You are on page 1of 12

J Appl Physiol

90: 1125–1136, 2001.

highlighted topics
Plasticity in Skeletal, Cardiac, and Smooth Muscle
Invited Review: Pathophysiology of cardiac muscle
contraction and relaxation as a result of alterations
in thin filament regulation
OLGA M. HERNANDEZ,1 PHILIPPE R. HOUSMANS,2 AND JAMES D. POTTER1
1
Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine,
Miami, Florida 33136; and 2Department of Anesthesiology, Mayo Foundation, Rochester, Minnesota 55905

Hernandez, Olga M., Philippe R. Housmans, and James D.


Potter. Invited Review: Pathophysiology of cardiac muscle contraction
and relaxation as a result of alterations in thin filament regulation. J
Appl Physiol 90: 1125–1136, 2001.—Cardiac muscle contraction depends
on the tightly regulated interactions of thin and thick filament proteins
of the contractile apparatus. Mutations of thin filament proteins (actin,
tropomyosin, and troponin), causing familial hypertrophic cardiomyopa-
thy (FHC), occur predominantly in evolutionarily conserved regions and
induce various functional defects that impair the normal contractile
mechanism. Dysfunctional properties observed with the FHC mutants
include altered Ca2⫹ sensitivity, changes in ATPase activity, changes in
the force and velocity of contraction, and destabilization of the contractile
complex. One apparent tendency observed in these thin filament muta-
tions is an increase in the Ca2⫹ sensitivity of force development. This
trend in Ca2⫹ sensitivity is probably induced by altering the cross-bridge
kinetics and the Ca2⫹ affinity of troponin C. These in vitro defects lead
to a wide variety of in vivo cardiac abnormalities and phenotypes, some
more severe than others and some resulting in sudden cardiac death.
familial hypertrophic cardiomyopathy; troponin; tropomyosin; actin;
myocardium

THIS BRIEF REVIEW WILL SUMMARIZE some recent insights Ca2⫹-binding subunit troponin C (TnC) of the troponin
into thin filament regulation of cardiac muscle contrac- complex, which, together with troponin I (TnI), tropo-
tion and relaxation. Several mutations in regulatory nin T (TnT), and tropomyosin (Tm), forms the regula-
proteins of the thin filament have been described that tory system of the contractile apparatus (21, 72, 79,
change the regulation of contraction and relaxation, 96). Contraction occurs when the myosin head in the
and, recently, their impact on in vivo and in vitro thick filament interacts with actin in the thin filament
contractility was studied. Within the framework of causing the two filaments to slide past each other. The
current knowledge, we will also offer some suggestions troponin complex in the thin filament regulates the
for directions for further research. actin-myosin interaction. Figure 1 summarizes the
changes in the thin filament during activation of con-
REGULATION OF CONTRACTION BY THIN
traction. In the absence of Ca2⫹, the troponin complex
FILAMENT PROTEINS
exists in a closely held conformation. TnI inhibits ac-
Contraction of vertebrate striated (skeletal and car- tin-myosin binding by interacting with actin-Tm and
diac) muscle is activated by the binding of Ca2⫹ to the fixing the troponin complex on actin through TnI’s
interaction with TnT (14, 15, 26, 68). This inhibitory
interaction of TnI with actin is completely eliminated
Address for reprint requests and other correspondence: J. D. Pot-
ter, Dept. of Molecular and Cellular Pharmacology, Univ. of Miami
when Ca2⫹ binds to the amino-terminal Ca2⫹-specific
School of Medicine, 1600 N.W. 10th Ave., Miami, FL 33136 (E-mail: sites (only site II in cardiac muscle) of TnC and the
jdpotter@miami.edu). carboxyl terminus of TnI dissociates from the actin-Tm
http://www.jap.org 8750-7587/01 $5.00 Copyright © 2001 the American Physiological Society 1125

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


1126 INVITED REVIEW

Fig. 1. Schematic diagram of the reg-


ulation of muscle contraction by tropo-
nin. TnC, TnI, and TnT, tropomyosin
C, I, and T, respectively. N, amino ter-
minus; C, carboxyl terminus (78a).

complex, which is then followed by the movement of cardiac TnT mutations have been associated with
Tm into a nonblocking position, allowing myosin to FHC: I79N, R92Q/W/L, R94L, A104V, F110I, R130C,
bind to actin (15, 26, 68). The exact function of TnT is ⌬E160, E163K/R, S179F, E244D, R278C, and a muta-
still somewhat controversial, but it is thought to stabi- tion that arises from abnormal splicing of intron 16
lize the troponin complex and to affect the Ca2⫹ sensi- (G13 A) (2, 18, 27, 36, 51, 58, 84, 86, 90, 91). Other
tivity of actomyosin ATPase activity and the level of mutations associated with FHC found in thin filament
ATPase activation and/or force development (23, 44, proteins are the cardiac Tm mutations A63V, K70T,
66, 68). The extended amino acid terminus of TnT may V95A, D175N, and E180G/V (10, 33, 57, 59, 71, 84, 87,
interact with actin and with the overlapping regions of 90); the TnI mutations R145G/Q, R162W, ⌬K183,
adjacent Tm molecules (28). TnC, an EF-hand protein, S199N, G203S, K206Q (34, 37), and an exon 8 deletion
transfers the Ca2⫹ signal, eliciting contraction to the mutant (55); and four missense mutations in actin (50,
fiber, whereas the main function of TnI is to inhibit the 65). Table 1 summarizes the clinical manifestations of
actomyosin ATPase activity (40). TnT directly inter- each of these mutations in ␣-Tm, actin, TnI, and TnT.
acts with Tm (66), TnI (66), and TnC (68). Conforma- The mechanism by which these mutations alter cardiac
tional changes in TnC affected by Ca2⫹ binding (re-
contraction and relaxation is still largely unknown,
viewed in Ref. 16) ultimately evoke the interactions
although some studies suggest that changes in myofi-
between the thin filament proteins. In addition, TnI
interaction with the other troponin subunits is affected brillar Ca2⫹ sensitivity can lead to diastolic dysfunc-
by TnI phosphorylation (1, 32, 62) and by the oxidation tion and sensitivity to dysrhythmias, which at times
state of Cys48 and Cys64 at the carboxyl terminus of cause sudden death. Tables 2 and 3 summarize the
TnI when interacting with TnT (9). results from in vitro studies and from studies on trans-
genic animals, respectively, that examine the mecha-
MUTATIONS OF THIN FILAMENT PROTEINS IN nisms of these mutations and pathologies associated
FAMILIAL HYPERTROPHIC CARDIOMYOPATHY with FHC. We will now highlight several key observa-
tions that relate to the mechanisms of FHC, in partic-
Familial hypertrophic cardiomyopathy (FHC) is an
autosomal dominant disease, characterized by left ven- ular those caused by TnT mutations.
tricular hypertrophy, myofibril disarray, and sudden TnT. Several mutations in TnT are responsible for
cardiac death (SCD). Numerous studies have shown mechanical abnormalities in the sarcomeric complex.
that FHC is caused by one of many missense or dele- Replication-defective recombinant adenovirus vectors
tion mutations in various genes that encode for ␤-my- for gene transfer of I79N and R92Q mutant cardiac
osin heavy chain (3, 13, 20, 24, 92, 93), ventricular TnT cDNAs into fully differentiated adult cardiac cells
myosin light chains 1 and 2 (12, 17, 67), myosin binding in primary culture show a regular periodic pattern of
protein C (88), titin (75), actin (50, 65), ␣-Tm (10, 33, immunolabeled TnT localization (74). In this system,
57, 59, 71, 84, 87, 90), TnT (2, 11, 36, 43, 51, 84–86, 89, unlike others mentioned below, direct force measure-
90), and TnI (34, 37, 55, 77). Whereas individuals with ments demonstrated marked desensitization of sub-
␤-myosin heavy chain mutations, in general, have a maximal Ca2⫹-activated tension (74). It was suggested
higher level of cardiac hypertrophy, those with TnT that the decrease in Ca2⫹ sensitivity may be attributed
mutations have less hypertrophy but a higher inci- to a change in the secondary structure caused by the
dence of SCD in young adults (90). To date, 15 human mutation of the incorporated protein (85) and that this

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


INVITED REVIEW 1127

Table 1. Clinical studies of FHC associated with thin filament mutations


Protein Mutation Phenotype Ref.

␣-Tm A63V Hypertrophy of left ventricle and progression to dilated cardiomyopathy; high risk of sudden 59, 94
death; ST segment depression and T-wave inversion
K70T Hypertrophy of left ventricle and progression to dilated cardiomyopathy; high risk of sudden death 59, 94
V95A Spanish-American Family; affected family members only; mild cardiac hypertrophy; high risk of 33
sudden death (11 of 13 deaths were sudden)
D175N Myocyte hypertrophy; myofibril disarray; interstitial fibrosis; varying degree of left ventricular 10
hypertrophy; favorable prognosis
Finnish Study indicating that 25% of HCM were due to ␣-Tm mutation 30
Heart failure and atrial fibrillation 59
FHC patients of diverse origin; only heterozygotes affected 83, 84
Left ventricular hypertrophy in affected individuals only 88
E180G FHC patients of diverse origin; only heterozygotes affected 84
E180V Early onset (14 yr) of hypertrophic nonobstructive cardiomyopathy; left ventricular hypertrophy 71
progressing to severe systolic dysfunction
Actin E99K FHC showing abnormal electrocardiograms and variable pathology 65
P164A De novo, early onset (17 mo) showing abnormal electrocardiograms 65
A295S FHC mostly asymptomatic; heterogeneous phenotype; abnormal electro- and echo-cardiograms; 50
septal hypertrophy and progressive dilation; ventricular tachycardia and diastolic dysfunction
A331P Case study; de novo; early onset (8 yr); abnormal electrocardiograms 65
Tnl R145G Ventricular hypertrophy associated with FHC 34
R145Q Ventricular hypertrophy 34
R162W Apical hypertrophy (apical HCM) 34
⌬K183 Apical FHC and variable ventricular hypertrophy; good prognosis 34
Variable pathology and prognosis with significant penetrance 37
S199N Caucasian family with left ventricular dysfunction, arrhythmia, dyspnoea, angina, and 49a
palpitations; apex hypertrophy, sudden death (2 of 9 deaths)
G203S Apical hypertrophy (apical HCM) 34
K206Q Ventricular hypertrophy associated with FHC 34
⌬Exon8 Associated with mild FHC (left ventricular hypertrophy) 55
TnT I79N Poor prognosis; left ventricular thickening and early sudden death; 75% penetrance; some 90
individuals with mutation unaffected
Associated with FHC 84, 85
R92Q Associated with FHC (affected individuals only); poor prognosis including left ventricular 90
thickening and early sudden death
R92W Minimal hypertrophy and low disease penetrance with high risk of sudden death and poor 51, 52
prognosis
Abnormal Q waves; larger left ventricle; impaired systolic function (asymmetric septal 36
hypertrophy); 11% with mild outflow obstruction
R92L French family with variable hypertrophy; no history of sudden death 8, 18
R94L Myocyte disarray and high risk of sudden death in the absence of myocardial hypertrophy 86
A104V Moderate left ventricular hypertrophy and risk of sudden death 58
F110I Variable hypertrophy; good prognosis; some with mutation unaffected 2
Not affected (small group, n ⫽ 5 from 2 families) 36
Gene dose effect; 4 of 9 heterozygotes with moderate HCM and 2 homozygotes with severe HCM 43
Associated with FHC; present in affected individuals 90
R130C 6 mutations in 3 families; left ventricular hypertrophy 36
⌬E160 Poor prognosis, including left ventricular thickening and early sudden death; associated with FHC; 90
present in affected individuals
Systolic dysfunction 36
E163K Associated with FHC; present in affected individuals 90
E163R Septal hypertrophy 36
S179F Case study; homozygous died at 17 yr with profound left and right ventricular hypertrophy; 26
heterozygous parents and 3 siblings unaffected
E244D Associated with FHC; present in affected individuals 90
R278C Case study of 57-yr-old male with FHC with left ventricular hypertrophy; assymptomatic at 47 10a
Associated with FHC, present in affected individuals 90
Int16(G1 3 A) Found in all affected family members 91
Poor prognosis, including left ventricular thickening with 75% penetrance; high risk of sudden 91
death
Found in affected and clinically unaffected family members with disease haplotype only 84
FHC, familial hypertrophic cardiomyopathy; HCM, hypertrophic cardiomyopathy; Tm, tropomyosin; TnI and TnT, troponin I and T, respectively.

change in structure could subsequently modify the ing and a high risk of sudden death (56, 91). A glycine
actin-Tm interaction of the myofibril (28). to alanine mutation in the intron 16 splice donor site of
The TnT mutation in the intron 16 splice donor site TnT was identified in all affected and three clinically
is associated with pronounced left ventricular thicken- unaffected adults of a family with a history of FHC

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


1128 INVITED REVIEW

Table 2. In vitro analysis of thin filament mutations associated with FHC


Protein Mutation Phenotype Condition Ref.

␣-Tm A63V Increases calcium sensitivity of force production Ad a


47
K70T Increases calcium sensitivity of force production Ada 47
D175N Increases velocity of actin translocation RFb 4
Increases calcium sensitivity Ada RFb MF 4, 7, 57
No change in force or velocity RFb 4, 7
Reduces actin affinity; conformational changes when switched from off to on state RFb 21
No effect on calcium sensitivity of force production Ada 47
E180G Weaker affinity for actin; increase velocity of actin translocation RFb 5
No change in force or velocity RFb 4
Increases calcium sensitivity RFb Adc 4, 47
Conformational changes when switched from off/on state RFb 21
Increase in force produced at submaximal calcium activation levels Ada 47
TnI R145G Reduces inhibition of actin-Tm-activated myosin ATPase activity; increases calcium RFb 11
sensitivity of ATPase regulation
Increases calcium sensitivity; suppresses maximum ATPase activity; reduces MFa 80
inhibitory effect of TnI (impairs actin-Tm-TnC interaction); reduces maximum
inhibition
R162W Reduces inhibition of actin-Tm-activated myosin ATPase activity; increases calcium RFb 11
sensitivity of ATPase regulation; stronger affinity for TnC in the presence of
calcium
TnT I79N Reduces isometric force by 25%; increases sliding speed which varied with ionic RFb 28
strength
Increases sliding speed RFe 42
Increases unloaded shortening velocity IM 78
May change actin-Tm interaction RFb 28
Reduces affinity/stability of troponin complex formation, inducing alternation of Ada 74
submaximal force generation
Reduces calcium sensitivity of force production Ada, IM 74, 78
Increases calcium sensitivity of force development SFb 54, 79
No change in calcium sensitivity or binding with Tn complex RFe 42
No change in cooperativity and maximum force SFb 54
Increases calcium sensitivity of ATPase activity without change in maximum level MFb 95
of ATPase activity; impairs inhibitory action of TnI
R92Q Time-dependent impairment of contractility (reduces fractional shortening and Adc 45
peak velocity of shortening)
a
Reduces affinity/stability of troponin complex formation, inducing alternation of Ad 74
submaximal force generation
Increases unloaded shortening velocity IM 78
Reduces calcium sensitivity of force production Ada IM 74, 78
Increases calcium sensitivity of force development SF RFb MFb
b
54, 79, 95
Possible abnormal protein folding due to altered solubility RFd 85
Potentiates maximum level of ATPase activity MFb 95
Reduces ATPase activation RFb 79
Reduces Tn-complex affinity RFb 95
No change in cooperativity and maximum force SFb 54
F110I No increase in calcium sensitivity SFb 61
Increases calcium sensitivity of force development SFb 79
Reduced activation of actin-Tm activated myosin-ATPase RFb
Increases maximum force without affecting cooperativity SFb 61
Reduced Tn affinity for actin-tropomyosin; possible abnormal protein folding due to RFd 85
altered solubility
⌬E160 Increases calcium sensitivity of ATPase activity; reduces calcium-dependent MFb 25
cooperativity
Reduces calcium sensitivity of force production IM 78
Increases TnC calcium affinity RFd 85
E163K Small change in calcium sensitivity; increases ATPase activation RFb 79
E244D Increases maximum force without affecting cooperativity; increases calcium SFb 61
sensitivity of force generation
Reduces Tn-complex affinity for actin-tropomyosin RFd 85
Potentiates maximum level of ATPase activity without calcium sensitization MFb 95
R278C Decreases maximal force and cooperativity SFb 53
Increases in calcium sensitivity of force development SFb 53, 79
Reduces ATPase inhibition RFb 79
Increases calcium sensitivity and potentiates maximum levels of ATPase activity MFb 95
Int16(G1 3 A) Reduced activation of actomyosin ATPase activity (with calcium) for both mutants RFb 56
(delEx16 and delEx15/16); reduces affinity for TnI (delEx16 and delEx15/16); no
change in calcium-independent inhibition or calcium-dependent activation
(delEx16 and delEx15/16)

Continued

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


INVITED REVIEW 1129

Table 2.—Continued
Protein Mutation Phenotype Condition Ref.
b
Increases calcium sensitivity of force generation; decreases cooperativity of SF 60
contraction-independent TnI phosphorylation (delEx16 and delEx15/16); reduces
calcium-activated maximum force in delEx15/16
Prevents switching off at low-calcium concentrations; reduces inhibition; increases RFb 69
velocity (delEx15/16)
b
Reduces activation and inhibition force including that of ATPase activation SF 79
(delEx15/16)
Reduces ATPase inhibition (delEx15/16) RFb
Reduces ATPase activation; reduced Tn affinity for actin-tropomyosin (delEx16 and RFd 85
delEx15/16)
Disruption of sarcomeric structure in 14–21% of cell; reduces calcium-activated IM 91
force of contraction
RF, reconstituted filament; IM, isolated myotubles from transfected primary quail myocytes; Ad, adenovirus infection of human protein;
SF, reconstituted skinned fiber. a Infection of rat cardiomyocytes; b human protein expressed in E. coli; c infection of feline cardiomyocytes;
d
bovine protein expressed in E. coli; e embryonic rat protein (rat I91N ⫽ Hu I79N) expressed in E. coli.

(84). This mutation results in two atypical truncated expressing low levels of truncated TnT (⬃5%) have
transcripts that translate into a short and a long pep- myocyte disarray, atrial hypertrophy, reduced ventric-
tide with a loss of 28 or 14 amino acids residues in the ular mass, and overall fewer and smaller cardiomyo-
carboxyl end plus 7 new amino acids in the shorter cytes (resulting in a smaller heart) than their wild-type
protein (90, 91). Actomyosin ATPase assays of troponin litter mates, whereas homozygous mice (twice the ex-
complexes reconstituted with the truncated proteins pression of truncated TnT) died within 24 h of birth
show that activation of the ATPase activity is greatly (81). The surviving transgenic mice showed diastolic
reduced for both mutants in the presence of Ca2⫹ (56). dysfunction and cardiac arrhythmias postexercise con-
The carboxyl terminus of TnT interacts with both TnC sistent with impaired contractility.
and TnI, and, predictably, these truncated proteins Some TnT mutations cause an increase in unloaded
have a lower affinity for TnI (56) and, when reconsti- myofilament sliding speed indicative of defective acto-
tuted in a troponin complex, for actin-Tm (85). This myosin cross-bridge activity and an increased or de-
change in the affinity of the truncated TnTs for partic- creased Ca2⫹ affinity (85). A faster myofilament sliding
ular proteins or filament complexes may account for speed was observed for I79N (42), R92Q (78), E244D,
the inadequate regulatory function. Transgenic mice and the intron 16 (G13 A) truncated TnT mutant,

Table 3. Transgenic models of FHC associated with thin filament mutations


Protein Mutation Phenotype Transgenic Ref.

␣-Tm D175N Slower contraction and relaxation; normal ventricular function but weaker response Mice 57
to exercise and ␤-adrenergic stimulation
Increased calcium sensitivity; variable myocyte disarray, hypertrophy, and fibrosis (Mouse ␣-Tm)
TnI R145G Myocyte disarray, interstitial fibrosis, premature death; increased calcium sensitivity, Mice 31
hypercontractility, diastolic dysfunction
TnT I79N No cardiac hypertrophy even with exercise, normal survival; large increase in calcium Mice 49
sensitivity of ATPase activity and force relaxation; increase in rate of force
activation and in rate of force relaxation; decreases maximal force/cross-section
area and ATPase activity; loss of sensitivity of pH-induced shifts in calcium-
dependent force and computer simulations, suggesting a decrease in apparent off-
rate of calcium from TnC and increase cross-bridge detachment rate g
R92Q Variable myocyte disarray and interstitial collagen content; reduced left ventricular Mice 41
ejection fraction; systolic and diastolic dysfunction
Normal systolic but dysfunctional diastolic; cardiomyocyte disarray; increases Mice 63
interstitial collagen
Smaller left ventricles; dose-dependent fibrosis and increased mitochondria; increases Mice 82
atrial natriuretic factor and ␤-MHC mRNAs; increased basal sarcomeric activation;
impaired relaxation and shorter sarcomere lengths; hypercontractility and diastolic
dysfunction
Int16(G1 3 A) Diastolic dysfunction and sudden death; cardiac arrhythmias postexercise; Rat (delEx16) 18
myofibrillar disarray, no hypertrophy (delEx15/16 produced no transgenics; high
frequency of resorption and still births)
Cardiomyopathy, smaller hearts, diastolic and milder systolic dysfunction Mice (mouse 81
␣-Tm)
Myofibril disarray, reduced number and size of myocytes
Expression ⬎5% lethal (delEx15/16)
Data are for human TnT unless otherwise stated.

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


1130 INVITED REVIEW

whereas ⌬E160 appears to decrease unloaded sliding When native TnT is replaced with mutant I79N TnT,
speed of the filament (85). The shift in sliding speed of an increased filament speed is not unexpected. Alter-
the I79N, R92Q, and ⌬E160 TnT mutants is also ac- ations in the actin interaction surface may allosteri-
companied by changes in Ca2⫹ sensitivity of force or cally change actomyosin binding and cross-bridge ki-
ATPase activity (reviewed in Ref. 85). netics. If changes in actomyosin interactions increase
The TnT-I79N mutation is of special interest because the rate of cross-bridge dissociation from actin, peak
it poses the highest risk of SCD in young adults (90). At isometric force will be reduced and maximal filament
present, there is no clear understanding of why this sliding velocity increases. The increased Ca2⫹ sensitiv-
TnT-I79N mutation is associated with increased SCD. ity of force in reconstituted skinned cardiac fibers in
Several investigators have demonstrated an in vitro which native TnT was replaced with TnT-I79N (79)
effect of the TnT-I79N mutation on the contractile and in skinned cardiac fibers of transgenic mice (49) is
properties of cardiac and skeletal muscle with conflict- not readily explained by changes in cross-bridge kinet-
ing results. Lin et al. (42), using rat cardiac TnT ics alone. The results from skinned cardiac fiber stud-
containing a mutation in an equivalent position to the ies, flash photolysis force transients, and force-pCa
TnT-I79N mutation in humans, showed that this mu- relations (49) were reproduced by computer simula-
tant TnT had a normal affinity for actin-Tm and con- tions that integrate Ca2⫹ binding to intracellular Ca2⫹
ferred normal Ca2⫹ sensitivity to actomyosin ATPase buffers and predict force generation based on a modi-
activity. The regulated thin filaments, however, moved fication of the model of Robertson et al. (73) and a
50% faster over heavy meromyosin (HMM) than con- two-state cross-bridge model (48) that utilized an ex-
trol filaments in an in vitro motility assay. Additional ponential dependence of the TnC off-rate for Ca2⫹
measurements utilizing the same system revealed that (Ca2⫹-specific site II) on force (29, 38). Simulations
HMM exerted reduced isometric force on single thin postulate that an increase in the apparent rate of
filaments reconstituted with the TnT-I79N mutant actomyosin cross-bridge detachment (g) was not suffi-
(28). Sweeney et al. (78) reported that TnT-I79N trans- cient to account for all experimental observations. The
fected quail skeletal muscle myotubules had decreased combination of an increased affinity of TnC for Ca2⫹
Ca2⫹ sensitivity of force production, whereas the un- and an increase in g reproduced all experimental ob-
loaded shortening velocity was increased by about two- servations in skinned fibers that contain the human
fold. An embryonic isoform of rat TnT-I79N expressed TnT-I79N. An increased g was also inferred by others
in adult rat cardiac myocytes induced a decreased Ca2⫹ from their in vitro motility assays (28, 42) or TnT-I79N
sensitivity of isometric force (74). Our results on transfected myotubes (28), and a consistent picture
TnT-I79N reconstituted porcine fibers (79) are in ac- regarding this mutation is emerging based on results
cord with Morimoto et al. (54), who demonstrated that from several different approaches. Moreover, the cross-
TnT-I79N reconstituted skinned rabbit trabeculae in- bridge effects brought about by this mutation are dis-
creased the Ca2⫹ sensitivity of contraction. A recent tinct from the calcium effects, and it is possible that the
study confirmed increased Ca2⫹ sensitivity of myofi- former arise from an alteration in the interactions
brillar ATPase activity of TnT-I79N reconstituted rab- between TnT, Tm, and actin, whereas the latter arise
bit cardiac myofibrils (95). Part of the disparity is likely from altered TnT and TnC interactions. The predicted
due to the different in vitro assays used by these increased affinity of TnC for Ca2⫹ still requires exper-
investigators, illustrating the need to study the effect imental verification.
of the mutations in an in vivo system. Compared with wild-type TnT, relaxation of force in
Until now, a transgenic model for the TnT-I79N has TnT-I79N containing myocardium is slowed by a de-
not been reported, although other mutant TnT trans- creased affinity of TnC for Ca2⫹; however, this effect is
genic mice have been described (Table 3). Miller et al. somewhat attenuated by an increase in g. The twitch
(49) examined a wild-type (Tg-WT) and two I79N- contraction operates from a pCa range of ⬃7.7 at rest
transgenic mouse lines (Tg-I79N) of human cardiac to a peak systolic value of 5.7. With the use of these
TnT (hcTnT) driven by a murine ␣-myosin heavy chain pCa values, simulated peak twitch force in steady-state
promoter. The levels of expression of either Tg-WT or conditions in fibers containing Tg-I79N was shown to
Tg-I79N, relative to mouse cTnT, were 71% (WT) or be higher than in fibers that contained Tg-WT (Fig. 2).
35% and 52% in the two I79N lines. Extensive charac- Simulations for intact living myocardium further pre-
terization of the Tg-I79N lines compared with Tg-WT dict a faster rise of force, a slower isometric relaxation,
and/or non-Tg mice demonstrated the following: nor- and an increased residual force or resting tension at
mal survival and no cardiac hypertrophy even with the onset of the next contraction. If twitch amplitudes
chronic exercise in all groups, large increases in the in both Tg-WT and Tg-I79N are the same or are nor-
Ca2⫹ sensitivity of ATPase activity and force in malized, isometric relaxation of Tg-I79N myocardium
skinned fibers, a substantial increase in the rate of is slower than in the WT, as observed in isovolumetri-
force activation and a small increase in the rate of force cally contracting isolated heart (Fig. 2) (35). The higher
relaxation in flash photolysis experiments, signifi- basal contractile state, the increased rate of contrac-
cantly lower maximal force/cross-sectional area and tion, and the slower relaxation (35) in TnT-I79N myo-
ATPase activity, and a loss of sensitivity to pH-induced cardium would have the following consequences. First,
shifts in the Ca2⫹ dependence of force correlated with the contractile reserve is decreased, and an increase in
hcTnT-I79N expression levels (49). heart rate and/or of contractility, such as after isopro-

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


INVITED REVIEW 1131

quired for its regulatory properties, or 4) produces


other effects. At least one of these conditions appears to
have an impact on thin filament regulation. Skinned
cardiac fiber reconstituted with the E244D TnT in-
creased the maximum force and increased the Ca2⫹
sensitivity of the force generated compared with wild-
type TnT (61). The E244D mutation is located in a
region of the carboxyl terminus of TnT that interacts
with Tm, and changes in TnT-Tm interaction may alter
actomyosin regulation. Indeed, the E224D mutation
was found to have a lower affinity for actin-Tm than
wild-type TnT (85). This mutation has been found in
affected individuals of families with FHC having poor
prognosis (90).
The F110I TnT mutation is associated with variable
cardiac morphologies and prognosis (2, 36, 43, 90). This
mutation, located in a region of TnT that interacts with
actin and Tm (16), also increased the maximum force
in skinned muscle fibers but did not influence the Ca2⫹
sensitivity of force generation (61).
Another TnT transgenic animal model reported by
Oberst et al. (63) was generated for the human cardiac
TnT-R92Q mutation using a murine cTnT promoter.
The level of expression in transgenic lines (wild-type
and R92Q) varied from 1 to 10% of the total cTnT pool.
Diastolic dysfunction and myocyte disarray were ob-
served in the mutant mice but not in wild-type mice
Fig. 2. Simulation of Ca2⫹ transients and force during twitches. (63). The same TnT-R92Q mutant was expressed in
Time courses are shown of the intracellular Ca2⫹ concentration transgenic mice, in which the level of R92Q expression
transient (top) and of the corresponding force (middle) for isometric varied from 30 to 92% (82). A murine cTnT cDNA and
twitches of intact ventricular myocardium during repetitive stimu- a rat ␣-myosin heavy chain promoter were used in the
lation at 400-ms intervals in steady-state control conditions for the
TnT transgenic wild-type (WT; solid lines) and for the TnT trans-
latter study. The R92Q hearts had a significant induc-
genic I79N mutation (dashed lines). Bottom: normalized force traces tion of atrial natriuretic factor and ␤-myosin heavy
for the same twitches, demonstrating a slower isometric relaxation chain transcripts, interstitial fibrosis, and mitochon-
in I79N myocardium than in WT. The TnT-I79N mutant data were drial pathology. Isolated cardiac myocytes from these
obtained by decreasing the apparent dissociation rate of Ca2⫹ from R92Q transgenic mice had increased basal sarcomeric
TnC from 300 to 88 s⫺1 and increasing the cross-bridge apparent
dissociation constant from 10 to 20 s⫺1. activation, impaired relaxation, and shorter sarcomere
lengths. Isolated working hearts showed hypercontrac-
tility and diastolic dysfunction, both of which are com-
terenol administration, would jeopardize relaxation mon findings in patients with FHC (82). Transgenic
and cause diastolic dysfunction. An increased contrac- mice expressing 30%, 67%, and 92% R92Q TnT show a
tility and heart rate would increase diastolic intra- dose-related induction and progression of atrial hyper-
cellular Ca2⫹ concentration, cause intracellular Ca2⫹ trophy but a constant and age-independent decrease in
overload, and dysrhythmias. These predictions seem to ventricular mass. Analogous to the cardiomyocytes and
hold true for Tg-I79N mice challenged with isoproter- hearts of the truncated TnT transgenic mice (above),
enol (35), which demonstrated an impaired inotropic the R92Q myocytes and hearts are smaller than their
response, relaxation impairment, and fatal dysrhyth- wild-type counterparts. The left ventricle contains both
mias. It is likely that these mechanisms contribute to hypertrophied and dying myocytes with abnormal his-
the mortality observed in patients with a TnT-I79N- topathologies, which are particularly prevalent in the
based FHC, especially in stimulated states of contrac- higher expressing mice (82). The mice expressing 67%
tility such as seen during vigorous exercise or during R92Q displayed higher heart rates and diastolic dys-
inotropic interventions. function, prolonged contraction and relaxation, and
The E244D mutation of TnT also appears to increase possible early activation of ATPase activity. The dia-
Ca2⫹ sensitivity (61). The TnT-E244D mutation has a stolic dysfunction is attributed to an increase in Ca2⫹
shorter side chain due to the absence of a methyl group sensitivity, as demonstrated in in vitro studies by Mo-
in the aspartic acid residue, yet the charge is con- rimoto et al. (54), Szczesna et al. (79), and Yanaga et al.
served. The spatial difference due to the lack of a (95) or a deregulation of intracellular Ca2⫹. It is rea-
methyl group becomes significant if it 1) alters TnT sonable to assume that a change to a more polar amino
secondary structure, 2) alters protein-protein interac- acid residue in the Tm-interacting domain of TnT will
tion within the troponin complex, 3) induces or elimi- affect troponin interactions as shown by a reduction of
nates structural changes in the troponin complex re- the stability of troponin complex formation containing

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


1132 INVITED REVIEW

R92Q (74, 79). In a recent paper (41), a murine ␣-my- and a risk of sudden death in individuals with FHC
osin heavy chain promoter was used to produce a (59, 94), was found to increase the Ca2⫹ sensitivity of
transgenic mouse expressing human cardiac TnT- force production in the isometric force recordings of
R92Q. The level of protein expression was relatively single adult rat myocytes that expressed this mutant
low, and the mutant mice demonstrated myocyte dis- ␣-Tm by means of an adenovirus vector (47). Three
array and excess interstitial collagen. Interestingly, other mutations associated with FHC, K70T, E180G,
none of these transgenic mice demonstrated significant and D175N, showed a smaller increase in Ca2⫹ sensi-
cardiac hypertrophy. tivity of force production, with the smallest increase
Exchange studies of TnT-R278C vastly increased reported for the D175N mutation (4, 47). The D175N
Ca2⫹ sensitivity, more than any other TnT mutation Tm mutation is associated with variable prognosis and
known hitherto (53, 79). Reconstituted skinned cardiac pathophysiologies; however, favorable prognosis in
fibers containing the R278C mutation in the globular some cases have been documented (10, 59, 87). The
carboxyl terminal domain of TnT greatly increases the D175N ␣-Tm mutation in transgenic mice causes im-
Ca2⫹ sensitivity of force development (79). TnT resi- paired contractility and relaxation and enhances Ca2⫹
dues 188–288, characterized as the T2 proteolytic frag- sensitivity (57). The increases in Ca2⫹ sensitivity of
ment having an unusually large number of positively force production, as reported by Michele et al. (47)
charged residues, have been reported to interact with (A63V ⬎ K70T ⬎ E180G ⬎⬎ D175N ⫽ wild type),
Tm, TnI, and TnC (reviewed in Refs. 16 and 70). The appear to be directly related to the severity of the
interaction of the TnT carboxyl terminus with Tm disease. Several lines of transgenic mice overexpress-
appears to be Ca2⫹ dependent (76). Mutation of a basic ing the D175N Tm mutation at ⬍40% and 60% of total
amino acid to a neutral in the carboxyl end of TnT may Tm myofibrillar content suggest that the degree of
induce steric changes within the troponin complex, physiological abnormalities associated with the muta-
altering the contacts between the regulatory subunits tion is dependent on the amount of mutant ␣-Tm in-
in response to Ca2⫹. Ohtsuki’s group (53) showed that corporated in the filaments (57). Mice expressing 60%
this TnT mutation induced a higher level of sub-half- of the mutant protein had reduced contraction and
maximum force as a result of a decrease in maximum relaxation rates during exercise compared with the
force and cooperativity, although only a slight increase mice expressing ⬍40% of the D175N protein or com-
in Ca2⫹ sensitivity was observed. pared with the wild-type littermates (57). The abnor-
TnI. The R145G TnI mutation identified in patients mal cardiac performance of the D175N transgenic mice
with FHC constitutes a change in charge in an evolu- was attributed to an increase in Ca2⫹ sensitivity,
tionarily conserved residue (34). We observed impair- which was observed in skinned fiber preparations. Pro-
ment in both ATPase assays and in the relaxation of tein-protein interactions within the troponin-Tm com-
force in skinned cardiac muscle preparations incorpo- plex may mediate the regulatory dysfunction because
rating the R145G mutation (Lang R, Zhao J, Hous- both D175N and E180G are within a region considered
mans PR, and Potter JD, unpublished observations). to interact with TnT, whereas A63V and K70T are in
Another group reported that actin-Tm-activated myo- repeat sequences and may alter Tm-actin interaction
sin S1 ATPase assays using troponin complexes recon- (reviewed in Refs. 6, 16, 70).
stituted with R145G TnI revealed no change in activa- ␣-Actin. In 1999, the first ␣-actin mutation linked to
tion but induced minimal inhibition and increased the FHC was reported in a Danish family. The A295S
Ca2⫹ sensitivity of regulation compared with wild-type ␣-actin mutation was found in 13 individuals with
TnI complexes (11). The switch from a positively different disease phenotypes and a variable age of
charged to a neutral residue in the R145G TnI muta- disease onset (as early as 4 yr old) (50). Only one
tion occurs in a region that interacts with the amino- individual was asymptomatic, whereas 3 of the 13 with
terminal domain of TnC and possibly actin (reviewed the mutation showed more severe phenotypes, which
in Ref. 16) and, consequentially, may alter the dynam- included pronounced hypertrophy, ventricular tachy-
ics or influence the regulatory mechanism of the tropo- cardia, and diastolic dysfunction (50). The only other
nin complex. We detected a slight increase in the ␣-he- report identifying cardiac actin mutations linked to
lical content in circular dichroism studies of the R145G FHC is a recent study describing the phenotypes of
mutant protein in addition to an increase in the affinity family members of three individuals with different
between TnC and R145G TnI in the presence of Ca2⫹ actin mutations. In this study, two de novo mutations
and Mg2⫹ vs. Mg2⫹ alone (Lang R, Zhao J, Housmans (P164A and A331P) were associated with early-onset
PR, and Potter JD, unpublished observations). The (8 yr and 17 mo of age, respectively) left ventricular
large change in the ability of this mutant TnI to inhibit hypertrophy and repolarization abnormalities (65).
contraction, combined with the increased Ca2⫹ sensi- The third actin mutation, E99K, was found to be auto-
tivity of contraction, would likely lead to severely im- somal dominant, with affected individuals having a
paired diastolic function. later onset of the disease, variable hypertrophy, and
␣-Tm. Six missense mutations in the cardiac ␣-Tm diastolic dysfunction (65). FHC-linked mutations in
have been linked to FHC (Table 1). In vitro analysis of other genes were excluded in both studies. The four
several missense mutations in the cardiac ␣-Tm gene missense cardiac actin mutations are located near or
have also shown increased Ca2⫹ sensitivity (see Table within regions that interact with other actin molecules,
2). The A63V mutation, associated with poor prognosis troponin components, or the myosin head and may

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


INVITED REVIEW 1133

either destabilize the sarcomeric structure or alter the stasis, and/or from other processes. It remains to be
force generation during the cross-bridge cycle of con- determined whether the occurrence of SCD results
traction. Defective myofilament regulation or function from an altered thin filament environment or whether
may be due to reduced or enhanced filament protein- additional primary affects of the point mutation act at
protein interaction caused by replacement of a neutral the level of the cardiac myocyte. Future work in this
or acidic amino acid with a polar or basic amino acid area should provide insights into the molecular mech-
(A295S and E99K missense mutations); a faulty actin- anisms responsible for the induction and progression of
actin interaction may affect calcium signal transduc- FHC.
tion by altering the Tn-Tm-actin interaction. A change
in the secondary structure of actin, possibly induced by This work was supported by National Institutes of Health Grants
AR-45391 (to J. D. Potter), HL-42325 (to J. D. Potter), and GM-36365
replacement or addition of the ␣-helical-destabilizing (to P. R. Housmans).
amino acid proline (as in the case of P164A and
A331P), could also conceivably alter the native envi-
REFERENCES
ronment in actin that interacts with itself or other
filament components and ultimately affect force gener- 1. Al-Hillawi E, Bhandari DG, Trayer HR, and Trayer IP. The
effects of phosphorylation of cardiac troponin-I on its interac-
ation and regulation. These cardiac actin mutations tions with actin and cardiac troponin-C. Eur J Biochem 228:
were only recently identified; therefore, little is known 962–970, 1995.
about their functional consequences. 2. Anan R, Shono H, Kisanuki A, Arima S, Nakao S, and
Tanaka H. Patients with familial hypertrophic cardiomyopathy
caused by a Phe110Ile missense mutation in the cardiac troponin
SUMMARY AND CONCLUSIONS
T gene have variable cardiac morphologies and a favorable prog-
nosis. Circulation 98: 391–397, 1998.
Thin filament proteins provide for a very tight reg- 3. Anan R, Shono H, and Tei C. Novel cardiac beta-myosin heavy
ulation of cardiac contraction and relaxation. Each of chain gene missense mutations (R869C and R870C) that cause
several mutations profoundly impacted basal contrac- familial hypertrophic cardiomyopathy. Hum Mutat 15: 584,
tility and the time course of relaxation. In most muta- 2000.
tions, Ca2⫹ sensitivity of force development was in- 4. Bing W, Knott A, Redwood C, Esposito G, Purcell I,
Watkins H, and Marston S. Effect of hypertrophic cardiomy-
creased; in some cases, filament sliding velocity was opathy mutations in human cardiac muscle ␣-Tm (Asp175Asn
also increased (Table 2). The myofilament is a highly and Glu180Gly) on the regulatory properties of human cardiac
interacting system in which even a slight change in troponin determined by in vitro motility assay. J Mol Cell Car-
interaction between any of the filament components diol 32: 1489–1498, 2000.
5. Bing W, Redwood CS, Purcell IF, Esposito G, Watkins H,
may alter the Ca2⫹ sensitivity of force generation. TnT and Marston SB. Effects of two hypertrophic cardiomyopathy
plays a key role in maintaining the Ca2⫹-dependent mutations in alpha-Tm, Asp175Asn and Glu180Gly, on Ca2⫹
competition of TnC and actin-Tm for the inhibitory regulation of thin filament motility. Biochem Biophys Res Com-
region of TnI (64). The biophysical consequences of one mun 236: 760–764, 1997.
amino acid substitution in a critical part of TnT will 6. Bonne G, Carrier L, Richard P, Hainque B, and Schwartz
K. Familial hypertrophic cardiomyopathy: from mutations to
alter the balance of power between the competing ac- functional defects. Circ Res 83: 580–593, 1998.
tin-Tm and TnC sites for the inhibitory region of TnI. 7. Bottinelli R, Coviello DA, Redwood CS, Pellegrino MA,
McKay et al. (46) illustrated the precision of filament Maron BJ, Spirito P, Watkins H, and Reggiani C. A mutant
protein interactions by describing a skeletal TnC mu- Tm that causes hypertrophic cardiomyopathy is expressed in
tant (without Ca2⫹-binding site I, similar to the cardiac vivo and associated with an increased calcium sensitivity. Circ
Res 82: 106–115, 1998.
TnC in that cardiac TnC has no functional Ca2⫹ site I), 8. Charron P, Dubourg O, Desnos M, Isnard R, Hagege A,
with a “delicate energetic balance” between Ca2⫹ and Millaire A, Carrier L, Bonne G, Tesson F, Richard P,
TnI, binding to the TnC regulatory domain in the Bouhour JB, Schwartz K, and Komajda M. Diagnostic value
“open” state. It is therefore not surprising that several of electrocardiography and echocardiography for familial hyper-
trophic cardiomyopathy in a genotyped adult population. Circu-
TnT mutations, and mutations of other thin filament lation 96: 214–219, 1997.
proteins, manifest themselves by altering both cross- 9. Chong PC and Hodges RS. Photochemical cross-linking be-
bridge kinetics (actomyosin dependent), and the Ca2⫹ tween rabbit skeletal troponin subunits. Troponin I-troponin T
affinity of TnC, as recently proposed (49). Impaired interactions. J Biol Chem 257: 11667–11672, 1982.
contractile function due to thin filament mutations 10. Coviello DA, Maron BJ, Spirito P, Watkins H, Vosberg HP,
Thierfelder L, Schoen FJ, Seidman JG, and Seidman CE.
would most likely contribute to the progression of com- Clinical features of hypertrophic cardiomyopathy caused by mu-
pensatory hypertrophy (FHC). tation of a “hot spot” in the ␣-tropomyosin gene. J Am Coll
Mutations in thin filament proteins give rise to vary- Cardiol 29: 635–640, 1997.
ing degrees of hypertrophy and incidences of SCD 10a.Elliott PM, D’Cruz L, and McKenna WJ. Late-onset hyper-
trophic cardiomyopathy caused by a mutation in the cardiac
(Table 1). One of the most puzzling questions regarding troponin T gene. N Engl J Med 341: 1855–1856, 1999.
the pathogenesis of TnT-related FHC is the mecha- 11. Elliott K, Watkins H, and Redwood CS. Altered regulatory
nism of SCD. Individuals with TnT-related FHC tend properties of human cardiac troponin I mutants that cause
to exhibit mild or no ventricular hypertrophy, yet ex- hypertrophic cardiomyopathy. J Biol Chem 275: 22069–22074,
perience a high frequency of sudden death at an early 2000.
12. Epstein ND. The molecular biology and pathophysiology of
age. These events could result from diastolic dysfunc- hypertrophic cardiomyopathy due to mutations in the ␤ myosin
tion, from the lack of pH sensitivity of Ca2⫹ sensitivity heavy chains and the essential and regulatory light chains. Adv
of force, from alterations in intracellular Ca2⫹ homeo- Exp Med Biol 453: 105–114, 1998.

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


1134 INVITED REVIEW

13. Epstein ND, Cohn GM, Cyran F, and Fananapazir L. Dif- Kuusisto J. The cardiac ␤-myosin heavy chain gene is not the
ferences in clinical expression of hypertrophic cardiomyopathy predominant gene for hypertrophic cardiomyopathy in the Finn-
associated with two distinct mutations in the beta-myosin heavy ish population. J Am Coll Cardiol 32: 1709–1716, 1998.
chain gene. A 908Leu3 Val mutation and a 403Arg3 Gln muta- 31. James J, Zhang Y, Osinska H, Sanbe A, Klevitsky R,
tion. Circulation 86: 345–352, 1992. Hewett TE, and Robbins J. Transgenic modeling of a cardiac
14. Farah CS, Miyamoto CA, Ramos CH, da Silva AC, Quaggio troponin I mutation linked to familial hypertrophic cardiomyop-
RB, Fujimori K, Smillie LB, and Reinach FC. Structural and athy. Circ Res 87: 805–811, 2000.
regulatory functions of the NH2- and COOH-terminal regions of 32. Jaquet K, Lohmann K, Czisch M, Holak T, Gulati J, and
skeletal muscle troponin I. J Biol Chem 269: 5230–5240, 1994. Jaquet R. A model for the function of the bisphosphorylated
15. Farah CS and Reinach FC. The troponin complex and regu- heart-specific troponin-I N-terminus. J Muscle Res Cell Motil 19:
lation of muscle contraction. FASEB J 9: 755–767, 1995. 647–659, 1998.
16. Filatov VL, Katrukha AG, Bulargina TV, and Gusev NB. 33. Karibe A, Bachinski LL, Arai AE, Tripodi D, Roberts R,
Troponin: structure, properties, and mechanism of functioning. and Fananapazir L. Familial hypertrophic cardiomyopathy
Biochemistry (Mosc) 64: 969–985, 1999. caused by a novel ␣-tropomyosin mutation (Val95Ala) is associ-
17. Flavigny J, Richard P, Isnard R, Carrier L, Charron P, ated with mild cardiac hypertrophy but a high incidence of
Bonne G, Forissier JF, Desnos M, Dubourg O, Komajda M, sudden death (Abstract). Circulation 100: I-619, 1999.
Schwartz K, and Hainque B. Identification of two novel mu- 34. Kimura A, Harada H, Park JE, Nishi H, Satoh M, Taka-
tations in the ventricular regulatory myosin light chain gene hashi M, Hiroi S, Sasaoka T, Ohbuchi N, Nakamura T,
(MYL2) associated with familial and classical forms of hypertro- Koyanagi T, Hwang TH, Choo JA, Chung KS, Hasegawa A,
phic cardiomyopathy. J Mol Med 76: 208–214, 1998. Nagai R, Okazaki O, Nakamura H, Matsuzaki M, Saka-
18. Forissier JF, Carrier L, Farza H, Bonne G, Bercovici J, moto T, Toshima H, Koga Y, Imaizumi T, and Sasazuki T.
Richard P, Hainque B, Townsend PJ, Yacoub MH, Faure Mutations in the cardiac troponin I gene associated with hyper-
S, Dubourg O, Millaire A, Hagege AA, Desnos M, Komajda trophic cardiomyopathy. Nat Genet 16: 379–382, 1997.
M, and Schwartz K. Codon 102 of the cardiac troponin T gene 35. Knollmann BC, Blatt SA, Horton K, deFreitas F, Miller T,
is a putative hot spot for mutations in familial hypertrophic Bell M, Housmans PR, Weissman NJ, Morand M, and
cardiomyopathy. Circulation 94: 3069–3073, 1996. Potter JD. Inotropic stimulation induces cardiac dysfunction in
19. Frey N, Franz WM, Gloeckner K, Degenhardt M, Muller M, transgenic mice expressing a troponin T (I79N) mutation linked
Muller O, Merz H, and Katus HA. Transgenic rat hearts to familial hypertrophic cardiomyopathy. J Biol Chem In press.
expressing a human cardiac troponin T deletion reveal diastolic 36. Koga Y, Toshima H, Kimura A, Harada H, Koyanagi T,
dysfunction and ventricular arrhythmias. Cardiovasc Res 47: Nishi H, Nakata M, and Imaizumi T. Clinical manifestations
254–264, 2000. of hypertrophic cardiomyopathy with mutations in the cardiac
20. Geisterfer-Lowrance AA, Kass S, Tanigawa G, Vosberg ␤-myosin heavy chain gene or cardiac troponin T gene. J Card
HP, McKenna W, Seidman CE, and Seidman JG. A molec-
Fail 2: S97–103, 1996.
ular basis for familial hypertrophic cardiomyopathy: a beta car-
37. Kokado H, Shimizu M, Yoshio H, Ino H, Okeie K, Emoto Y,
diac myosin heavy chain gene missense mutation. Cell 62: 999–
Matsuyama T, Yamaguchi M, Yasuda T, Fujino N, Ito H,
1006, 1990.
and Mabuchi H. Clinical features of hypertrophic cardiomyop-
21. Gergely J. Molecular switches in troponin. Adv Exp Med Biol
athy caused by a Lys183 deletion mutation in the cardiac tropo-
453: 169–176, 1998.
nin I gene. Circulation 102: 663–669, 2000.
22. Golitsina N, An Y, Greenfield NJ, Thierfelder L, Iizuka K,
38. Landesberg A and Sideman S. Coupling calcium binding to
Seidman JG, Seidman CE, Lehrer SS, and Hitchcock-
troponin C and cross-bridge cycling in skinned cardiac cells.
DeGregori SE. Effects of two familial hypertrophic cardio-
Am J Physiol Heart Circ Physiol 266: H1260–H1271, 1994.
myopathy-causing mutations on ␣-tropomyosin structure and
function. Biochemistry 36: 4637–4642, 1997. [Corrigenda. Bio- 40. Leavis PC and Gergely J. Thin filament proteins and thin
chemistry 38: March 23, 1999, p. 3850.] filament-linked regulation of vertebrate muscle contraction.
23. Greaser ML and Gergely J. Reconstitution of troponin activity CRC Crit Rev Biochem 16: 235–305, 1984.
from three protein components. J Biol Chem 246: 4226–4233, 41. Lim DS, Oberst L, McCluggage M, Youker K, Lacy J,
1971. DeMayo F, Entman ML, Roberts R, Michael LH, and Mar-
24. Gruver EJ, Fatkin D, Dodds GA, Kisslo J, Maron BJ, ian AJ. Decreased left ventricular ejection fraction in transgenic
Seidman JG, and Seidman CE. Familial hypertrophic cardio- mice expressing mutant cardiac troponin T-Q(92), responsible
myopathy and atrial fibrillation caused by Arg663His ␤-cardiac for human hypertrophic cardiomyopathy. J Mol Cell Cardiol 32:
myosin heavy chain mutation. Am J Cardiol 83: 13H–18H, 1999. 365–374, 2000.
25. Harada K, Takahashi-Yanaga F, Minakami R, Morimoto S, 42. Lin D, Bobkova A, Homsher E, and Tobacman LS. Altered
and Ohtsuki I. Functional consequences of the deletion muta- cardiac troponin T in vitro function in the presence of a mutation
tion ⌬Glu160 in human cardiac troponin T. J Biochem (Tokyo) implicated in familial hypertrophic cardiomyopathy. J Clin In-
127: 263–268, 2000. vest 97: 2842–2848, 1996.
26. Hinkle A, Goranson A, Butters CA, and Tobacman LS. 43. Lin T, Ichihara S, Yamada Y, Nagasaka T, Ishihara H,
Roles for the troponin tail domain in thin filament assembly and Nakashima N, and Yokota M. Phenotypic variation of familial
regulation. A deletional study of cardiac troponin T. J Biol Chem hypertrophic cardiomyopathy caused by the Phe(110)3 Ile mu-
274: 7157–7164, 1999. [Corrigenda. J Biol Chem 274: October, tation in cardiac troponin T. Cardiology 93: 155–162, 2000.
29 1999, p. 31750.] 44. Malnic B, Farah CS, and Reinach FC. Regulatory properties
27. Ho CY, Lever HM, DeSanctis R, Farver CF, Seidman JG, of the NH2- and COOH-terminal domains of troponin T. ATPase
and Seidman CE. Homozygous mutation in cardiac troponin T: activation and binding to troponin I and troponin C. J Biol Chem
implications for hypertrophic cardiomyopathy. Circulation 102: 273: 10594–10601, 1998.
1950–1955, 2000. 45. Marian AJ, Zhao G, Seta Y, Roberts R, and Yu QT. Expres-
28. Homsher E, Lee DM, Morris C, Pavlov D, and Tobacman sion of a mutant (Arg92Gln) human cardiac troponin T, known to
LS. Regulation of force and unloaded sliding speed in single thin cause hypertrophic cardiomyopathy, impairs adult cardiac myo-
filaments: effects of regulatory proteins and calcium. J Physiol cyte contractility. Circ Res 81: 76–85, 1997.
(Lond) 524: 233–243, 2000. 46. McKay RT, Saltibus LF, Li MX, and Sykes BD. Energetics of
29. Housmans PR and Wanek LA. Effects of halothane, enflu- the induced structural change in a Ca2⫹ regulatory protein: Ca2⫹
rane, and isoflurane on measurements of Ca2⫹ by calcium elec- and troponin I peptide binding to the E41A mutant of the
trode and aequorin luminescence. Anal Biochem 284: 60–64, N-domain of skeletal troponin C. Biochemistry 39: 12731–12738,
2000. 2000.
30. Jaaskelainen P, Soranta M, Miettinen R, Saarinen L, 47. Michele DE, Albayya FP, and Metzger JM. Direct, conver-
Pihlajamaki J, Silvennoinen K, Tikanoja T, Laakso M, and gent hypersensitivity of calcium-activated force generation pro-

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


INVITED REVIEW 1135

duced by hypertrophic cardiomyopathy mutant ␣-tropomyosins effect of a mutant cardiac troponin T on cardiac structure and
in adult cardiac myocytes. Nat Med 5: 1413–1417, 1999. function in transgenic mice. J Clin Invest 102: 1498–1505, 1998.
48. Mikane T, Araki J, Kohno K, Nakayama Y, Suzuki S, 64. Ohtsuki I. Calcium ion regulation of muscle contraction: the
Shimizu J, Matsubara H, Hirakawa M, Takaki M, and regulatory role of troponin T. Mol Cell Biochem 190: 33–38,
Suga H. Mechanism of constant contractile efficiency under 1999.
cooling inotropy of myocardium: simulation. Am J Physiol Heart 65. Olson TM, Doan TP, Kishimoto NY, Whitby FG, Ackerman
Circ Physiol 273: H2891–H2898, 1997. MJ, and Fananapazir L. Inherited and de novo mutations in
49. Miller T, Szczesna D, Housmans PR, Zhao J, deFreitas F, the cardiac actin gene cause hypertrophic cardiomyopathy. J
Gomes AV, Culbreath L, McCue J, Wang Y, Xu Y, Kerrick Mol Cell Cardiol 32: 1687–1694, 2000.
WG, and Potter JD. Abnormal contractile function in trans- 66. Perry SV. Troponin T: genetics, properties and function. J Mus-
genic mice expressing an FHC-linked troponin T (179N) muta- cle Res Cell Motil 19: 575–602, 1998.
tion. J Biol Chem In press. 67. Poetter K, Jiang H, Hassanzadeh S, Master SR, Chang A,
49a.Mogensen J, Klausen IC, Egebald H, Baandrup U, and Dalakas MC, Rayment I, Sellers JR, Fananapazir L, and
Borglum AD. Sudden cardiac death in familial hypertrophic Epstein ND. Mutations in either the essential or regulatory
cardiomyopathy is associated with a novel mutation in the tro- light chains of myosin are associated with a rare myopathy in
ponin I gene. Circulation 100: I-618, 1999. human heart and skeletal muscle. Nat Genet 13: 63–69, 1996.
50. Mogensen J, Klausen IC, Pedersen AK, Egeblad H, Bross 68. Potter JD, Sheng Z, Pan BS, and Zhao J. A direct regulatory
P, Kruse TA, Gregersen N, Hansen PS, Baandrup U, and role for troponin T and a dual role for troponin C in the Ca2⫹
Borglum AD. Alpha-cardiac actin is a novel disease gene in regulation of muscle contraction. J Biol Chem 270: 2557–2562,
familial hypertrophic cardiomyopathy. J Clin Invest 103: R39– 1995.
R43, 1999. 69. Redwood C, Lohmann K, Bing W, Esposito GM, Elliott K,
51. Moolman JC, Corfield VA, Posen B, Ngumbela K, Seidman Abdulrazzak H, Knott A, Purcell I, Marston S, and
C, Brink PA, and Watkins H. Sudden death due to troponin T Watkins H. Investigation of a truncated cardiac troponin T that
mutations. J Am Coll Cardiol 29: 549–555, 1997. causes familial hypertrophic cardiomyopathy: Ca2⫹ regulatory
52. Moolman-Smook JC, De Lange WJ, Bruwer EC, Brink PA, properties of reconstituted thin filaments depend on the ratio of
and Corfield VA. The origins of hypertrophic cardiomyopathy- mutant to wild-type protein. Circ Res 86: 1146–1152, 2000.
causing mutations in two South African subpopulations: a 70. Redwood CS, Moolman-Smook JC and Watkins H. Proper-
unique profile of both independent and founder events. Am J ties of mutant contractile proteins that cause hypertrophic car-
Hum Genet 65: 1308–1320, 1999. diomyopathy. Cardiovasc Res 44: 20–36, 1999.
53. Morimoto S, Nakaura H, Yanaga F, and Ohtsuki I. Func- 71. Regitz-Zagrosek V, Erdmann J, Wellnhofer E, Raible J,
tional consequences of a carboxyl terminal missense mutation and Fleck E. Novel mutation in the ␣-tropomyosin gene and
Arg278Cys in human cardiac troponin T. Biochem Biophys Res transition from hypertrophic to hypocontractile dilated cardio-
Commun 261: 79–82, 1999. myopathy. Circulation 102: E112–E116, 2000.
54. Morimoto S, Yanaga F, Minakami R, and Ohtsuki I. Ca2⫹- 72. Reinach FC, Farah CS, Monteiro PB, and Malnic B. Struc-
sensitizing effects of the mutations at Ile-79 and Arg-92 of tural interactions responsible for the assembly of the troponin
troponin T in hypertrophic cardiomyopathy. Am J Physiol Cell complex on the muscle thin filament. Cell Struct Funct 22:
Physiol 275: C200–C207, 1998. 219–223, 1997.
55. Morner S, Richard P, Kazzam E, Hainque B, Schwartz K, 73. Robertson SP, Johnson JD, and Potter JD. The time-course
and Waldenstrom A. Deletion in the cardiac troponin I gene in of Ca2⫹ exchange with calmodulin, troponin, parvalbumin, and
a family from northern Sweden with hypertrophic cardiomyop- myosin in response to transient increases in Ca2⫹. Biophys J 34:
athy. J Mol Cell Cardiol 32: 521–525, 2000. 559–569, 1981.
56. Mukherjea P, Tong L, Seidman JG, Seidman CE, and 74. Rust EM, Albayya FP, and Metzger JM. Identification of a
Hitchcock-DeGregori SE. Altered regulatory function of two contractile deficit in adult cardiac myocytes expressing hyper-
familial hypertrophic cardiomyopathy troponin T mutants. Bio- trophic cardiomyopathy-associated mutant troponin T proteins.
chemistry 38: 13296–13301, 1999. J Clin Invest 103: 1459–1467, 1999.
57. Muthuchamy M, Pieples K, Rethinasamy P, Hoit B, Grupp 75. Satoh M, Takahashi M, Sakamoto T, Hiroe M, Marumo F,
IL, Boivin GP, Wolska B, Evans C, Solaro RJ, and Wiec- and Kimura A. Structural analysis of the titin gene in hyper-
zorek DF. Mouse model of a familial hypertrophic cardiomyop- trophic cardiomyopathy: identification of a novel disease gene.
athy mutation in ␣-tropomyosin manifests cardiac dysfunction. Biochem Biophys Res Commun 262: 411–417, 1999.
Circ Res 85: 47–56, 1999. 76. Schaertl S, Lehrer SS, and Geeves MA. Separation and
58. Nakajima-Taniguchi C, Matsui H, Fujio Y, Nagata S, characterization of the two functional regions of troponin in-
Kishimoto T, and Yamauchi-Takihara K. Novel missense volved in muscle thin filament regulation. Biochemistry 34:
mutation in cardiac troponin T gene found in Japanese patient 15890–15894, 1995.
with hypertrophic cardiomyopathy. J Mol Cell Cardiol 29: 839– 77. Solaro RJ. Troponin I, stunning, hypertrophy, and failure of the
843, 1997. heart. Circ Res 84: 122–124, 1999.
59. Nakajima-Taniguchi C, Matsui H, Nagata S, Kishimoto T, 78. Sweeney HL, Feng HS, Yang Z, and Watkins H. Functional
and Yamauchi-Takihara K. Novel missense mutation in ␣-tro- analyses of troponin T mutations that cause hypertrophic car-
pomyosin gene found in Japanese patients with hypertrophic diomyopathy: insights into disease pathogenesis and troponin
cardiomyopathy. J Mol Cell Cardiol 27: 2053–2058, 1995. function. Proc Natl Acad Sci USA 95: 14406–14410, 1998.
60. Nakaura H, Morimoto S, Yanaga F, Nakata M, Nishi H, 78a.Szczesna D and Potter JD. The role of troponin in the Ca2⫹-
Imaizumi T, and Ohtsuki I. Functional changes in troponin T regulation of skeletal muscle contraction. In: Molecular Interac-
by a splice donor site mutation that causes hypertrophic cardio- tions of Actin, edited by dasRemedios CG and Thomas DD.
myopathy. Am J Physiol Cell Physiol 277: C225–C232, 1999. Heidelberg, Germany. In press.
61. Nakaura H, Yanaga F, Ohtsuki I, and Morimoto S. Effects 79. Szczesna D, Zhang R, Zhao J, Jones M, Guzman G, and
of missense mutations Phe110Ile and Glu244Asp in human Potter JD. Altered regulation of cardiac muscle contraction by
cardiac troponin T on force generation in skinned cardiac muscle troponin T mutations that cause familial hypertrophic cardiomy-
fibers. J Biochem (Tokyo) 126: 457–460, 1999. opathy. J Biol Chem 275: 624–630, 2000.
62. Noland, TA Jr, Guo X, Raynor RL, Jideama NM, Avery- 80. Takahashi-Yanaga F, Morimoto S, and Ohtsuki I. Effect of
hart-Fullard V, Solaro RJ, and Kuo JF. Cardiac troponin I Arg145Gly mutation in human cardiac troponin I on the ATPase
mutants. Phosphorylation by protein kinases C and A and reg- activity of cardiac myofibrils. J Biochem (Tokyo) 127: 355–357,
ulation of Ca2⫹-stimulated MgATPase of reconstituted actomy- 2000.
osin S-1. J Biol Chem 270: 25445–25454, 1995. 81. Tardiff JC, Factor SM, Tompkins BD, Hewett TE, Palmer
63. Oberst L, Zhao G, Park JT, Brugada R, Michael LH, Ent- BM, Moore RL, Schwartz S, Robbins J, and Leinwand LA.
man ML, Roberts R, and Marian AJ. Dominant-negative A truncated cardiac troponin T molecule in transgenic mice

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.


1136 INVITED REVIEW

suggests multiple cellular mechanisms for familial hypertrophic 89. Watkins H, MacRae C, Thierfelder L, Chou YH, Frenneaux
cardiomyopathy. J Clin Invest 101: 2800–2811, 1998. M, McKenna W, Seidman JG, and Seidman CE. A disease
82. Tardiff JC, Hewett TE, Palmer BM, Olsson C, Factor SM, locus for familial hypertrophic cardiomyopathy maps to chromo-
Moore RL, Robbins J, and Leinwand LA. Cardiac troponin T some 1q3. Nat Genet 3: 333–337, 1993.
mutations result in allele-specific phenotypes in a mouse model 90. Watkins H, McKenna WJ, Thierfelder L, Suk HJ, Anan R,
for hypertrophic cardiomyopathy. J Clin Invest 104: 469–481, O’Donoghue A, Spirito P, Matsumori A, Moravec CS, Seid-
1999. man JG, et al. Mutations in the genes for cardiac troponin T
83. Thierfelder L, MacRae C, Watkins H, Tomfohrde J, Wil- and ␣-tropomyosin in hypertrophic cardiomyopathy. N Engl
liams M, McKenna W, Bohm K, Noeske G, Schlepper M, J Med 332: 1058–1064, 1995.
Bowcock A, et al. A familial hypertrophic cardiomyopathy 91. Watkins H, Seidman CE, Seidman JG, Feng HS, and
locus maps to chromosome 15q2. Proc Natl Acad Sci USA 90: Sweeney HL. Expression and functional assessment of a trun-
6270–6274, 1993. cated cardiac troponin T that causes hypertrophic cardiomyopa-
84. Thierfelder L, Watkins H, MacRae C, Lamas R, McKenna thy. Evidence for a dominant negative action. J Clin Invest 98:
W, Vosberg HP, Seidman JG, and Seidman CE. ␣-Tropomy- 2456–2461, 1996.
osin and cardiac troponin T mutations cause familial hypertro- 92. Watkins H, Thierfelder L, Anan R, Jarcho J, Matsumori A,
phic cardiomyopathy: a disease of the sarcomere. Cell 77: 701– McKenna W, Seidman JG, and Seidman CE. Independent
712, 1994. origin of identical beta cardiac myosin heavy-chain mutations in
85. Tobacman LS, Lin D, Butters C, Landis C, Back N, Pavlov hypertrophic cardiomyopathy. Am J Hum Genet 53: 1180–1185,
D, and Homsher E. Functional consequences of troponin T 1993.
mutations found in hypertrophic cardiomyopathy. J Biol Chem 93. Watkins H, Thierfelder L, Hwang DS, McKenna W, Seid-
274: 28363–28370, 1999. man JG, and Seidman CE. Sporadic hypertrophic cardiomy-
86. Varnava A, Baboonian C, Davison F, de Cruz L, Elliott opathy due to de novo myosin mutations. J Clin Invest 90:
PM, Davies MJ, and McKenna WJ. A new mutation of the 1666–1671, 1992.
cardiac troponin T gene causing familial hypertrophic cardio- 94. Yamauchi-Takihara K, Nakajima-Taniguchi C, Matsui H,
myopathy without left ventricular hypertrophy. Heart 82: Fujio Y, Kunisada K, Nagata S, and Kishimoto T. Clinical
621–624, 1999. implications of hypertrophic cardiomyopathy associated with
87. Watkins H, Anan R, Coviello DA, Spirito P, Seidman mutations in the ␣-tropomyosin gene. Heart 76: 63–65, 1996.
JG, and Seidman CE. A de novo mutation in ␣-tropomyosin 95. Yanaga F, Morimoto S, and Ohtsuki I. Ca2⫹ sensitization and
that causes hypertrophic cardiomyopathy. Circulation 91: 2302– potentiation of the maximum level of myofibrillar ATPase activity
2305, 1995. caused by mutations of troponin T found in familial hypertrophic
88. Watkins H, Conner D, Thierfelder L, Jarcho JA, MacRae cardiomyopathy. J Biol Chem 274: 8806–8812, 1999.
C, McKenna WJ, Maron BJ, Seidman JG, and Seidman CE. 96. Zot AS and Potter JD. The effect of [Mg2⫹] on the Ca2⫹
Mutations in the cardiac myosin binding protein-C gene on dependence of ATPase and tension development of fast skeletal
chromosome 11 cause familial hypertrophic cardiomyopathy. muscle. The role of the Ca2⫹-specific sites of troponin C. J Biol
Nat Genet 11: 434–437, 1995. Chem 262: 1966–1969, 1987.

Downloaded from www.physiology.org/journal/jappl (046.161.058.175) on February 25, 2019.

You might also like