You are on page 1of 14

PHYSIOLOGY IN MEDICINE

In collaboration with
The American Physiological Society, Thomas E. Andreoli, MD, Editor

Calcium Currents and Arrhythmias:


Insights from Molecular Biology
Stephen R. Shorofsky, MD, PhD, and C. William Balke, MD

Calcium channels are critical to normal cardiac function. They ulum and is responsible for the release of the Ca that activates
are involved in the generation and conduction of the action contraction. Oscillatory behavior of this channel influences the
potential and in contraction. Three surface membrane channels sarcolemmal membrane, causing delayed aftercontractions and
have been identified. The L-type Ca channel is most abundant arrhythmias such as those seen in digoxin toxicity. The on-go-
and is responsible for Ca entry into the cell that triggers contrac- ing molecular characterization of these channels will enhance
tion. T-type Ca channels are most prevalent in the conduction our knowledge of their normal function and dysfunction in
system and are probably involved in automaticity. A newly de- disease states, leading to the development of new therapeutic
scribed TTX-sensitive calcium current may be important in agents to treat arrhythmias and contractile dysfunction. Am J
“boosting” or enhancing conduction and contraction. The Med. 2001;110:127–140. 䉷2001 by Excerpta Medica, Inc.
main intracellular Ca channel resides in the sarcoplasmic retic-

C
alcium channels are wonderfully complex pro- cent data concerning the molecular structure, function,
teins with vitally important roles in cardiac func- and regulation of these channels and identifies areas
tion. The opening of calcium (Ca) channels is in- where modulation of Ca channel function may have an
volved in pacemaker depolarization, supports conduc- important role in the generation of arrhythmias and in
tion through the atrioventricular (AV) node, and the progression and treatment of cardiac diseases.
maintains the distinctive plateau of the cardiac action po-
tential. In addition, they serve as the primary gatekeepers
for Ca entry into cells, thus transducing the electrical sig-
nals at the surface of the myocyte into the biochemical NOMENCLATURE
and mechanical events that result in a contraction. It is no Heart cells actually contain at least five types of Ca cur-
wonder, therefore, that Ca channels have attracted in- rents, three of which (L-type, T-type, and the channels
tense interest over the last three decades. responsible for the newly described current (ICaTTX) are
Ion channels in the cardiac cell membrane are respon-
expressed on the surface membrane, and two (the sarco-
sible for the generation and control of the action poten-
plasmic reticulum Ca release channel and the IP3 recep-
tial. Thus, alterations in channel function often lead to
tor) in internal membranes. The Table summarizes the
abnormal electrical activity and arrhythmias. In fact, ge-
nomenclature, localization, and salient physiological
netic defects in cardiac sodium and potassium channels
roles of each Ca channel type.
have been shown to cause the congenital long QT syn-
When clinicians speak of the Ca channel in the heart,
drome and its resultant arrhythmia, torsades de pointes
they usually mean the L-type Ca channel. This is not only
(1). To date, there has not been a specific arrhythmia that
because of the fact that it exists in large quantities in myo-
results from an identified Ca channel defect in humans,
possibly because Ca channels have such a crucial role in cytes and was first recorded in 1967 in cardiac Purkinje
cardiac function that significant abnormalities in channel fibers (2), but also because these channels function as the
function are lethal. This review, however, focuses on re- receptors for a number of clinically useful drugs, includ-
ing nifedipine, verapamil, and diltiazem.
This review concentrates on the L- and T-type Ca
From the Departments of Physiology and Medicine, University of channels, because they play the most obvious and prom-
Maryland School of Medicine, Baltimore, Maryland. inent role in cardiac arrhythmogenesis. The sarcoplasmic
Requests for reprints should be addressed to C. William Balke, De- reticulum (SR) Ca release channel, the IP3 receptor, and
partment of Physiology, University of Maryland School of Medicine,
Howard Hall, Room 525, 660 West Redwood Street, Baltimore, Mary- the recently reported Ca-permeable Na channel (ICaTTX)
land 21201-1541. are reviewed briefly.

䉷2001 by Excerpta Medica, Inc. 0002-9343/01/$–see front matter 127


All rights reserved. PII S0002-9343(00)00586-6
Calcium Currents and Arrhythmias/Shorofsky and Balke

Table 1. Cardiovascular Calcium Channels


Calcium Channel Type Location Ligands Physiological Roles
L-type calcium channel Sarcolemma Dihydropyridines Atrioventricular nodal conduction
Benzothiazepines Slow response upstroke
Phenylalkylamines Excitation–contraction coupling
Plateau depolarization
T-type calcium channel Sarcolemma Mibefradil Pacemaker depolarization
Cell growth and differentiation?
ICa(TTX) channel Sarcolemma Tetrodotoxin Augmenting depolarization
Excitation–contraction coupling?
Calcium release channel (ryanodine SR Ryanodine SR calcium release
receptor)
IP3 receptor SR Inositol triphosphate Modulation of SR Ca release ?
Other functions
SR ⫽ sarcoplasmic reticulum.

L-TYPE CALCIUM CHANNELS L-type Ca channels also play potentially important


roles in arrhythmia generation and their treatment. Ca
Physiological and Pathophysiological Roles channels are capable of mediating the inward current that
Sarcolemmal Ca channels constitute the major pathway
underlies some types of early afterdepolarizations
for Ca entry into the cell. The existence of a Ca-selective
(EADs) that cause triggered arrhythmias and torsades de
current in heart muscle has been recognized for more
pointes in animals (6,7). As mentioned earlier, there has
than 25 years (2). Two distinct types of channels with
not been a demonstration in humans of a Ca channel
high selectivity for Ca ions (L-type and T-type) are now
defect causing torsades de pointes, or any other arrhyth-
known to coexist in the surface membranes of heart cells
mia thought to be the result of EADs. Ca channels can
(3,4). The two types of channels can be distinguished on
also support slow conduction in reentrant circuits, espe-
the basis of their gating properties (ie, their pattern of
cially in depolarized tissue, although this mechanism ap-
opening and its voltage dependence) and their pharma-
cological sensitivity. We first consider the “traditional” pears to predominate in only a small minority of patients
Ca current of heart muscle, which is sensitive to modula- with inducible ventricular tachycardia. By their effects on
tion by a variety of drugs. This current flowing through automaticity and AV node conduction, L-type Ca chan-
so-called L-type channels (L for large and long lasting) nels may be involved in the generation of certain auto-
(4) is well characterized with respect to its role in cardiac matic arrhythmias [eg, repetitive monomorphic right
excitability, arrhythmogenesis, and excitation– contrac- ventricular outflow tachycardia (RVOT) and some atrial
tion coupling. These channels are fairly plentiful, being tachycardias], and modulation of these Ca channels is
present in some 30,000 copies per ventricular myocyte useful in treating many supraventricular tachycardias
(5). (eg, AV nodal reentry, atrial fibrillation, and atrial flut-
The L-type Ca channel is important in normal and ter).
abnormal cardiac excitation. Ca current supports excita- An appreciation of the role of Ca channels in excita-
tion in the sinoatrial (SA) and atrioventricular (AV) tion– contraction coupling is important in understand-
nodes as well as conduction through the AV node. Inter- ing the potential side effects of Ca channel blockers.
estingly, the AV node is the only site in the body where Ca Ringer (8) first realized in 1883 that Ca ions must be
channels actually conduct an excitatory impulse; this dis- present in the extracellular medium for the heart to gen-
tinctive feature, as well as use-dependent drug binding erate force. Even though the amount of Ca entering the
explains why Ca channel blockers preferentially suppress cell during a single action potential through L-type chan-
AV nodal conduction. In working heart muscle (atria and nels appears to be too small to account fully for contrac-
ventricles) and in specialized conducting tissue, the prin- tile activation (9), Ca influx through these channels is a
cipal excitatory inward current underlying the action po- necessary prerequisite for the initiation of contraction
tential plateau flows through L-type Ca channels. Every- (10 –12).
thing else remaining equal, an increase in Ca current pro- Several organic L-type channel inhibitors exist. The
longs depolarization and thereby increases the height and classic members of this drug family come from three
duration of the action potential plateau. Conversely, chemical groups: the phenylalkylamines (eg, verapamil),
blockade of L-type channels shortens the action potential the benzothiazepines (eg, diltiazem), and dihydropyri-
in working myocardium and renders nodal tissue inex- dines (eg, nifedipine, nitrendipine, and amlodipine) (13).
citable. Exposure to any of the Ca channel antagonists can abolish

128 February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110


Calcium Currents and Arrhythmias/Shorofsky and Balke

Figure 1. Schematic model of the transmembrane topology of the ␣1 subunit of the L-type Ca channel. The P loops in each repeat
are indicated by the thick lines. [Reproduced with permission from W.B. Saunders Company, Cardiac Electrophysiology, 3rd ed.
(147).]

contractile force in isolated myocardial preparations by the ␣1 subunit of the L-type Ca channel (the presumed
interrupting the flux of Ca into the cell by means of L-type pattern of assembly with the other subunits is dia-
Ca channels. There is good evidence that the high-affinity grammed in Figure 2). The overall protein, deduced from
binding sites for these compounds are located on the ␣1 the cDNAs of various isoforms cloned to date (including
subunit of the L-type Ca channel (14), the structure of that from heart), is more than 2,000 amino acids long and
which is discussed below. consists of four internally homologous domains (I to IV).
Structure–function Relationship Each domain is believed to span the membrane six times
Channels are pore-forming proteins that open and close (S1 to S6). The fourth such segment (S4) in each domain
in response to defined stimuli. The processes whereby is distinguished by the repetition, at every third position,
channels open and close are collectively known as gating, of positively charged amino acids (lysine or arginine)
whereas permeation describes how a given channel con- (16,18). Because Na and Ca channels open in response to
ducts ions when it is open. The conceptual distinction depolarization, elucidation of the S4 structure immedi-
between gating on the one hand and permeation on the ately suggested a physical correlate for the voltage sensor
other predates the molecular biology era. Now that many involved in activation gating. Site-directed mutagenesis
channels have been cloned, expressed, and mutated, spe- to modify the net charge in S4 in Na channels alters the
cific and separate structures have been identified that me- voltage dependence of activation, verifying the prediction
diate gating and permeation. Among biological voltage- that S4 functions as a voltage sensor (19). The ␣1 subunit
dependent ion channels, those selective for potassium (K contains the binding sites for Ca channel blocking drugs,
channels) are the smallest and, not coincidentally, the including the dihydropyridines, benzothiazepines, and
most thoroughly characterized (15). Despite the fact that phenylalkylamines (14). The cloned ␣1 subunit can be
channels selective for Na or Ca are larger and more com- expressed in Xenopus oocytes, dysgenic myotubes, or
plex than K channels, several prominent features have mammalian tissue culture cells and is capable of conduct-
emerged. ing Ca currents (20 –22). These studies established that
L-type Ca channels were first purified from skeletal the fundamental properties of the channel allowing for
muscle and consist of five subunits: ␣1 (165 kD), ␣2 (130 selective permeation of Ca across the membrane reside
kD) linked to the ␦ subunit (28 kD) by a disulfide bond, ␤ entirely in the ␣1 subunit. Beyond these basic observa-
(55 kD), and ␥ (32 kD) (16,17). The primary sequence of tions, much remains to be learned about the specific sites
the ␣1 subunit was then determined from the cDNA se- in the L-type Ca channel that underlie its distinctive gat-
quence encoding the protein in rabbit skeletal muscle ing and permeation properties. Expression of chimeric ␣1
(14). Remarkable homology exists between the ␣1 sub- constructs in dysgenic mouse muscle has revealed that
unit of the Ca channel and the pore-forming (␣) subunit the loop between domains II and III is important in iso-
of voltage-dependent Na channels, both having four ho- form-specific excitation– contraction coupling (22),
mologous domains composed of six transmembrane seg- whereas domain I confers features of activation gating
ments each (16). Figure 1 shows a schematic diagram of (23).

February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110 129


Calcium Currents and Arrhythmias/Shorofsky and Balke

Figure 2. Schematic representation of the regulatory subunits and their putative relationship to the ␣1 subunit of the L-type Ca
channel. [Reproduced with permission from W.B. Saunders Company, Cardiac Electrophysiology, 3rd ed. (147).]

Na and Ca channels enter a long-lived nonconducting A confluence of biophysical and molecular biological
state during maintained depolarization, a gating process data has defined regions and residues in the Na channel ␣
known as “inactivation” (24,25), which is not altered by subunit that are important in forming the permeation
changes in S4 (19). Unlike S4, no feature of Na or Ca pathway. A region of striking sequence conservation oc-
channels suggests a role in inactivation merely from in- curs between the fifth and sixth membrane-spanning re-
spection of the primary sequence. Nevertheless, mu- gions (S5 to S6), also called the P loop (P for pore or
tagenesis has revealed that the cytoplasmic linker be- permeation) (26). There is now solid evidence, from elec-
tween domains III and IV, which has been highly con- trophysiological analysis of mutant Na and K channels,
served during evolution, forms at least part of the that the P loops from each of the four domains come
inactivation gate of Na channels (19). Thus, two regions together in the center of the protein to line the pore
of the Na channel are clearly implicated in gating: S4 (15,27). The P loops have been proposed to form ␤-hair-
senses voltage and initiates activation gating, and the pins that form a ␤-barrel structure, but the precise con-
III–IV linker figures prominently in inactivation. Muta- formation is not yet known.
tions in either region do not grossly influence pore prop- Ca channels are very selective. Typically, the perme-
erties. The functional roles of S4 and of the III-IV linker in ability of the channel to Ca is a thousand-fold higher than
the Ca channel have yet to be investigated, partly because the permeability to K or Na (28). Yet, a single Ca channel
the Ca channel clones have not been available as long as can conduct more than 106 Ca ions per second across the
those for the Na channel, but also because Ca channels cell membrane (29). To explain how a Ca channel recon-
have proved to be considerably more difficult to express ciles the conflicting demands of maintaining a large con-
in surrogate cells. Nevertheless, the fact that Na channels ductance in the face of exquisite selectivity, models have
are so similar in their overall molecular architecture gives been proposed in which Ca binds with high affinity at two
good reason to predict the likely functions of the homol- or more sites down the permeation pathway (30,31).
ogous regions of the Ca channel. High-affinity binding makes the channel selective, while

130 February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110


Calcium Currents and Arrhythmias/Shorofsky and Balke

the presence of multiple binding sites enables a high second membrane protein called G protein (for its ability
throughput rate: simultaneous occupancy by two or to bind and to split guanosine triphosphate [GTP]). The
more Ca ions accelerates flux resulting from electrostatic activated ␣ subunit of the G protein in turn stimulates the
ion–ion repulsion. The model handily explains the ability membrane-bound enzyme adenylate cyclase. Biological
of Ca channels to conduct large fluxes of monovalent membranes contain several types of G proteins (45,46).
cations when divalent cations are absent (30,31). Re- The type that mediates stimulation of adenylate cyclase
markably, this model and others (32) were proposed (Gs) carries the subscript s to distinguish it from others of
without any knowledge of the actual protein sequence of inhibitory (Gi) or unrelated (Go) function. The enzyme
any voltage-dependent ion channels. adenylate cyclase catalyzes the formation of cyclic aden-
The P loops from Na and Ca channels are largely con- osine 3⬘,5⬘-monophosphate (cAMP) from ATP, tending
served but differ in the presence of acidic residues in re- to increase the cellular content of cAMP (43). Cyclic AMP
peats III and IV. Substituting acidic residues in these po- regulates a number of cellular functions by promoting
sitions of the Na channel produces the essential features selective protein phosphorylation, initiated by the bind-
of permeation in Ca channels (monovalent permeability ing of cAMP to the regulatory subunit of cAMP-depen-
at very low [Ca], Ca selectivity at higher [Ca]) without dent protein kinase (protein kinase A). The binding of
grossly changing other properties of the channel, such as cAMP liberates the catalytic subunit of the enzyme, which
gating kinetics (33). While confirming that the S5–S6 re- then phosphorylates a variety of cellular proteins (47).
gion in each repeat of the Na channel is an integral part of The cardiac L-type Ca channel appears to be a particu-
the permeation pathway, these results speak only indi- larly important substrate for phosphorylation by protein
rectly to the question of how the Ca channel itself forms kinase A, based on evidence primarily from functional
the pore. More direct confirmation for the role of S5–S6 studies (44,48,49).
in Ca channel permeation comes from recent mutagene- Ca channels, when phosphorylated by ␤-adrenergic
sis studies that suggest that a ring of highly conserved stimulation, are considerably more active than in the
glutamate residues in the P loops confers Ca selectivity nonphosphorylated state, thus producing an increase in
(34 –36). the Ca current during each action potential (44,48,50).
Dihydropyridine (DHP) Ca channel ligands bind spe- An increase in Ca current can occur by several different
cifically to L-type Ca channels and, like local anesthetics means when one examines the underlying single-channel
(LA) acting on Na channels, do so in a voltage-dependent events. Macroscopic current (I) is the product of the
manner. Bean (37) demonstrated that voltage protocols number of channels in the membrane available for open-
that favor Ca channel inactivation markedly enhance the ing (N), the probability that a channel will open (Po), and
potency of channel block by nitrendipine; he invoked the the current carried by a single, open channel at that volt-
modulated receptor hypothesis to explain the results, age (i), or I ⫽ N 䡠 Po 䡠 i. Activation of the cAMP-depen-
postulating a 3,000-fold higher affinity for the inactivated dent phosphorylation pathway results in an increase in
channel availability (seen as an increase in the number of
state than for the resting state. In agreement with this
openings during a depolarizing pulse) and an increase in
interpretation, Kamp and Miller (38,39) showed that ra-
the probability of opening. The latter is manifested both
diolabeled DHPs bind much more avidly during surface
as an increased frequency of openings during a depolar-
membrane depolarization, either in intact cells or in
izing pulse (51) and as an augmentation of the number of
membrane vesicles. Numerous other studies have
ultralong openings (52). Under control conditions, sin-
pointed out the importance of the gating-related channel
gle-channel openings are quite brief and often do not
state in the binding not only of DHPs but also of benzo-
occur at all during depolarizations to various test poten-
thiazepines and phenylalkylamines to L-type Ca channels
tials. In contrast, exposure to 1 ␮M isoproterenol (a
(40,41). Virtually nothing is known about the structural
␤-adrenergic agonist) increases channel availability (as
basis for the interaction; particularly intriguing is the fact
evidenced by a reduction in the number of blank sweeps)
that photoaffinity-labeling experiments place the DHP
and increases the occurrence of a long-opening mode of
binding site at the external face of the channel (in contact
the channel.
with III-S5–S6 and with the external end of IV-S6), far
Thus, a complex chain of molecular events produces
from any putative intracellular inactivation gate (14).
an increase in L-type Ca current and ultimately enhanced
Regulation Ca delivery to the cytoplasm. The increase in Ca current
Beta-adrenergic stimulation. Sympathetic stimulation increases conduction through the AV node and increases
causes an increase in heart rate and augmented contrac- the firing rate of normal and subsidiary pacemakers. Ab-
tility. L-type Ca channels figure prominently in mediat- normal slow conduction will be accelerated to the extent
ing these effects of ␤-adrenergic stimulation (42– 44). that it is mediated by Ca channels, thus altering the like-
When an agonist binds to the cardiac ␤ receptors, a series lihood of reentry.
of events takes place. The occupied ␤ receptor activates a Recently it has become clear that G proteins can di-

February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110 131


Calcium Currents and Arrhythmias/Shorofsky and Balke

rectly influence the activity of ion channels, even in the cellular concentrations of Mg-ATP on L-type Ca currents
absence of second messengers, such as cAMP. Exposure with the predominant mechanism for regulating Ca
to a purified preparation of activated Gs ␣ subunits in- channels, ␤-adrenergic receptor activation, and cAMP-
creases the likelihood that L-type Ca channels will open in dependent phosphorylation. In a series of elegant studies,
bilayers (53). Although such cell-free experiments are they demonstrated that a direct relationship exists be-
consistent with the idea that part of the ␤-adrenergic tween intracellular Mg-ATP levels and L-type Ca current
stimulation of Ca channels is mediated directly by G pro- amplitude, which was independent of phosphorylation.
teins, the fact that cAMP and its derivatives mimic the Elucidation of the phosphorylation-independent
effects of ␤ agonists on Ca currents in intact cells implies pathway does not diminish the importance of cAMP-de-
that G proteins are not required. Nevertheless, the recog- pendent phosphorylation as the primary mechanism for
nition of direct ion channel gating by G proteins opens increasing Ca channel activity in the heart. However, it
the novel possibility of drugs that might act on this G does stimulate speculation as to the supplementary role
protein link, rather than on the more conventional sec- of Mg-ATP in linking metabolism to Ca influx. As
ond-messenger–mediated cascade. pointed out by Goldhaber et al (61), cytosolic Ca tran-
sients are reduced in ventricular myocytes during meta-
Regulation by intracellular magnesium and Mg-ATP. bolic inhibition as a result of both a reduction in L-type
Although often taken for granted, the importance of Ca current and impaired coupling between the Ca cur-
magnesium (Mg) in the control of cell physiology cannot rent and SR Ca release, even though the caffeine-releas-
be overemphasized. Whether the cell is using energy sub- able pool of Ca is actually greater than in the control
strates, pumping up or dissipating ion gradients across a condition. Similarly, Taniguchi et al (62) reported that
membrane, contracting, or synthesizing nucleic acids and Ca current depression induced by metabolic inhibition
proteins, Mg participates in the process. The vital re- could be reversed by injection of ATP. During times of
quirement for Mg and Mg-nucleotide complexes in sup- metabolic stress, the ability of channels to sense Mg-ATP
porting enzyme activity has long been recognized. More levels may serve as a protective mechanism to reduce the
recently, a growing body of work has elucidated a number metabolic demand associated with Ca removal by cutting
of mechanisms whereby ion channels can sense and re- back on the trigger for Ca release. This type of stopgap
spond to Mg and Mg nucleotides. regulation would be carried out at no energy cost to the
L-type Ca channels are subject to several modulatory cell and would function even when the phosphorylation
actions of Mg. Direct channel blocking effects of both pathway is in high gear. One situation in which this may
extracellular and intracellular Mg have been observed occur is during ischemia, when cytosolic Mg increases,
(54). Furthermore, the rate of inactivation of inward Ba Mg-ATP decreases (63), and the sympathetic nerves are
or Na currents through L-type Ca channels has been re- in overdrive. The overall picture of ion channel activity at
ported to increase, and the amplitude of the currents to this time may include inhibition of the SR Ca release
decrease, when intracellular Mg was raised from 0.3 to 3 channels (64,65), activation of ATP-sensitive K channels,
mM in frog cardiomyocytes (55). When Ca is the charge and perhaps a reduction in Ca current as a result of both
carrier, raising intracellular Mg also reduces the ampli- the decrease in Mg-ATP and the increase in Mg.
tude of Ca current in guinea pig cardiomyocytes (56). In
the frog, this effect is accentuated by prior channel phos-
phorylation, perhaps by altering the affinity of the Mg
binding site (55). T-TYPE CALCIUM CHANNELS
Although the role of Mg-ATP in the cAMP-mediated Physiological and Pathophysiological Roles
phosphorylation of Ca channels has been recognized, rel- T-type Ca channels are found in cardiac and vascular
atively little attention has been paid to investigating the smooth muscle. They can be distinguished from L-type
effects of Mg-ATP itself on L-type Ca channels. Previous channels on the basis of their distinctive biophysical char-
studies have shown that the inclusion of Mg-ATP in suf- acteristics (gating and permeation properties), relative
ficient quantities in intracellular solutions can retard the distribution, differential pharmacology, and structure
rate of “rundown” of Ca current (57). In addition, a pos- (66). Compared with L-type channels, T-type channels
itive correlation between intracellular Mg-ATP and the open at significantly more negative membrane potentials
amplitude of cardiac L-type Ca currents has been re- that are near the resting potential, rapidly inactivate
ported (58). Both these effects have been assumed to be (hence, T for transient), slowly deactivate, and have a low
related to the importance of Mg-ATP as a substrate for conductance (hence, T for tiny) (3,4,66).
phosphorylation. In the heart, T-type Ca channels are expressed in high-
Exploiting the Mg binding properties of the photosen- density in SA and AV nodal tissue (67) and Purkinje cells
sitive compounds [DM-nitrophen (59) and caged Mg], (68), and are relatively sparse or absent in most atrial and
O’Rourke et al (60) compared the effects of various intra- ventricular cells. This distribution is consistent with their

132 February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110


Calcium Currents and Arrhythmias/Shorofsky and Balke

putative role in pacemaker impulse formation. T-type nels may make to cardiac electrical and mechanical func-
channels probably do not contribute to the action poten- tion.
tial upstroke of cardiac ventricular cells, because their
Structure-function Relationship
relatively small and transient current component is dom-
Until recently, information regarding the structure of T-
inated by the coincident and much larger classical Na
type Ca channels was inferential and indirect. Augmen-
current (69). However, T-type channels may play a role in
tation of T-type current by exposure to neuraminidase
action potential generation, particularly in cell types that
(87) suggested that the molecules are glycosylated and
lack a high density of Na channels, such as vascular
that such glycosylation might modulate function. Other
smooth muscle (70).
structural features were implied from the comparison
T-type channels have been implicated in cell growth
with L-type channels. Despite important differences in
under both physiological and pathophysiological condi-
single-channel conductance (T-type channels are about
tions. T-type channels are abundant in the heart during
one third to one half as large as L-type channels) (88),
fetal development (71) and during rapid growth in the
their selectivity for Ca over monovalents is comparable,
early postnatal period (72). In addition, cardiac T-type
and both types of channel can conduct monovalent cat-
Ca channel density is increased under several conditions
ions well when divalent cations are absent (88). On the
that are associated with myocyte growth, including rats
with growth-hormone–secreting tumors (73), ventricu- other hand, T-type channels do not exhibit differences in
lar cells from failing cardiomyopathic hamsters (74), and inactivation depending on the ion that carries the charge,
endothelin-1 induced hypertrophy in neonatal rat ven- unlike L-type channels, which display prominent Ca-me-
tricular cells (75). Experimentally induced pressure over- diated inactivation (4,88). Finally, an important struc-
load hypertrophy in cats induced a re-expression of T- tural difference in the outer channel mouth regions is
type Ca channels in ventricular cells that normally do not suggested by the fact that T-type channels do not bind
have any demonstrable T-type Ca current (76). Further- dihydropyridines (3,4).
more, pretreatment with a selective inhibitor of T-type Perez-Reyes et al (89) recently cloned the first member
channels, mibefradil, reduces intimal proliferation after (␣1G) of a new family of Ca channels in rat, mouse, and
experimentally induced vascular injury in rats (77). Of human neuronal tissue that is distinct from all previously
interest, long-term treatment with mibefradil in a rat cloned Ca channels. They localized the gene encoding
model of chronic heart failure was associated with an in- human ␣1G to human chromosome 17q22 and the
crease in survival (78). Although the precise mechanism mouse locus to distal mouse chromosome 11. Functional
for this response is as yet unknown, the reappearance of expression studies in Xenopus oocytes provided compel-
the fetal phenotype may be a marker for the transition of ling evidence that ␣1G was, indeed, a T-type Ca channel
cardiac hypertrophy to heart failure and may contribute on the basis of its negative voltage range of activation, fast
to the increased incidence of sudden death and pump inactivation, slow deactivation, and tiny unitary conduc-
failure characteristic of the failing heart. tance of approximately 7.5 pS. On the heels of this land-
Compared with the well-studied L-type Ca channel, mark report, Perez-Reyes et al cloned a second member
the pharmacology of the T-type channels is less well char- of this family of T-type Ca channels (␣1H) from human
acterized. Several drugs and inorganic compounds affect heart (90). The cDNA of ␣1H was consistent with the
T-type channels, including amiloride (79) and its deriva- general structure of a voltage-gated Ca channel, and there
tive 3,4-dichlorobenzamil (80), verapamil, diltiazem was a high degree of homology with human and mouse
(81), flunarizine (81), and tetradrine. Nickel and cad- ␣1G. Specifically, both ␣1H and ␣1G are predicted to
mium also block T-type channels. However, all of these have a four-domain structure with conserved pore loops
agents also act on L-type Ca channels and, therefore, are and voltage sensors. In addition, ␣1H lacks both an iden-
not useful for dissecting out the effects of selective T-type tifiable motif for ␤ subunit binding and a Ca binding
channel modulation on cardiac electrical or mechanical domain that would confer Ca-dependent inactivation. In
activity. Recently, a novel Ca channel antagonist, mibe- contrast to human ␣1G, human ␣1H was mapped to a
fradil, has been shown to inhibit both L- and T-type different chromosome, human chromosome 16p13.3.
channels with a marked selectivity for the T-type channel Both ␣1G and ␣1H were found in brain and heart. How-
(82– 85). It preferentially affects the coronary vasculature ever, ␣1G was most abundant in brain, and ␣1H was
relative to the myocardium. In contrast to other Ca chan- most abundant in heart. Importantly, heterologous ex-
nel antagonists, its vasodilatory effects are not associated pression of ␣1H in HEK cells produced Ca currents with
with negative inotropic or conduction abnormalities, but the biophysical hallmarks of native T-type channels in-
it did cause sinus slowing. Although its clinical utility has cluding a single-channel conductance for barium of ap-
been compromised by serious drug interactions (86), it proximately 5.3 pS and a sensitivity to mibefradil at the
remains potentially useful as a research tool for a detailed concentration reported for native T-type Ca channels.
characterization of the contribution that T-type Ca chan- The identification of multiple genes encoding T-type Ca

February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110 133


Calcium Currents and Arrhythmias/Shorofsky and Balke

channels represents a major advance in the study of volt- agents that are targeted to the classical cardiac Na chan-
age-gated ion channels. The availability of recombinant nel.
T-type channels provides the tools necessary to ade- Because ICa(TTX) is permeable to Ca as well as Na, it
quately separate the contributions of T- and L-type Ca may also play a role in providing the Ca trigger for SR Ca
channels to cardiac function and to identify T-type Ca release by one of two possible mechanisms. 1) Na perme-
channel–related pathophysiology. ation of ICa(TTX) could elevate the concentration of intra-
cellular Na in a “restricted space” near the sarcolemmal
Regulation
membrane and thereby activate Ca influx by means of the
T-type Ca channels are not prominently regulated by
Na/Ca exchanger (ie, “reverse mode”) to a level that con-
␤-adrenergic mechanisms (91) but do exhibit a promi-
tributes to the trigger for SR Ca release. 2) Ca permeation
nent up-regulation with norepinephrine (92), phenyl-
of ICa(TTX) could contribute directly to the trigger for SR
ephrine (an ␣-adrenergic agonist) (93), and extracellular
Ca release.
ATP (94). Endothelin has also been found to augment Ca
Apart from this initial characterization and the dem-
entry through T-type channels in heart cells (75). These
onstration that the current is augmented by ␤-adrenergic
agents only modestly alter L-type currents. Developmen-
stimulation (96,97), no information is yet available re-
tal changes in the relative densities of L-type and T-type
garding the relative permeabilities, structure, and regula-
channel expression have been deduced (94,95) and may
tion of ICa(TTX). Importantly, ICa(TTX) may represent a
turn out to be clinically important in distinguishing
mechanisms of pediatric versus adult automaticity. In the distinct new Na (or Ca) channel isoform or a differen-
cultured chick myocyte model, developmentally young tially regulated or otherwise posttranslationally modified
cells exhibit large T-type currents (94). The mammalian Na (or Ca) channel.
correlates of these changes remain to be fully defined.
Interestingly, Nuss and Houser (76) have observed that SARCOPLASMIC RETICULUM CALCIUM
T-type currents are enhanced in hypertrophic ventricular
cells from the cat, and this may represent an electrophys-
RELEASE CHANNELS (RYANODINE
iological example of the general truism that cardiac hy- RECEPTORS)
pertrophy turns back the developmental clock by activat- Physiological and Pathophysiological Roles
ing a “program” of fetal gene expression. The Ca release channels of the sarcoplasmic reticulum
(SR) are entirely intracellular; they mediate the rapid ef-
flux of Ca from the SR lumen into the cytosol during each
TETRODOTOXIN-SENSITIVE CALCIUM cardiac cycle. In the scientific literature, SR Ca release
CHANNELS (ICa(TTX)) channels are often called ryanodine receptors because
they bind the plant-derived toxin ryanodine with nano-
Recently, a new and functionally distinct inward current
molar affinity.
component has been identified in rat ventricular cells
The SR has been generally accepted as the predominant
(96). The channels that carry this current are permeable
source of activator Ca for activation of the myofilaments
to both Na and Ca and are blocked by tetrodotoxin
and contraction in both skeletal and cardiac muscle
(TTX), hence their designation as ICa(TTX). This new
(98,99). However, ultrastructural comparison with skel-
component displays different kinetics, different voltage
etal muscle reveals that cardiac SR occupies a much
ranges for both activation and inactivation, and different
smaller fraction of the intracellular volume (100). Fur-
permeability properties from classical sodium channels
thermore, the mechanism of signal transduction cou-
and L- and T-type Ca channels. Specifically, ICa(TTX) ac-
pling the cell membrane and the terminal cisternae of the
tivates over a more negative voltage range than classical
SR is still incompletely understood. As complicating fac-
Na and T-type channels. Therefore, it may act to amplify
tors, substantial differences have been reported regarding
the depolarization delivered by Purkinje fibers or adja-
the contribution of SR Ca in atrial as opposed to ventric-
cent ventricular cells and so provide the immediate trig-
ular tissue, between mammalian and lower vertebrate
ger for the generation of the cardiac action potential. To
hearts, and even among ventricles from different mam-
the extent that ICa(TTX) triggers the cardiac action poten-
malian species (101).
tial, it may be important for cardiac arrhythmias and
their control. In addition, pharmacological modulation Structure–function Relationship
of ICa(TTX) would be predicted to affect the timing and Ca release channel proteins are the largest biological
conduction of the cardiac action potential without com- channels yet described; they are so large that the shadows
promising the action potential itself (ie, Vmax or over- of their cytoplasmic protrusions can be visualized in rou-
shoot potential). Consequently, this pharmacological tine electron micrographs as the “foot” processes that
strategy would be expected to have fewer and less severe span the gap between the SR and surface membranes. In
side effects compared with traditional antiarrhythmic contrast to membrane structures embedded in the sarco-

134 February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110


Calcium Currents and Arrhythmias/Shorofsky and Balke

lemma, functional proteins located in the SR are often titatively to account for the Ca transients recorded in
studied under experimental conditions where the integ- mammalian muscle. Thus, several lines of evidence point
rity of the cell has to be disrupted by “skinning” myocar- to the SR as the primary site of origin for activator Ca in
dial cells or by preparing vesicular membrane fractions mammalian heart muscle (113).
from the SR. At the intact cellular level, several agents that Interestingly, the SR seems to lose its capability to reg-
inhibit SR Ca release markedly reduce contractility, and ulate diastolic Ca tightly when the cell is “Ca overloaded.”
drugs that facilitate SR Ca release activate contraction. Under these conditions, the SR releases Ca spontaneously
Functional studies (10,11) suggest that in the intact cell, and asynchronously (114,115). The resultant Ca oscilla-
Ca release from the SR is triggered by the increase of tions produce delayed afterdepolarizations (DADs) ob-
intracellular free Ca concentration ([Ca]) initially pro- served when the Ca loading of the cell is excessive, as in
duced by the L-type Ca current. Such observations sup- digitalis toxicity, and may result in lethal arrhythmias
port the mechanism of Ca-induced Ca release from the (116).
SR, first described in skinned preparations (102). Several At the molecular level, biochemical techniques and re-
recent studies have advanced our understanding of the cent single channel studies have provided more direct
quantitative relationship (or the gain) between the trigger evidence to improve our understanding of SR function
Ca provided by the L-type Ca current and activator Ca and pharmacology. The Ca release channel serves as the
provided by the SR Ca release channel (103,104). They pathway for the sudden systolic delivery of Ca from the
have shown that SR Ca release is controlled by events SR to the cytosol, triggered by the increment in cytosolic
localized to the region of the L-type Ca channels and SR Ca itself, and facilitated by the presence of ATP. This
Ca release channels (ryanodine receptors), and these molecule has been purified from skeletal and cardiac
events may be very different from those seen macroscop- muscle SR (117–119) by several laboratories using proce-
ically in the whole cell. dures that exploit the high affinity of this channel to the
Several recent experimental studies provide support plant alkaloid, ryanodine. The ryanodine-binding pro-
for this local coupling of the L-type Ca channel and the SR tein has been characterized electrophysiologically as a
Ca release channel. 1) Immunocytochemical and ultra- channel passing Ca ions and, with a several-fold lower
structural studies demonstrate the close physical proxim- permeability, potassium ions. It can be blocked by the
ity and clustering of L-type Ca channels and ryanodine dye, ruthenium red. Ryanodine changes the properties of
receptors. In ventricular cells, L-type Ca channels were the channel in a complex manner, producing long-lasting
closely associated with ryanodine receptors at the T tu- openings of low conductance at nanomolar ryanodine
bules (105). In atrial cells, which lack T tubules, L-type Ca concentrations. Thus, it causes a continuous leakage of
channels and ryanodine receptors also co-localized to the Ca out of the SR, which consequently loses its ability to
sarcolemma. 2) Spontaneous, spatially localized non- store and release Ca phasically in the intact cell.
propagating transient elevations in [Ca] (ie, Ca sparks) The SR Ca release channel has been cloned from both
have been observed in resting single cardiac cells (106) skeletal and cardiac muscle (120). The cardiac cDNA is
and in intact cardiac muscle (107). 3) López-López et al 16,532 base pairs in length and encodes a protein of 4,969
(108,109) and others (110,111) have shown that SR Ca amino acids that is 66% identical with the skeletal muscle
release was controlled locally (ie, Ca sparks) by L-type Ca gene product. Four of these identical subunits are be-
current in voltage-clamped rat ventricular cells. Ca sparks lieved to assemble to form the functional Ca release chan-
evoked by membrane depolarization behave in a stochas- nel spanning the SR membrane. The molecule is domi-
tic manner (108 –110) and have a voltage and time de- nated by a huge N-terminal domain that appears to form
pendence similar to that of the L-type Ca channel (109).
the “foot processes” that span the cytosolic gap between
4) Ca sparks do not appear to be elicited by Ca entry by
the SR and the transverse tubules in skeletal muscle. The
means of the reverse mode of the Na/Ca exchanger under
putative channel resides entirely within the carboxy ter-
certain experimental conditions (109). 5) Consistent
minal end of the molecule. One interesting difference be-
with the immunocytochemical and ultrastructural stud-
ies, Ca sparks do not occur randomly throughout the cell tween the skeletal and cardiac muscle isoforms arises
but localize to the region of the T tubule and the junc- from a difference in phosphorylation sites (121); the car-
tional SR (112) where L-type Ca channels and SR Ca re- diac receptor has a multifunctional Ca-calmodulin– de-
lease channels co-localize (105). Thus, there is a prepon- pendent protein kinase site that is absent in the skeletal
derance of evidence that Ca entry by means of L-type Ca muscle isoform, hinting at possible differences in regula-
channels is the major trigger of SR Ca release and that this tory mechanisms (see below).
trigger Ca “grades” the release of SR Ca from a single or Because ryanodine acts effectively to dissociate the SR
cluster of ryanodine receptors in close proximity. Finally, from excitation– contraction coupling (122,123), it has
other sources of trigger Ca including Na/Ca exchange and been widely used as an experimental tool (101,124,125).
voltage per se have not been shown to be sufficient quan- A homologous mode of action has been proposed for

February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110 135


Calcium Currents and Arrhythmias/Shorofsky and Balke

high (millimolar) concentrations of caffeine, which in- fects of ␣-adrenergic stimulation are usually quite small
duce a contracture in heart muscle preparations owing to and differ among species (132,133).
a sudden release of Ca, causing Ca depletion of the SR. Angiotensin II receptors are present on cardiac myo-
The anthracycline cytostatic agent, doxorubicin, has been cytes, and exposure to angiotensin is known to increase
proposed to affect cardiac contractility by such a mecha- cellular IP3 levels (134). Thus, it seems plausible that the
nism (126,127). In contrast to ryanodine, caffeine and postulated arrhythmogenic effects of angiotensin II in
doxorubicin exert a number of additional effects. A spe- heart failure may at least partly be attributable to IP3 ele-
cific antiarrhythmic drug has not yet been found to act by vation, possibly as a result of a feedback effect of elevated
interfering with SR Ca release, although this pathway has intracellular [Ca] on ion channels in the surface mem-
brane.
emerged as a promising target for rational drug design
IP3 receptors are found in higher density in specialized
now that the specific molecular components are becom-
cardiac conducting tissue (135) and Purkinje cells (136),
ing increasingly well defined.
consistent with a possible role in cardiac excitability, and
Regulation in intercalated disks (137), suggesting a potential role in
Despite a wealth of biophysical data under nonphysi- intercellular communication. In addition, IP3 receptors
ological conditions (with channels reconstituted in lipid have been implicated in cell growth, differentiation, and
bilayers and conducting monovalent cations), little is cer- apoptosis (138,139).
tain about the physiological factors that regulate SR Ca
Structure–function Relationship
channel function in living cells. Presumably, Ca activates
IP3 receptors are formed of four similar or identical sub-
the channel and initiates Ca-induced Ca release. Another
units of 315,000 molecular weight (2,749 amino acid res-
regulatory factor that most surely plays a prominent
idues) arranged in a quatrefoil pattern similar to that de-
pathophysiological role in ischemia is ATP; both ATP
scribed for the ryanodine receptor. Several subunits have
and, to a lesser extent, Mg-ATP potentiate channel flux by
been cloned from cerebellar cDNA libraries (140). Hy-
increasing open probability. In ischemia, intracellular
dropathy analysis predicts that 80% of the peptide forms
ATP depletion would tend to decrease Ca release by
a large cytoplasmic domain, followed by six to eight
means of the ryanodine receptors, perhaps contributing
transmembrane domains per subunit. The membrane-
to ischemic contractile failure (which temporally pre-
spanning regions probably form the ion conductance
cedes the onset of electrical inexcitability).
pathway. It is not yet clear whether the cardiac receptor
represents the same gene product, a splice variant, or an
entirely different gene from the cerebellar isoform(s).
IP3 RECEPTORS Regulation
Physiological and Pathophysiological Roles Little is known about the regulation of IP3 receptors aside
Like ryanodine receptors, inositol triphosphate (IP3) re- from the reported effects of angiotensin II and ␣-adren-
ceptors are integral SR membrane proteins that mediate ergic stimulation. IP3 receptors are both autophosphory-
the efflux of Ca from this intracellular store. IP3 receptors lated (141) and phosphorylated by several protein ki-
are found predominantly in smooth muscle, where they nases, including PKA (142), PKC, and Ca-calmodulin-
appear to mediate drug-induced receptor-activated SR dependent kinase II (143). In addition, IP3 has two
Ca release. Although IP3 receptors are also found in the potential tyrosine phosphorylation sites (144). In T lym-
myocardium, less is known about their functional role in phocytes, tyrosine phosphorylation of IP3 appears to
heart. In contrast to observations made in other tissues modulate intracellular Ca release and may modulate in-
(128,129), it appears unlikely that Ca release in cardiac SR tracellular Ca levels (138,145). However, this putative
is primarily triggered by the second messenger IP3, al- role has yet to be demonstrated in cardiac cells.
though a modulatory role cannot be excluded. Although
it appears that IP3 is not involved in normal cardiac exci-
tation– contraction coupling, it may have a role in the CONCLUSIONS
maintenance of diastolic tension and in the physiological
modulation of contractility in response to a variety of Calcium channels are critical to normal cardiac function.
drugs and hormones. IP3 is produced by hydrolysis of the They are important in impulse generation, conduction,
membrane phospholipid phosphatidylinositol-biphos- maintaining the plateau phase of the action potential, and
phate, catalyzed by the enzyme phospholipase C. A vari- contraction. At least three different surface membrane
ety of agonists can activate phospholipase C in the heart channels exist and are preferentially distributed in vari-
(130). In particular, the molecular basis of the positive ous areas of the heart, giving a clue as to their function.
inotropic effect of ␣-adrenergic stimulation might in- The L-type Ca channel is the most abundant type in all
volve this pathway (131). Nevertheless, the inotropic ef- cardiac cells and is responsible for the entry of Ca into the

136 February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110


Calcium Currents and Arrhythmias/Shorofsky and Balke

cell that triggers contraction. Inhibitors of L-type Ca different constituents of the blood on the contraction of the heart.
channels will depress conduction through the AV node, J Physiol. 1883;4:29 – 42.
9. Fabiato A, Baumgarten CM. Methods for detecting calcium re-
thus making them useful drugs for the treatment of many lease from the sarcoplasmic reticulum of skinned cardiac cells and
supraventricular arrhythmias. T-type Ca channels are the relationships between calculated transsarcolemmal calcium
prevalent in the conduction system and are probably in- movements and calcium release. In: Sperelakis N, ed. Physiology
volved in automaticity. Enhanced function of these chan- and Pathophysiology of the Heart. Boston: The Hague, Martinus
nels may lead to an increase in automaticity, as is ob- Nijhoff Publishing, 1984:215–254.
10. Beuckelmann DJ, Wier WG. Mechanism of release of calcium
served in repetitive monomorphic ventricular tachycar-
from sarcoplasmic reticulum of guinea-pig cardiac cells. J Physiol.
dia and some forms of atrial tachycardias. The newly 1988;405:233–255.
described ICa(TTX) activates over a voltage range more 11. Näbauer M, Callewaert G, Cleeman L, Morad M. Regulation of
negative than the sodium channel and thus may be im- calcium release is gated by calcium current, not gating charge, in
portant in “boosting” or enhancing conduction and con- cardiac myocytes. Science. 1989;243:800 – 803.
traction. The main intracellular Ca channel resides in the 12. Valdeolmillos M, O’Neill SCO, Smith GL, Eisner DA. Calcium-
induced calcium release activates contraction in intact cardiac
sarcoplasmic reticulum and is responsible for the ulti- cells. Pflügers Arch. 1989;413:676 – 678.
mate release of the Ca that activates the contractile appa- 13. Hosey MM, Lazdunski M. Calcium channels: molecular pharma-
ratus. Oscillatory behavior of this channel influences the cology, structure and regulation. J Membr Biol. 1988;104:81–105.
sarcolemmal membrane, causing delayed aftercontrac- 14. Catterall WA, Striessnig J. Receptor sites for Ca channel antago-
tions and arrhythmias, such as those seen in digoxin tox- nists. Trends Pharmacol Sci. 1992;13:256 –262.
15. Miller C. 1990: Annus mirabilis of potassium channels. Science.
icity. The molecular characterization of each of these
1991;252:1092–1096.
channels has begun, and important structural data al- 16. Catterall WA. Structure and function of voltage-sensitive ion
ready has been obtained. The continued molecular deter- channels. Science. 1988;242:50 – 61.
mination of these channels and their regulation will en- 17. Takahashi M, Seagar MJ, Jones JF, et al. Subunit structure of di-
hance our knowledge of their normal function and dys- hydropyridine-sensitive calcium channels from skeletal muscle.
function in disease states, leading to the development of Proc Natl Acad Sci USA. 1987;83:5378 –5482.
18. Noda M, Shimizu S, Tanabe T, et al. Primary structure of Electro-
new therapeutic agents to treat arrhythmias and contrac- phorus electricus sodium channel deduced from cDNA sequence.
tile dysfunction. Nature. 1984;312:121–127.
19. Stühmer W, Conti F, Suzuki H, et al. Structural parts involved in
activation and inactivation of the sodium channel. Nature. 1989;
339:597– 603.
ACKNOWLEDGMENT 20. Mikami A, Imoro K, Tanabe T, et al. Primary structure and func-
The authors apologize in advance to our many colleagues whose tional expression of the cardiac dihydropyridine-sensitive calcium
outstanding scientific contributions to our understanding of channel. Nature. 1989;340:230 –233.
the general field of cardiac calcium channels could not be in- 21. Perez-Reyes E, Kim HS, Lacerda AE, et al. Induction of calcium
cluded in this concise review because of the inevitable space currents by the expression of the ␣1 subunit of the dihydropyri-
limitations. The authors also thank all past and present mem- dine receptor from skeletal muscle. Nature. 1989;340:233–236.
bers of their laboratories for the numerous and valuable contri- 22. Tanabe T, Beam KG, Adams BA, et al. Regions of the skeletal
butions to this review. muscle dihydropyridine receptor critical for excitation–
contraction coupling. Nature. 1990;346:567–572.
23. Tanabe T, Adams BA, Numa S, Beam KG. Repeat I of the dihy-
dropyridine receptor is critical in determining calcium channel
REFERENCES activation kinetics. Nature. 1991;352:800 – 803.
1. El-Sherif N, Turitto G. The long QT syndrome and torsade de 24. Hille B. Ionic Channels of Excitable Membranes, 2nd ed. Sunder-
pointes. PACE. 1999;22:91–110. land, MA: Sinauer Associates, 1992.
2. Reuter H. The dependence of slow inward current in Purkinje 25. Lawrence JH, Yue DT, Rose WC, Marban E. Sodium channel in-
fibres on the extracellular calcium-concentration. J Physiol. 1967; activation from closed states in guinea-pig ventricular myocytes.
192:479 – 492. J Physiol. 1991;443:629 – 650.
3. Bean BP. Two kinds of calcium channels in canine atrial cells. 26. Tomaselli GF, Backx PH, Marban E. Permeation in voltage-gated
J Gen Physiol. 1985;86:1–30. ion channels. Circ Res. 1993;72:491– 496.
4. Nilius B, Hess P, Lansman JB, Tsien RW. A novel type of cardiac 27. Backx PH, Yue DT, Lawrence JH, et al. Molecular localization of
calcium channel in ventricular cells. Nature. 1985;316:443– 446. an ion-binding site within the pore of mammalian sodium chan-
5. Rose WC, Balke CW, Wier WG, Marban E. Macroscopic and uni- nels. Science. 1992;257:248 –251.
tary properties of physiological ion flux through L-type Ca chan- 28. Hess P, Lansman JB, Tsien RW. Calcium channel selectivity for
nels in guinea-pig heart cells. J Physiol. 1992;456:267–284. divalent and monovalent actions. Effect of ionic radius on the
6. Marban E, Robinson SW, Wier WG. Mechanisms of arrhythmo- rates of ion entry and exit. J Gen Physiol. 1986;88:293.
genic delayed and early afterdepolarizations in ferret ventricular 29. Yue DT, Marban E. Permeation in the dihydropyridine-sensitive
muscle. J Clin Invest. 1986;78:1185–1192. calcium channel: multi-ion occupancy but no anomalous mole-
7. January CT, Riddle JM. Early afterdepolarizations: mechanism of fraction effect between Ba2⫹ and Ca2⫹. J Gen Physiol. 1990;95:
induction and block. A role for L-type Ca2⫹ current. Circ Res. 911–939.
1989;64:977–990. 30. Hess P, Tsien RW. Mechanism of ion permeation through calcium
8. Ringer S. A further contribution regarding the influence of the channels. Nature. 1984;309:453– 456.

February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110 137


Calcium Currents and Arrhythmias/Shorofsky and Balke

31. Hadley RW, Hume JR. An intrinsic potential-dependent inactiva- gle calcium channels by Cd2⫹, Mg2⫹, and Ca2⫹. Voltage and con-
tion mechanism associated with calcium channels in guinea-pig centration dependence of calcium entry into the pore. J Gen
myocytes. J Physiol. 1987;389:205–222. Physiol. 1986;88:321–347.
32. Tsien RW, Hess P, McCleskey EW, Rosenberg RL. Calcium 55. Hartzell HC, White RE. Effects of magnesium on inactivation of
channels: mechanism of selectivity, permeation, and block. Annu the voltage-gated calcium current in cardiac myocytes. J Physiol.
Rev Biophys Biophys Chem. 1987;16:265–290. 1989;94:745–767.
33. Heinemann SH, Terlau H, Stühmer W, et al. Calcium channel 56. Agus ZS, Kelepouris E, Dukes I, Morad M. Cytosolic magnesium
characteristics conferred on the sodium channel by single muta- modulates calcium channel activity in mammalian ventricular
tions. Nature. 1992;356:441– 443. cells. Am J Physiol. 1989;256:C452–C455.
34. Kim M-S, Morii T, Sun L-X, et al. Structural determinants of ion 57. Belles B, Malécot CO, Hescheler J, Trautwein W. “Run-down” of
selectivity in brain calcium channel. FEBS Letters. 1993;318:145– the Ca current during long whole-cell recordings in guinea pig
148. heart cells: role of phosphorylation and intracellular calcium.
35. Tang S, Mikala G, Bahinski A, et al. Molecular localization of ion Pflügers Arch. 1988;411:353–360.
selectivity sites within the pore of a human cardiac L-type calcium 58. Irisawa H, Kokubun S. Modulation by intracellular ATP and cyclic
channel. J Biol Chem. 1993;286:13026 –13029. AMP of the slow inward current in isolated single ventricular cells
36. Yang J, Ellinor PT, Sather WA, et al. Molecular determinants of Ca of the guinea-pig. J Physiol. 1983;338:321–337.
selectivity and ion permeation in L-type Ca channels. Nature. 59. Kaplan JH, Ellis-Davies GCR. Photolabile chelators for the rapid
1993;366:158 –161. photorelease of divalent cations. Proc Natl Acad Sci USA. 1988;85:
37. Bean BP. Nitrendipine block of cardiac calcium channels: high- 6571– 6575.
affinity binding to the inactivated state. Proc Natl Acad Sci USA. 60. O’Rourke B, Backx PH, Marban E. Phosphorylation-independent
1984;81:6388 – 6392. modulation of L-type calcium channels by magnesium-nucleotide
38. Kamp TJ, Miller RJ. Voltage-dependent nitrendipine binding to complexes. Science. 1992;257:245–248.
cardiac sarcolemmal vesicles. Mol Pharmacol. 1987;32:278 –285. 61. Goldhaber JI, Parker JM, Weiss JN. Mechanisms of excitation–
39. Kamp TJ, Miller RJ. Voltage-dependent binding of dihydropyri- contraction coupling failure during metabolic inhibition in guin-
dine calcium channel blockers to guinea-pig ventricular myocytes. ea-pig ventricular myocytes. J Physiol. 1991;443:371–386.
J Pharmacol Exp Ther. 1988;247:1240 –1247. 62. Taniguchi J, Noma A, Irisawa H. Modification of the cardiac ac-
40. Pelter D, Trautwein W, McDonald TF. Calcium channel block and tion potential by intracellular injection of adenosine triphosphate
recovery from block in mammalian ventricular muscle treated and related substances in guinea pig single ventricular cells. Circ
with organic channel inhibitors. Pflügers Arch. 1982;394:97–105. Res. 1983;53:131–139.
41. Lee KS, Tsien RW. Mechanism of calcium channel blockade by 63. Borchgrevink PC, Bergan AS, Bakoy OE, Jynge P. Magnesium and
verapamil, D600, diltiazem and nitrendipine in single dialysed reperfusion of ischemic rat heart assessed by 31P-NMR. Am J
heart cells. Nature. 1982;902:790 –794. Physiol. 1989;256:H195–H204.
42. Reuter H, Scholz H. The regulation of calcium conductance of 64. Meissner G, Henderson JS. Rapid calcium release from cardiac
cardiac muscle by adrenaline. J Physiol. 1977;246:49 – 62. sarcoplasmic reticulum vesicles is dependent on Ca2⫹ and is mod-
43. Tsien RW. Cyclic AMP and contractile activity in heart. Adv Cyclic ulated by Mg2⫹, adenine nucleotide, and calmodulin. J Biol Chem.
Nucleotide Res. 1977;8:363– 420. 1987;262:3065–3073.
44. Brum G, Osterrieder W, Trautwein W. ␤-Adrenergic increase in 65. Venkatesh N, Lamp ST, Weiss JN. Sulfonylureas, ATP-sensitive
the calcium conductance of cardiac myocytes studied with the K⫹ channels, and cellular K⫹ loss during hypoxia, ischemia, and
patch clamp. Pflügers Arch. 1984;401:111–118. metabolic inhibition in mammalian ventricle. Circ Res. 1991;69:
45. Neer EJ, Clapham DE. Roles of G protein subunits in transmem- 623– 637.
brane signaling. Nature. 1988;333:129 –134. 66. Bean BP. Classes of calcium channels in vertebrate cells. Annu Rev
46. Robishaw JD, Foster KA. Role of G proteins in the regulation of Physiol. 1989;51:367–384.
the cardiovascular system. Annu Rev Physiol. 1989;51:229 –244. 67. Hagiwara N, Irisawa H, Kameyama M. Contribution of the two
47. Tada M, Katz AM. Phosphorylation of the sarcoplasmic reticulum types of calcium currents to the pacemaker potentials of rabbit
and the sarcolemma. Annu Rev Physiol. 1982;44:401– 423. sino-atrial node cells. J Physiol. 1988;395:233–253.
48. Kameyama M, Hescheler J, Hofmann F, Trautwein W. Modula- 68. Shorofsky SR, January CT. L- and T-type Ca2⫹ channels in canine
tion of Ca current during the phosphorylation cycle in the guinea cardiac Purkinje cells. Single-channel demonstration of L-type
pig heart. Pflügers Arch. 1986;407:123–128. Ca2⫹ window current. Circ Res. 1992;70:456 – 464.
49. Hescheler J, Mieskes G, Rüegg JC, et al. Effects of a protein phos- 69. Katz AM. Molecular biology of calcium channels in the cardiovas-
phatase inhibitor, okadaic acid, on membrane currents of isolated cular system. Am J Cardiol. 1997;80:171–221.
guinea pig myocytes. Pflügers Arch. 1988;412:248 –252. 70. Bogdanov KY, Ziman BD, Spurgeon HA, Lakatta EG. L- and T-
50. Meinenz T, Nawrath H, Scholz H. Dibutyryl cyclic AMP and type calcium currents differ in finch and rat ventricular cardiomy-
adrenaline increase contractile force and 45Ca uptake in mamma- ocytes. J Mol Cell Cardiol. 1995;27:2581–2593.
lian cardiac muscle. Naunyn Schmiedebergs Arch Pharmacol. 1973; 71. Bkaily G, Sculptoresunu A, Jacques D, et al. Apamin, a highly
277:107–122. potent fetal L-type Ca2⫹ current blocker in single heart cells. Am J
51. Tsien RW, Bean BP, Hess P, et al. Mechanisms of calcium channel Physiol. 1992;262:H463–H471.
modulation by ␤-adrenergic agents and dihydropyridine calcium 72. Xu X, Best PM. Postnatal changes in T-type calcium current den-
agonists. J Mol Cell Cardiol. 1986;18:691–710. sity in rat atrial myocytes. J Physiol. 1992;454:657– 672.
52. Yue DT, Herzig S, Marban E. ␤-Adrenergic stimulation of calcium 73. Xu X, Best PM. Increase in T-type calcium current in atrial myo-
channels occurs by potentiation of high-activity gating modes. cytes from adult rats with growth hormone secreting tumors. Proc
Proc Natl Acad Sci USA. 1990;87:753–757. Natl Acad Sci USA. 1990;87:4655– 4659.
53. Yatani A, Codina J, Imoto Y, et al. A G protein directly regulates 74. Sen L, Smith TW. T-type Ca2⫹ channels are abnormal in geneti-
mammalian cardiac calcium channels. Science. 1987;238:1288 – cally determined cardiomyopathic hamster hearts. Circ Res. 1994;
1292. 75:149 –155.
54. Lansman JB, Hess P, Tsien RW. Blockade of current through sin- 75. Furukawa T, Ito H, Nitta J, et al. Endothelin-1 enhances calcium

138 February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110


Calcium Currents and Arrhythmias/Shorofsky and Balke

entry through T-type calcium channels in cultured neonatal rat 97. Balke CW, Goldman L, Aggarwal R, Shorofksy SR. Whether “slip-
ventricular myocytes. Circ Res. 1992;71:1242–1253. mode conductance” occurs. Science. 1999;284:711.
76. Nuss HB, Houser SR. T-type Ca2⫹ current is expressed in hyper- 98. Huxley AF, Taylor RE. Local activation of striated muscle fibres.
trophied adult feline left ventricular myocytes. Circ Res. 1993;73: J Physiol. 1958;144:426 – 441.
777–782. 99. Schneider MF, Chandler WK. Voltage dependent charge move-
77. Schmitt R, Clozel J-P, Iberg N, Bühler FR. Mibefradil prevents ment in skeletal muscle: a possible step in excitation– contraction
neointima formation after vascular injury in rats. Possible role of coupling. Nature. 1973;242:244 –246.
the blockade of the T-type voltage-operated calcium channel. Ar- 100. Fawcett DW, McNutt NS. The ultrastructure of the cat myocar-
terioscler Vasc Biol 1995;15:1161–1165. dium. I. Ventricular papillary muscle. J Cell Biol. 1969;42:1– 45.
78. Mulder P, Richard V, Compagnon P, et al. Increased survival after 101. Bers D. Ca influx and sarcoplasmic reticulum Ca release in cardiac
long-term treatment with mibefradil, a selective T-channel cal- muscle activation during postrest recovery. Am J Physiol. 1985;
cium antagonist, in heart failure. J Am Coll Cardiol. 1997;29:416 – 248:H366 –H381.
421. 102. Fabiato A. Calcium-induced release of calcium from the cardiac
79. Tang C-M, Presser F, Morad M. Amiloride selectively blocks the sarcoplasmic reticulum. Am J Physiol. 1983;245:C1–C14.
low threshold (T) calcium channel. Science. 1988;240:213–215. 103. Wier WG, Egan TM, López-López JR, Balke CW. Local control of
80. Suarez-Kurtz G, Kaczorowski GJ. Effects of dichlorbenzamil on excitation– contraction coupling in rat heart cells. J Physiol. 1994;
calcium currents in clonal GH3 pituitary cells. J Pharmacol Exp 474:463– 471.
Ther. 1988;247:248 –253. 104. Wier WG, López-López JR, Shacklock PS, Balke CW. Calcium
81. Tytgat J, Vereecke J, Carmeliet E. Differential effects of verapamil waves, gradients and oscillations. In: Calcium Signaling in Cardiac
and flunarizine on cardiac L-type and T-type Ca channels. Nau- Muscle Cells. Chichester: Wiley, 1995:146 –164.
nyn Schmiedebergs Arch Pharmacol. 1988;337:690 – 692. 105. Lewis-Carl SL, Felix K, Caswell AH, et al. Immunolocalization of
82. Mishra SK, Hermsmeyer K. Selective inhibition of T-type Ca2⫹ sarcolemmal dihydropyridine receptor and sarcoplasmic reticular
channels by Ro 40-5967. Circ Res. 1994;75:144 –148. triadin and ryanodine receptor in rabbit ventricle and atrium.
83. Bezprozvanny I, Tsien RW. Voltage-dependent blockade of di- J Cell Biol. 1995;129:673– 682.
verse types of voltage-gated Ca2⫹ channels expressed in Xenopus 106. Cheng H, Lederer WJ, Cannell MB. Calcium sparks: elementary
events underlying excitation– contraction coupling in heart mus-
oocytes by the Ca2⫹ channel antagonist mibefradil (Ro 40-5967).
cle. Science. 1993;262:740 –744.
Mol Pharmacol. 1995;48:540 –549.
107. Wier WG, ter Keurs HEDJ, Marban E, et al. Ca2⫹ “Sparks” and
84. Mehrke G, Zong XG, Flockerzi V, Hofmann F. The Ca2⫹ channel
waves in intact ventricular muscle resolved by confocal imaging.
blocker Ro 40-5967 blocks differently T-type and L-type Ca2⫹
Circ Res. 1997;81:462– 469.
channels. J Pharmacol Exp Ther. 1994;271:1483–1488. 108. López-López JR, Shacklock PS, Balke CW, Wier WG. Local, sto-
85. Clozel J-P, Ertel EA, Ertel SI. Discovery and main pharmacological chastic release of Ca2⫹ in voltage-clamped rat heart cells: visual-
properties of mibefradil (Ro 40-5967), the first selective T-type ization with confocal microscopy. J Physiol. 1994;480:21–29.
calcium channel blocker. J Hypertens. 1997;15:S17–S25. 109. López-López JR, Shacklock PS, Balke CW, Wier WG. Local cal-
86. Mullins ME, Horowitz BZ, Linden DH, et al. Life-threatening in- cium transients triggered by single L-type calcium channel cur-
teraction of mibefradil and beta-blockers with dihydropyridine rents in cardiac cells. Science. 1995;268:1042–1045.
calcium channel blockers. JAMA. 1998;280:157–158. 110. Cannell MB, Cheng H, Lederer WJ. The control of calcium release
87. Yee HF, Weiss JN, Langer GA. Neuraminidase selectively en- in heart muscle. Science. 1995;268:1045–1049.
hances the transient (T) calcium current in isolated cardiac myo- 111. Santana LF, Cheng H, Gómez AM, Cannell MB, Lederer WJ. Re-
cytes. Am J Physiol. 1989;256:C1267–C1272. lation between the sarcolemmal Ca2⫹ current and Ca2⫹-sparks
88. Hess P. Elementary properties of cardiac calcium channels: a brief and local control theories for cardiac excitation– contraction cou-
review. Can J Physiol Pharmacol. 1988;66:1218 –1223. pling. Circ Res. 1996;78:166 –171.
89. Perez-Reyes E, Cribbs LL, Daud A, et al. Molecular characteriza- 112. Shacklock PS, Wier WG, Balke CW. Local Ca2⫹-transients (Ca2⫹-
tion of a neuronal low-voltage-activated T-type calcium channel. sparks) originate at transverse tubules in rat heart cells. J Physiol.
Nature. 1998;391:896 –900. 1995;487:601– 608.
90. Cribbs LL, Lee J-H, Yang J, et al. Cloning and characterization of 113. Jorgensen AO, Broderick R, Somlyo AP, Somlyo AV. Two struc-
␣1H from human heart, a member of the T-type Ca2⫹ channel turally distinct calcium storage sites in rat cardiac sarcoplasmic
gene family. Circ Res. 1998;83:103–109. reticulum: an electron microprobe analysis study. Circ Res. 1988;
91. Tytgat J, Nilius B, Vereecke J, Carmeliet E. The T-type Ca channel 63:1060 –1069.
in guinea-pig ventricular myocytes is insensitive to isoproterenol. 114. Wier WG, Kort AA, Stern MD, et al. Cellular calcium fluctuations
Pflügers Arch. 1988;411:704 –706. in mammalian heart: direct evidence from noise analysis of ae-
92. Tseng GN, Boyden PA. Multiple types of Ca currents in single quorin signals in Purkinje fibers. Proc Natl Acad Sci USA. 1983;80:
canine Purkinje cells. Circ Res. 1989;65:1735–1750. 7367–7371.
93. Alvarez J, Vassort G. Properties of the low threshold Ca current in 115. Wier WG, Cannell MB, Berlin JK, et al. Cellular and subcellular
single frog atrial cardiomyocytes. A comparison with the high heterogeneity of [Ca2⫹], in single heart cells revealed by Fura-2.
threshold Ca current. J Gen Physiol. 1992;100:519 –545. Science. 1987;235:325–328.
94. Kawano S, DeHaan RL. Low-threshold current is major calcium 116. Eisner DA, Valdeolmillos M. A study of intracellular calcium os-
current in chick ventricle cells. Am J Physiol. 1989;256:H1505– cillations in sheep cardiac Purkinje fibres measured at the single
H1508. cell level. J Physiol. 1986;372:539 –556.
95. Alvarez J, Vassort G. Cardiac T-type Ca current. Pharmacology 117. Inui M, Saito A, Fleischer S. Isolation of the ryanodine receptor
and emphasis on its roles in cardiac tissues. J Cardiovasc Electro- from cardiac sarcoplasmic reticulum and identity with the feet
physiol. 1994;5:376 –393. structures. J Biol Chem. 1987;262:15637–15642.
96. Aggarwal R, Shorofsky SR, Goldman L, Balke CW. Tetrodotoxin- 118. Smith JS, Imagawa T, Ma J, et al. Purified ryanodine receptor from
blockable calcium currents in rat ventricular myocytes; a third rabbit skeletal muscle is the calcium-release channel of sarcoplas-
type of cardiac cell sodium current. J Physiol. 1997;505:353–369. mic reticulum. J Gen Physiol. 1988;92:1–26.

February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110 139


Calcium Currents and Arrhythmias/Shorofsky and Balke

119. Anderson K, Lai FA, Liu QY, et al. Structural and functional char- 133. Hescheler J, Nawrath H, Tang M, Trautwein W. Adrenoceptor-
acterization of the purified cardiac ryanodine receptor-Ca2⫹ re- mediated changes of excitation and contraction in ventricular
lease channel complex. J Biol Chem. 1989;264:1329 –1335. heart muscle from guinea-pigs and rabbits. J Physiol. 1988;397:
120. Otsu K, Willard HF, Khanna VK, et al. Molecular cloning of cDNA 657– 670.
encoding the Ca2⫹ release channel (ryanodine receptor) of rabbit 134. Baker KM, Boot GW, Dostal DE. Cardiac actions of angiotensin II:
cardiac muscle sarcoplasmic reticulum. J Biol Chem. 1990;265: role of an intracardiac renin-angiotensin system. Annu Rev
13472–13483. Physiol. 1992;54:227–241.
121. Witcher DR, Strifler BA, Jones LR. Cardiac-specific phosphoryla- 135. Borgatta L, Watras J, Katz AM, Ehrlich B. Regional differences in
tion site for multifunctional Ca/calmodulin-dependent protein calcium-release channels from heart. Proc Natl Acad Sci USA.
kinase is conserved in the brain ryanodine receptor. J Biol Chem. 1991;88:2486 –2489.
1992;267:4963– 4967. 136. Gorza L, Schiaffino S, Volpe P. Inositol 1,4,5-trisphosphate recep-
122. Sutko JL, Willerson JT, Templeton JH, et al. Ryanodine: its alter- tor in heart: evidence for its concentration in Purkinje myocytes of
ation of cat papillary muscle contractile state and responsiveness the conduction system. J Cell Biol. 1993;121:345–353.
to inotropic interventions and a suggested mechanism of action. 137. Kijima Y, Saito A, Jetton TL, et al. Different intracellular localiza-
J Pharmacol Exp Ther. 1979;209:37– 47. tion of inositol 1,4,5-trisphosphate and ryanodine receptors in
123. Sutko JL, Kenyon JL. Ryanodine modification of cardiac muscle cardiomyocytes. J Biol Chem. 1993;268:3499 –3506.
responses to potassium-free solutions. Evidence for inhibition of 138. Marks AR. Intracellular calcium-release channels: regulators of
sarcoplasmic reticulum calcium release. J Gen Physiol. 1983;82: cell life and death. Am J Physiol. 1997;272:H597–H605.
385– 404. 139. Katz AM. Calcium channel diversity in the cardiovascular system.
124. Stemmer P, Akera T. Concealed positive force-frequency relation- J Am Coll Cardiol. 1996;28:522–529.
ships in rat and mouse cardiac muscle revealed by ryanodine. Am J 140. Furuichi T, Yoshikawa S, Miyawaki A, et al. Primary structure and
Physiol. 1986;251:H1106 –H1110. functional expression of the inositol 1,4,5-trisphosphate-binding
125. Marban E, Kusuoka H, Yue DT, et al. Maximal Ca2⫹-activated protein P400. Nature. 1989;342:32–38.
force elicited by tetanization of ferret papillary muscle and whole 141. Ferris CD, Cameron AM, Bredt DS, et al. Autophosphorylation of
heart: mechanism and characteristics of steady state contractile inositol 1,4,5-trisphosphate receptors. J Biol Chem. 1992;267:
activation in intact myocardium. Circ Res. 1986;59:262–269. 7036 –7041.
126. Zorzato F, Salviati G, Facchinetti T, Volpe P. Doxorubicin induces 142. Ferris CD, Cameron AM, Bredt DS, et al. Inositol 1,4,5-trisphos-
calcium release from terminal cisternae of skeletal muscle. A study phate receptor is phosphorylated by cyclic AMP-dependent pro-
on isolated sarcoplasmic reticulum and chemically skinned fibers. tein kinase at serines 1755 and 1589. Biochem Biophys Res Com-
J Biol Chem. 1985;260:7349 –7355. mun. 1991;175:192–198.
127. Hagane K, Akera T, Berlin JR. Doxorubicin: mechanism of car- 143. Ferris CD, Huganir RL, Bredt DS, et al. Inositol trisphosphate
diodepressant actions in guinea pigs. J Pharmacol Exp Ther. 1988; receptor: phosphorylation by protein kinase C and calcium calm-
246:655– 661. odulin-dependent protein kinases in reconstituted lipid vesicles.
128. Berridge MJ, Irvine RF. Inositol triphosphate, a novel second mes- Proc Natl Acad Sci USA. 1991;88:2232–2235.
senger in cellular signal transduction. Nature. 1984;312:315–321. 144. Harnick DH, Jayaraman T, Go L, et al. The human type 1 inositol
129. Ehrlich RE, Watras J. Inositol 1,4,5-triphosphate activates a chan- 1,4,5-trisphosphate receptor from T lymphocytes: structure, localiza-
nel from smooth muscle sarcoplasmic reticulum. Nature. 1988; tion and phosphorylation. J Biol Chem. 1995;270:2833–2840.
336:583–586. 145. Jayaraman T, Ondrias K, Ondriasova E, Marks AR. Regulation of
130. Grupp G. Selective updates on mechanisms of action of positive the inositol 1,4,5-trisphosphate receptor by tyrosine phosphory-
inotropic agents. Mol Cell Biochem. 1987;76:97–112. lation. Science. 1996;272:1492–1494.
131. Mügge A. Alpha-Adrenozeptoren am Myokard: Vorkommen und 146. Balke CW, Gold MR. Excitation-contraction coupling in the nor-
funktionelle Bedeutung. Klin Wochenschr. 1985;63:1087–1097. mal and failing heart. Heart Dis Stroke. 1993;2:150 –155.
132. Jakob H, Nawrath H, Rupp J. Adrenoceptor-mediated changes of 147. Balke CW, Marbán E, O’Rourke B. Calcium channel structure,
action potential and force of contraction in human isolated ven- formation, and regulation. In: Zipes, Jalife, eds. Cardiac Electro-
tricular heart muscle. Br J Pharmacol. 1988;94:584 –590. physiology, 3rd ed. City: Philadelphia: Saunders, 1999:8 –21.

140 February 1, 2001 THE AMERICAN JOURNAL OF MEDICINE威 Volume 110

You might also like