Sie sind auf Seite 1von 17

6.

04 Cardiac Physiology and Pharmacology


R J Sommer, Bates College, Lewiston, ME, USA
2010 Elsevier Ltd. All rights reserved.

6.04.1 Introduction 51
6.04.2 Myocardial Excitation and Contraction 51
6.04.2.1 Generation and Spread of Excitation 51
6.04.2.2 Ionic Basis of Cardiac Action Potentials 53
6.04.2.3 ExcitationContraction Coupling 54
6.04.2.4 Relaxation of Contraction 55
6.04.3 Autonomic Regulation of Cardiac Excitation and Contraction 56
6.04.3.1 Sympathetic Modulation of the Heartbeat 56
6.04.3.2 Parasympathetic Modulation of the Heartbeat 57
6.04.4 Compartmentation 58
6.04.5 Classification and Mechanisms of Arrhythmias 59
6.04.5.1 Abnormal Impulse Generation 59
6.04.5.2 Abnormal Impulse Conduction 62
6.04.5.3 Genetic Basis of Arrhythmias 62
6.04.6 Classification and Mechanisms of Action of Antiarrhythmic Agents 63
6.04.6.1 Conventional Antiarrhythmic Agents 64
6.04.6.1.1 Class I and III antiarrhythmic agents 64
6.04.6.1.2 Class II antiarrhythmic agents 64
6.04.6.1.3 Class IV antiarrhythmic agents 64
6.04.6.2 Novel Antiarrhythmic Agents and Approaches 64
6.04.6.2.1 Atrial-selective agents 64
6.04.6.2.2 Gap junction modulators 64
6.04.6.2.3 Preventing heart failure and arrhythmias 65
6.04.6.2.4 Future directions 66
References 66

6.04.1 Introduction content of the first edition of this chapter, written by


Sheu and Colecraft (1997), has been retained; how-
Under normal circumstances, the hearts rate and force ever, significant revisions and additions have been
of contraction is tightly regulated to meet the circula- made to include dramatic advancements in under-
tory demands of the body. The sympathetic and standing the molecular biology of ion channels, the
parasympathetic nervous systems are primary regula- autonomic receptor signaling pathways, and the
tors of cardiac output but a number of integrated genetic basis of some forms of arrhythmias.
systems of the body including the renal system, adre-
nal glands, vascular endothelium, immune system, and
cardiac tissue itself play important roles in regulating 6.04.2 Myocardial Excitation and
circulation and hydroelectrolytic homeostasis. This Contraction
chapter describes the cellular and molecular basis of
myocardial excitationcontraction coupling, explains 6.04.2.1 Generation and Spread of
how the autonomic nervous system regulates excita- Excitation
tioncontraction coupling, and examines emerging The ability of the heart to contract is intrinsic. Unlike
information on the mechanisms of arrhythmias and neurons and skeletal muscle fibers, cardiac muscle
the new pharmacologic approaches being taken to does not depend upon nervous system stimulation to
treat arrhythmias and chronic heart failure. Much of initiate an action potential (AP). Instead, autorhyth-
the organizational structure and some informational mic cells of the intrinsic cardiac conduction system

51
52 Introduction to Cardiovascular Biology

generate and distribute impulses throughout the heart detailed review of the SA node, the interested reader
in a coordinated fashion from the atria to the ventri- is referred to Dobrzynski et al. (2007).
cles. In a healthy heart, the sinoatrial (SA) node, Once an AP has been initiated in the SA node, gap
located at the junction between the superior vena junction channels allow ions to pass rapidly to adjacent
cava and the right atrium, sets the pace for the heart, cells, directly transmitting the current through both
generating an AP approximately 75 times every the conducting cells of the internodal pathway and the
minute. However, in the absence of extrinsic neural contractile cells of the left and right atria. Atria and
and hormonal factors the SA node generates approxi- ventricles abut one another but are not connected by
mately 100 APs per minute. The pacemaker cells of gap junctions. Thus, the atrioventricular (AV) node,
the SA node are not homogenous in terms of their rate located immediately above the tricuspid valve, is the
of AP generation and the exact position of the leading only electrical connection between the upper and
pacemaking site within the node shifts depending on the lower chambers (Figure 1). Within the AV node,
conditions such as autonomic nerve stimulation the rate of impulse propagation is slowed, at least in
(Boyett et al. 2000; Schuessler et al. 1996). For a part, due to differential expression of the number of

0
3
Superior SA node 4
vena cava

Atrium

AV node
Overshoot
1
0 2

3
mV
Phase 0
Tricuspid Purkinje 4
valve
100 Resting potential
Mitral
valve

Ventricle

R
Action potential phases
0: Upstroke
1: Early-fast repolarization
2: Plateau T
3: Repolarization
4: Diastole ECG P

Q S
PR QT
200 ms
Figure 1 Schematic diagram of cardiac action potentials (APs) in different regions of the heart. The cumulative electrical
activity of the heart is manifested on the body surface as the ECG. The P wave is generated by atrial depolarization, the QRS
by ventricular muscle depolarization, and the T wave by ventricular repolarization. Action potential phases: 0, upstroke; 1,
early fast repolarization; 2, plateau; 3, repolarization; 4, diastole resting potential. (Mason, J. W.; Hondeghem, L. M. Ann.
N.Y. Acad. Sci. 1984, 432, 162176.)
Cardiac Physiology and Pharmacology 53

gap junction channels, their cellular location, and the membrane potential of atrial and ventricular myo-
specific type of connexin proteins that form the gap cytes and Purkinje fibers range from 70 to 95 mV.
junctions (Desplantez et al. 2007). This delay allows SA node and AV node cells have resting membrane
time for ventricular filling. From the AV node, cardiac potentials of 50 to 60 mV. Resting potentials of
APs are rapidly conducted through the AV bundle SA and AV node cells are not stable but exhibit
(Bundle of His) to the right and left bundle branches spontaneous, slow depolarization that, upon reaching
to a rapidly conducting network of Purkinje fibers that a threshold voltage, trigger an influx of Ca2, further
supply the ventricular myocardium (Figure 1). Again, depolarizing the cell. This depolarization is the fir-
the excitation is transferred from the Purkinje network ing of an AP. In myocytes, as opposed to node cells,
to the myocardial cells by means of gap junctions the upstroke (phase 0) of an AP results from a rapid
between the fiber terminals and the ventricular myo- influx of Na (Figure 2).
cytes, resulting in the rapid and nearly simultaneous Immediately following this influx of positive
activation of the two ventricles. ions, the cell begins to return to its resting voltage
The cumulative electrical activity of the heart can (repolarization) by moving K out of the cell, which
be recorded by electrodes placed on the body surface creates multiple time-dependent K currents
generating an electrocardiogram (ECG). An ECG is a (Figure 2). Transient-outward (Ito) and ultra-rapid
composite of all the APs generated by the cells of the (IKur) K currents are primarily responsible for
cardiac conduction system and the contracting myo- rapid early repolarization (phase 1) of the AP.
cytes at a given time. Note that the APs from differing During phase 2, Ca2 enters the myocyte through
cardiac cell types have different characteristic shapes, voltage-gated L-type Ca2 channels (CaV1.2) and
which when combined form a series of three deflection initiates mechanical contraction of the myocyte. At
waves per heartbeat recorded on the ECG (Figure 1). this time, inward Ca2 (ICaL) and outward K cur-
Primarily, the P wave is generated by depolarization of rents are relatively balanced to form the AP plateau
the atria while the QRS complex results from depolar- (phase 2). Slower rectifying currents (IKr and IKs),
ization of the ventricles. The T wave coincides with created by the movement of K outward by hERG
ventricular repolarization. The QT interval spans the (KV11.1) and KVLQT1 (KV7.1) K channels, med-
period from the beginning of ventricular depolarization iate terminal repolarization (phase 3) and bring the
to the end of repolarization, and thus reflects the dura- cell back to resting potential (phase 4). The resting
tion of ventricular APs. It is important to consider that a potential is maintained in myocardial cells by IK1
single type of abnormality detected by an ECG, such as and IKACh currents until an influx of ions from a
a long QT interval, could be the manifestation of one of neighboring cell triggers the next AP.
several diverse causes such as overstimulation of the No AP trigger is needed in pacemaker cells
sympathetic nervous system, loss of function mutation because the pacemaker current, If, spontaneously
in hERG K channels (KV11.1), or gain of function depolarizes node cells during phase 4. When first
mutation in Na channels (NaV1.5). discovered, If was labeled the funny current for a
number of its unusual features. If is activated at
hyperpolarized voltages (40 to 50 mV), has
6.04.2.2 Ionic Basis of Cardiac Action
mixed permeability to Na and K, and is activated
Potentials
by cAMP (Accili et al. 2002; DiFrancesco and Borer
Differential concentrations of ions (primarily Na, 2007). A relatively new family of ion channels,
K, Ca2, and Cl) inside versus outside of cardiac the hyperpolarization-activated cyclic nucleotide-
cells result in electrochemical gradients across the gated (HCN) channels, carries the If current and
cell membrane that are necessary for normal cardiac have become novel targets for the development of
function. At rest, the Na, Ca2 exchanger (NCX) pure heart rate-lowering drugs such as Ivabradine
and Na, K ATPase pump maintain higher extra- (Tardif 2007).
cellular concentrations of Na and Ca2 and a higher Cardiac cells are at least partially protected from
intracellular concentration of K. Cardiac cells are early reexcitation because they cannot undergo
impermeable to Na and Ca2, but are permeable to depolarization during a refractory period that exists
K. As a result, positively charged K ions flow for most of the time from initial depolarization to
slowly out of the cell (current designated as IK1), final repolarization (termed the AP duration, APD)
and keep the interior of cardiac cells negatively (Figure 2). However, this also means that any nor-
charged relative to the exterior. The resting mal change in heart rate requires a change in APD.
54 Introduction to Cardiovascular Biology

Early Ito
Plateau (2)
repolarization (1)
ICaL
IKur
IKs
0
IKr

Final

mV
INa Upstroke (0) repolarization (3)
IKACh
IK1
IKACh Pacemaker
IK1 If function
APD
80
Resting (4) 0.2 sec NCX
IK1
INa
Ito
IKur
ICaL
IKr
IKs
NCX
If
IKACh

Figure 2 Ion currents during an action potential (AP) for a typical atrial or ventricular cell. Outward currents are indicated
with an arrow pointing up. Inward currents are indicated with an arrow pointing down. The timing of each current in relation
to the AP is shown. APD, action potential duration; NCX, Na/Ca2 exchanger. (Nattel, S.; Carlsson, L. Nat. Rev. Drug Discov.
2006, 5, 10341049.)

Depolarization is dominated by a single type of ion (cAMP)-dependent protein kinase (PKA), Ca2/
channel (Na in the case of atrial and ventricle cells, calmodulin-dependent protein kinase (CaMKII), pro-
Ca2 in the case of the SA and AV node cells); tein phosphatases 1 and 2A, and A-kinase anchoring
however, plateau and repolarization phases result protein (AKAP) (Bers 2006; Marx et al. 2000).
from complex interactions between many inward Calstabin 2 stabilizes RyR2 in a closed state until
depolarizing and outward repolarizing currents PKA-mediated phosphorylation of RyR2 facilitates
carried by various types of ion channels the dissociation of calstabin 2, increasing SR Ca2
(Figure 2). This multifaceted system of repolariza- release (Brillantes et al. 1994). The elevation in the free
tion provides many points for fine control of APD; intracellular Ca2 concentration is directly responsible
however, it also exposes many targets to perturba- for initiating the interaction of thin actin and thick
tion by disease or drugs. myosin filaments which leads to muscle contraction
(Figure 3). The thin filaments contain two regulatory
proteins, tropomyosin and troponin (Tn), in addition to
6.04.2.3 ExcitationContraction Coupling
actin. There are three Tn proteins each of which has a
2
Ca is the central regulator of excitationcontraction unique function: Tn C binds Ca2, Tn I affects -
coupling. The influx of Ca2 via voltage-dependent tropomyosin so as to prevent the interaction of actin
L-type Ca2 channels during phase 2 of the AP triggers and myosin, and Tn T attaches the other two Tn units
the release of large amounts of Ca2 from the sarco- to tropomyosin. In the absence of Ca2, Tn works
plasmic reticulum (SR). This process is termed Ca2- through tropomyosin to prevent the interaction
induced Ca2-release (CICR) and is mediated by between actin and myosin. The elevated Ca2 concen-
ryanodine receptors (RyR2) of the SR (for review, see tration from CICR results in the diffusion of Ca2 to the
Bers 2002). RyR2s are Ca2 release channels, ligand- myofibrils where it binds to the Tn C subunit of the
activatedby Ca2, and are found within a macromole- Tntropomyosin complex. This causes a conforma-
cular complex that includes calmodulin, calstabin 2 tional change in the Tn molecule that shifts the
(FKBP12.6), cyclic adenosine monophosphate tropomyosin strand, exposing the myosin-binding
Cardiac Physiology and Pharmacology 55

Strain

Plasma membrane Integrin

Ca2+
Troponin C
Troponin T
Troponin I Actin

Thin filament
-Tropomyosin

Myosin-binding -Myosin
protein C heavy chain
Thick filament
PEVK region

Calsarcin 1
T-cap Calcineurin

MLP
Titin
cGMP
PDES -Actinin
PKG
Z-disk

Figure 3 Myofilament and structural proteins of a cardiac sarcomere. Ca2 binds regulatory sites on Tn C, resulting in a
conformational change in Tn I which releases -tropomyosin from its position of blocking actin and myosin binding. Activated
myosin heads strongly attach to actin filaments, forming multiple cross bridge attachments that contract the myofilament by
sliding the actin filament toward the center of the sarcomere. Mechanical stimuli are transduced by clustered membrane
integrins that couple to proteins of the Z-disk (shaded) that bind contractile filaments. cGMP, cyclic GMP; MLP, muscle LIM
protein; PDE5, phosphodiesterase 5; PKG, cGMP-dependent protein kinase; T-cap, titin cap. (Mudd, J. O.; Kass, D. A. Nature
2008, 451, 919928.)

sites on the actin, allowing actin and myosin to inter- most likely mediated by calmodulin binding to the
act. The binding of myosin heads to actin filaments pore-forming region of the L-type Ca2 channel
forms multiple cross bridge attachments that con- (Bodi et al. 2005). Second, a combination of RyR2
tract the myofilament by sliding the actin filament inactivation and partial depletion of SR Ca2 stores
toward the center of the sarcomere (Figure 3). turns off Ca2 release from the SR (Bers 2002). Third,
the Ca2-ATPase pump of the SR (SERCA2)
removes most of the elevated Ca2 from the cyto-
6.04.2.4 Relaxation of Contraction plasm. SERCA2 uses ATP to pump Ca2 back into
Relaxation of myocardial contraction is achieved the SR where it is reversibly buffered by the Ca2-
by multiple processes which serve to reduce the cyto- binding protein, calsequestrin.
solic concentration of Ca2 back to resting levels. SERCA2 activity is regulated by the phosphory-
First, Ca2 influx is mostly terminated by inactivation lation state of the protein phospholamban (PLB), an
of ICaL during the plateau (phase 2) of the AP. L-type SR membrane protein (Simmerman and Jones 1998).
Ca2 channels undergo Ca2-dependent inactivation, Phosphorylation of PLB by any of several kinases
56 Introduction to Cardiovascular Biology

relieves PLBs inhibition of SERCA2, enhancing receptors ( - and -AR) (Hartzell 1998). However, it is
Ca2 sequestration in the SR. Fourth, proteins pre- signaling of -ARs, via one of the most well-known
sent on the sarcolemma serve to transport Ca2 out molecular pathways, that regulates heart rate (chrono-
of the cell. The primary transport system for the tropy), contractility (inotropy), and relaxation
efflux of Ca2 from myocardial cells is the NCX (lusitropy). Initially, signaling of -ARs was thought
which uses the Na gradient to transport three Na to be a simple linear cascade, initiated primarily by 1-
ions into and one Ca2 ion out of the cell (Sheu and AR stimulation, but it is now recognized to be a com-
Blaustein 1992). Finally, there are also the ATP- plex network that utilizes both 1-AR and 2-ARs and
dependent Ca2 pump (Carafoli 1987) and mito- has cross talk with numerous other pathways
chondrial Ca2 uniporter (Gunter et al. 1994; Maack (Saucerman and McCulloch 2006). The classical
and ORourke 2007) that move small amounts of understanding of -AR signaling is that norepinephr-
Ca2 outside of the cell and into the mitochondria, ine, or other -AR agonist, binding leads to -AR
respectively. As the cytosolic concentration of Ca2 activation of the stimulatory G protein, Gs, facilitating
decreases, Ca2 dissociates from Tn C, reestablishing exchange of GTP for GDP and dissociation of the
the tropomyosin blockade of actin and myosin bind- subunit from the subunits. The activated Gs sub-
ing, leading to muscle relaxation and diastole. unit then directly interacts with all isoforms of adenylyl
cyclase (AC) expressed in cardiac tissue (Smit and
Iyengar 1998) to stimulate production of cAMP and
6.04.3 Autonomic Regulation of activation of PKA. PKA-dependent phosphorylation
Cardiac Excitation and Contraction of numerous cardiac proteins, including L-type Ca2
channels (CaV1.2) (Hulme et al. 2003), Na channels
6.04.3.1 Sympathetic Modulation of the
(NaV1.5) (Herfst et al. 2004), RyR2 (Marx et al. 2000),
Heartbeat
PLB (Simmerman and Jones 1998), and Tn 1 (Kentish
Sympathetic innervation is found throughout the heart et al. 2001) increases both contractile and relaxation
and acts via stimulation of both - and -adrenergic responses (Figure 4). To increase heart rate you must

NE, epinephrine, isoproterenol

1-AR 2-AR sarcolemma


AC AC
ICa Gs GRK2 Gs GRK2 Gi PI3K

-arrestin -arrestin
CaMKII cAMP PDE4D cAMP PDE4D ERK Akt

apoptosis PDE PKA PDE PKA apoptosis


CREB
If RyR TnI PLB ICa

ICER
transcription
chronotropy lusitropy inotropy
CaN NFATc hypertrophy nucleus

Figure 4 Functional consequences of cardiac -AR signaling. Historically, cardiac -AR signaling was known to act only
through the 1-AR-AC-cAMP-PKA signaling axis (bold arrows) to acutely regulate heart rate (chronotropy), contractility
(inotropy), and relaxation rate (lusitropy). Recent evidence additionally demonstrates an important role for 2-AR signaling
and long-term cardiac remodeling including regulation of hypertrophy and apoptosis. AC, adenylylcyclase; CaMKII, Ca2/
calmodulin-dependent kinase II; cAMP, cyclic adenosine monophosphate; CaN, calcineurin; CREB, cAMP response
element-binding factor; ICa, Ca2 current; ERK, extracellular-signal-regulated kinase; ICER, inducible cAMP early repressor;
GRK2, G-protein receptor kinase 2; Gi, G inhibitory protein; Gs, G stimulatory protein; NFATc, nuclear factor of activated
T cells; PDE, phosphodiesterase; PDE4D, phosphodiesterase 4D; PI3K, phosphatidyl-inositol 3-OH-kinase; PKA, cAMP-
dependent protein kinase; PLB, phospholamban; RyR, ryanodine receptor; TnI, troponin I. (Saucerman, J. J.; McCulloch,
A. D. Ann. N.Y. Acad. Sci. 2006, 1080, 348361.)
Cardiac Physiology and Pharmacology 57

cycle through contracting the heart and relaxing the 6.04.3.2 Parasympathetic Modulation of
heart faster. The duality of increasing both contraction the Heartbeat
and relaxation is achieved by the diverse effectors of
The stimulatory effects of the sympathetic nervous
the -AR signaling pathway. Phosphorylation of L-
system are generally opposed by the parasympa-
type Ca2 channels increases the inward flow of Ca2
thetic nervous system. The parasympathetic left
(ICaL) and phosphorylation of PLB enhances SR Ca2
and right vagus nerves terminate in intracardiac
stores, so more Ca2 is delivered to the myofilaments
ganglia that go onto richly innervate atrial myocar-
during subsequent APs. Phosphorylation of PLB and dium, SA and AV nodes, and the ventricular
Tn I increases the rates of SR Ca2 uptake and dis- conduction system (Randall and Wurster 1993).
sociation of Ca2 from Tn C, respectively, enhancing Although less dense, there is significant parasympa-
the rate of relaxation. thetic innervation of the ventricular myocardium
The classical -AR activation pathway (Gs-AC- (Standish et al. 1994) and muscarinic acetylcholine
cAMP production) can also increase heart rate receptors (mAChR), primarily of the M2 subtype,
independently of phosphorylation (Figure 4). are expressed throughout all areas of the heart,
Increased cAMP levels increase If which increases including the ventricles (Loffelholz and Pappano
pacemaker cells spontaneous depolarization rates 1985). However, following mAChR activation, atrial
(Accili et al. 2002). cAMP directly binds to a cyclic and node cells readily display decreased contractile
nucleotide-binding domain on the cytoplasmic C- and impulse initiation and propagation responses,
terminus of the HCN channel, which carries If. whereas, ventricular myocyte response is evident,
HCN channels have opening kinetics consistent or at least more prominent, only in the presence of
with a model of combined allosteric and voltage- elevated cAMP levels and/or sympathetic ( -AR)
dependent gating (Altomare et al. 2001). This model stimulation (Harvey and Belevych 2003). These
proposes that HCN channels can be in an open or tissue-specific differences are thought to be
closed state, and each of the channels four voltage mediated by different mechanisms that follow one
sensors can be in a reluctant or willing state. The of two general paradigms. The first mechanism, in
probability of the channel being in the open state SA node and atrial cells, involves mAChRs directly
increases each time one voltage sensor switches to coupled to inward rectifying K channels (KACh)
the willing state and cAMP binds preferentially to via PTX-sensitive G proteins (Go/Gi family)
open channels and locks them in an open state (Tamargo et al. 2004). Upon mAChR activation, G
(Altomare et al. 2001; DiFrancesco 1999). protein subunits activate IKACh by directly bind-
In just the last several years, non-classical -AR ing the cytoplasmic N- and C-termini of KACh
signaling pathways that are cAMP/PKA-indepen- (Tamargo et al. 2004). Activation of IKACh hyperpo-
dent have been discovered. These new forms of larizes the membrane potential, slowing the
-AR signaling often cause long-term effects such spontaneous firing rate of the pacemaker cells and
as cardiac hypertrophy and apoptosis (Figure 4) decreasing atrial cell contraction. The second
(Saucerman and McCulloch 2006). One non- mechanism, found throughout the heart, involves
classical signaling pathway is both dependent and indirect regulation of ion channel activity through
independent of PKA. It is Ca2-calmodulin- modulation of cAMP levels (Harvey and Belevych
dependent kinase II (CaMKII) which is activated 2003). mAChR activation decreases cAMP synthesis
by PKA-induced increases in intracellular Ca2 and increases cAMP degradation by direct inhibi-
and can also be activated as a result of Gs acting tion of AC subtypes 5 and 6 (AC5/6) by subunit of
directly on Ca2 channels (Yatani et al. 1987). Go/Gi and increased production of phosphodiester-
CaMKII is an important mediator of cardiac myo- ase (PDE) via production of nitric oxide and cyclic
cyte apoptosis (Zhu et al. 2003) and is an attractive GMP, respectively (Figure 5a) (Harvey and
therapeutic target for heart failure (HF) Belevych 2003).
(Saucerman and McCulloch 2006). Unlike 1- Paradoxically, under a variety of conditions,
ARs, 2-ARs signal via Gs and G inhibitory (Gi) mAChR activation has been reported to increase
proteins and increase extracellular-signal-regu- strength of contraction and/or cause rebound
lated kinase (ERK) phosphorylation, a likely increases in heart rate. One potential explanation is
cause of myocyte hypertrophy (Figure 4) (Baillie that mAChRs differentially regulate different iso-
et al. 2003; Molkentin and Dorn 2001). forms of AC (Harvey and Belevych 2003). It is
58 Introduction to Cardiovascular Biology

AC
(a) s 5/6 i/o M2
i/o
ATP
CM Ca2+
cAMP
PKA 5-AMP NOS3

PDE2 NO L-arginine

sGC

cGMP GTP
ATP PO4

stimulatory
Ca2+ inhibitory

AC AC
(b) 1 s 5/6 i/o M2 4/7 s 1
i/o
ATP ATP
cAMP cAMP
PKA

ATP PO4

stimulatory
Ca2+ inhibitory

Figure 5 Proposed signaling pathways responsible for mAChR (M2) inhibition of cAMP-dependent ion channel responses
(a) and both inhibitory and stimulatory responses through differential regulation of specific AC isoforms (b). (Harvey, R. D.;
Belevych, A. E. Brit. J. Pharmacol. 2003, 139, 10741084.)

known that subunit of Gi inhibits AC5 and AC6 but 6.04.4 Compartmentation
not group 2 ACs (AC2, AC4, and AC7) (Smit and
Iyengar 1998; Sunahara et al. 1996). Furthermore, In recent years, the importance of cellular compart-
AC5 and AC6 are inhibited by subunit, whereas ments has become apparent. We now know the
group 2 ACs are stimulated by subunit binding plasma membrane is a heterogeneous environment,
(Hanoune et al. 1997). Belevych and colleagues pro- with lipid rafts and microdomains. Caveolae, or little
pose that the net response to mAChR activation is a caves of membrane invaginations, and the larger
balance between inhibition and facilitation of cAMP- membrane invaginations of the transverse (T) tubu-
dependent responses mediated by specific isoforms of lar system of ventricular myocytes, located near the
AC (Figure 5b) (Harvey and Belevych 2003). This SR terminal cisternae, form microdomains or com-
hypothesis is supported, in part, by the observation partments that concentrate ions and/or effectors of
that AC5 null mice have elevated heart rates, indi- signal transduction systems (Balijepalli et al. 2006;
cating that more parasympathetic inhibition, rather Saucerman and McCulloch 2006). Interestingly,
than sympathetic stimulation, is mediated by AC5 2-ARs are almost exclusively located in caveolae,
(Okumura et al. 2003). but 1-ARs are not (Rybin et al. 1999). Using green
Cardiac Physiology and Pharmacology 59

fluorescent protein-based technology, real-time ima- 6.04.5 Classification and


ging of living cells has demonstrated discrete Mechanisms of Arrhythmias
localization patterns for several second messengers,
such as Ca2, cAMP, and PKA (Zhang et al. 2002). Arrhythmias can be classified based on their properties
Using this technique, -AR stimulation was observed at the molecular, cellular, or clinical levels (Table 1).
to induce gradients of cAMP in the regions of T In general, arrhythmias occur because of abnormal
tubule membranes (Zaccolo and Pozzan 2002). impulse generation and/or abnormal impulse conduc-
Furthermore, computational modeling has predicted tion. Both abnormalities may result from intrinsic
that concentrations of Ca2 in the T tubule micro- heart disease, some drug exposures, or inherited
domain may be 100 to 1000 times that of the bulk disorders. This section discusses important mechan-
cytosol (Shannon et al. 2004). Compartmentation of isms of abnormal impulse generation and conduction.
Ca2 likely explains the observation that Ca2- It also includes a section on the genetic basis of
dependent signaling pathways are not themselves arrhythmias. Work in this area has identified particu-
triggered by alterations in Ca2 concentration that lar molecules whose dysfunction leads to arrhythmias
occur during myocardial contraction and relaxation (Table 1) and moved the entire field from empirically
(Mudd and Kass 2008). derived, often less than satisfactory, treatments toward
Functional compartmentation, or localization mechanism-based therapeutics that hold great poten-
of particular signaling and regulatory molecules tial (Knollmann and Roden 2008).
near one another in multi-protein complexes, is
accomplished by a number of scaffolding and adap-
tor proteins (Pawson and Nash 2003; Pawson and 6.04.5.1 Abnormal Impulse Generation
Scott 1997). Not only do these proteins ensure that
upon activation the enzyme is near its specific Triggered activity is an abnormality of impulse gen-
eration in which impulses are initiated by oscillations
targets (spatial regulation), but also carry shut-off
in membrane potential which arise following the
proteins (temporal regulation) and/or means to
upstroke of a preceding action potential. These oscil-
integrate the signal with other pathways (cross-
lations in membrane potential are referred to as
talk regulation) (Beene and Scott 2007). Two
afterdepolarizations and are of two types: early
scaffolding proteins with particular importance for
afterdepolarizations (EADs) or delayed afterdepolar-
the cardiovascular system are the A-kinase anchor-
izations (DADs). EADs occur during the plateau or
ing proteins (AKAPs) and -arrestins. More than 50
repolarization phases (phase 2 or 3) of the AP while
AKAPs exist with significant structural diversity,
DADs occur after the completion of repolarization.
yet all bind PKA and other signaling enzymes to When afterdepolarizations are large enough to reach
form multi-protein complexes at specific subcellu- a threshold value, they result in the generation of
lar sites (Beene and Scott 2007). The first AKAP triggered APs. Thus, in contrast to automaticity in
discovered was found in the macromolecular which the rhythm can be generated in the absence of
complex of RyR2 (Marx et al. 2000) and facilitates any preceding electrical activity, triggered rhythms
SR Ca2 release. It is also known that AKAP15 require at least one AP preceding them (Waldo and
directly anchors PKA to L-type Ca2 channels Wit 1993).
and is needed for -AR-stimulated-phosphoryla- Experimentally, EADs have been induced in
tion of L-type Ca2 channels and subsequent preparations exposed to hypoxia, hypercapnia, and
increase in ICaL (Hulme et al. 2003). More than 20 high levels of catecholamines which are all conditions
proteins have been shown to be targeted by - present in the ischemic or infarcted myocardium
arrestins (Lefkowitz and Shenoy 2005). (Binah and Rosen 1992). Triggered APs during the
Importantly for the heart, -arrestin acts as a scaf- plateau and early repolarizing phases have an upstroke
fold that enables ERK phosphorylation through Gi carried by ICaL, due to inactivation of the fast inward
signaling in response to 2-AR stimulation Na channels at depolarized potentials. In contrast,
(Figure 2) (Perry et al. 2002) and allows for the triggered APs occurring during late phase 3 of repo-
dynamic movement of active phosphodiesterase larization have upstrokes caused by the fast inward
4D (PDE4D) from the cytosol to -ARs to create INa, due to the partial reactivation of the fast Na
a localized sink for cAMP degradation and thus channels at repolarized membrane potentials. The
dampen PKA activity (Houslay et al. 2007). amplitude of EADs and thus their likelihood of
Table 1 Mechanism-based approach to antiarrhythmic drug therapy

Phenotype
Disease- Possible acquired Potential mechanism-
Mechanism Molecular Cellular Clinical associated genes syndrome specific drug therapy

Failure of channel closing Destabilized Increased APD, Long QT interval (mostly SCN5A, CAV3, Drug-induced LQTS Blocker of Na
or inactivation Na-channel EADs at rest) SCN4B channels
inactivation
Destabilized DAD-mediated Catecholaminergic RYR2 Arrhythmia in heart Stabilizers of ryanodine
ryanodine automaticity, polymorphic VT CASQ2 failure, channels; blocker
channels premature Atrial and ventricular phenocopied by of Ca2-activated
Ca2 release arrhythmia, bradycardia digitalis intoxication arrhythmogenic
from SR processes
Abnormal L-type Increased APD, Timothy syndrome (extreme CACNA1C Drug-induced LQTS Blocker of Ca2
Ca2-channel EADs LQTS and extracardiac channels
inactivation effects)
Increased repolarizing K Failure of fast IKr Decreased APD SQTS (with AF and VF) KCNH2 (HERG) AF induced by vagal Blocker of affected
current inactivation stimulation K channel
IKs activated at KCNQ1
more negative
voltages
Increased IK1 KCNJ2
secondary
to failure of
normal inward
rectification
Increased non-selective ND Decreased AF ND AF Blocker of stretch-
stretch current APD, DADs activated channels
(such as spider
toxin MTx-4)
Decreased current as Decreased Na Loss of AP Increased PR and SCN5A SCD during therapy Activator of peak Na
a result of abnormal current dome in QRS duration (slow with Na-channel current; blocker of
channel gating or epicardial conduction), ST-segment blockers, especially transient outward
decreased cell-surface cells elevation, VF (Brugada in ischemic heart current
protein expression syndrome) disease
Decreased Isolated conduction-system
slope of AP disease (PR and QRS
upstroke prolongation, bundle
branch block, AV nodal
block
Decreased K Increased APD, LQTS (often with exertion) KCNQ1, KCNH2, Drug-induced LQTS Activator of affected
current EADs KCNE1, K channel
KCNE2, KCNJ2
ND AF KCNA5 AF
Decreased L- Decreased APD Combined SQTS and CACNA1C, ND Activator of peak Ca2
type Ca2 Brugada syndrome CACNB2 current
current
Decreased ND AF GJAS Some VT in ischemic Activation of gap
connexin heart disease junction
channel conductance
current
Decreased Slowed Sinus-node dysfunction HCN4 Bradycardia Activation of
pacemaker pacemaker pacemaker current
current rate
Abnormal targeting of Ca2 ND DADs AF, VF, bradycardia, type 4 ANK2 Drug-induced LQTS ND
control proteins LQTS
Cardio-myopathic lesions, Cytoskeletal- ND Dilated cardiomyopathy, LMNA, DMD, Viral carditis ND
associated with protein heart block, AF, VF DES, SGCD,
arrhythmias dysfunction ACTN2, VCL,
CLP, LDB3
Sarcomere- EADs or DADs Mostly hypertrophic cardio- MYH7, MYBPC3, Acquired cardiac
protein in some cases myopathy: myofibrillar TNNT2, TNNI3, hypertrophy
dysfunction disarray, nonconcentric TPM1, MYL2,
ventricular hypertrophy, MYL3, ACTC1,
AF, VT TTN
Desmosome- ND Arrhythmogenic right PKP2, DSP, JUP, ND
protein ventricular DSG2, DSC2,
dysfunction cardiomyopathy DES
Metabolism and Cytoplasmic Hypertrophic LAMP2, PRKAG2 WolffParkinson
lysosomal aggregates cardiomyopathy, White syndrome
protein due to ventricular preexcitation
dysfunction defective
lysosomes

The molecular, cellular, and clinical phenotypes associated with fundamental molecular defects are indicated, together with the genetic and acquired arrhythmia syndromes that probably have
the same pathophysiology. This information has led to specific interventions that are predicted or have been established to be antiarrhythmic. ACTC1, cardiac 2-actinin; AF, atrial fibrillation;
ANK2, ankyrin 2; APD, action potential duration; AV, atrioventricular; CACNA1C, L-type Ca2 channel 1C subunit; CACNB2, L-type Ca2 channel 2-subunit; CASQ2, cardiac calsequestrin 2;
CAV3 caveolin 3; CLP cardiac LIM protein; DAD, delayed afterdepolarization; DES, desmin; DMD, dystrophin; DSC2, desmocollin 2; DSG2, desmoglein 2; DSP, desmoplakin; EAD, early
afterdepolarization; GJA5, gap junction protein- 5 (also known as connexin 40); HCN4, hyperpolarization-activated cyclic-nucleotide-gated K channel 4; JUP, junction plakoglobin (also known
as -catenin); KCN, K channel; LAMP2, lysosomal-associated membrane protein 2; LDB3, LIM-domain-binding 3; LMNA, lamin A; LQTS, long-QT syndrome (QT-interval prolongation and
torsades de pointes-type polymorphic VT); MYBPC3, cardiac myosin-binding protein C; MYH7, -myosin heavy chain; MYL, myosin light chain; ND, not determined; PKP2, plakophilin 2;
PRKAG2, AMP-activated protein kinase- 2; SCGD, -sarcoglycan; SCN, voltage-gated Na channel; SR, sarcoplasmic reticulum; SQTS, short-QT syndrome (QT-interval shortening associated
with AF and VF); TNNI3, cardiac troponin I; TNNT2, cardiac troponin T; TPM1, -tropomyosin; TTN, titin; VCL, vinculin; VF, ventricular fibrillation; VT, ventricular tachycardia. (Knollman, B. C.;
Roden, D. M. Nature 2008, 451, 929936.
62 Introduction to Cardiovascular Biology

initiating triggered arrhythmias are enhanced by slow (Morady 1999). Random reentry occurs with no fixed
heart rates (Damiano and Rosen 1984). Triggered circuit but travels from cell to cell as soon as the
rhythms are commonly subdivided into those occur- neighboring cell repolarizes. Atrial fibrillation and
ring when the heart rate is slow (long QT-interval- ventricular fibrillation (AF and VF) are often caused
associated arrhythmias) and when it is rapid (catecho- by random reentry mechanisms. Atrial flutter, mono-
laminergic polymorphic ventricular tachycardia). morphic ventricular tachycardias, AV nodal reentrant
DADs are transient depolarizations occurring tachycardia, and sinus node reentrant tachycardia are
after repolarization of the AP and have been caused by ordered reentry mechanisms.
observed under a number of experimental conditions, Underlying cellular and molecular mechanisms of
all of which are associated with an increase in intra- reentry include a wide range of conditions from
cellular Ca2. These conditions include cardiac acquired injury or ischemia, to inherited congenital
glycoside intoxication, elevated extracellular Ca2 structural defects and channelopathies, to changes
concentration, and administration of catecholamines in the expression levels of gap junction or structural
(Ferrier 1977). When the heart rate is high, subthres- proteins (Wolf and Berul 2006). Any condition that
hold DADs can build to reach the threshold for firing slows conduction in a subset of myocytes, such as an
a triggered AP. Thus, tachycardias triggered by elevated resting membrane potential that partially or
DADs can be expected after an increase in sponta- completely inactivates fast Na channels requiring
neous or pacing-induced rate (Binah and Rosen propagation to depend on the more slow L-type
1992). This property of DADs demonstrates an Ca2 channels, can result in an AP arriving at neigh-
important difference between normal automaticity boring cells post their refractory period and be
in Purkinje fibers and DAD-induced triggered treated as a new impulse for propagation.
activity. While rapid pacing and overdrive stimula-
tion suppress normal automaticity, DAD-induced
6.04.5.3 Genetic Basis of Arrhythmias
arrhythmias undergo overdrive excitation such that
pacing at a rate faster than the triggered activity Studying relatively rare genetic diseases that cause
results in an increase in the rate of the arrhythmia. familial cardiomyopathies and/or arrhythmias have
The underlying ionic mechanisms responsible for helped define molecular constituents and underlying
DADs have been extensively studied. Abnormalities mechanisms of common arrhythmias (Knollmann
in the sequestration and release of Ca2 by the SR and Roden 2008; Nattel and Carlsson 2006). For
are important factors contributing to elevated intra- example, long-QT syndrome (LQTS) was first
cellular Ca2 and increased risk of DADs (Bers 2002, described in the 1950s as a familial disease with
2006). Most ventricular tachycardias in HF patients long QT-interval, normal heart structure, and
are initiated by altered Ca2 handling in the myo- increased risk of sudden cardiac death (SCD). Most
cytes, including SR Ca2 leak and activation of NCX investigators thought the condition resulted from an
(Bers 2006). autonomic imbalance (Rosen 1998). Today, we know
that loss-of-function mutations in ion channel
subunits underlying IKr, IKs, IK1 and gain-of-function
6.04.5.2 Abnormal Impulse Conduction
mutations in INa and ICaL increase APD and can
Normally in the intact heart, the impulse generated by cause life-threatening polymorphic ventricular
the SA node dies out after sequentially activating the tachycardia known as torsade de pointes (Nattel and
atria and the ventricles via the AV conduction system Carlsson 2006; Sanguinetti and Tristani-Firouzi
because it encounters refractory tissue that it has just 2006). The most common mutation responsible
excited and also because it meets the inexcitable for this disease is found in the gene HERG
fibrous annulus. Reentrant excitation occurs when (KCNQ1), which codes the -subunit of the channel
there is continuous excitation in an electrical circuit that conducts the slow delayed rectifier K current,
without independent triggering of each beat. IKs (Curren et al. 1995). This and other mutations
Reentrant circuits can be small (micro-reentry) or that inactivate inward K currents delay the ability
large (macro-reentry), and require heterogeneity in of the cell to return to normal resting potential,
the electrophysiological properties of tissue in the delaying repolarization and increasing APD. Gain-
circuit, such as differences in refractory periods or in of-function in Na channel activity (often incom-
conduction velocity. Stable reentrant circuits are ana- plete inactivation) results in a small but persistent
tomical structures and can often be cured by ablation inward current during phase 2 that prolongs the APD
Cardiac Physiology and Pharmacology 63

(Bennett et al. 1995). Discovering that different mole- (Vaughan Williams 1970). Class I agents block the
cular mechanisms could be responsible for LQTS inward sodium current, INa, responsible for the AP
explained why only some patients responded to upstroke (phase 0) and are divided into subclasses
treatment with Na channel blocking agents, and based on their ability to alter APD; class II agents
ushered in a new era of mechanism-based approaches block -AR receptors; class III agents prolong APD,
to antiarrhythmic drug therapy (Table 1) primarily through blocking the late phase 3 repolar-
(Knollmann and Roden 2008). izing currents, IKr and IKs; and class IV agents block
In addition to inherited disease, several common L-type calcium channels (Table 2). Although widely
medications can prolong the QT interval and induce used, this classification system is not ideal. Many
torsade de pointes (De Bruin et al. 2005). Torsade de antiarrhythmic drugs have multiple actions and do
pointes is frequent in patients treated with the antiar- not fit into a single class. For example, amiodarone
rhythmic drug quinidine (as high as 9%), but occurs prolongs APD by potently inhibiting the outward
in about 1 out of 120 000 patients treated with the rapid K current, IKr, but also inhibits sodium chan-
non-antiarrhythmic drugs that can induce the effect nel, -adrenergic receptor, and calcium channel
(Haverkamp et al. 2000; Vitola et al. 1998). Although actions (Singh and Hauswirth 1974; Singh and
rare in non-antiarrhythmic drugs, the risk level of Vaughan Williams 1970). Thus, amiodarone could
torsade de pointes is unacceptable for patients being be categorized into any one of the four Vaughan
treated for non-life-threatening conditions and sev- WilliamsSingh classes. Lack of class specificity and
eral agents (cisapride, sertindole, grepafloxacin, additional concerns about the Vaughan Williams
terfenadine, and astemizole) were restricted in their Singh classification system are discussed in detail by
use or completely removed from the market members of the Sicilian Task Force (Members of the
(Sanguinetti and Tristani-Firouzi 2006). These Sicilian Gambit 1994; Rosen 1998; Task Force of the
structurally diverse agents all bind and block hERG Working Group on Arrhythmias of the European
channels (Sanguinetti and Tristani-Firouzi 2006). It Society of Cardiology 1991). They gathered in 1991
is now common practice for pharmaceutical compa- to devise a new antiarrhythmic drug classification
nies to screen compounds for hERG channel binding, system. Their proposal, the Sicilian Gambit, empha-
ability to alter hERG current in cells, or ability to sizes individual drug characteristics (actions on ion
alter QT intervals in animals during drug develop-
channels, autonomic and purinergic receptors, and
ment (Sanguinetti and Mitcheson 2005).
clinical effects), rather than class groupings, in order
to promote treatment based on known mechanisms of
6.04.6 Classification and arrhythmic disease and drug action (Members of the
Mechanisms of Action of Sicilian Gambit 1994; Rosen 1998; Task Force of the
Antiarrhythmic Agents Working Group on Arrhythmias of the European
Society of Cardiology 1991). Unfortunately, their
Antiarrhythmic drugs are commonly classified by the approach produced a complex system that has lim-
Vaughan WilliamsSingh system, which groups ited the Sicilian Gambits usefulness as a means of
drugs according to their electrophysiologic effects classification (Reiffel 1997). However, the Sicilian

Table 2 Vaughan WilliamsSingh antiarrhythmic drug classification system with prototypical


agents listed for each class

Class Action Agents

Ia Sodium channel antagonists Quinidine, procainamide, disopyramide


Prolong action potential duration (APD)
Ib Less potent sodium channel antagonists Lidocaine, mexiletine
No change or shortening of APD
Ic Potent sodium channel antagonists Propafenone, flecainide, moricizine
No change or mild APD prolongation
II -Adrenergic receptor antagonists Propranolol, esmolol
III Prolong APD Amiodarone, sotalol, dofetilide
IV Calcium channel antagonists Verapamil, diltiazem
64 Introduction to Cardiovascular Biology

Gambits original intent of target-specific, mechan- 6.04.6.1.3 Class IV antiarrhythmic agents


ism-based drug development and use is now being Ca2 channel antagonists are used most often to treat
realized as many of the ion channel subunits that hypertension and angina, but are also effective against
mediate cardiac currents have been identified, the treatment of a wide variety of supraventricular
cloned, and physiologically characterized. tachycardias. There are three chemical classes of cal-
cium channel antagonists: phenylalkylamines (e.g.,
verapamil), benzothiazepines (e.g., diltiazem), and
dihydropyridines (e.g., nifedipine). Each class binds
6.04.6.1 Conventional Antiarrhythmic
to distinct, high affinity binding sites in close proxi-
Agents
mity to one another on the 1 subunit of L-type Ca2
6.04.6.1.1 Class I and III antiarrhythmic channels (for review see Striessnig et al. 1998). The 1
agents subunit forms the conduction pore, voltage sensor, and
Only class I and III agents (Na and K channel gating apparatus of the channel (Bodi et al. 2005). Four
blockers) are effective in terminating or preventing separate L-type 1 subunit genes have been identified:
recurrences of AF (Levy 2001), the most common CaV1.1 ( 1S), 1.2 ( 1C), 1.3 ( 1D), and 1.4 ( 1F). Only
form of sustained arrhythmia. However, these agents CaV1.2 is expressed at high levels in cardiac myocytes
can themselves induce life-threatening ventricular and may explain the differential responses of skeletal,
arrhythmias. The Cardiac Arrhythmia Suppression smooth and cardiac muscle to class IV agents
Trial (CAST), terminated early in 1989 because (Catterall et al. 2005).
patients being treated with potent Na channel block-
ers, encainide and flecainide, had two- to three-fold
6.04.6.2 Novel Antiarrhythmic Agents and
increases in mortality versus placebo controls (CAST
Approaches
Investigators 1989), and several subsequent studies
(SWORD (SWORD Investigators 1996), AFFIRM 6.04.6.2.1 Atrial-selective agents
(AFFIRM Investigators 2002), RACE (RACE Currently available antiarrhythmic agents target the
Investigators 2002)) have shown no mortality benefit same channels (primarily IKr) in atrial and ventricular
to restoring normal sinus rhythm in AF patients with cells and it is their action in the ventricles as opposed
either class I or class III antiarrhythmic agents. It is to the atria that can lead to lethal arrhythmias. Newer
likely that the toxicities and proarrhythmic risks of agents, such as XEN-D0101 (Xention), that specifi-
the agents negate the benefits of chronically suppres- cally target Kv1.5 subunits, found in atrial but not
sing the AF (Morrow et al. 2007). Out of necessity, class ventricular cells (Feng et al. 1997) and carry the
I and III antiarrhythmic agents continue to be used for ultrarapid delayed rectifier current (IKur), are
both acute and chronic cessation of atrial and ventricu- promising compounds to treat AF without causing
lar fibrillations, but novel pharmaceutical agents and ventricular arrhythmias (Figure 6) (Nattel and
non-pharmacological approaches, such as ablation ther- Carlsson 2006). Another selective target is the acet-
apy and implantable cardioverter defibrillators, are ylcholine-related K current (IKACh) carried by
increasing (Bardy et al. 2005; Morady 1999). Kir3.1/3.4 heterotetramers (Figure 6). Normally
inactive until stimulated by the parasympathetic sys-
6.04.6.1.2 Class II antiarrhythmic agents tem, IKACh is constitutively active and contributes to
-AR blocking agents have well-established effects basal inward rectifier K current in chronic AF
on arrhythmias that involve the AV node (Task (Dobrev et al. 2005). IKACh is atrial selective and has
Force of the Working Group on Arrhythmias of the no effect on ventricular current or APD (Cha et al.
European Society of Cardiology 1991). Blocking 2006). IKACh blockers, NIP-141 and 142, are
sympathetic stimulation decreases conduction velo- currently in pre-clinical trials (Hashimoto et al.
city and increases refractoriness of conduction cells 2007; Matsuda et al. 2006).
as well as decreasing myocardial cell contraction rate.
-ARs antagonists have become important agents in 6.04.6.2.2 Gap junction modulators
the prevention of many forms of heart disease, Gap junctions are aggregates of voltage-gated, cellcell
including arrhythmias (Ellison and Gandhi 2005). channels, with each channel constructed from two
See Section 6.04.6.2.3 for more information on the hemichannels and each hemichannel comprised of six
use of -AR blocking agents to slow the progression connexin subunits (Desplantez et al. 2007). There are at
of HF to reduce arrhythmia formation. least three different connexin types expressed in the
Cardiac Physiology and Pharmacology 65

IKATP IKr IKs IKACh Ito IKur IK1 If


Na+
Kir6.2 hERG KvLGT1 Kir3.1 Kv4.3 Kv1.5 Kir2.1 HCN4

M2 G-protein
SUR2A Kir3.4 KChlp
MiRP1 KCR1 minK
+ + + + +
2.2
K K K K K K+ K+ K+

Clamikalant Azimilide NIP-141 XEN-D0101 Ivabradine


withdrawn Phase III NIP-142 Phase I Phase III
Preclinical

Vernakalant Dronedarone Mibefradil


Phase II/III Phase III withdrawn GsMTx4
Preclinical Rotigaptide
INa ICaL ICaT Phase II
+
NaV1.5 CaV1.2 CaV3.1/3.2 Connexin

Ahnak SAC SAC

Gap junction
Na+ Ca2+ Ca2+ +

Figure 6 Schematic of cardiac cells depicting ion channel subunits and transporters that are targeted by investigational
drugs. As indicated, drugs are in preclinical, phase II, phase III, or have been withdrawn from clinical trials. SAC, stretch
activated channel. (Nattel, S.; Carlsson, L. Nat. Rev. Drug Discov. 2006, 5, 10341049.)

heart. Connexin 45 is primarily found in SA and AV ventricular tachycardias induced by non-reentrant


node cells. Connexin 40 and 43 are not found in node mechanisms (triggered mechanisms such as DADs,
cells, but are highly expressed in the ventricular con- EADs, and abnormal automaticity) (Pogwizd 1994;
duction system and atrial myocytes. Only connexin 43 Pogwizd et al. 1998). Thus, systolic dysfunction and
is expressed at high levels in ventricle myocytes arrhythmias are not mutually exclusive diseases and
(Desplantez et al. 2007). The tissue-specific distribution use of -AR antagonists to slow the progression of
of connexins makes them attractive targets for novel HF also decreases the risk of arrhythmias.
drug development. Furthermore, alterations in gap The classical view of -AR signaling as a linear
junction kinetics are known to slow conduction velocity pathway (see Section 6.04.3.1) led to the use of inotropic
and promote AF (Kanno and Saffitz 2001). Accordingly, agents to treat both acute and chronic HF. Dobutamine,
drugs that enhance gap junction conduction could be a -AR agonist, is still a mainstay of acute HF manage-
good agents for AF management (Mazzini and ment (Gauthier et al. 2008); however, several large,
Monahan 2008; Nattel and Carlsson 2006). well-controlled clinical trials have demonstrated that
Rotigaptide is a peptide stabilizer of gap junction con- the opposite treatment, -AR antagonism, is clearly the
duction during ischemia (Xing et al. 2003) but does not best treatment for chronic HF (Ellison and Gandhi
affect ischemia-induced focal ventricular tachycardia or 2005; Ghali and Lindenfeld 2008). Two to four
triggered activity in dogs (Xing et al. 2005). Additional thousand patients in each of the Metoprolol
studies will continue to determine whether gap junction Controlled-Release/Extended-Release Randomized
enhancers are clinically viable antiarrhythmic agents. Intervention Trial (MERIT-HF Investigators 1999),
the Cardiac Insufficiency Bisoprolol Study II (CIBIS-
6.04.6.2.3 Preventing heart failure and II Investigators 1999), and the Carvedilol Prospective
arrhythmias Randomized Cumulative Survival trial (Packer et al.
Many late-stage HF patients also experience 2001) demonstrated a 3435% mortality reduction in
arrhythmias and sudden cardiac death due to -AR antagonist-treated versus placebo-controlled
66 Introduction to Cardiovascular Biology

groups. Today, the American Heart Association and Boyett, M. R.; Honjo, H.; Kodama, I. Cardiovasc. Res. 2000, 47,
658687.
American College of Cardiology recommends the use Brillantes, A. B.; Ondrias, K.; Scott, A.; Kobrinsky, E.;
of -AR antagonists in all patients with symptomatic Ondriasova, E.; Moschella, M. C.; Jayaraman, T.; Landers, M.;
left ventricular dysfunction, yet only one-third of eligi- Ehrlich, B. E.; Marks, A. R. Cell 1994, 77, 513523.
Carafoli, E. Annu. Rev. Biochem. 1987, 56, 395433.
ble patients may be actually receiving -AR antagonist Catterall, W. A.; Perez-Reyes, E.; Snutch, T. P.; Striessnig, J.
therapy for HF (Ansari et al. 2003). The use of -AR Pharmacol. Rev. 2005, 57, 411425.
antagonists to manage HF has not been embraced Cha, T. J.; Ehrlich, J. R.; Chartier, D.; Qi, X. Y.; Xiao, L.; Nattel, S.
Circulation 2006, 113, 17301737.
because it appears counterintuitive to the classical CIBIS-II Investigators. Lancet 1999, 353, 913.
understanding of -AR function (Ellison and Gandhi Curren, M. E.; Splawski, I.; Timothy, K. W.; Vincent, G. M.;
2005). Recent advances in our knowledge of -AR Green, E. D.; Keating, M. T. Cell 1995, 80, 795803.
Damiano, B. P.; Rosen, M. R. Circulation 1984, 69, 10131025.
signaling have revealed a much more complex signaling De Bruin, M. L.; Pettersson, M.; Meyboom, R. H.; Hoes, A. W.;
network than previously believed (Figure 4) and Leufkens, H. G. Eur. Heart J. 2005, 26, 590597.
should have a great impact on our understanding and Desplantez, T.; Dupont, E.; Severs, N. J.; Weingart, R. J.
Membr. Biol. 2007, 218, 1328.
treatment of HF and other cardiac diseases (Saucerman DiFrancesco, D. J. Physiol. (London) 1999, 515, 367376.
and McCulloch 2006). DiFrancesco, D.; Borer, J. S. Drugs 2007, 67 (Suppl. 2), 1524.
Dobrev, D.; Friedrich, A.; Voigt, N., et al. Circulation 2005, 112,
36973706.
6.04.6.2.4 Future directions Ellison, K. E.; Gandhi. G. Drugs 2005, 65 (6), 787797.
Knollmann and Roden (2008) articulated the opti- Feng, J.; Wible, B.; Li, G.-R.; Wang, Z.; Nattel, S. Circ. Res.
mism of the field that increased knowledge of specific 1997, 80, 572579.
Ferrier, G. R. Prog. Cardiovasc. Dis. 1977, 19, 459474.
molecular targets important to normal and dysfunc- Gauthier, N.; Anselm, A. H.; Haddad, H. Curr. Opin. Cardiol.
tional cardiac physiology will (1) translate into new, 2008, 23 (2), 134140.
improved therapies for arrhythmias and other forms Ghali, J. K.; Lindenfeld, J. Expert Rev. Cardiovasc. Ther. 2008, 6
(4), 555565.
of heart disease; (2) lead to the identification of Gunter, T. E.; Gunter, K. K.; Sheu, S. S.; Gavin, C. E. Am. J.
genetic markers that will improve cardiovascular dis- Physiol. 1994, 267, C313C339.
ease screening and preventative measures in the Hanoune, J.; Pouille, Y.; Tzavara, E.; Shen, T.; Lipskaya, L.;
Miyamoto, N.; Suzuki, Y.; Defer, N. Mol. Cell. Endocrinol.
general population; (3) identify common polymorph- 1997, 128, 179194.
isms that will lead to personalized medicine for the Hartzell, H. C. Prog. Biophys. Mol. Biol. 1988, 52, 165247.
most effective treatment for individual patients; and Harvey, R. D.; Belevych, A. E.; Br. J. Pharmacol. 2003, 139,
10741084.
(4) allow development of pharmaceutical drugs with- Haverkamp, W.; Breithardt, G.; Camm, A. J.; Janse, M. J.;
out unintended cardiovascular toxicity. Rosen, M. R.; Antzelevitch, C.; Escande, D.; Franz, M.;
Malik, M.; Moss, A., et al. Eur. Heart J. 2000, 21, 12161231.
Herfst, L. J.; Rook, M. B.; Jongsma, H. J. J. Mol. Cell. Cardiol.
2004, 36, 185193.
References Hoffman, B. F.; Cranefield, P. F. Am. J. Med. 1964, 37, 670684.
Houslay, M. D.; Baillie, G. S.; Maurice, D. H. Circ. Res. 2007,
Accili, E. A.; Proenza, C.; Baruscotti, M.; DiFrancesco, D. News 100, 950966.
Physiol. Sci. 2002, 17, 3237. Hulme, J. T.; Lin, T. W.-C.; Westenbroek, R. E.; Scheuer, T.;
Altomare, C.; Bucchi, A.; Camatini, E.; Baruscotti, M.; Catterall, W. A. PNAS 2003, 100 (22), 1309313098.
Viscomi, C.; Moroni, A.; DiFrancesco, D. J. Gen. Physiol. Kanno, S.; Saffitz, J. E. Cardiovasc. Pathol. 2001, 10, 169177.
2001, 17, 519532. Kentish, J. C.; McCloskey, D. T.; Layland, J.; Palmer, S.;
Ansari, M.; Shlipak, M.; Heidenreich, P.; Van Ostaeyen, D.; Leiden, J. M.; Martin, A. F.; Solaro, R. J. Cir. Res. 2001, 88,
Pohl, E. C.; Browner, W. S.; Massie, B. M. Circulation 2003, 10591065.
107 (22), 27992804. Knollmann, B. C.; Roden, D. M. Nature 2008, 451, 929936.
Atrial Fibrillation-up Investigation of Rhythm Management Lefkowitz, R. J.; Shenoy, S. K. Science 2005, 308, 512515.
(AFFIRM) Investigators. N. Engl. J. Med. 2002, 347, Levy, S. Am. Heart J. 2001, 141, S15S21.
18251833. Loffelholz, K.; Pappano, A. J. Pharmacol. Rev. 1985, 37, 124.
Baillie, G. S.; Soos, A.; McPhee, I.; Gall, I.; Perry, S. J.; Maack, C.; ORourke, B. Basic Res. Cardiol. 2007, 102,
Lefkowitz, R. J.; Houslay, M. D. Proc. Natl. Acad. Sci. USA 369392.
2003, 100, 940945. Marx, S. O.; Reiken, S.; Hisamatsu, Y.; Jayaraman, T.;
Balijepalli, R. C.; Foell, J. D.; Hall, D. D.; Hell, J. W.; Kamp, T. J. Burkhoff, D.; Rosemblit, N.; Marks, A. R. Cell 2000, 101,
Proc. Natl. Acad. Sci. USA 2006, 103, 75007505. 365376.
Beene, D. L.; Scott, J. D. Curr. Opin. Cell Biol. 2007, 19, 192198. Mason, J. W.; Hondeghem, L. M. Ann. N.Y. Acad. Sci. 1984,
Bennett, P. B.; Yazawa, K.; Makita, N.; George, A. L. Nature 432, 162176.
1995, 376, 683685. Mazzini, M. J.; Monahan, K. M. Heart Rhythm 2008, 5 (6),
Bers, D. M. Nature 2002, 415, 198205. 526531.
Bers, D. M. Physiology 2006, 21, 380387. Members of the Sicilian Gambit, Eds.; Antiarrhythmic Therapy:
Binah, O.; Rosen, M. R. Circulation 1992, 85, 125131. A Pathophysiologic Approach. Futura Publishing Co., Inc.:
Bodi, I.; Mikala, G.; Koch, S. E.; Akhter, S. A.; Schwartz, A. J. Armonk, NY, 1994.
Clin. Invest. 2005, 115 (12), 33063317. MERIT-HF Investigators. Lancet 1999, 353, 20012007.
Cardiac Physiology and Pharmacology 67

Molkentin, J. D.; Dorn, I. G. Annu. Rev. Physiol. 2001, 63, Sheu, S.; Colecraft, H. M. In Cardiovascular
391426. Toxicology; Bishop, S. P., Kerns, W. D., Eds.; Elsevier:
Morady, F. N. Engl. J. Med. 1999, 340, 534544. 1997; Vol. 6, Chapter 5, pp 5572.
Morrow, J. P.; Cannon, C. P.; Reifel, J. A. Am. Heart J. 2007, Simmerman, H. K. B.; Jones, L. R. Physiol. Rev. 1998, 78,
154, 824829. 921947.
Mudd, J. O.; Kass, D. A. Nature 2008, 451, 919928. Singh, B. N.; Hauswirth, O. Am. Heart J. 1974, 87, 367382.
Nattel, S.; Carlsson, L. Rev. Drug Discov. 2006, 10341049. Singh, B. N.; Vaughan Williams, E. M. Br. J. Pharmacol. 1970,
Okumura, S.; Kawabe, J.-I.; Yatani, A.; Takagi, G.; Lee, M.-C.; 39, 657667.
Hong, C.; Liu, J.; Takagi, I.; Sadoshima, J.; Vatner, D. E., Smit, M. J.; Iyengar, R. Phosphoprotein Res. 1998, 32, 121.
et al. Circ. Res. 2003, 93, 364371. Standish, A.; Enquist, L. W.; Schwaber, J. S. Science 1994, 263,
Packer, M.; Coats, A. J.; Fowler, M. B.; Katus, H. A.; Krum, H.; 232234.
Mohacsi, P.; Rouleau, J. L.; Tendera, M.; Castaigne, A.; Striessnig, J.; Grabner, M.; Mitterdorfer, J.; Hering, S.;
Roecker, E. B., et al. N. Engl. J. Med. 2001, 22, 16511658. Sinnegger, M. J.; Glossmann, H. Trends Pharmacol. Sci.
Pawson, T.; Nash, P. Science 2003, 300, 445452. 1998, 19 (3), 108115.
Pawson, T.; Scott, J. D. Science 1997, 278, 20752080. Sunahara, R. K.; Dessauer, C. W.; Gilman, A. G. Annu. Rev.
Perry, S. J.; Baillie, G. S.; Kohout, T. A.; McPhee, I.; Pharmacol. Toxicol. 1996, 36, 461480.
Magiera, M. M.; Ang, K. L.; Miller, W. E.; McLean, A. J.; Survival With Oral d-Sotalol (SWORD) Investigators. Lancet
Conti, M.; Houslay, M. D., et al. Science 2002, 298, 834836. 1996, 348, 712.
Pogwizd, S. M. Circulation 1994, 90, 14411458. Tamargo, J.; Caballero, R.; Gomez, R.; Valenzuela, C.;
Pogwizd, S. M.; McKenzie, J. P.; Cain, M. E. Circulation 1998, Delpon, E. Cardiovasc. Res. 2004, 62, 933.
98, 24042414. Tardif, J. C. Drugs 2007, 67 (Suppl. 2), 3541.
Randall, W. C.; Wurster, R. D. In Vagal Control of the Heart: Task Force of the Working Group on Arrhythmias of the
Experimental Basis and Clinical Implications; Levy, M. N., European Society of Cardiology. Circulation 1991, 84,
Schwartz, P. J., Eds.; Futura: New York, 1993; pp 2132. 18311851.
Rate Control Versus Electrical Cardioversion for Persistent Vaughan Williams, E. M. In Symposium on Cardiac
Atrial Fibrillation Investigators. N. Engl. J. Med. 2002, 347, Arrhythmias; Sandoe, E., Flensted-Jensen, E., Olesen, K. H.,
18341840. Eds.; Astra: Sweden, 1970; pp 449472.
Reiffel, J. A. Acc. Curr. J. Rev. 1997, 6 (5), 3234. Vitola, J.; Vukanovic, J.; Roden, D. M. J. Cardiovasc.
Rosen, M. R. Card. Electrophysiol. Rev. 1998, 2, 109114. Electrophysiol. 1998, 9, 11091113.
Rybin, V. O.; Xu, X.; Steinberg, S. F. Cir. Res. 1999, 84, 980988. Waldo, A. L.; Wit, A. L. Lancet 1993, 341, 11891193.
Sanguinetti, M. C.; Mitcheson, J. S. Trends Pharmacol. Sci. Wolf, C. M.; Berul, C. I. J. Cardiovasc. Electrophysiol. 2006, 17,
2005, 26, 119124. 446455.
Sanguinetti, M. C.; Tristani-Firouzi, M. Nature 2006, 440, 463469. Xing, D.; Kjlbye, A. L.; Nielsen, M. S.; Petersen, J. S.;
Saucerman, J. J.; McCulloch, A. D. Ann. N.Y. Acad. Sci. 2006, Harlow, K. W.; Holstein-Rathlou, N. H.; Martins, J. B. J.
1080, 348361. Cardiovasc. Electrophysiol. 2003, 14, 510520.
Schuessler, R. B.; Boineau, J. P.; Bromberg, B. I. J. Cardiovasc. Xing, D.; Kjlbye, A. L.; Petersen, J. S.; Martins, J. B. Am. J.
Electrophysiol. 1996, 7, 263274. Physiol. Heart Circ. Physiol. 2005, 288, H511H516.
Shannon, T. R.; Wang, F.; Puglisi, J.; Weber, C.; Bers, D. M. Yatani, A.; Codina, J.; Imoto, Y.; Reves, J. P.; Birnbaumer, L.;
Biophys. J. 2004, 87, 33513371. Brown, A. M. Science 1987, 238, 12881292.
Sheu, S.; Blaustein, M. P. In The Heart and Cardiovascular Zaccolo, M.; Pozzan, T. Science 2002, 295, 17111715.
System; Fozzard, H. A., Haber, E., Jennings, R. B., et al., Zhang, J.; Campbell, R. E.; Ting, A. Y.; Tsien, R. Y. Nat. Rev.
Eds.; Raven Press: New York, 1992; pp 903943. Mol. Cell Biol. 2002, 3, 906918.

Das könnte Ihnen auch gefallen