J Mol Cell Cardiol 18: 499-510, 1986.
BETTS TR, HO SY, SANCHEZ-QUINTANA D, ROBERTS PR, ANDERSON RH, MORGAN JM: Three-
dimensional mapping of right atrial activation during sinus rhythm and its relationship to endocardial
architecture. J Cardiovasc Electrophysiol 13: 1152-1159, 2002.
S16 Sedmera and Gourdie Vol. 63
BOYDEN PA, HIROSE M, DUN W: Cardiac Purkinje cells. Heart Rhythm 7: 127-135, 2010.
BRESSAN M, LIU G, MIKAWA T: Early mesodermal cues assign avian cardiac pacemaker fate potential in a tertiary
heart field. Science 340: 744-748, 2013.
BURGGREN WW, CHRISTOFFELS VM, CROSSLEY DA 2ND, ENOK S, FARRELL AP, HEDRICK MS, HICKS
JW, JENSEN B, MOORMAN AF, MUELLER CA, SKOVGAARD N, TAYLOR EW, WANG T:
Comparative cardiovascular physiology: future trends, opportunities and challenges. Acta Physiol: 2013 (in
press).
CHENG G, LITCHENBERG WH, COLE GJ, MIKAWA T, THOMPSON RP, GOURDIE RG: Development of the
cardiac conduction system involves recruitment within a multipotent cardiomyogenic lineage. Development
126: 5041-5049, 1999.
CHOI BR, SALAMA G: Optical mapping of atrioventricular node reveals a conduction barrier between atrial and nodal
cells. Am J Physiol 274: H829-H845, 1998.
CHRISTOFFELS VM, HOOGAARS WM, MOORMAN AF: Patterning and development of the conduction system of
the heart: origins of the conduction system in development. In: Heart Development and Regeneration.
ROSENTHAL N, HARVEY RP (eds), Elsevier, London, 2010, pp 171-194.
COPPEN SR, SEVERS NJ, GOURDIE RG: Connexin45 (alpha 6) expression delineates an extended conduction
system in the embryonic and mature rodent heart. Dev Genet 24: 82-90, 1999.
DAVIES F: The conducting system of the bird's heart. J Anat 64: 129-146, 1930.
DAVIES F, FRANCIS ET, KING TS: The conducting (connecting) system of the crocodilian heart. J Anat 86: 152-161,
1952.
DAVIES PJ, DONALD JA, CAMPBELL G: The distribution and colocalization of neuropeptides in fish cardiac
neurons. J Auton Nerv Syst 46: 261-272, 1994.
DE JONG F, OPTHOF T, WILDE AA, JANSE MJ, CHARLES R, LAMERS WH, MOORMAN AF: Persisting zones
of slow impulse conduction in developing chicken hearts. Circ Res 71: 240-250, 1992.
EFIMOV IR, FAHY GJ, CHENG Y, VAN WAGONER DR, TCHOU PJ, MAZGALEV TN: High-resolution
fluorescent imaging does not reveal a distinct atrioventricular nodal anterior input channel (fast pathway) in the
rabbit heart during sinus rhythm. J Cardiovasc Electrophysiol 8: 295-306, 1997.
ELISKA O: Purkynje fibers of the heart conduction system--history and the present time (in Czech). Cas Lek Cesk 145:
329-335, 2006.
FAZEL AR, SUMIDA H, SCHULTE BA, THOMPSON RP: Lectin histochemistry of the embryonic heart: fucose-
specific lectin binding sites in developing rats and chicks. Am J Anat 184: 76-84, 1989.
FEDOROV VV, GLUKHOV AV, CHANG R, KOSTECKI G, AFEROL H, HUCKER WJ, WUSKELL JP, LOEW
LM, SCHUESSLER RB, MOAZAMI N, EFIMOV IR: Optical mapping of the isolated coronary-perfused
human sinus node. J Am Coll Cardiol 56: 1386-1394, 2010.
GORZA L, SCHIAFFINO S, VITADELLO M: Heart conduction system: a neural crest derivative? Brain Res 457:
360-366, 1988.
GORZA L, VETTORE S, VITADELLO M: Molecular and cellular diversity of heart conduction system myocytes.
Trends Cardiovasc Med 4: 153-159, 1994.
GOURDIE RG: A map of the heart: gap junctions, connexin diversity and retroviral studies of conduction myocyte
lineage. Clin Sci (Lond) 88: 257-262, 1995.
GOURDIE RG, SEDMERA D: Letter by Gourdie and Sedmera regarding article, "abnormal conduction and
morphology in the atrioventricular node of mice with atrioventricular canal-targeted deletion of Alk3/Bmpr1a
receptor". Circulation 118: e106; author reply e107, 2008.
GOURDIE RG, GREEN CR, SEVERS NJ, THOMPSON RP: Immunolabelling patterns of gap junction connexins in
the developing and mature rat heart. Anat Embryol 185: 363-378, 1992.
GOURDIE RG, GREEN CR, SEVERS NJ, ANDERSON RH, THOMPSON RP: Evidence for a distinct gap-junctional
phenotype in ventricular conduction tissues of the developing and mature avian heart. Circ Res 72: 278-289,
1993.
GOURDIE RG, MIMA T, THOMPSON RP, MIKAWA T: Terminal diversification of the myocyte lineage generates
Purkinje fibers of the cardiac conduction system. Development 121: 1423-1431, 1995.
2014 Purkinje Fibers in Vertebrates S17
GOURDIE RG, WEI Y, KIM D, KLATT SC, MIKAWA T: Endothelin-induced conversion of embryonic heart muscle
cells into impulse-conducting Purkinje fibers. Proc Natl Acad Sci USA 95: 6815-6818, 1998.
GOURDIE RG, HARRIS BS, BOND J, EDMONDSON AM, CHENG G, SEDMERA D, O'BRIEN TX, MIKAWA T,
THOMPSON RP: His-Purkinje lineages and development. Novartis Found Symp 250: 110-122; discussion
122-114, 276-119, 2003a.
GOURDIE RG, HARRIS BS, BOND J, JUSTUS C, HEWETT KW, O'BRIEN TX, THOMPSON RP, SEDMERA D:
Development of the cardiac pacemaking and conduction system. Birth Defects Res C Embryo Today 69: 46-57,
2003b.
HO SY, ANDERSON RH, SANCHEZ-QUINTANA D: Atrial structure and fibres: morphologic bases of atrial
conduction. Cardiovasc Res 54: 325-336, 2002.
HYER J, JOHANSEN M, PRASAD A, WESSELS A, KIRBY ML, GOURDIE RG, MIKAWA T: Induction of
Purkinje fiber differentiation by coronary arterialization. Proc Natl Acad Sci USA 96: 13214-13218, 1999.
JAMES TN, SHERF L: Specialized tissues and preferential conduction in the atria of the heart. Am J Cardiol 28:
414-427, 1971.
JENSEN B, BOUKENS BJ, POSTMA AV, GUNST QD, VAN DEN HOFF MJ, MOORMAN AF, WANG T,
CHRISTOFFELS VM: Identifying the evolutionary building blocks of the cardiac conduction system. PLoS
One 7: e44231, 2012.
KEITH A, FLACK MW: The form and nature of the musclar connections between the primary divisions of the
vertebrate heart. J Anat Physiol 41: 172-189, 1907.
KIRBY ML: Cardiac Development. Oxford University Press, New York, 2007, pp 273.
KOMURO H, SAKAI T, HIROTA A, KAMINO K: Conduction pattern of excitation in the amphibian atrium assessed
by multiple-site optical recording of action potentials. Jpn J Physiol 36: 123-137, 1986.
KOPRLA EC: The ultrastructure of alligator conductive tissue: an electron microscopic study of the sino-atrial node.
Acta Physiol Hung 69: 71-84, 1987.
MEILHAC SM, KELLY RG, ROCANCOURT D, ELOY-TRINQUET S, NICOLAS JF, BUCKINGHAM ME:
A retrospective clonal analysis of the myocardium reveals two phases of clonal growth in the developing
mouse heart. Development 130: 3877-3889, 2003.
MIQUEROL L, MEYSEN S, MANGONI M, BOIS P, VAN RIJEN HV, ABRAN P, JONGSMA H, NARGEOT J,
GROS D: Architectural and functional asymmetry of the His-Purkinje system of the murine heart. Cardiovasc
Res 63: 77-86, 2004.
MIQUEROL L, MORENO-RASCON N, BEYER S, DUPAYS L, MEILHAC SM, BUCKINGHAM ME, FRANCO D,
KELLY RG: Biphasic development of the mammalian ventricular conduction system. Circ Res 107: 153-161,
2010.
MOMMERSTEEG MT, HOOGAARS WM, PRALL OW, DE GIER-DE VRIES C, WIESE C, CLOUT DE,
PAPAIOANNOU VE, BROWN NA, HARVEY RP, MOORMAN AF, CHRISTOFFELS VM: Molecular
pathway for the localized formation of the sinoatrial node. Circ Res 100: 354-362, 2007.
OOSTHOEK PW, VIRAGH S, LAMERS WH, MOORMAN AF: Immunohistochemical delineation of the conduction
system. II: The atrioventricular node and Purkinje fibers. Circ Res 73: 482-491, 1993.
PALLANTE BA, GIOVANNONE S, FANG-YU L, ZHANG J, LIU N, KANG G, DUN W, BOYDEN PA, FISHMAN
GI: Contactin-2 expression in the cardiac Purkinje fiber network. Circ Arrhythm Electrophysiol 3: 186-194,
2010.
RADDATZ E: Response of the embryonic heart to hypoxia and reoxygenation: an in vitro model. Exp Clin Cardiol 2:
128-134, 1997.
RENTSCHLER S, YEN AH, LU J, PETRENKO NB, LU MM, MANDERFIELD LJ, PATEL VV, FISHMAN GI,
EPSTEIN JA: Myocardial Notch signaling reprograms cardiomyocytes to a conduction-like phenotype.
Circulation 126: 1058-1066, 2012.
ROBB JS: Comparative Basic Cardiology. Grune & Stratton, New York, 1965, pp 602.
RYU S, YAMAMOTO S, ANDERSEN CR, NAKAZAWA K, MIYAKE F, JAMES TN: Intramural Purkinje cell
network of sheep ventricles as the terminal pathway of conduction system. Anat Rec (Hoboken) 292: 12-22,
2009.
S18 Sedmera and Gourdie Vol. 63
SEDMERA D, RECKOVA M, DEALMEIDA A, SEDMEROVA M, BIERMANN M, VOLEJNIK J, SARRE A,
RADDATZ E, MCCARTHY RA, GOURDIE RG, THOMPSON RP: Functional and morphological evidence
for a ventricular conduction system in the zebrafish and Xenopus heart. Am J Physiol Heart Circ Physiol 284:
H1152-H1160, 2003.
SEDMERA D, RECKOVA M, BIGELOW MR, DEALMEIDA A, STANLEY CP, MIKAWA T, GOURDIE RG,
THOMPSON RP: Developmental transitions in electrical activation patterns in chick embryonic heart. Anat
Rec A Discov Mol Cell Evol Biol 280: 1001-1009, 2004.
SEDMERA D, WESSELS A, TRUSK TC, THOMPSON RP, HEWETT KW, GOURDIE RG: Changes in activation
sequence of embryonic chick atria correlate with developing myocardial architecture. Am J Physiol Heart Circ
Physiol 291: H1646-H1652, 2006.
SUMA K: Sunao Tawara: a father of modern cardiology. Pacing Clin Electrophysiol 24: 88-96, 2001.
TAKEBAYASHI-SUZUKI K, YANAGISAWA M, GOURDIE RG, KANZAWA N, MIKAWA T: In vivo induction of
cardiac Purkinje fiber differentiation by coexpression of preproendothelin-1 and endothelin converting
enzyme-1. Development 127: 3523-3532, 2000.
TAWARA S: Das Reizleitungssystem des Säugetierherzens: eine anatomisch-pathologische Studie über das
Atrioventrikularbündel und die Purkinjeschen Fäden. Verlag von Gustav Fischer, Jena, 1906, pp 252.
THOMPSON RP, LINDROTH JR, ALLEN AJ, FAZEL AR: Cell differentiation birthdates in the embryonic rat heart.
Ann NY Acad Sci 588: 446-448, 1990.
VICENTE-STEIJN R, PASSIER R, WISSE LJ, SCHALIJ MJ, POELMANN RE, GITTENBERGER-DE GROOT AC,
JONGBLOED MR: Funny current channel HCN4 delineates the developing cardiac conduction system in
chicken heart. Heart Rhythm 8: 1254-1263, 2011.
VUILLEMIN M, PEXIEDER T, WONG YM, THOMPSON RP: A two-step alignment method for 3D computer-aided
reconstruction based on fiducial markers and applied to mouse embryonic hearts. Eur J Morphol 30: 181-193,
1992.
... The LSI values at which VT was terminated in three patients were 4.6, 4.6, and 4.7. The number of RF applications was 12 (6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16)(17)(18)(19)(20)(21)(22). In three of the four patients, linear ablation was performed at 3 mm (2-7 mm) above the ventricular earliest activation site (EAS) along the recorded tags of P1 and P2, and the number of RF applications was 4 (4-7) and the distance from the EAS or the good pace map site was 10 (10-14) mm (Figs. 3, 4, 5 and 6). ...
... Studies have reported that the P2 recorded in the left ventricular endocardial surface, which recorded sharp potential by mapping catheters, is not critical for the VT circuit [13,14]. The circuit of verapamil-sensitive ventricular LPF-VT may be the left ventricular myocardium or Purkinje fibers within the left ventricular myocardium [13][14][15][16]. Echocardiography performed the day after RFA showed no change in the echo density in the left ventricular inferior to midapical septal area. ...
Article
Full-text available
In verapamil-sensitive left posterior fascicular ventricular tachycardia (LPF-VT), radiofrequency catheter ablation (RFA) is performed targeting mid-to-late diastolic potential (P1) and presystolic potential (P2) during tachycardia. This study included four patients who had undergone electrophysiological study (EPS) and pediatric patients with verapamil-sensitive LPF-VT who had undergone RFA using high-density three-dimensional (3D) mapping. The included patients were 11–14 years old. During EPS, right bundle branch block and superior configuration VT were induced in all patients. VT mapping was performed via the transseptal approach. P1 and P2 during VT were recorded in three of the four patients. All patients initially underwent RFA via the transseptal approach. In three patients, P1 during VT was targeted, and VT was terminated. The lesion size indices in which VT was terminated were 4.6, 4.6, and 4.7. For one patient whose P1 could not be recorded, linear ablation was performed perpendicularly in the area where P2 was recorded during VT. Among the three patients in whom VT was terminated, linear ablation was performed in two to eliminate the ventricular echo beats. In all patients, VT became uninducible in the acute phase and had not recurred 8–24 months after RFA. High-density 3D mapping with an HD Grid Mapping Catheter allows recording of P1 and P2 during VT and may improve the success rate of RFA in pediatric patients with verapamil-sensitive LPF-VT.
... Only a slight slurring, rather than a broad notch, was observed in leads I, aVL, V5 or V6, and the QRS duration of the bundle branch pattern was on average 86.6 ± 12.1 ms, instead of 135-150 ms, as proposed by Strauss et al. and the current societies' guidelines (Strauss and Selvester, 2009;McDonagh et al., 2021;Glikson et al., 2022). This may be due to the specific anatomy of the Purkinje fiber system, which is distributed subendocardially in humans but transmurally in pigs (Sedmera and Gourdie, 2014;Elbrønd et al., 2023). It therefore must be taken into account that the hemodynamic consequences of LBBB and its treatment by CSP may differ from the situation in the human heart. ...
Article
Full-text available
Background Electrocardiographic (ECG) features of left bundle branch (LBB) block (LBBB) can be observed in up to 20%–30% of patients suffering from heart failure with reduced ejection fraction. However, predicting which LBBB patients will benefit from cardiac resynchronization therapy (CRT) or conduction system pacing remains challenging. This study aimed to establish a translational model of LBBB to enhance our understanding of its pathophysiology and improve therapeutic approaches. Methods Fourteen male pigs underwent radiofrequency catheter ablation of the proximal LBB under fluoroscopy and ECG guidance. Comprehensive clinical assessments (12-lead ECG, bloodsampling, echocardiography, electroanatomical mapping) were conducted before LBBB induction, after 7, and 21 days. Three pigs received CRT pacemakers 7 days after LBB ablation to assess resynchronization feasibility. Results Following proximal LBB ablation, ECGs displayed characteristic LBBB features, including QRS widening, slurring in left lateral leads, and QRS axis changes. QRS duration increased from 64.2 ± 4.2 ms to 86.6 ± 12.1 ms, and R wave peak time in V6 extended from 21.3 ± 3.6 ms to 45.7 ± 12.6 ms. Echocardiography confirmed cardiac electromechanical dyssynchrony, with septal flash appearance, prolonged septal-to-posterior-wall motion delay, and extended ventricular electromechanical delays. Electroanatomical mapping revealed a left ventricular breakthrough site shift and significantly prolonged left ventricular activation times. RF-induced LBBB persisted for 3 weeks. CRT reduced QRS duration to 75.9 ± 8.6 ms, demonstrating successful resynchronization. Conclusion This porcine model accurately replicates the electrical and electromechanical characteristics of LBBB observed in patients. It provides a practical, cost-effective, and reproducible platform to investigate molecular and translational aspects of cardiac electromechanical dyssynchrony in a controlled and clinically relevant setting.
... The electrical signals from the sinoatrial node propagate to the atrioventricular node to cause the ventricles to pump blood, via a network of Purkinje fibers (Sedmera & Gourdie, 2014;Romero et al, 2016). Analyses have shown that a helical continuum of fibers, together with their associated transmural turning from the outer wall to the inner wall, can explain the point-to-point time to arrival conduction wave propagation maps in the heart wall, under a model of anisotropic diffusion (Young & Panfilov, 2010). ...
Article
Full-text available
Coordinated cardiomyocyte contraction drives the mammalian heart to beat and circulate blood. No consensus model of cardiomyocyte geometrical arrangement exists, due to the limited spatial resolution of whole heart imaging methods and the piecemeal nature of studies based on histological sections. By combining microscopy and computer vision, we produced the first-ever three-dimensional cardiomyocyte orientation reconstruction across mouse ventricular walls at the micrometer scale, representing a gain of three orders of magnitude in spatial resolution. We recovered a cardiomyocyte arrangement aligned to the long-axis direction of the outer ventricular walls. This cellular network lies in a thin shell and forms a continuum with longitudinally arranged cardiomyocytes in the inner walls, with a complex geometry at the apex. Our reconstruction methods can be applied at fine spatial scales to further understanding of heart wall electrical function and mechanics, and set the stage for the study of micron-scale fiber remodeling in heart disease.
... In a healthy heart, depolarization [10] during each heartbeat occurs in a precise sequence starting at the sinoatrial node and passing through the atria, atrioventricular node, bundle of His fibers, and purkinje [11]. , eventually spreading into the ventricles in a descending and contralateral direction. ...
Article
Full-text available
Cardiovascular ailment (CVD)is a main reason of morbidity and mortality worldwide. WHO reviews that there are almost 1 crore 20 Lakhs of deaths due to coronary heart illnesses. Early detection and correct prognosis of CVDs are important for powerful remedy and prevention. Monitoring the affected person for twenty-four hrs isn't continually feasible as it calls for plenty of understanding and time. Heart disorder remedy or prognosis are very complicated, specifically in growing countries or poor nations. The clinical enterprise has a big quantity of statistics and is constantly utilized by researchers to increase advancing technology and generation to limit sizeable quantity of deaths because of coronary heart sicknesses. A good buy of statistics mining and ML strategies or algorithms are to be had to fetch the statistics from databases and use this fetched statistics to expect the coronary heart sicknesses very accurately. This paper offers a comparative study of those 3 deep learning architectures for arrhythmia detection using electrocardiogram (ECG) signals. The overall performance of the models become evaluated using standard metrics together with accuracy, sensitivity, and specificity.
... Cx43 is an important component of gap-junction channels located at the intercalated discs where it mediates cardiomyocyte contraction by regulating intercellular ion propagation in cardiac muscle. [38][39][40] Interestingly, Cx43 redistribution to the lateral membrane of cardiomyocytes has been shown to contribute to altered cardiac electrical conduction, arrhythmias, and reduced contractile function of the heart. 41 Cx43 remodeling has also been associated with many cardiac diseases including Duchenne muscular dystrophy-related arrhythmogenesis, 42,43 hypertrophic cardiomyopathy, and heart failure. ...
Article
Full-text available
Background Integrin α7β1 is a major laminin receptor in skeletal and cardiac muscle. In skeletal muscle, integrin α7β1 plays an important role during muscle development and has been described as an important modifier of skeletal muscle diseases. The integrin α7β1 is also highly expressed in the heart, but its precise role in cardiac function is unknown. Mutations in the integrin α7 gene ( ITGA7 ) have been reported in children with congenital myopathy. Methods and Results In this study, we described skeletal and cardiac muscle pathology in Itga7 −/− mice and 5 patients from 2 unrelated families with ITGA7 mutations. Proband in family 1 presented a homozygous c.806_818del [p.S269fs] variant, and proband in family 2 was identified with 2 intron variants in the ITGA7 gene. The complete absence of the integrin α7 protein in muscle supports the ITGA7 mutations are pathogenic. We performed electrocardiography, echocardiography, or cardiac magnetic resonance imaging, and histological biopsy analyses in patients with ITGA7 deficiency and Itga7 −/− mice. The patients exhibited cardiac dysrhythmia and dysfunction from the third decade of life and late‐onset respiratory insufficiency, but with relatively mild limb muscle involvement. Mice demonstrated corresponding abnormalities in cardiac conduction and contraction as well as diaphragm muscle fibrosis. Conclusions Our data suggest that loss of integrin α7 causes a novel form of adult‐onset cardiac dysfunction indicating a critical role for the integrin α7β1 in normal cardiac function and highlights the need for long‐term cardiac monitoring in patients with ITGA7 ‐related congenital myopathy.
Article
Of the monomorphic ventricular tachycardias, there are 4 specific tachycardias related to the Purkinje system: 1) idiopathic verapamil-sensitive fascicular ventricular tachycardia (FVT); 2) non-re-entrant FVT; 3) bundle branch re-entry and interfascicular re-entry; and 4) Purkinje-mediated VT in structural heart disease. Verapamil-sensitive FVT is classified into 4 types according to the location of the circuit: 1) left posterior type; 2) left anterior type; 3) left upper septal type;and 4) reverse type. And, in the left anterior and posterior types, there are septal and papillary muscle subtypes. Although macro-re-entry has been reported to be the mechanism underlying verapamil-sensitive FVT, recording the entire circuit is challenging. One possible reason is that the Purkinje-muscle junction may penetrate the myocardial layer as a part of the circuit. The Purkinje network may thus play an important role in the initiation and maintenance of ventricular fibrillation. Further, it has been reported that the development and the abnormalities of the Purkinje system are associated with the arrhythmogenesis of ventricular fibrillation. Furthermore, it has been reported that catheter ablation of trigger ventricular premature complexes, and/or "de-networking" of the Purkinje system, can be used as electrical bailout therapy. There is a hypothesis that the intramural Purkinje system is involved in the generation of J waves. Nevertheless, as there are still unresolved issues that must be debated and accurately analyzed, this review aims to discuss the solved and unsolved questions related to Purkinje-related arrhythmias.
Article
Unlabelled: The Purkinje fibers convey the electrical impulses at much higher speed than the working myocardial cells. Thus, the distribution of the Purkinje network is of paramount importance for the timing and coordination of ventricular activation. The Purkinje fibers are found in the subendocardium of all species of mammals, but some mammals also possess an intramural Purkinje fiber network that provides for relatively instantaneous, burst-like activation of the entire ventricular wall, and gives rise to an rS configuration in lead II of the ECG. Aim: To relate the topography of the horse heart and the distribution and histology of the conduction system to the pattern of ventricular activation as a mechanism for the unique electrical axis of the equine heart. Methods: The morphology and distribution of the cardiac conduction system was determined by histochemistry. The electrical activity was measured using ECG in the Einthoven and orthogonal configuration. Results: The long axis of the equine heart is close to vertical. Outside the nodal regions the conduction system consisted of Purkinje fibers connected by connexin 43 and long, slender parallel running transitional cells. The Purkinje fiber network extended deep into the ventricular walls. ECGs recorded in an orthogonal configuration revealed a mean electrical axis pointing in a cranial-to-left direction indicating ventricular activation in an apex-to-base direction. Conclusion: The direction of the mean electrical axis in the equine heart is determined by the architecture of the intramural Purkinje network, rather than being a reflection of ventricular mass.
Article
Full-text available
The inaugural Kjell Johansen lecture in the Zoophysiology Department of Aarhus University (Aarhus, Denmark), afforded the opportunity for a focused workshop comprising comparative cardiovascular physiologists to ponder some of the key unanswered questions in the field. Discussions were centered around three themes. The first considered function of the vertebrate heart in its various forms in extant vertebrates, with particular focus on the role of intracardiac shunts, the trabecular ("spongy") nature of the ventricle in many vertebrates, coronary blood supply and the building plan of the heart as revealed by molecular approaches. The second theme involved the key unanswered questions in the control of the cardiovascular system, emphasizing autonomic control, hypoxic vasoconstriction and developmental plasticity in cardiovascular control. The final theme involved poorly understood aspects of the interaction of the cardiovascular system with the lymphatic, renal and digestive systems. Having posed key questions around these three themes, it is increasingly clear that an abundance of new analytical tools and approaches will allow us to learn much about vertebrate cardiovascular systems in the coming years. This article is protected by copyright. All rights reserved.
Article
Full-text available
Setting the Pace The heart beats rhythmically throughout life. Highly specialized cardiac pacemaker cells control the timing of this beating. Bressan et al. (p. 744 , published online 21 March) identified the embryonic location of the pacemaker precursors in early avian development and traced the cells throughout their incorporation into the heart. The events that establish the pacemaker lineage occur prior to the initiation of heart formation, and are governed, at least in part, by a class of Wnt signaling molecules.
Article
Full-text available
The endothermic state of mammals and birds requires high heart rates to accommodate the high rates of oxygen consumption. These high heart rates are driven by very similar conduction systems consisting of an atrioventricular node that slows the electrical impulse and a His-Purkinje system that efficiently activates the ventricular chambers. While ectothermic vertebrates have similar contraction patterns, they do not possess anatomical evidence for a conduction system. This lack amongst extant ectotherms is surprising because mammals and birds evolved independently from reptile-like ancestors. Using conserved genetic markers, we found that the conduction system design of lizard (Anolis carolinensis and A. sagrei), frog (Xenopus laevis) and zebrafish (Danio rerio) adults is strikingly similar to that of embryos of mammals (mouse Mus musculus, and man) and chicken (Gallus gallus). Thus, in ectothermic adults, the slow conducting atrioventricular canal muscle is present, no fibrous insulating plane is formed, and the spongy ventricle serves the dual purpose of conduction and contraction. Optical mapping showed base-to-apex activation of the ventricles of the ectothermic animals, similar to the activation pattern of mammalian and avian embryonic ventricles and to the His-Purkinje systems of the formed hearts. Mammalian and avian ventricles uniquely develop thick compact walls and septum and, hence, form a discrete ventricular conduction system from the embryonic spongy ventricle. Our study uncovers the evolutionary building plan of heart and indicates that the building blocks of the conduction system of adult ectothermic vertebrates and embryos of endotherms are similar.
Article
The mechanisms responsible for atrioventricular (AV) delay remain unclear, in part due to the inability to map electrical activity by conventional microelectrode techniques. In this study, voltage-sensitive dyes and imaging techniques were refined to detect action potentials (APs) from the small cells comprising the AV node and to map activation from the "compact" node. Optical APs (124) were recorded from 5 x 5 mm (∼0.5-mm depth) AV zones of perfused rabbit hearts stained with a voltage-sensitive dye. Signals from the node exhibited a set of three spikes; the first and third (peaks I and III) were coincident with atrial (A) and ventricular (V) electrograms, respectively. The second spike (peak II) represented the firing of midnodal (N) and/or lower nodal (NH) cell APs as indicated by their small amplitude, propagation pattern, location determined from superimposition of activation maps and histological sections of the node region, dependence on depth of focus, and insensitivity to tetrodotoxin (TTX). AV delays consisted of τ1 (49.5 ± 6.59 ms, 300-ms cycle length), the interval between peaks I and II (perhaps AN to N cells), and τ2 (57.57 ± 5.15 ms), the interval between peaks II and III (N to V cells). The conductance time across the node was 10.33 ± 3.21 ms, indicating an apparent conduction velocity (ΘN) of 0.162 ± 0.02 m/s (n = 9) that was insensitive to TTX. In contrast, τ1 correlated with changes in AV node delays (measured with surface electrodes) caused by changes in heart rate or perfusion with acetylcholine. The data provide the first maps of activation across the AV node and demonstrate that ΘN is faster than previously presumed. These findings are inconsistent with theories of decremental conduction and prove the existence of a conduction barrier between the atrium and the AV node that is an important determinant of AV node delay.
Article
This chapter focuses on the cellular origin of the conduction system components and the molecular genetic mechanisms that may control their phenotype and position within the developing heart. It discusses the connection between heart cell precursor pools, which in a temporal pattern form the heart, and the genesis of the conduction system components. The distinct components of the cardiac conduction system of the heart are essentially myocardial. They are innervated by cardiac ganglia largely derived from neural crest. In addition, a large fraction of cells in the mature conduction system is noncardiac, and insulating layers of fibrous tissue are found around conduction system components, such as the SAN and AV bundle. These noncardiac cell types are derived from the epicardium (fibroblasts), endocardium, neural crest (neural innervations), and other sources, although their origins have not been defined in detail. Although these nerves and fibrous tissues are important, or even a prerequisite, for conduction system formation and function, the cardiomyocytes are essential for impulse generation and propagation. Furthermore, in the embryo the functional myocardial conduction system is not yet innervated and interstitial fibroblast and fibrous tissues in association with the conduction system are sparse or absent. Studies of the development of the conduction system components strongly suggest that they originate from myocardial precursors, which in turn are derived from mesoderm and pericardial wall mesenchyme.
Article
Notch signaling has previously been shown to play an essential role in regulating cell fate decisions and differentiation during cardiogenesis in many systems including Drosophila, Xenopus, and mammals. We hypothesized that Notch may also be involved in directing the progressive lineage restriction of cardiomyocytes into specialized conduction cells. In hearts where Notch signaling is activated within the myocardium from early development onward, Notch promotes a conduction-like phenotype based on ectopic expression of conduction system-specific genes and cell autonomous changes in electrophysiology. With the use of an in vitro assay to activate Notch in newborn cardiomyocytes, we observed global changes in the transcriptome, and in action potential characteristics, consistent with reprogramming to a conduction-like phenotype. Notch can instruct the differentiation of chamber cardiac progenitors into specialized conduction-like cells. Plasticity remains in late-stage cardiomyocytes, which has potential implications for engineering of specialized cardiovascular tissues.
Chapter
The heartbeat is initiated and coordinated by a multi-component set of specialized muscle tissues collectively referred to as the pacemaking and conduction system. Over the last few years, impetus has gathered into unravelling the cellular and molecular processes that regulate differentiation and integration of this essential cardiac network. One focus of our collective work has been the developmental history of cells comprising His–Purkinje tissues of the conduction system. This interest in part arose from studies of the expression of connexins in periarterial Purkinje fibres of the chick heart. Using lineage-tracing strategies, including those based on replication-defective retroviruses and adenoviruses, it has been shown that conduction cells are derived from multipotent, cardiomyogenic progenitors in the tubular heart. Moreover, heterogeneity within myocardial clones has indicated that the elaboration of the conduction system in the chick embryo occurs by progressive, localized recruitment from within this pool of cardiomyogenic cells. Cell birth dating has revealed that inductive conscription of cells to central elements of the conduction system (e.g. the His bundle) precedes recruitment to the peripheral components of the network (i.e. subendocardial and periarterial Purkinje fibres). Birth dating studies in rodents suggest an analogous recruitment process is occurring in this species. In addition to summarizing earlier work, this chapter provides information on ongoing studies of cell–cell signalling and transcriptional mechanisms that may regulate the development of His–Purkinje tissues.
Article
Background: Purkinje cells (PCs) comprise the most distal component of the cardiac conduction system, and their unique electrophysiological properties and the anatomic complexity of the Purkinje fiber network may account for the prominent role these cells play in the genesis of various arrhythmic syndromes. Methods and results: Differential transcriptional profiling of murine Purkinje fibers and working ventricular myocytes was performed to identify novel genes expressed in PCs. The most highly enriched transcript in Purkinje fibers encoded Contactin-2 (Cntn2), a cell adhesion molecule critical for neuronal patterning and ion channel clustering. Endogenous expression of Cntn2 in the murine ventricle was restricted to a subendocardial network of myocytes that also express beta-galactosidase in CCS-lacZ transgenic mice and the connexin40 gap junction protein. Both Cntn2-lacZ knockin mice and Cntn2-EGFP BAC transgenic reporter mice confirmed expression of Cntn2 in the Purkinje fiber network, as did immunohistochemical staining of single canine Purkinje fibers. Whole-cell patch-clamp recordings and measurements of Ca(2+) transients in Cntn2-EGFP(+) cells revealed electrophysiological properties indicative of PCs and distinctive from those of cardiac myocytes, including prolonged action potentials and frequent afterdepolarizations. Conclusions: Cntn2 is a novel marker of the specialized cardiac conduction system. Endogenous expression of Cntn2 as well as Cntn2-dependent transcriptional reporters provides a new tool through which Purkinje cell biology and pathophysiology can now more readily be deciphered. Expression of a contactin family member within the CCS may provide a mechanistic basis for patterning of the conduction system network and the organization of ion channels within Purkinje cells.