Calcium Signaling Series: Review

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Review

Calcium Signaling Series


Donald M. Bers, Guest Editor

Calcium Signaling and Transcriptional


Regulation in Cardiomyocytes
Matthias Dewenter,* Albert von der Lieth,* Hugo A. Katus, Johannes Backs

Abstract: Calcium (Ca2+) is a universal regulator of various cellular functions. In cardiomyocytes, Ca2+ is the central
element of excitation–contraction coupling, but also impacts diverse signaling cascades and influences the regulation
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

of gene expression, referred to as excitation–transcription coupling. Disturbances in cellular Ca2+-handling and


alterations in Ca2+-dependent gene expression patterns are pivotal characteristics of failing cardiomyocytes, with
several excitation–transcription coupling pathways shown to be critically involved in structural and functional
remodeling processes. Thus, targeting Ca2+-dependent transcriptional pathways might offer broad therapeutic
potential. In this article, we (1) review cytosolic and nuclear Ca2+ dynamics in cardiomyocytes with respect to
their impact on Ca2+-dependent signaling, (2) give an overview on Ca2+-dependent transcriptional pathways in
cardiomyocytes, and (3) discuss implications of excitation–transcription coupling in the diseased heart.   (Circ Res.
2017;121:1000-1020. DOI: 10.1161/CIRCRESAHA.117.310355.)
Key Words: calcium ■ calcium-calmodulin dependent protein kinase II ■ calcineurin ■ calmodulin
■ excitation transcription coupling

C alcium (Ca2+) is a ubiquitous key signaling element in all


eukaryotic cell types. It regulates diverse essential physi-
ological processes such as cell differentiation, proliferation
are just beginning to understand how the multiple ET coupling
pathways are differentially activated, how they interrelate, and
how the multiple targets in this complex network relatively
and motility, apoptosis, secretion, excitation, contraction, and contribute to failure of cardiomyocytes.
neuronal plasticity.1 In the heart, the most obvious crucial role This review gives an overview on Ca2+-dependent tran-
of Ca2+ is its involvement in electric activity and cardiac con- scriptional mechanisms in cardiomyocytes and their role in
tractility, acting as the central player of excitation–contraction cardiac disease. First, we discuss cytosolic and nuclear Ca2+
(EC) coupling.2 Besides being directly involved in EC cou- dynamics as the basis for the regulation of Ca2+-dependent
pling, Ca2+ is an important element in various signaling cas- signaling. Then we consider the key players and pathways in-
cades, regulating the activity of diverse downstream effectors. volved in ET coupling in cardiomyocytes. Finally, we review
Thereby, Ca2+ can exert acute effects but also influence the implications of ET coupling in the diseased heart and give an
regulation of gene expression, called excitation–transcription outlook on possible therapeutic perspectives.
(ET) coupling. These ET coupling pathways play a critical
role not only in cardiac homeostasis but also in cardiac disease
Ca2+ Dynamics in Cardiomyocytes
development, since diseased cardiomyocytes show vast altera-
tions in Ca2+-handling and Ca2+-dependent transcriptional pat- Cytosolic Ca2+
terns.3,4 Among these pathways, especially signaling cascades Given the central role of Ca2+ in controlling EC coupling, a
involving CaMKII (Ca2+/calmodulin-dependent kinase II) and strict regulation of Ca2+-handling is crucial for cardiac func-
the Ca2+/calmodulin-dependent serine/threonine phosphatase tion. Ca2+ transport mechanisms in cardiomyocytes include
calcineurin have been extensively characterized on their role both Ca2+ cycling between the cytosol and the extracellular
in cardiac hypertrophy and remodeling processes.5,6 Yet, we space and Ca2+ cycling between the cytosol and intracellular

From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University,
Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.).
*These authors contributed equally to this article.
Correspondence to Johannes Backs, MD, Department Molecular Cardiology and Epigenetics, University Hospital Heidelberg, Im Neuenheimer Feld
669, 69120 Heidelberg, Germany. E-mail Johannes.backs@med.uni-heidelberg.de
© 2017 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.117.310355

1000
Dewenter et al   Calcium Signaling and Transcription   1001

the cytosol. The 2 main mechanisms are Ca2+ transport back


Nonstandard Abbreviations and Acronyms
into the SR by SERCA (SR Ca2+ ATPase) and Ca2+ extrusion
ANGII angiotensin 2 from the cell via NCX (Na+/Ca2+ exchanger) in the plasma
AP-1 activator protein 1 membrane. A small amount of Ca2+ is also removed by Ca2+
AR adrenergic receptor ATPases in the plasma membrane and by mitochondrial Ca2+
ATF1 activating transcription factor 1 uniporters.10
CaMKII Ca2+/calmodulin-dependent kinase II These Ca2+ cycling processes are finely tuned in response
CAMTA calmodulin-binding transcription activator to extra- or intracellular stimuli by various regulators that
CnA calcineurin subunit A affect the activity state of Ca2+-handling proteins. Central
CnB Ca2+-binding regulatory subunit B
modulators of cardiomyocyte Ca2+-handling proteins are PKA
(protein kinase A) and CaMKII. PKA is activated in response
CREB cAMP response element binding protein
to sympathetic stimulation of β-AR (β-adrenergic receptors)
EC excitation–contraction
and phosphorylates LTCC, RyR, and the SERCA inhibitor
ET excitation–transcription
PLN (phospholamban), thereby increasing Ca2+ influx, SR
ET-1 endothelin-1
Ca2+ release, and SR Ca2+ reuptake. CaMKII shares common
HDAC histone deacetylase
targets with PKA and is also activated in response to sym-
HP1 heterochromatin protein 1
pathetic activation; however, CaMKII-dependent mechanisms
IGF-1 insulin-like growth factor 1 have been suggested to exert more long-term effects on Ca2+
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

IP3 inositol 1,4,5-trisphosphate cycling.4 These kinase-mediated effects on EC coupling pro-


IRF8 interferon regulatory factor 8 teins are counterbalanced by protein phosphatases, especially
LTCC L-type Ca2+ channel protein phosphatase 1 and 2A.11 In addition, the activity state
MCIP1 myocyte-enriched calcineurin-interacting protein 1 of Ca2+-handling proteins and also of their upstream regula-
MEF2 myocyte enhancer factor 2 tory kinases can be modulated by multiple other posttrans-
NCX Na+/Ca2+ exchanger lational modifications, such as oxidation, nitrosylation, and
NFAT nuclear factor of activated T cells glycosylation.12–15
NFκB nuclear factor κB Besides the above-mentioned elements of Ca2+-handling,
NPC nuclear pore complex other Ca2+ cycling proteins have been characterized in car-
PKA protein kinase A diomyocytes that are suggested to play a less pivotal role in
PKC protein kinase C the classical concept of EC coupling but are implicated in
PLC phospholipase C Ca2+-dependent signaling mechanisms; in particular, IP3 (ino-
PLN phospholamban sitol 1,4,5-trisphosphate) receptors, TRP (transient receptor
RyR ryanodine receptor potential) channels, and SOCE (store-operated Ca2+ entry)
SERCA SR Ca2+ ATPase controlled by STIM1 (stromal interaction molecule 1). IP3
SOCE store-operated Ca2+ entry
receptors are Ca2+ channels activated by IP3, which is gener-
SR sarcoplasmic reticulum
ated through PLC (phospholipase C)–dependent hydrolysis of
phosphatidylinositol-4,5-bisphosphate after Gq protein–cou-
SRF serum response factor
pled receptor stimulation.16 Typical activators of IP3 signaling
STIM1 stromal interaction molecule 1
in cardiomyocytes are ET-1 (endothelin-1), catecholamines,
TRP transient receptor potential
and ANGII (angiotensin 2), which presumably modulate spe-
TXNIP thioredoxin-interacting protein
cialized pools of Ca2+ via this mechanism.17 Localized prefer-
ably in the perinuclear region and the nuclear envelope, IP3
Ca2+ stores, in particular the sarcoplasmic reticulum (SR). A receptors are suggested to play a major role in local nuclear
critical cellular microdomain for cytosolic Ca2+ cycling is the Ca2+ cycling. Further details in this regard will be discussed
dyadic junction, where invaginations of the plasma membrane below. TRP channels are ubiquitously expressed nonselective
(T-tubules) and the SR are in close proximity.7 On depolariza- cation channels with variable Ca2+ permeability. In cardiomy-
tion of the cell membrane, LTCC (L-type Ca2+ channels; also ocytes, TRP channels, especially TRPCs, have been suggested
known as dihydropyridine receptors) in the plasma membrane to act as fine-tuners of Ca2+ cycling and were demonstrated
are activated, allowing Ca2+ to enter the cytosol. This Ca2+ en- to control Ca2+-dependent signaling in response to neurohu-
try raises local Ca2+ concentration from 0.1 to >10 μmol/L moral stimulation.18–20 Besides the activation via Gq protein–
in the junctional cleft, which triggers the activation of Ca2+ coupled receptor stimulation (receptor-operated Ca2+ entry),
release channels (RyR [ryanodine receptors]) in the SR mem- TRPCs also participate in SOCE.21 SOCE involving STIM1
brane, a process often referred to as Ca2+-induced Ca2+ release. and Ca2+ channel Orai1 has been characterized as a key ele-
The SR Ca2+ release raises local cleft Ca2+ to >100 µmol/L and ment of Ca2+-dependent signaling in nonexcitable cells.22 In
global cytosolic Ca2+ concentration from 0.1 µmol/L during cardiomyocytes, STIM1-dependent Ca2+ entry was observed
diastole to 1 µmol/L during systole.8 This increase in cytosolic to coexist with the global Ca2+ transients. Increasing evidence
Ca2+ causes contraction via Ca2+-binding–induced conforma- indicates that STIM1 is important for Ca2+ response signal-
tional changes in the troponin–tropomyosin complex, which ing within microdomains, inducing changes in cardiac tran-
allows the myofilaments actin and myosin to slide past one scriptional profiles.23 Apart from association with Orai1 and
another.9 For relaxation to occur, Ca2+ must be removed from TRPC channels, recent data suggest that STIM1 interacts with
1002  Circulation Research  September 29, 2017

several other Ca2+ cycling proteins, including LTCC, SERCA, showed that even Ca2+ puffs in the perinuclear area within 3
plasma membrane Ca2+ ATPases, and RyR.24–27 µm of the nuclear envelope can lead to an increase in nuclear
This highly complex system of cardiomyocyte Ca2+ cy- Ca2+ while leaving the rest of the cytosol unaffected.42 This
cling on the one hand allows global continuous intracellular clearly states the importance of the perinuclear SR when
Ca2+ oscillations and on the other hand controls local pools considering nuclear Ca2+ regulation and already implies that
of Ca2+ within spatial microdomains, and the regulation of nuclear Ca2+ is influenced by many aspects beyond whole-cell
nuclear Ca2+ adds an additional layer of complexity. Ca2+ oscillation.
However, passive diffusion is only 1 aspect of nuclear
Nuclear Ca2+ Ca2+ cycling. Ion diffusion can also be subject to regulation
The nucleus is a subcellular compartment surrounded by 2 at the level of NPCs. Ambient Ca2+ and ATP are essential for
phospholipid bilayers, referred to as the nuclear envelope. the maintenance of NPC diffusion capacity; thus, decreasing
This nuclear envelope separates cytoplasm from nucleoplasm Ca2+ or ATP in the area surrounding the NPC slows down its
and is structurally and functionally subdivided. The outer nu- conductance, but not to the extent of a full diffusion block-
clear membrane faces the cytosol and merges with the mem- ade.43 Furthermore, the distribution and position of NPCs on
brane of the SR. Therefore, both the outer nuclear membrane the nuclear envelope, as well as their distance to nuclear Ca2+
and the SR membrane share the same characteristics on their channels on the outer and inner nuclear membrane, seem to
composition, and the perinuclear space, which is in between have an important effect on local Ca2+ diffusion.44 The amount
the outer nuclear membrane and the inner nuclear membrane, or density of NPCs in the nuclear envelope can vary between
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

resembles the SR in ion and protein content.28,29 In contrast 10 and 20 pores/µm2 in vertebrates, and, thus, total Ca2+ diffu-
to the outer nuclear membrane, the inner nuclear membrane, sion capacity varies accordingly.44,45
which is directed to the nucleoplasm, shows a distinct com- Functional and structural evidence further indicates that
position unique to the nucleus. The nuclear envelope forms nuclear Ca2+ can be actively controlled. IP3 receptor type 2,
nuclear invaginations that reach deep into the nucleus, creat- the predominant subtype in cardiomyocytes, is concentrated
ing complex structures that in their entirety are termed the nu- in the perinuclear area and can be found on the junctional
clear reticulum.28,30 In close proximity to the nuclear envelope, SR, the outer and the inner nuclear membrane.46 These nu-
T-tubules reaching deep into the cytosol and mitochondria can clear IP3 receptors seem to account for the widely described
be found, constituting microdomains for Ca2+ cycling and sig- observation of a diastolic nucleoplasmic-to-cytoplasmic
nal responsiveness.31 [Ca2+] gradient.37 Experiments on isolated nuclei, permea-
The nuclear membranes are poorly permeable to ions and bilized, and intact cardiac myocytes consistently show that
water-soluble molecules. Transport and diffusion is facilitat- the activation of IP3 receptors preferably increases nuclear
ed by nuclear pore complexes (NPCs), which are distributed Ca2+.46,47 IP3, generated after stimulation of Gq protein–
throughout the nuclear envelope.32,33 Proteins and RNAs ≤39 coupled receptors in the plasma membrane, acts not only
nm in diameter are actively shuttled in and out of the nucleus on plasmalemmal IP3 receptors but can also diffuse to the
by a transport system associated with the NPC.34,35 Ions and nucleus and activate IP3 receptors in the outer and inner nu-
molecules smaller than 9 nm and 40 kDa can diffuse through clear membrane.48,49 In addition, there is increasing evidence
the NPC.34 Thus, the NPCs function as both barriers and selec- that IP3 can also be directly generated at the inner nuclear
tive filters for the substances trafficking between nucleoplasm membrane and, therefore, selectively act on nuclear IP3 re-
and cytoplasm. ceptors to increase nuclear Ca2+, especially in response to
In cardiomyocytes, every cytosolic Ca2+ transient is ac- ET-1, ANGII, and α-adrenergic stimuli.47,50–52 Notably, ET-1
companied by a nuclear Ca2+ transient.36 However, nuclear receptors, ANGII receptors, and α-AR have been identified
Ca2+ transients, compared with cytosolic transients, seem not only on the plasma membrane but also on the nuclear
to have a slower and delayed upstroke, a lower peak, and a envelope.50–53
prolonged return back to baseline.37,38 These findings led Several more receptors were proposed to play a role
to the hypothesis that cytosolic Ca2+ passively diffuses into in nuclear Ca2+ regulation. For example, it has been dem-
the nucleus during systole, thereby causing the nuclear Ca2+ onstrated that activation of the IGF-1 (insulin-like growth
transients. Interestingly, to date none or only inconsiderable factor 1) receptor, a receptor tyrosine kinase localized deep
numbers of primary active pumps for Ca2+ reuptake have been within the T-tubules in proximity to the nucleus, leads
found on the inner nuclear membrane.33,39 Therefore, the idea to PLC-dependent production of perinuclear IP3.54 This
is that Ca2+ mostly diffuses out of the nucleus via NPCs and is consequently causes a selective increase in nuclear Ca2+.
then taken up by SERCA on the outer nuclear membrane and Moreover, the existence of β1- and β3-AR on the nuclear
the junctional SR, by nearby mitochondria or is extruded by membrane was demonstrated,55 suggesting that β-adrenergic
NCX on T-tubules in proximity to the nuclear envelope.31,40,41 signaling might have an influence on nuclear [Ca2+] within
This would also explain the observation that the nuclear base- microdomains via localized perinuclear PKA activation
line [Ca2+] increases with higher beating frequencies. Because and subsequent phosphorylation of PKA target structures.56
the decay of nucleoplasmic Ca2+ transients is slower compared Although there is substantial evidence for the influence of
with cytoplasmic Ca2+ transients, Ca2+ noticeably builds up nuclear β-ARs on transcriptional processes, the exact mech-
within the nucleus when diastole is shortened.37 anisms involved remain unknown.57 Another suggested reg-
Nuclear Ca2+ not only follows whole-cell Ca2+ oscilla- ulator of nuclear Ca2+-handling is the RyR. It is primarily
tion but also small perinuclear Ca2+ release events. Lipp et al located on the SR and only to a lesser extent on the outer
Dewenter et al   Calcium Signaling and Transcription   1003

nuclear membrane.58 Intranuclear RyR localization remains Ca2+-Dependent Regulation of Gene


controversial, yet it was described for neonatal cardiac myo- Expression in Cardiomyocytes
cytes.58 Perinuclear RyRs, however, were reported to play a Several Ca2+-dependent pathways are described that modu-
pivotal role in generation of Ca2+ waves originating from the late gene expression by signal transduction to transcriptional
perinuclear region.59 regulators. In cardiomyocytes, well-characterized signaling
In conclusion, nuclear Ca2+ is basically subject to cyto- patterns in this regard involve calmodulin, CaMKII, and cal-
solic Ca2+ oscillations. The mere passive character of cytosolic cineurin. Figure 1 illustrates the major findings summarized
to nuclear Ca2+ diffusion is, however, enriched by different in this section.
modifying mechanisms incorporating influences and specific
pathways of various origins. Moreover, nuclear, perinuclear, Calmodulin
and cytosolic structures, such as nuclear invaginations, NPCs, Calmodulin is a highly conserved Ca2+ sensor protein in eu-
perinuclear reticulum, T-tubules, and the distribution of, for karyotic cells with no innate enzymatic function. It is a small
example, receptors, channels, and pumps, create Ca2+ micro- 16 kDa protein that consists of a C-terminal and an N-terminal
domains that offer a vast array of possibilities for spatially and lobe with 2 Ca2+-binding EF-hands each. Different Ca2+ af-
temporally restricted Ca2+ control. finities of the N-terminal and C-terminal EF-hands and intra-
Thus, cytosolic and nuclear Ca2+ cycling are part of a molecular structures cooperatively facilitate a differentiated
highly sophisticated system which controls Ca2+-dependent response to a broad range in Ca2+ concentrations. When Ca2+
signaling by modifications of global Ca2+ oscillations as well binds to the lobes, conformational changes occur and thereby
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

as Ca2+ regulation within microdomains. In the following, we protein interaction sites are released.60 To date, >300 pro-
focus on pathways that link cardiomyocyte Ca2+ cycling with teins have been shown to bind to calmodulin. Various bind-
transcriptional regulation. ing motifs and mechanisms of interaction have been identified

Figure 1. Schematic diagram of the Ca2+-dependent pathways mediating transcriptional regulation in cardiomyocytes. For details,
see main text. CaM indicates calmodulin; CAMKII, Ca2+/calmodulin-dependent kinase II; CAMTA, calmodulin-binding transcriptional
activator; CaN, calcineurin; cPKC, conventional protein kinase C; cPKCct, conventional protein kinase C c terminal; CREB, cAMP
response element binding protein; DAG, diacylglycerol; H3S10, histone 3 serine 10; HDAC4, histone deacetylase 4; HDAC5, histone
deacetylase 5; HSF1, heat shock factor 1; IκB, inhibitor of κB; IKK, inhibitor of κB kinase; INM, inner nuclear membrane; IP3, inositol
1,4,5-trisphosphate; IP3R, inositol 1,4,5-trisphosphate receptor; LTCC, L-type Ca2+ channel; MEF2, myocyte enhancer factor 2; NFAT,
nuclear factor of activated T cells; NFκB, nuclear factor κB; NPC, nuclear pore complex; ONM, outer nuclear membrane; RyR, ryanodine
receptor; SRF, serum response factor; STIM1, stromal interaction molecule 1; and TRPC, classical transient receptor potential cannel.
1004  Circulation Research  September 29, 2017

resulting in local regulation of target activity at very different to mediate CREB activation in response to neurohumoral
Ca2+ concentrations.61,62 stimulation by ET-1 and phenylephrine.77 In addition, ATF1
Besides its regulatory function in activating Ca2+-sensitive (activating transcription factor 1), that shares huge homology
enzymes such as CaMKII and calcineurin, calmodulin exerts with CREB and that is also modulated by cAMP/PKA, can be
effects on gene expression via CAMTA (calmodulin-binding phosphorylated by CaMKII, remarkably only at its activation
transcriptional activator).63 In humans, 2 different CAMTA site Ser-63 (corresponding to Ser-133 of CREB).78 However,
genes, CAMTA1 and CAMTA2, have been characterized. up to now the specific functional consequences of CaMKII-
Their expression seems to be most prominent in heart and dependent modulation of these cAMP-regulated transcription
brain tissue.64 In the heart, CAMTA2 raised interest because of factors in cardiac myocytes remain elusive.79
its noteworthy ability to induce the expression of atrial natri- The functional role of other CaMKII-dependent tran-
uretic peptide—a surrogate parameter for cardiac hypertrophy scription factor activation events has been more specifically
and marker of heart failure–related diseases. Mechanistically, characterized, for example, on apoptosis and inflammation.
it was shown that CAMTA2 binds to the homeobox pro- Interestingly, whereas the CaMKIIδ splice variant CaMKIIδC
tein NKX2-5, which is also implicated in hypertrophic re- has been suggested to mediate stress-induced proapoptotic
sponse, and coactivates NKX2-5–dependent transcription. effects via classical mitochondrial pathways,80,81 the splice
Furthermore, the Olson laboratory demonstrated that HDAC5 variant CaMKIIδB seems to play an antiapoptotic role.
(histone deacetylase 5) represses CAMTA2 through a direct Mechanistically, CaMKIIδB has been suggested to activate
interaction. With PKD (protein kinase D)–dependent HDAC5 the transcription factor heat shock factor 1 by phosphorylation
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

phosphorylation and consequent nuclear export, CAMTA2 is at Ser-230, inducing increased transcription of cell-protective
released and executes its coactivating function.63,65 Therefore, factor inducible heat shock protein 70.82 This putatively pro-
CAMTA is not only directly regulated by Ca2+ via binding of tective role of the CaMKIIδB splice variant is supported by
Ca2+/calmodulin but also indirectly via its interaction with the observation that it is required for binding of transcription
HDAC5. The relevance of CAMTA2 in human was only re- factor GATA4 to the promotor region of antiapoptotic protein
cently confirmed by showing that certain genetic polymor- Bcl-2 and consequently is crucial for reducing doxorubicin-
phisms in the coding region of CAMTA2 alter the risk of mediated cardiotoxicity.83
developing cardiac hypertrophy.66 Emerging evidence points to an important role of CaMKII
CaMKII in inflammation. For instance, CaMKIIδ has been described
CaMKII is a serine/threonine kinase with the isoforms CaMKIIδ to mediate activation of the transcription factor NFκB (nu-
and CaMKIIγ being expressed in the heart. The splice vari- clear factor κ B).84,85 The underlying mechanism most likely
ant CaMKIIδC primarily localizes within the cytosol, whereas involves a more indirect pathway via inhibitor of κB kinase
CaMKIIδB has a nuclear localization sequence. CaMKII be- activation and subsequent phosphorylation and degradation
comes activated upon Ca2+/calmodulin binding with a Kd ≈50 of Inhibitor of κB, leading to nuclear translocation of NFκB.
nmol/L.67 An important regulatory mechanism of CaMKII activ- This pathway has been investigated, for example, in the set-
ity dynamics is its autophosphorylation, which is sensitive to the ting of ischemia/reperfusion injury, suggesting a detrimental
frequency and duration of Ca2+ spikes.68 Autophosphorylation at proinflammatory role of CaMKIIδ.86 In the same setting of
Thr 286/287 enhances the affinity of CaMKII toward calmodu- ischemia/reperfusion injury, we found that CaMKII induces
lin and prevents autoinhibition of CaMKII, thereby maintaining transcription and secretion of the chemokines CCL2 and
enzyme activity independent of Ca2+/calmodulin binding.69,70 CCL3 from cardiomyocytes, thereby triggering an intrinsic
Analogous to autophosphorylation, several other posttransla- chemoattractant cardiomyocyte signaling cascade that is as-
tional modifications, such as oxidation, glycosylation, and ni- sociated with scar formation and cardiac fibrosis.87
trosylation, were described to further increase CaMKII activity In addition, CaMKII was demonstrated to interact with
after initial opening by Ca2+/calmodulin binding.15,71,72 CaMKII SRF (serum response factor), a transcription factor implicated
exerts its molecular effects by binding and phosphorylation of in cardiac integrity as genetic models have revealed.88,89 It has
target proteins. Regarding the regulation of transcriptional pro- been shown that CaMKII phosphorylates SRF at Ser103 and
cesses, CaMKII has been shown to phosphorylate transcription Thr160 in skeletal muscle90; however, the functional role of
factors, epigenetic regulators, and histones. these events in cardiomyocytes is not clear yet. Furthermore,
To date, several transcription factors that are activated in myocardin, a transcriptional coactivator of SRF that has been
a CaMKII-dependent manner have been identified. CREB causatively associated with cardiac hypertrophy,91 is activated
(cAMP response element binding protein) is a ubiquitously via Ca2+-dependent signaling involving CaMKII.92
expressed nuclear transcription factor involved in cardiac in- CaMKII modulates ion currents not only by direct interac-
tegrity, inflammation processes, and metabolic signaling.73–75 tion and phosphorylation of ion channels but also by transcrip-
CREB activity is regulated via various signaling pathways, and tional regulation. Studies reported that CaMKII is critical for
especially PKA-mediated phosphorylation of Ser-133 has been β-AR–stimulated upregulation of NCX1 by transcription fac-
shown to be critical for its activation. CaMKII either increases tor AP-1 (activator protein 1) activation.93 Moreover, CaMKII
activation of CREB by phosphorylation of Ser-133 or decreas- has been demonstrated to repress the expression of LTCC by
es CREB activity by phosphorylation of Ser-142, suggesting activating downstream regulatory element binding transcrip-
a modulatory role beside cAMP/PKA-dependent CREB regu- tion factor DREAM, thus constituting a negative feedback
lation.76 Notably, CaMKII has recently been demonstrated mechanism on Ca2+ influx.94
Dewenter et al   Calcium Signaling and Transcription   1005

CaMKII may also affect transcriptional patterns on a more HDAC4-dependent repression of MEF2 is diminished, re-
global scale by regulating transcription factor MeCP2 (meth- sulting in enhanced MEF2-dependent transcriptional activ-
yl CpG binding protein 2). MeCP2, which was implicated ity.103 Aside from MEF2, HDAC4 also acts as a repressor
in regulating the genome-wide chromatin state, was shown of the above-mentioned SRF, indicating a more complex
to be phosphorylated by CaMKII in neurons.95,96 Notably, functional consequence of CaMKII-mediated HDAC4 shut-
MeCP2 is also expressed in the heart, where its relevance in tling.106,107 Via HDAC4 binding, CaMKII is also capable of
human and experimental heart failure was recently demon- regulating HDAC5. Although HDAC5 alone is not respon-
strated.97 Looking at these studies, it is worth considering that sive to CaMKII, hetero-oligomerization of HDAC5 with
CaMKII signaling to MeCP2 may depict another important HDAC4 enables CaMKII to phosphorylate HDAC5, result-
Ca2+-dependent aspect of transcriptional regulation in cardiac ing in nuclear export and subsequent activation of MEF2.108
myocytes. CaMKII-regulated nuclear export of HDAC4 seems to affect
Besides the phosphorylation of transcription factors, transcriptional patterns even further. For example, it has been
CaMKII is also involved in chromatin modification. On the demonstrated that methylation of H3K9 at the promoter site
one hand, CaMKIIδB can directly phosphorylate histones, of atrial natriuretic peptide depends on HDAC4 shuttling.
in particular histone 3 at Ser10 (H3S10). This phosphoryla- Mechanistically, HP1 (heterochromatin protein 1) and histone
tion event has been associated with induction of hypertrophic methyltransferase SUV39H1 associate with class IIa HDACs
growth, based on increased chromatin accessibility for prohy- to build a corepressor complex.109 HDAC4, when shuttled out
pertrophic gene regulation.98,99 It will be interesting to see in of the nucleus, releases the corepressor complex and, there-
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

the future whether CaMKII-mediated histone phosphorylation fore, prevents further methylation. Subsequently, the H3K9
at H3S10 or at other potential histone phosphorylation sites site becomes demethylated, which relieves chromatin conden-
directly results in the regulation of specific gene programs. sation and facilitates gene expression.110
Chromatin immunoprecipitation of CaMKII-phosphorylated Calcineurin
histones followed by massive genomic sequencing might Calcineurin (protein phosphatase 2B) is a serine/threonine
enable one to study this fascinating possibility. On the other phosphatase that is structurally composed of 2 subunits: CnA
hand, CaMKII has been identified to interact with chroma- (calmodulin-binding catalytic subunit A) and CnB (Ca2+-
tin modifying enzymes, in particular HDAC4. HDAC4 is a binding regulatory subunit B).111 Like CaMKII, calcineurin is
class IIa HDAC that itself has only a weak deacetylase activ- activated by binding of Ca2+/calmodulin, notably with a much
ity and exerts transcriptional regulation by binding of tran- higher affinity (Kd<<1 nmol/L).67 The classical calcineurin-
scription factors or recruitment of other chromatin modifying dependent transcriptional regulation pathway involves NFAT
enzymes.100 One of the critical transcription factors regulated (nuclear factor of activated T cells). On cytosolic dephosphor-
by HDAC4 is MEF2 (myocyte enhancer factor 2). MEF2 ac- ylation by calcineurin, NFAT is imported into the nucleus,
tivation has been implicated in cardiac remodeling, fetal gene where it acts in conjunction with transcription factor GATA4.
reprogramming, and inflammation,101,102 and HDAC4 acts This calcineurin-dependent pathway has been elegantly dem-
as a repressor in this scenario. CaMKII has been identified onstrated to play a crucial role in cardiac development and in
as a specific kinase for HDAC4, since HDAC4 has a unique the adult cardiac hypertrophic response.112,113 Calcineurin has
docking site for CaMKII that is absent in other HDACs. also been observed to regulate NFAT directly in the nucleus.
Nuclear CaMKIIδB-dependent phosphorylation of HDAC4 Mechanistically, nuclear calcineurin was shown to suppress
(at Ser-467 and Ser-632) promotes chaperone 14-3-3 bind- exportin protein Crm1 binding to NFAT, with nuclear NFAT
ing and subsequent nuclear export of HDAC4, whereas cy- consequently accumulating.114,115
tosolic CaMKIIδC-dependent phosphorylation of HDAC4 Downstream effectors of calcineurin signaling may also
blocks nuclear import (Figure 2).103–105 As a consequence, include MEF2, as this is implied by findings from skeletal

Figure 2. Proposed interplay between


Ca2+/calmodulin-dependent kinase II
and calcineurin and their downstream
signaling pathways in cardiomyocytes.
CaMKII indicates Ca2+/calmodulin
dependent kinase II; CaN, calcineurin;
HDAC4, histone deacetylase 4; MEF2,
myocyte enhancer factor 2; Mrj,
mammalian relative of DnaJ; and NFAT,
nuclear factor of activated T cells.
1006  Circulation Research  September 29, 2017

muscle cells, neurons, and lymphocytes.116–118 As an under- 1 µmol/L.134 Otherwise, it is regulated by its endogenous in-
lying mechanism, it was proposed that calcineurin is able to hibitor calpastatin. Although some physiological functions
dephosphorylate MEF2, thereby blocking its sumoylation and have been demonstrated, calpain is mainly described to cleave
consequently promoting MEF2-dependent transcription.119 In proteins in pathological settings. In regard to cardiomyocytes,
addition, it was shown that calcineurin possesses the ability especially ischemia reperfusion settings and heart failure con-
to enhance MEF2 transcriptional activity via a calcineurin/ ditions depict situations of Ca2+ overload where calpain was
mAKAP/MEF2 complex formation.120 Another possible path- shown to be more active.135
way involves prominent calcineurin downstream target NFAT, Calpain was described to cleave PKCα, the most abundant
since Blaeser et al described activated calcineurin to induce PKC isoform in the heart, in its V3 hinge region, thereby sepa-
the formation of a MEF2/NFATc2 transcriptional complex.118 rating the C-terminal catalytic from the N-terminal regulatory
Irrespective of the exact mechanism, the functional improve- domain.136–138 Accordingly, without this regulatory domain
ment of MEF2 antagonization in a calcineurin-induced heart permanently inhibiting the catalytic function, the C-terminal
failure model argues for a relevant calcineurin-dependent domain is constitutively active. Furthermore, the C-terminal
MEF2 effect also in cardiomyocytes.121 Yet, one has to con- fragment of PKCα gains the ability to phosphorylate addition-
sider that MEF2 activation in this model of calcineurin over- al targets and to translocate to the nucleus. Here, the catalytic
expression may also be a secondary phenomenon during the fragment leads to HDAC5 phosphorylation, which results in
development of heart failure, which is not based on a direct nuclear export of HDAC5, and consequent activation of MEF2-
signaling of calcineurin to MEF2. Another aspect of inter- dependent genes.139 Aside from PKC, 2 other prominent cleav-
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

relation between the HDAC/MEF2 axis and NFAT was em- age targets of calpain have been described in regard to their
phasized by Dai et al,122 showing that NFATc3 is negatively influence on transcriptional regulation. For the phosphatase
regulated by class II HDACs through the DnaJ-Related Factor calcineurin, calpain was reported to cleave the autoinhibitory
Mrj. Notably, calcineurin activity can be negatively regulated domain, which results in translocation of calcineurin to the
by direct CaMKII-dependent phosphorylation, causing dimin- nucleus, causing increased and prolonged NFAT-dependent
ished NFAT translocation.123 This phenomenon has been dem- gene transcription.140 In addition, the calpain-mediated calci-
onstrated by us to account for hypertrophic cardiac growth neurin cleavage product was described to be independent of
in CaMKIIδ/CaMKIIγ double knockout (KO) mice.124 It has Ca2+/calmodulin binding and, therefore, constitutively active
moreover been suggested that calcineurin-dependent dephos- in its function.141 Moreover, calpain cleavage of LTCC was
phorylation of CaMKII at its autophosphorylation site vice proposed. Its distal C-terminal fragment was shown to local-
versa constitutes another negative crosstalk pathway between ize to the nucleus and alter transcriptional activity.142 Via this
these 2 enzymes.125 This pathway may also involve a more mechanism, the LTCC, for example, autoregulates its own ex-
indirect mechanism via calcineurin-dependent dephosphory- pression in cardiomyocytes.143
lation of protein phosphatase 1 inhibitor-1.126 Thereby, protein
phosphatase 1 activity is increased, which would consequently Direct Effects of Ca2+
reduce autophosphorylation of CaMKII.127 Figure 2 illustrates An interesting finding in terms of direct interaction between
the potential interplay between CaMKII and calcineurin and Ca2+ and chromatin is the observation that certain GC se-
their downstream signaling pathways. quences in the DNA have the ability to bind Ca2+, which leads
In addition to CaMKII, calcineurin via NFAT was also to local changes in the DNA structure.144 The physiological
reported to have an impact on the transcriptional coactivator relevance of this finding is still unclear, yet one has to consider
myocardin.128 Li et al129 could further show that NFAT binds that Ca2+, locally enhanced in nuclear microdomains, might
to the promotor region of myocardin, induces its expression, bind to a very specific region of the DNA and recruit Ca2+-
and synergistically with myocardin enhances expression of dependent enzymes to spatially modify chromatin structure
the LTCC α subunit. and potentially change transcription.
Beyond the regulation of NFAT, calcineurin was revealed
to interact with the transcription factor nuclear factor Y and ET Coupling in the Diseased Heart
affect gene expression of TXNIP (thioredoxin-interacting pro-
Ca2+ Cycling in Heart Failure
tein). Nuclear factor Y binds to the CCAAT element in promo-
Defects in cellular Ca2+-handling and alterations in Ca2+-
tor regions, and depending on the particular DNA sequence
dependent signaling pathways are molecular hallmarks of
either is an activator or repressor of gene expression.130 It
heart failure. The underlying mechanisms of cardiac dysfunc-
has been proposed that calcineurin dephosphorylates nuclear
tion may vary depending on the different causes and stages of
factor Y, therefore, decreases its affinity to the DNA and at-
the disease, yet several consistent findings on the dysregula-
tenuates its transcriptional repression of one of its targets—
tion of Ca2+ cycling have been characterized. On the one hand,
TXNIP.131 TXNIP, an endogenous inhibitor of antioxidant
structural changes in Ca2+-handling microdomains regularly
thioredoxin, is considered to be a relevant player in control-
ling energy metabolism and reactive oxygen species in cardiac occur such as decreases in T-tubule and nuclear invagination
pathologies.132,133 density and rearrangements of the nuclear transport machin-
ery.33,145–148 On the other hand, several Ca2+-handling proteins
Calpain show altered expression patterns and activity states. In par-
The nonlysosomal cysteine protease calpain gets only acti- ticular, defective SR Ca2+-handling is considered an important
vated at relatively high ambient Ca2+ concentrations of about aspect of heart failure pathophysiology. On a molecular level,
Dewenter et al   Calcium Signaling and Transcription   1007

impaired SERCA function, based on reduced protein expres- cycling protein modification versus effects on transcriptional
sion and increased SERCA inhibition by PLN, and increased regulation components is challenging. However, deciphering
open probability of RyR, based on hyperphosphorylation by the role of nuclear versus cytosolic CaMKII might deliver in-
PKA and CaMKII, seem to be causative.149 Therefore, SR sights into distinct mechanisms. By comparative approach, it
Ca2+ load is decreased because of diminished SR filling and was shown that overexpression of both splice variants induces
increased diastolic RyR Ca2+ leak. As a consequence of defec- activation of MEF2, mechanistically via phosphorylation of
tive SR Ca2+ cycling and SR Ca2+ depletion, Ca2+ transients HDAC4, whereas SR Ca2+ cycling is only affected by cyto-
in failing cardiomyocytes show distinct alterations: the am- solic CaMKIIδC.105 Accordingly, CaMKIIδ KO mice are pro-
plitude decreases, the time to peak prolongs, the rate of Ca2+ tected from cardiac remodeling upon pressure overload.162,163
removal is reduced, and the resting [Ca2+] is elevated.150 NCX In our studies on CaMKIIδ KO mice, we found a marked
is increased in expression and activity in various heart failure reduction in CaMKII-dependent HDAC4 phosphorylation
conditions151 and may influence these alterations in different and attenuated induction of the fetal gene program, while we
ways. Shifting the balance of cytosolic Ca2+ removal toward could not see any genotype-related disturbances in intracellu-
enhanced Ca2+ extrusion via NCX might further impair SR lar Ca2+ transients under either unstressed conditions or in the
Ca2+ load. In contrast, NCX can switch into reversed mode at presence of transverse aortic constriction.163 These results in-
high intracellular [Na+] as observed in heart failure, causing dicate that effects on Ca2+-handling are not absolutely required
Ca2+ influx which may contribute to diastolic Ca2+ overload.150 for the protective CaMKIIδ KO–dependent phenotype. In
Besides these global Ca2+ disturbances, distinct local changes contrast, Ling et al demonstrated in a different CaMKIIδ KO
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

in Ca2+ have been observed in heart failure. Ljubojevic et al mouse model that the increased SR Ca2+ leak after transverse
demonstrated that nuclear Ca2+ transients display pathological aortic constriction is completely abolished via prevention of
patterns earlier in disease development than the cytosolic Ca2+ RyR2-S2814 hyperphosphorylation, suggesting that restored
transients,33 suggesting an important role for Ca2+-dependent SR Ca2+ might underlie the beneficial effects of CaMKIIδ
maladaptive gene program activation. Consistent with this ob- deletion.162 However, attempts to rescue the cardiomyopathic
servation, changes in nucleocytoplasmic trafficking processes phenotype in CaMKIIδC TG mice by restoring SR function
were observed to precede cardiac functional deterioration in to our knowledge failed to date. SR-targeted CaMKII inhibi-
experimental heart failure.148 Moreover, increasing evidence tion indeed significantly reduced the SR Ca2+ leak but accel-
indicates that also other Ca2+ channels, less implicated in EC erated the development of cardiac remodeling in the TG.158
coupling, significantly contribute to disturbances in Ca2+- In addition, restoring SR load in CaMKIIδC TG by genetic
dependent pathways, as IP3 receptors and several TRPC KO of PLN showed no rescue effect but resulted in exacerba-
channels have been demonstrated to be upregulated in experi- tion of cardiac dysfunction and mortality in the TG.157 These
mental and human heart failure.152–155 findings strongly indicate that even cytosolic CaMKIIδC ex-
Changes in Ca2+-Dependent Transcriptional erts maladaptive effects in heart failure via SR-independent
Regulation in Heart Failure mechanisms.
Ca2+-dependent transcriptional pathways involving key regu- The critical involvement of the class IIa HDAC/MEF2 axis
lators such as CaMKII and calcineurin are overactive in the in the induction of cardiac hypertrophy and remodeling has
diseased heart and suggested to drive structural changes and also been demonstrated in genetic mouse models. HDAC9 KO
functional deterioration by inducing remodeling processes. mice are sensitized to hypertrophic signals including calcineu-
Table 1 gives an overview on in vivo studies on proteins in- rin overexpression, show an increased activation of the fetal
volved in these pathways with respect to their impact on cardi- gene program, and display a hypersensitive MEF2 activity.171
ac hypertrophy, cardiac function, cardiac fibrosis, and cardiac Similar findings have been reported for HDAC5 KO mice.170
inflammation. Regarding MEF2, characterization of MEF2A and MEF2C
CaMKII activity is consistently increased in human and in TG revealed a dosage-dependent cardiomyopathic phenotype
experimental heart failure,156,202 and activation of CaMKII has and reduction in ventricular performance,172 as well as an in-
been observed in response to multiple neurohumoral signals creased susceptibility to calcineurin overexpression-induced
that are enhanced in heart failure, including ANGII, ET-1, α- hypertrophy.121 In addition, MEF2D overexpression induces
AR, and β-AR agonists.71,80,203–209 This neurohumoral activation pathological cardiac remodeling and drives the fetal gene
of CaMKII has been linked to the induction of hypertrophic program, whereas MEF2D KO are protected against remod-
and fetal gene programs.204,210–212 In addition, CaMKII was eling and exhibit attenuated fetal gene activation in response
shown to act as a critical downstream mediator of noncanoni- to pressure overload and chronic β-adrenergic stimulation.101
cal Wnt signaling via dishevelled, which represents a widely Calcineurin is overactive not only in human heart failure
unrecognized but central pathway in myocardial remodeling but also in compensated cardiac hypertrophy.213,214 Notably,
processes.166 A causative role of CaMKII in heart failure devel- IRF8 (interferon regulatory factor 8), which acts as an in-
opment has been derived from cardiomyocyte-specific genetic hibitor of calcineurin downstream target NFATc1, has been
mouse models.87,124,162,163,165 Cardiac-specific overexpression of described to be downregulated in dilated and hypertrophic
cytosolic CaMKIIδC and nuclear CaMKIIδB induces dilated cardiomyopathy.215 In vitro data consistently indicate that
cardiomyopathy and cardiac hypertrophy, respectively.156,161 pharmacological and genetic calcineurin inhibition at-
Given the multiple targets of CaMKII in cardiomyocytes, tenuates cardiomyocyte hypertrophy induced by neurohu-
specifically estimating the relevance of effects based on Ca2+ moral stimulation.113,204,216 This antihypertrophic effect was
1008  Circulation Research  September 29, 2017

Table 1.  Overview on In Vivo Studies Investigating Ca2+-Dependent Signaling Proteins Involved in Transcriptional Pathways
With Respect to Their Impact on Cardiac Hypertrophy, Cardiac Function, Cardiac Fibrosis, and Cardiac Inflammation
Protein/Mutant Genetic Pharmacological/ Cardiac
Intervention Cardiac Growth Cardiac Function Cardiac Fibrosis
Name Model Peptide-Based Inhibition Inflammation
CaMKIIδC ↑(156,157)
none ↓↓(156–160) ↑(156)
TG ↑↑(159,160)
MI ↓↓(86) ↑(86)
CaMKIIδB TG none ↑( )161
↓( )161

CaMKIIδ TAC ↓( 162,163


) ↑(162) ↓(162,163)
KO MI ↑(85) ↓(85)
ISO →( ) 164
↑( )164
↓( )
164

CaMKIIδ/ TAC →( 124,165


) ↑( 124,165
) ↓(124,165)
CaMKIIγ
MI →(87) ↑(87) ↓↓(87) ↓(87)
DKO
ISO →(124) ↓↓(124)
DVL TG ↓( )166
↑( )166
↓(166)
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

CaMKII MI ↑(167) ↓(168)


AC3-I TG CnA TG →(169) ↑(169)
ISO ↓(167) ↑(167)
KN93 MI ↑(167)
HDAC5 none ↑(170)
KO TAC ↑(170)
ISO →(170)
HDAC5/
CnA (constitutively KO/TG none ↑↑↑(170)*
active CnA)
HDAC9 none ↑(171)
KO TAC ↑(171)
ISO →(170)
HDAC9/
CnA (constitutively KO/TG none ↑↑↑(171)*
active CnA)
HDAC5/
DKO none ↑(170)
HDAC9
MEF2A ↑↑(172)
none ↓↓(172)
TG ↑(121)
TAC ↑(172) ↓↓(172)
MEF2C TG none ↑(172) ↓↓(172)
MEF2D TG none ↓(101) ↑↑(101)
MEF2D TAC ↓(101) ↑(101) ↓↓(101)
KO
ISO ↓( )101
↑( )101

CnA (constitutively →(121,175)


active form) ↑↑(113,121,152,173–177)
TG none ↓(173,178,179) ↑(113,178)
↑(178–180) ↓↓(174,177)
CnAß TAC ↓( )181

KO ISO ↓(181)
ANG ↓(181)
dnCnA TG TAC ↓(182) ↓(182)

(Continued )
Dewenter et al   Calcium Signaling and Transcription   1009

Table 1. Continued
Protein/Mutant Genetic Pharmacological/ Cardiac
Intervention Cardiac Growth Cardiac Function Cardiac Fibrosis
Name Model Peptide-Based Inhibition Inflammation
CaN →(183–186) ↑(188)
TAC →(187)
↓(187–190) →(189)
→(191) →(192,193)
MI →(191,192)
CsA ↓(192–194) ↓(191,194)
↓(113,177,195)
CnA TG ↑↑(177)
↓↓(113,177,195)
ANGII ↓(196)
→(184,185)
TAC ↓↓(197)
FK506 ↓(197)
CnA TG ↓↓(195)
TAC ↓(198)
ΔCain TG
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

ISO ↓(198)
TAC ↓(198)
ΔAKAP TG
ISO ↓(198)
TAC ↓(199) →(199) →(199)
MI ↓( )
200
↑( )
200
↓↓(200)
MCIP1 TG
CnA TG ↓(174) ↑↑(174)
ISO ↓(174)
Zaki-4β TG TAC ↓(201) →(201)
NFATc3 Δ317 TG none ↑( )
113
↑(113)
NFATc3 TAC ↓(173)
KO CnA TG ↓(173) →(173)
ANGII ↓( )
173

NFATc4 TG none ↑(177)


NFATc4 KO CnA TG →(173)
CAMTA2 TG none ↑↑(65)
CAMTA2 TAC ↓(65)
KO ISO ↓(65)
ANGII ↓(65) ↓(65)
Calpain1 KO MI ↑( )
137

ANGII indicates chronic angiotensin II infusion; AKAP, A-kinase anchoring protein; CaMKII, Ca /calmodulin-dependent kinase II; CAMTA, calmodulin-binding
2+

transcriptional activator; CaN, calcineurin; CnA, calcineurin subunit A; CsA, cyclosporin A; DKO, double knockout; dnCnA, dominant negative calcineurin subunit
A; DVL, dishevelled; HDAC, histone deacetylase; ISO, chronic isoprenaline infusion; KO, knockout; MCIP, myocyte-enriched calcineurin-interacting protein; MEF2,
myocyte enhancer factor 2; MI, myocardial infarction; NFAT, nuclear factor of activated T cells; PE+ANGII, chronic application of phenylephrine and angiotensin
II; TAC, transverse aortic constriction; TG, transgenic overexpression; ↑, <2-fold increase compared with respective control; ↑↑, >2-fold increase; ↑↑↑, >3-fold
increase (*Compared with the wild-type control group); →, no significant alterations; ↓, <50% decrease; and ↓↓, >50% decrease.

confirmed in various in vivo models.181,187,195,198 Further evi- revealing that NFATc3 deletion protects against calcineurin
dence for a causative role of calcineurin and its downstream overexpression-induced, pressure overload–induced, and
effector NFAT in heart failure was obtained from cardiac- ANGII-induced cardiac hypertrophy.173
specific genetic mouse models. Calcineurin and NFAT3 TG There are, however, some inconsistent findings about the
develop cardiac hypertrophy and heart failure, with NFAT role of the calcineurin/NFAT pathway in physiological ver-
and GATA4 synergistically activating the fetal gene pro- sus pathological hypertrophy because several studies also
gram.113 Of note, calcineurin TG mice were shown to ex- suggest calcineurin-dependent pathways to be involved in
hibit cardiac remodeling despite increased SR function.217 nonmaladaptive growth. Interestingly, calcineurin upregula-
The critical involvement of NFAT in cardiac hypertrophy tion is more pronounced in compensated cardiac hypertrophy
was additionally investigated by studying NFATc3 KO mice, compared with heart failure.214 Eto et al reported calcineurin
1010  Circulation Research  September 29, 2017

upregulation in adaptive cardiac hypertrophy upon exercise striking, but not so the evidence for its role in pathological re-
training in rats.218 In line with this finding, calcineurin inhi- modeling and contractile dysfunction. Also the time course of
bition by MCIP1 (myocyte-enriched calcineurin-interacting calcineurin activation during the compensated versus the de-
protein 1) overexpression in mice has been shown to attenuate compensated phase of heart failure supports this perspective.
exercise-induced cardiac hypertrophy.174 Moreover, calcineu- For instance, our laboratory showed that calcineurin activation
rin activity was suggested to be a requirement for adaptive hy- precedes CaMKII activation upon the induction of pathologi-
pertrophy during pregnancy.219 Another observation that might cal pressure overload and is, in contrast to CaMKII, down-
challenge the view of calcineurin as an overall detrimental ET regulated during the transition to heart failure.124 These data
coupling player is related to its role in dilated cardiomyopa- may indicate that increased calcineurin/NFAT activity plays
thy. KO mice lacking the stress responsive isoform of calci- an important role in compensated and early cardiac hypertro-
neurin exhibit enhanced cardiomyocyte apoptosis and cardiac phy rather than in long-term cardiac remodeling processes.
dysfunction in a genetic mouse model of muscle LIM protein The observation that calcineurin is upregulated in exercise-
deficiency. Conversely, expression of activated calcineurin induced hypertrophy and required for adaptive cardiac growth
improves function and adverse remodeling in this model, sug- supports from our point of view this conclusion.
gesting a rather protective role in this scenario.175 Taken together, it will be important in the future to detect
Also findings in the context of the interrelation between dynamic changes of different Ca2+-dependent signaling mol-
CaMKII and calcineurin signaling indicate calcineurin ac- ecules in the course of heart failure development but also to
tivation in nonpathological cardiac growth. In line with the increase our understanding of the distinct Ca2+ sources that
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

above-mentioned detrimental role of overactivated CaMKII, account for the differential regulation in adaptive versus mal-
CaMKIIδ/CaMKIIγ double KO mice are protected against adaptive signaling cascades.
cardiac dysfunction and interstitial fibrosis upon pressure Increasing evidence indicates that IP3 receptors, TRPC
overload. However, these mice do develop cardiac hypertro- channels, and SOCE dysregulation significantly contribute to
phy, but remarkably without signs of pathological remodel- detrimental enhancement of the above-mentioned transcrip-
ing, establishing a dissociation of cardiac hypertrophy on the tional pathways in heart failure. Table 2 gives an overview
one hand and pathological remodeling on the other hand. In on in vivo studies investigating these Ca2+-handling proteins
the same study, these mice also show an increased activity with respect to their impact on cardiac hypertrophy, cardiac
of endogenous calcineurin and exaggerated cardiac hyper- function, cardiac fibrosis, and the suggested involvement in
trophy upon exercise, again without signs of heart failure. transcriptional regulation.
Mechanistically, calcineurin activation was shown to account IP3R2 TG mice develop cardiac hypertrophy in re-
for cardiac growth in this model because of completely dimin- sponse to chronic isoproterenol infusion, Gαq overexpres-
ished CaMKII-dependent phosphorylation at the inhibitory sion, and exercise stimulation, which is blocked via genetic
calcineurin phosphosite Ser-411 in the CaMKIIδ/CaMKIIγ calcineurinAβ deletion.17 Moreover, local nuclear envelope
double KO mice. Notably, this striking effect was only ob- Ca2+ release via IP3 receptors in response to ET-1 has been
served in the CaMKIIδ/CaMKIIγ double KO model, which shown to affect the HDAC5/MEF2 axis independent of the
lacks any measurable CaMKII activity in the heart, but not global Ca2+ transients.231
in the CaMKII single KO models, because CaMKIIδ and Also Ca2+ entry via TRPC channels has been linked to
CaMKIIγ are able to compensate for each other.124 calcineurin activation. TRPC3 TG develop hypertrophy and
In contrast to the indicated crucial involvement of the cal- contractile dysfunction in response to pressure overload,
cineurin/NFAT pathway activation in adaptive cardiac growth, which is abrogated in a calcineurinAβ KO background.225 In
Hainsey et al reported no effect of calcineurin inhibitor cyclo- addition, overexpression of TRPC6 was demonstrated to in-
sporin A on exercise-induced cardiac hypertrophy in wild-type duce cardiomyopathy and accelerate cardiac remodeling, ac-
mice.220 Other data even suggest that cardiac antiremodeling companied by an increase in NFAT-dependent expression of
effects of exercise training involve deactivation of the calci- the β-myosin heavy chain.152 Of note, Seo et al demonstrated
neurin/NFAT pathway.221 Consistent with the latter, Wilkins et that only combined deletion of TRPC3 and TRPC6 reduces
al also observed a decrease in NFAT activity upon exercise.222 the hypertrophic response to pressure overload, whereas indi-
An aspect that should be taken into consideration when vidual gene deletion is not protective.226 Moreover, studies on
trying to explain the discrepancies on the role of calcineurin dominant-negative TRPC3, TRPC6, and TRPC4 revealed that
in nonmaladaptive growth versus decompensated heart failure inhibition of TRPCs reduces the activity of the calcineurin/
is the applied genetic model. The studies investigating calci- NFAT pathway and attenuates hypertrophic response in vitro
neurin TG mice used a constitutively active form of calcineu- and in vivo.228 Interestingly, protection against neurohumor-
rin for the transgenic overexpression. These mice show severe al-induced cardiac hypertrophy in TRPC1/C4 KO mice was
cardiac remodeling accompanied by contractile dysfunction shown to involve a reduction not only in calcineurin activity
and increased collagen deposition. However, the fact that the but also in MEF2-dependent gene activation.227
overexpressed calcineurin is constitutively active might ac- Recent experimental data also suggest SOCE to play an
count for some divergences in the results compared with stud- important role in activating transcriptional pathways involved
ies investigating the modulation of endogenous calcineurin. in cardiomyocyte hypertrophy. STIM1-dependent Ca2+ signal-
Taking all of the studies on genetic and pharmacological cal- ing was revealed to activate the calcineurin/NFAT pathway
cineurin and NFAT inhibition into account (Table 1), the rele- and CaMKII signaling in vitro.232,233 Hulot et al emphasized
vance of the calcineurin/NFAT axis for cardiac hypertrophy is the in vivo relevance of STIM1 in cardiac hypertrophy,
Dewenter et al   Calcium Signaling and Transcription   1011

Table 2.  Overview on In Vivo Studies Investigating Ca2+-Handling Proteins With Respect to Their Impact on
Cardiac Hypertrophy, Cardiac Function, Cardiac Fibrosis, and the Suggested Involvement in Transcriptional
Pathways
Protein/Mutant
Name Genetic Model Intervention Cardiac Growth Cardiac Function Cardiac Fibrosis Suggested Pathway
IP3R2 none ↑(17) →(17)
TG TAC ↑(17) Calcineurin-NFAT17
ISO ↑(17)
IP3R2 KO TAC →(223) →(223)
TRPC1 Calcineurin-
KO TAC ↓(224) ↑(224) ↓(224)
NFATc3224
TRPC3 none ↑(225) ↓(225)
TG TAC ↑(225) ↓(225) Calcineurin-NFAT225
PE+ANGII ↑( )
225

TRPC3 KO TAC →(226) →(226)


Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

TRPC6 none ↑(152)


TG Calcineurin-NFAT152
TAC ↑(152) ↓(152)
TRPC6 KO TAC →(226) →(226)
TRPC3/ →(227)
TRPC6 TAC ↑(226)
↓(226)
DKO Calcineurin-NFAT226
ISO →(227)
ANG →(227)
TRPC1/ TAC ↓(227) ↑(227) ↓(227) Calcineurin-NFAT227
TRPC4
DKO ISO ↓(227)
CaMKII-MEF2227
ANG ↓(227)
dnTRPC3 TAC ↓(228) ↑(228) ↓(228)
TG Calcineurin-NFAT228
PE+ANGII ↓(228)
dnTRPC4 TG TAC ↓(228) Calcineurin-NFAT228
dnTRPC6 TAC ↓(228) ↑(228) ↓(228)
TG Calcineurin-NFAT228
PE+ANGII ↓(228)
STIM1 none ↑↑(229) ↓(229) Calcineurin-NFAT229
TAC ↑( )
229
↓( )
229

TG
ISO ↑( )
229
CaMKII-MEF2229
PE+ANGII ↑(229) ↑↑(229)
STIM1 none →(23,230) ↓(23,230) ↑(230) FAK-Akt23
KO
TAC ↓( )
23
→( ) 23
ERK1/223
ANGII indicates chronic angiotensin II infusion; CaMKII, Ca2+/calmodulin dependent kinase II; DKO, double knockout; dnTRPC, dominant
negative transient receptor potential C; IP3R, inositol-1,4,5-trisphosphate (IP3) receptor; ISO, chronic isoprenaline infusion; KO, knockout; MEF2,
myocyte enhancer factor 2; NFAT, nuclear factor of activated T cells; PE+ANGII, chronic application of phenylephrine and angiotensin II; STIM1,
Stromal interaction molecule-1; TAC, transverse aortic constriction; TG, transgenic overexpression; TRPC, transient receptor potential C; ↑, <2-
fold increase compared with respective control; ↑↑, >2-fold increase; →, no significant alterations; ↓, <50% decrease; and ↓↓, >50% decrease.

demonstrating that shRNA-mediated STIM1 gene silencing contractility and rearrangements of cytoskeletal structures
protects against cardiac growth in response to pressure over- under unstressed conditions.23 The important role of STIM1 in
load, associated with a reduction in nuclear NFATc3 trans- cardiac integrity was further illustrated by Collins et al, dem-
location.234 Conversely, it was shown that overexpression of onstrating that cardiac-restricted deletion of STIM1 leads to
STIM1 results in cardiac hypertrophy and contractile dysfunc- increased ER stress, mitochondrial disorganization, and pro-
tion.229 The cardiac-specific STIM1 KO mice from Parks et al gressive cardiac remodeling in mice.230 This shapes an overall
exhibited similar results with regard to attenuation of hyper- complex picture of STIM1-dependent cellular effects with
trophic response, but additionally presented impaired baseline broad implications that are up to now still poorly understood.
1012  Circulation Research  September 29, 2017

In conclusion, Ca2+-dependent transcriptional regula- Mice with this mutation exhibit pathological SR Ca2+ release
tors are evidently involved in heart failure pathophysiology. events and reduced SR Ca2+ load. Interestingly, this seems to
The majority is overactive in the state of heart disease and in primarily result in an increase in arrhythmia susceptibility
experimental in vivo models, and numerous studies empha- but causes only a slight decrease in contractile function with
sized that Ca2+-dependent ET effectors and pathways are able aging. Thus, it seems evident that CaMKII-dependent RyR
to dramatically change the cardiac phenotype. Nevertheless, phosphorylation at the 2814 site causes pathological altera-
whereas some few downstream targets are well characterized, tions in SR Ca2+-handling and arrhythmias. However, a clear
most of them remain elusive just as the interaction of different picture with respect to the pathogenesis of all aspects of heart
Ca2+-dependent pathways and the question whether 1 path- failure including contractile dysfunction cannot yet be drawn.
way is solely maladaptive or does also possess some adaptive Given the complexity of RyR regulation in a macromolecular
functions. complex with multiple posttranslational modifications, target-
ing proteins and interaction partners, future research will need
Therapeutic Implications
to shape an integrated view on RyR regulation, causes of RyR
Current pharmacological treatment options in heart failure (be-
dysfunction, and their relevance in the pathogenesis of heart
ta-blockers, angiotensin-converting enzyme inhibitors, angio-
failure.
tensin receptor blockers, and aldosterone inhibitors) are in line
Because decreased Ca2+ uptake into the SR is a character-
with the paradigm of neurohumoral overactivation. Indeed,
istic observation in heart failure, another promising approach
angiotensin, endothelin, and catecholamines are reportedly
may be enhancement of SR Ca2+ reuptake to rescue patho-
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

upregulated in heart failure,235–238 and exposure to excessive


logically altered Ca2+ cycling. In this regard, gene therapy ap-
amounts has detrimental effects on the cellular level.239–241
proaches may offer yet unexplored possibilities. Especially
Conversely, neurohumoral blockade effectively enhances heart
SERCA, SUMO-1, and S100A1 vectors proved to have bene-
function, improves clinical outcome, and reduces hospitaliza-
tion.242,243 Despite this, the overall mortality of heart failure is ficial effects on heart failure in animal models.255–257 However,
still high. In addition, tolerability of the optimal doses of stan- the outcome of the latest clinical trial for adeno-associated
dard medications is often limited because of contraindications, virus 1–delivered SERCA gene therapy (CUPID 2) could not
interactions, and adverse effects. This underlines the necessity yet successfully establish this approach in the clinical set-
to find novel and relevant therapeutic targets. ting.258 Although these results do not argue against SERCA as
Different neurohumoral stimuli were indeed shown to af- a promising target, ongoing difficulties with the gene therapy
fect cellular Ca2+, and neurohumoral inhibition consequently approach regarding vector design, delivery methods, and is-
improves Ca2+-handling, possibly thereby contributing to sues with humoral immunity need to be solved to use gene
clinical amelioration of contractility.244,245 The fact that Ca2+ is therapy for the re-expression of downregulated Ca2+-handling
regulated by a multitude of pathways, and the finding that Ca2+ proteins such as SERCA or S100A1.259
itself affects various cellular functions such as excitation, con- Moreover, the major Ca2+-dependent signaling enzymes,
traction, metabolism, and transcription, turns it into a nexus CaMKII and calcineurin, are focus of attempts to pharmaco-
of signaling cascades in cardiomyocytes. On the background logically treat heart failure. In case of CaMKII, it has been
of altered Ca2+-handling in heart failure, it was thus proposed demonstrated that induced genetic KO slows down the pro-
that restoring Ca2+ cycling back to the physiological state is a gression of heart failure development in response to pressure
promising therapeutic goal. However, defining a potent target overload, herewith providing further evidence for its thera-
turned out to be rather challenging. peutic potential.165 In addition, recent findings suggest that
As mentioned above, increased SR Ca2+ leak is 1 char- CaMKII blockade would complement the effect of the current
acteristic finding in failing hearts; thus, restabilization of the standard therapy.159 However, given the fundamental role of
RyR is thought to be a rationale concept.246,247 Still, the func- CaMKII in various physiological cellular functions involving,
tional role and the relative impact of the PKA- and CaMKII- for example, synaptic plasticity,260 fertility,261 and immuno-
dependent phosphorylation of RyR remain controversial. logic memory,262 pharmacologically inhibiting global CaMKII
Studies on PKA-phospho-resistant RyR mutant mice could might presumably be accompanied by serious adverse ef-
not consistently demonstrate the relevance of this phospho- fects. Thus, for a successful application, the issues of selec-
site for SR Ca2+ leak or cardiac function in experimental heart tive organ targeting and CaMKII isoform specificity have to
failure models.248–250 In contrast, the association of CaMKII- be addressed as well as the issue of target-specific inhibition,
dependent RyR phosphorylation with Ca2+ sparks is well es- which requires a deeper understanding of the critical CaMKII
tablished.251,252 Findings on RyR mutants that prevent or mimic downstream mechanisms in heart failure. Inhibitor develop-
CaMKII-dependent phosphorylation offered insights into the ment programs with focus on myocardial CaMKII have been
functional relevance of this phosphorylation site at S2814. launched by several companies,263 but the way into clinical
The phospho-resistant S2814A mutation was shown to inhibit application is not clear to date.
Ca2+ leakage from the SR, improve SR Ca2+ load, and to be Cardiac calcineurin is predominantly described to be in-
protective in the setting of transverse aortic constriction.253 In volved in pathological settings. However, this picture is con-
contrast, however, this RyR mutation had no protective im- troversial and has been challenged repeatedly, as discussed
pact on cardiac dysfunction upon myocardial infarction.253 above. Referring to Table 1, the evidence for calcineurin be-
Conversely, the S2814D mutation depicts a constitutively ac- ing involved in cardiac hypertrophy is indeed overwhelming,
tivated form of the CaMKII-dependent RyR phosphosite.254 but especially with regard to contractile function, the evidence
Dewenter et al   Calcium Signaling and Transcription   1013

for the benefit of calcineurin inhibition is not consistent. In 3. Frey N, McKinsey TA, Olson EN. Decoding calcium signals involved
in cardiac growth and function. Nat Med. 2000;6:1221–1227. doi:
part, this could be related to different models of experimental
10.1038/81321.
heart failure, inhibitor strategies, doses, treatment time points, 4. Bers DM. Calcium cycling and signaling in cardiac myocytes. Annu Rev
and treatment periods used in the various studies. These in- Physiol. 2008;70:23–49. doi: 10.1146/annurev.physiol.70.113006.100455.
consistencies might also explain why, despite the accessibility 5. Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracel-
lular signalling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600. doi:
of established pharmaceutical compounds, translation of the 10.1038/nrm1983.
findings around calcineurin inhibition into the clinics was not 6. Backs J, Olson EN. Control of cardiac growth by histone acety-
successful yet. A recent phase III trial with single application lation/deacetylation. Circ Res. 2006;98:15–24. doi: 10.1161/01.
of CsA in patients after myocardial infarction somewhat de- RES.0000197782.21444.8f.
7. Ibrahim M, Gorelik J, Yacoub MH, Terracciano CM. The structure
picts this translational dilemma and shows no improvement and function of cardiac T-tubules in health and disease. Proc Biol Sci.
in clinical outcomes or ventricular remodeling compared with 2011;278:2714–2723. doi: 10.1098/rspb.2011.0624.
the control group.264 8. Bers DM, Guo T. Calcium signaling in cardiac ventricular myocytes. Ann
N Y Acad Sci. 2005;1047:86–98. doi: 10.1196/annals.1341.008.
Even though the recent clinical trials following the prem- 9. Lehman W, Craig R, Vibert P. Ca(2+)-induced tropomyosin movement
ise to rescue cellular Ca2+ cycling did not yet meet the ex- in Limulus thin filaments revealed by three-dimensional reconstruction.
pectations, and also pharmacological inhibition of the major Nature. 1994;368:65–67. doi: 10.1038/368065a0.
Ca2+-dependent CaMKII and calcineurin pathways could 10. Bers DM. Cardiac excitation-contraction coupling. Nature. 2002;415:198–
205. doi: 10.1038/415198a.
not be translated into clinical application to date, these Ca2+- 11. Heijman J, Dewenter M, El-Armouche A, Dobrev D. Function and regula-
regulated enzymes were convincingly shown to be important tion of serine/threonine phosphatases in the healthy and diseased heart. J
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

players in cardiac pathologies and, therefore, their cascades Mol Cell Cardiol. 2013;64:90–98. doi: 10.1016/j.yjmcc.2013.09.006.
12. Burgoyne JR, Mongue-Din H, Eaton P, Shah AM. Redox signaling in
present promising therapeutic targets. Concurrently, these cardiac physiology and pathology. Circ Res. 2012;111:1091–1106. doi:
findings illustrate the major problems we face when dealing 10.1161/CIRCRESAHA.111.255216.
with the paradigm of globally disturbed Ca2+ cycling and Ca2+- 13. Sun J, Picht E, Ginsburg KS, Bers DM, Steenbergen C, Murphy

dependent signaling pathways, that is, deciphering (1) how E. Hypercontractile female hearts exhibit increased S-nitrosylation
of the L-type Ca2+ channel alpha1 subunit and reduced ischemia/
Ca2+-dependent pathways influence different pathological reperfusion injury. Circ Res. 2006;98:403–411. doi: 10.1161/01.
processes at varied time points, (2) how specifically and se- RES.0000202707.79018.0a.
lectively these Ca2+-dependent pathways mediate maladaptive 14. Tétreault MP, Bourdin B, Briot J, Segura E, Lesage S, Fiset C, Parent L.
Identification of glycosylation sites essential for surface expression of the
features, and (3) how Ca2+-dependent pathways interrelate.
CaVα2δ1 subunit and modulation of the cardiac CaV1.2 channel activity.
To circumvent the complexity of CaMKII or calcineu- J Biol Chem. 2016;291:4826–4843. doi: 10.1074/jbc.M115.692178.
rin signaling and their time-dependent and isoform-specific 15. Erickson JR, Pereira L, Wang L, Han G, Ferguson A, Dao K, Copeland
peculiarities, another therapeutic approach would be to sys- RJ, Despa F, Hart GW, Ripplinger CM, Bers DM. Diabetic hyperglycae-
mia activates CaMKII and arrhythmias by O-linked glycosylation. Nature.
tematically search for downstream effectors, elucidate their 2013;502:372–376. doi: 10.1038/nature12537.
pathophysiological relevance, and focus on the targets solely 16. Berridge MJ. Inositol trisphosphate and calcium signalling. Nature.

mediating maladaptive features. Pharmacological interven- 1993;361:315–325. doi: 10.1038/361315a0.
17. Nakayama H, Bodi I, Maillet M, DeSantiago J, Domeier TL, Mikoshiba
tions further downstream bear the potential to offer several
K, Lorenz JN, Blatter LA, Bers DM, Molkentin JD. The IP3 receptor
advantages like a higher specificity in the mechanism of ac- regulates cardiac hypertrophy in response to select stimuli. Circ Res.
tion and, therefore, a lower possibility of on-target adverse 2010;107:659–666. doi: 10.1161/CIRCRESAHA.110.220038.
effects. Given the various disease causes of heart failure and 18. Vennekens R. Emerging concepts for the role of TRP channels in the
cardiovascular system. J Physiol. 2011;589:1527–1534. doi: 10.1113/
the complex interplay between disturbed signaling pathways, jphysiol.2010.202077.
deciphering nodal points of maladaptive Ca2+-dependent sig- 19. Yue Z, Xie J, Yu AS, Stock J, Du J, Yue L. Role of TRP channels in the
naling might finally lay the foundation for effective novel cardiovascular system. Am J Physiol Heart Circ Physiol. 2015;308:H157–
therapeutic interventions. H182. doi: 10.1152/ajpheart.00457.2014.
20. Eder P, Molkentin JD. TRPC channels as effectors of cardiac hypertrophy.
Circ Res. 2011;108:265–272. doi: 10.1161/CIRCRESAHA.110.225888.
Acknowledgments 21. Liao Y, Plummer NW, George MD, Abramowitz J, Zhu MX, Birnbaumer
We thank Dr Christiane Vettel for her valuable suggestions. L. A role for Orai in TRPC-mediated Ca2+ entry suggests that a
TRPC:Orai complex may mediate store and receptor operated Ca2+
entry. Proc Natl Acad Sci USA. 2009;106:3202–3206. doi: 10.1073/
Sources of Funding pnas.0813346106.
H.A. Katus and J. Backs were supported by grants from the DZHK 22. Tojyo Y, Morita T, Nezu A, Tanimura A. Key components of store-operat-
(Deutsches Zentrum für Herz-Kreislauf-Forschung—German Centre ed Ca2+ entry in non-excitable cells. J Pharmacol Sci. 2014;125:340–346.
for Cardiovascular Research) and by the BMBF (German Ministry of 23. Parks C, Alam MA, Sullivan R, Mancarella S. STIM1-dependent Ca(2+)
Education and Research). microdomains are required for myofilament remodeling and signaling in
the heart. Sci Rep. 2016;6:25372. doi: 10.1038/srep25372.
24. Park CY, Shcheglovitov A, Dolmetsch R. The CRAC channel activator
Disclosures STIM1 binds and inhibits L-type voltage-gated calcium channels. Science.
None. 2010;330:101–105. doi: 10.1126/science.1191027.
25. Sampieri A, Zepeda A, Asanov A, Vaca L. Visualizing the store-oper-
ated channel complex assembly in real time: identification of SERCA2
References as a new member. Cell Calcium. 2009;45:439–446. doi: 10.1016/j.
1. Berridge MJ, Lipp P, Bootman MD. The versatility and universal- ceca.2009.02.010.
ity of calcium signalling. Nat Rev Mol Cell Biol. 2000;1:11–21. doi: 26. Krapivinsky G, Krapivinsky L, Stotz SC, Manasian Y, Clapham DE.
10.1038/35036035. POST, partner of stromal interaction molecule 1 (STIM1), targets STIM1
2. Bers DM. Excitation-Contraction Coupling and Cardiac Contractile to multiple transporters. Proc Natl Acad Sci USA. 2011;108:19234–19239.
Force. Dordrecht; The Netherlands: Kluwer Academic Publishers; 2001. doi: 10.1073/pnas.1117231108.
1014  Circulation Research  September 29, 2017

27. Stiber J, Hawkins A, Zhang ZS, Wang S, Burch J, Graham V, Ward CC, 48. Allbritton NL, Meyer T, Stryer L. Range of messenger action of calcium
Seth M, Finch E, Malouf N, Williams RS, Eu JP, Rosenberg P. STIM1 ion and inositol 1,4,5-trisphosphate. Science. 1992;258:1812–1815.
signalling controls store-operated calcium entry required for development 49. Remus TP, Zima AV, Bossuyt J, Bare DJ, Martin JL, Blatter LA, Bers
and contractile function in skeletal muscle. Nat Cell Biol. 2008;10:688– DM, Mignery GA. Biosensors to measure inositol 1,4,5-trisphosphate
697. doi: 10.1038/ncb1731. concentration in living cells with spatiotemporal resolution. J Biol Chem.
28. Fricker M, Hollinshead M, White N, Vaux D. Interphase nuclei of many 2006;281:608–616. doi: 10.1074/jbc.M509645200.
mammalian cell types contain deep, dynamic, tubular membrane-bound 50. Boivin B, Chevalier D, Villeneuve LR, Rousseau E, Allen BG.

invaginations of the nuclear envelope. J Cell Biol. 1997;136:531–544. Functional endothelin receptors are present on nuclei in cardiac ven-
29. Wu X, Bers DM. Sarcoplasmic reticulum and nuclear envelope are one tricular myocytes. J Biol Chem. 2003;278:29153–29163. doi: 10.1074/
highly interconnected Ca2+ store throughout cardiac myocyte. Circ Res. jbc.M301738200.
2006;99:283–291. doi: 10.1161/01.RES.0000233386.02708.72. 51. Tadevosyan A, Maguy A, Villeneuve LR, Babin J, Bonnefoy A, Allen
30. Malhas A, Goulbourne C, Vaux DJ. The nucleoplasmic reticulum: form and BG, Nattel S. Nuclear-delimited angiotensin receptor-mediated signaling
function. Trends Cell Biol. 2011;21:362–373. doi: 10.1016/j.tcb.2011.03.008. regulates cardiomyocyte gene expression. J Biol Chem. 2010;285:22338–
31. Escobar M, Cardenas C, Colavita K, Petrenko NB, Franzini-Armstrong 22349. doi: 10.1074/jbc.M110.121749.
C. Structural evidence for perinuclear calcium microdomains in car- 52. Wu SC, Dahl EF, Wright CD, Cypher AL, Healy CL, O’Connell TD.
diac myocytes. J Mol Cell Cardiol. 2011;50:451–459. doi: 10.1016/j. Nuclear localization of a1A-adrenergic receptors is required for signal-
yjmcc.2010.11.021. ing in cardiac myocytes: an “inside-out” a1-AR signaling pathway. J Am
32. Schermelleh L, Carlton PM, Haase S, Shao L, Winoto L, Kner P, Burke Heart Assoc. 2014;3:e000145.
B, Cardoso MC, Agard DA, Gustafsson MG, Leonhardt H, Sedat JW. 53. Wright CD, Chen Q, Baye NL, Huang Y, Healy CL, Kasinathan S,

Subdiffraction multicolor imaging of the nuclear periphery with 3D O’Connell TD. Nuclear alpha1-adrenergic receptors signal activated
structured illumination microscopy. Science. 2008;320:1332–1336. doi: ERK localization to caveolae in adult cardiac myocytes. Circ Res.
10.1126/science.1156947. 2008;103:992–1000. doi: 10.1161/CIRCRESAHA.108.176024.
33. Ljubojevic S, Radulovic S, Leitinger G, et al. Early remodeling of peri- 54. Ibarra C, Vicencio JM, Estrada M, et al. Local control of nuclear calcium
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

nuclear Ca2+ stores and nucleoplasmic Ca2+ signaling during the devel- signaling in cardiac myocytes by perinuclear microdomains of sarcolem-
opment of hypertrophy and heart failure. Circulation. 2014;130:244–255. mal insulin-like growth factor 1 receptors. Circ Res. 2013;112:236–245.
doi: 10.1161/CIRCULATIONAHA.114.008927. doi: 10.1161/CIRCRESAHA.112.273839.
34. Görlich D, Kutay U. Transport between the cell nucleus and the cyto- 55. Boivin B, Lavoie C, Vaniotis G, Baragli A, Villeneuve LR, Ethier N, Trieu
plasm. Annu Rev Cell Dev Biol. 1999;15:607–660. doi: 10.1146/annurev. P, Allen BG, Hébert TE. Functional beta-adrenergic receptor signalling on
cellbio.15.1.607. nuclear membranes in adult rat and mouse ventricular cardiomyocytes.
35. Panté N, Kann M. Nuclear pore complex is able to transport macromol- Cardiovasc Res. 2006;71:69–78. doi: 10.1016/j.cardiores.2006.03.015.
ecules with diameters of about 39 nm. Mol Biol Cell. 2002;13:425–434. 56. Ljubojevic S, Bers DM. Nuclear calcium in cardiac myocytes. J Cardiovasc
doi: 10.1091/mbc.01-06-0308. Pharmacol. 2015;65:211–217. doi: 10.1097/FJC.0000000000000174.
36. Tanaka H, Kawanishi T, Matsuda T, Takahashi M, Shigenobu K.
57. Vaniotis G, Del Duca D, Trieu P, Rohlicek CV, Hébert TE, Allen BG.
Intracellular free Ca2+ movements in cultured cardiac myocytes as Nuclear β-adrenergic receptors modulate gene expression in adult rat
shown by rapid scanning confocal microscopy. J Cardiovasc Pharmacol. heart. Cell Signal. 2011;23:89–98. doi: 10.1016/j.cellsig.2010.08.007.
1996;27:761–769. 58. Guatimosim S, Amaya MJ, Guerra MT, Aguiar CJ, Goes AM, Gómez-
37. Ljubojević S, Walther S, Asgarzoei M, Sedej S, Pieske B, Kockskämper Viquez NL, Rodrigues MA, Gomes DA, Martins-Cruz J, Lederer WJ,
J. In situ calibration of nucleoplasmic versus cytoplasmic Ca²+ concen- Leite MF. Nuclear Ca2+ regulates cardiomyocyte function. Cell Calcium.
tration in adult cardiomyocytes. Biophys J. 2011;100:2356–2366. doi: 2008;44:230–242. doi: 10.1016/j.ceca.2007.11.016.
10.1016/j.bpj.2011.03.060. 59. Guo A, Cala SE, Song LS. Calsequestrin accumulation in rough endo-
38. Hohendanner F, McCulloch AD, Blatter LA, Michailova AP. Calcium plasmic reticulum promotes perinuclear Ca2+ release. J Biol Chem.
and IP3 dynamics in cardiac myocytes: experimental and computational 2012;287:16670–16680. doi: 10.1074/jbc.M112.340927.
perspectives and approaches. Front Pharmacol. 2014;5:35. doi: 10.3389/ 60. Strynadka NC, James MN. Crystal structures of the helix-loop-helix calci-
fphar.2014.00035. um-binding proteins. Annu Rev Biochem. 1989;58:951–998. doi: 10.1146/
39. Humbert JP, Matter N, Artault JC, Köppler P, Malviya AN. Inositol
annurev.bi.58.070189.004511.
1,4,5-trisphosphate receptor is located to the inner nuclear membrane vin- 61. Sorensen AB, Søndergaard MT, Overgaard MT. Calmodulin in a heart-
dicating regulation of nuclear calcium signaling by inositol 1,4,5-trispho- beat. FEBS J. 2013;280:5511–5532. doi: 10.1111/febs.12337.
sphate. Discrete distribution of inositol phosphate receptors to inner and 62. Maier LS, Bers DM. Calcium, calmodulin, and calcium-calmodu-

outer nuclear membranes. J Biol Chem. 1996;271:478–485. lin kinase II: heartbeat to heartbeat and beyond. J Mol Cell Cardiol.
40. Lui PP, Chan FL, Suen YK, Kwok TT, Kong SK. The nucleus of HeLa 2002;34:919–939.
cells contains tubular structures for Ca2+ signaling with the involvement 63. Finkler A, Ashery-Padan R, Fromm H. CAMTAs: calmodulin-binding
of mitochondria. Biochem Biophys Res Commun. 2003;308:826–833. transcription activators from plants to human. FEBS Lett. 2007;581:3893–
41. Jaconi M, Bony C, Richards SM, Terzic A, Arnaudeau S, Vassort G, Pucéat 3898. doi: 10.1016/j.febslet.2007.07.051.
M. Inositol 1,4,5-trisphosphate directs Ca(2+) flow between mitochondria 64. Long C, Grueter CE, Song K, Qin S, Qi X, Kong YM, Shelton JM,
and the endoplasmic/sarcoplasmic reticulum: a role in regulating cardiac Richardson JA, Zhang CL, Bassel-Duby R, Olson EN. Ataxia and
autonomic Ca(2+) spiking. Mol Biol Cell. 2000;11:1845–1858. Purkinje cell degeneration in mice lacking the CAMTA1 transcription
42. Lipp P, Thomas D, Berridge MJ, Bootman MD. Nuclear calcium signal- factor. Proc Natl Acad Sci USA. 2014;111:11521–11526. doi: 10.1073/
ling by individual cytoplasmic calcium puffs. EMBO J. 1997;16:7166– pnas.1411251111.
7173. doi: 10.1093/emboj/16.23.7166. 65. Song K, Backs J, McAnally J, Qi X, Gerard RD, Richardson JA, Hill
43. Shahin V, Danker T, Enss K, Ossig R, Oberleithner H. Evidence for Ca2+- JA, Bassel-Duby R, Olson EN. The transcriptional coactivator CAMTA2
and ATP-sensitive peripheral channels in nuclear pore complexes. FASEB stimulates cardiac growth by opposing class II histone deacetylases. Cell.
J. 2001;15:1895–1901. doi: 10.1096/fj.00-0838com. 2006;125:453–466. doi: 10.1016/j.cell.2006.02.048.
44. Martins TV, Evans MJ, Wysham DB, Morris RJ. Nuclear pores enable sus- 66. Song WH, Lin YH, Sun K, Zhang YH, Song Y, Hou LB, Zhang CN, Hui
tained perinuclear calcium oscillations. BMC Syst Biol. 2016;10:55. doi: RT, Chen JZ. A functional variant in the coding region of CAMTA2 is
10.1186/s12918-016-0289-9. associated with left ventricular hypertrophy by affecting the activation
45. Grossman E, Medalia O, Zwerger M. Functional architecture of the nu- of Nkx2.5-dependent transcription. J Hypertens. 2016;34:942–949. doi:
clear pore complex. Annu Rev Biophys. 2012;41:557–584. doi: 10.1146/ 10.1097/HJH.0000000000000873.
annurev-biophys-050511-102328. 67. Bers DM. Ca²⁺-calmodulin-dependent protein kinase II regulation of car-
46. Zima AV, Bare DJ, Mignery GA, Blatter LA. IP3-dependent nuclear Ca2+ diac excitation-transcription coupling. Heart Rhythm. 2011;8:1101–1104.
signalling in the mammalian heart. J Physiol. 2007;584:601–611. doi: doi: 10.1016/j.hrthm.2011.01.030.
10.1113/jphysiol.2007.140731. 68. Schulman H, Anderson ME. Ca/calmodulin-dependent protein kinase II
47. Luo D, Yang D, Lan X, Li K, Li X, Chen J, Zhang Y, Xiao RP, Han Q, in heart failure. Drug Discov Today Dis Mech. 2010;7:e117–e122. doi:
Cheng H. Nuclear Ca2+ sparks and waves mediated by inositol 1,4,5-tri- 10.1016/j.ddmec.2010.07.005.
sphosphate receptors in neonatal rat cardiomyocytes. Cell Calcium. 69. Meyer T, Hanson PI, Stryer L, Schulman H. Calmodulin trapping by calci-
2008;43:165–174. doi: 10.1016/j.ceca.2007.04.017. um-calmodulin-dependent protein kinase. Science. 1992;256:1199–1202.
Dewenter et al   Calcium Signaling and Transcription   1015

70. Lai Y, Nairn AC, Gorelick F, Greengard P. Ca2+/calmodulin-dependent 88. Parlakian A, Charvet C, Escoubet B, Mericskay M, Molkentin JD, Gary-
protein kinase II: identification of autophosphorylation sites responsible Bobo G, De Windt LJ, Ludosky MA, Paulin D, Daegelen D, Tuil D, Li Z.
for generation of Ca2+/calmodulin-independence. Proc Natl Acad Sci Temporally controlled onset of dilated cardiomyopathy through disrup-
USA. 1987;84:5710–5714. tion of the SRF gene in adult heart. Circulation. 2005;112:2930–2939.
71. Erickson JR, Joiner ML, Guan X, et al. A dynamic pathway for calci- doi: 10.1161/CIRCULATIONAHA.105.533778.
um-independent activation of CaMKII by methionine oxidation. Cell. 89. Werfel S, Jungmann A, Lehmann L, Ksienzyk J, Bekeredjian R, Kaya
2008;133:462–474. doi: 10.1016/j.cell.2008.02.048. Z, Leuchs B, Nordheim A, Backs J, Engelhardt S, Katus HA, Müller OJ.
72. Gutierrez DA, Fernandez-Tenorio M, Ogrodnik J, Niggli E. NO-dependent Rapid and highly efficient inducible cardiac gene knockout in adult mice
CaMKII activation during β-adrenergic stimulation of cardiac muscle. using AAV-mediated expression of Cre recombinase. Cardiovasc Res.
Cardiovasc Res. 2013;100:392–401. doi: 10.1093/cvr/cvt201. 2014;104:15–23. doi: 10.1093/cvr/cvu174.
73. Fentzke RC, Korcarz CE, Lang RM, Lin H, Leiden JM. Dilated cardio- 90. Flück M, Booth FW, Waxham MN. Skeletal muscle CaMKII enriches
myopathy in transgenic mice expressing a dominant-negative CREB in nuclei and phosphorylates myogenic factor SRF at multiple sites.
transcription factor in the heart. J Clin Invest. 1998;101:2415–2426. doi: Biochem Biophys Res Commun. 2000;270:488–494. doi: 10.1006/
10.1172/JCI2950. bbrc.2000.2457.
74. Altarejos JY, Montminy M. CREB and the CRTC co-activators: sensors 91. Xing W, Zhang TC, Cao D, Wang Z, Antos CL, Li S, Wang Y, Olson
for hormonal and metabolic signals. Nat Rev Mol Cell Biol. 2011;12:141– EN, Wang DZ. Myocardin induces cardiomyocyte hypertrophy. Circ Res.
151. doi: 10.1038/nrm3072. 2006;98:1089–1097. doi: 10.1161/01.RES.0000218781.23144.3e.
75. Wen AY, Sakamoto KM, Miller LS. The role of the transcription fac- 92. Li M, Wang N, Gong HQ, Li WZ, Liao XH, Yang XL, He HP, Cao DS,
tor CREB in immune function. J Immunol. 2010;185:6413–6419. doi: Zhang TC. Ca²⁺ signal-induced cardiomyocyte hypertrophy through
10.4049/jimmunol.1001829. activation of myocardin. Gene. 2015;557:43–51. doi: 10.1016/j.
76. Sun P, Enslen H, Myung PS, Maurer RA. Differential activation of CREB gene.2014.12.007.
by Ca2+/calmodulin-dependent protein kinases type II and type IV in- 93. Mani SK, Egan EA, Addy BK, Grimm M, Kasiganesan H, Thiyagarajan
volves phosphorylation of a site that negatively regulates activity. Genes T, Renaud L, Brown JH, Kern CB, Menick DR. beta-Adrenergic receptor
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

Dev. 1994;8:2527–2539. stimulated Ncx1 upregulation is mediated via a CaMKII/AP-1 signaling


77. Subedi KP, Son MJ, Chidipi B, Kim SW, Wang J, Kim KH, Woo SH, pathway in adult cardiomyocytes. J Mol Cell Cardiol. 2010;48:342–351.
Kim JC. Signaling pathway for endothelin-1- and phenylephrine-induced doi: 10.1016/j.yjmcc.2009.11.007.
cAMP response element binding protein activation in rat ventricular my- 94. Ronkainen JJ, Hänninen SL, Korhonen T, Koivumäki JT, Skoumal R,
ocytes: role of inositol 1,4,5-trisphosphate receptors and CaMKII. Cell Rautio S, Ronkainen VP, Tavi P. Ca2+-calmodulin-dependent protein
Physiol Biochem. 2017;41:399–412. doi: 10.1159/000456422. kinase II represses cardiac transcription of the L-type calcium channel
78. Shimomura A, Ogawa Y, Kitani T, Fujisawa H, Hagiwara M. Calmodulin- alpha(1C)-subunit gene (Cacna1c) by DREAM translocation. J Physiol.
dependent protein kinase II potentiates transcriptional activation through 2011;589:2669–2686. doi: 10.1113/jphysiol.2010.201400.
activating transcription factor 1 but not cAMP response element-binding
95. Skene PJ, Illingworth RS, Webb S, Kerr AR, James KD, Turner DJ,
protein. J Biol Chem. 1996;271:17957–17960.
Andrews R, Bird AP. Neuronal MeCP2 is expressed at near histone-
79. Li B, Dedman JR, Kaetzel MA. Nuclear Ca2+/calmodulin-dependent pro-
octamer levels and globally alters the chromatin state. Mol Cell.
tein kinase II in the murine heart. Biochim Biophys Acta. 2006;1763:1275–
2010;37:457–468. doi: 10.1016/j.molcel.2010.01.030.
1281. doi: 10.1016/j.bbamcr.2006.09.029.
96. Buchthal B, Lau D, Weiss U, Weislogel JM, Bading H. Nuclear cal-
80. Zhu WZ, Wang SQ, Chakir K, Yang D, Zhang T, Brown JH, Devic E,
cium signaling controls methyl-CpG-binding protein 2 (MeCP2) phos-
Kobilka BK, Cheng H, Xiao RP. Linkage of beta1-adrenergic stimulation
phorylation on serine 421 following synaptic activity. J Biol Chem.
to apoptotic heart cell death through protein kinase A-independent activa-
2012;287:30967–30974. doi: 10.1074/jbc.M112.382507.
tion of Ca2+/calmodulin kinase II. J Clin Invest. 2003;111:617–625. doi:
97. Mayer SC, Gilsbach R, Preissl S, et al. Adrenergic repression of the epigen-
10.1172/JCI16326.
etic reader MeCP2 facilitates cardiac adaptation in chronic heart failure.
81. Zhu W, Woo AY, Yang D, Cheng H, Crow MT, Xiao RP. Activation of
Circ Res. 2015;117:622–633. doi: 10.1161/CIRCRESAHA.115.306721.
CaMKIIdeltaC is a common intermediate of diverse death stimuli-induced
98. Awad S, Kunhi M, Little GH, Bai Y, An W, Bers D, Kedes L, Poizat
heart muscle cell apoptosis. J Biol Chem. 2007;282:10833–10839. doi:
C. Nuclear CaMKII enhances histone H3 phosphorylation and remodels
10.1074/jbc.M611507200.
82. Peng W, Zhang Y, Zheng M, Cheng H, Zhu W, Cao CM, Xiao RP. chromatin during cardiac hypertrophy. Nucleic Acids Res. 2013;41:7656–
Cardioprotection by CaMKII-deltaB is mediated by phosphoryla- 7672. doi: 10.1093/nar/gkt500.
tion of heat shock factor 1 and subsequent expression of inducible 99. Awad S, Al-Haffar KM, Marashly Q, Quijada P, Kunhi M, Al-Yacoub N,
heat shock protein 70. Circ Res. 2010;106:102–110. doi: 10.1161/ Wade FS, Mohammed SF, Al-Dayel F, Sutherland G, Assiri A, Sussman
CIRCRESAHA.109.210914. M, Bers D, Al-Habeeb W, Poizat C. Control of histone H3 phosphoryla-
83. Little GH, Saw A, Bai Y, Dow J, Marjoram P, Simkhovich B, Leeka J, tion by CaMKIIδ in response to haemodynamic cardiac stress. J Pathol.
Kedes L, Kloner RA, Poizat C. Critical role of nuclear calcium/calmod- 2015;235:606–618. doi: 10.1002/path.4489.
ulin-dependent protein kinase IIdeltaB in cardiomyocyte survival in car- 100. Lehmann LH, Worst BC, Stanmore DA, Backs J. Histone deacetylase
diomyopathy. J Biol Chem. 2009;284:24857–24868. doi: 10.1074/jbc. signaling in cardioprotection. Cell Mol Life Sci. 2014;71:1673–1690.
M109.003186. doi: 10.1007/s00018-013-1516-9.
84. Kashiwase K, Higuchi Y, Hirotani S, Yamaguchi O, Hikoso S, Takeda T, 101. Kim Y, Phan D, van Rooij E, Wang DZ, McAnally J, Qi X, Richardson
Watanabe T, Taniike M, Nakai A, Tsujimoto I, Matsumura Y, Ueno H, JA, Hill JA, Bassel-Duby R, Olson EN. The MEF2D transcription fac-
Nishida K, Hori M, Otsu K. CaMKII activates ASK1 and NF-kappaB tor mediates stress-dependent cardiac remodeling in mice. J Clin Invest.
to induce cardiomyocyte hypertrophy. Biochem Biophys Res Commun. 2008;118:124–132. doi: 10.1172/JCI33255.
2005;327:136–142. doi: 10.1016/j.bbrc.2004.12.002. 102. Clark RI, Tan SW, Péan CB, Roostalu U, Vivancos V, Bronda K, Pilátová
85. Ling H, Gray CB, Zambon AC, Grimm M, Gu Y, Dalton N, Purcell M, Fu J, Walker DW, Berdeaux R, Geissmann F, Dionne MS. MEF2
NH, Peterson K, Brown JH. Ca2+/Calmodulin-dependent protein ki- is an in vivo immune-metabolic switch. Cell. 2013;155:435–447. doi:
nase II δ mediates myocardial ischemia/reperfusion injury through 10.1016/j.cell.2013.09.007.
nuclear factor-κB. Circ Res. 2013;112:935–944. doi: 10.1161/ 103. Backs J, Song K, Bezprozvannaya S, Chang S, Olson EN. CaM kinase II
CIRCRESAHA.112.276915. selectively signals to histone deacetylase 4 during cardiomyocyte hyper-
86. Gray CB, Suetomi T, Xiang S, Mishra S, Blackwood EA, Glembotski CC, trophy. J Clin Invest. 2006;116:1853–1864. doi: 10.1172/JCI27438.
Miyamoto S, Westenbrink BD, Brown JH. CaMKIIδ subtypes differen- 104. Little GH, Bai Y, Williams T, Poizat C. Nuclear calcium/calmodulin-de-
tially regulate infarct formation following ex vivo myocardial ischemia/ pendent protein kinase IIdelta preferentially transmits signals to histone
reperfusion through NF-κB and TNF-α. J Mol Cell Cardiol. 2017;103:48– deacetylase 4 in cardiac cells. J Biol Chem. 2007;282:7219–7231. doi:
55. doi: 10.1016/j.yjmcc.2017.01.002. 10.1074/jbc.M604281200.
87. Weinreuter M, Kreusser MM, Beckendorf J, et al. CaM kinase II me- 105. Zhang T, Kohlhaas M, Backs J, Mishra S, Phillips W, Dybkova N, Chang
diates maladaptive post-infarct remodeling and pro-inflammatory S, Ling H, Bers DM, Maier LS, Olson EN, Brown JH. CaMKIIdelta
chemoattractant signaling but not acute myocardial ischemia/reper- isoforms differentially affect calcium handling but similarly regulate
fusion injury. EMBO Mol Med. 2014;6:1231–1245. doi: 10.15252/ HDAC/MEF2 transcriptional responses. J Biol Chem. 2007;282:35078–
emmm.201403848. 35087. doi: 10.1074/jbc.M707083200.
1016  Circulation Research  September 29, 2017

106. Davis FJ, Gupta M, Camoretti-Mercado B, Schwartz RJ, Gupta MP. 126. El-Armouche A, Bednorz A, Pamminger T, Ditz D, Didié M, Dobrev D,
Calcium/calmodulin-dependent protein kinase activates serum response Eschenhagen T. Role of calcineurin and protein phosphatase-2A in the reg-
factor transcription activity by its dissociation from histone deacety- ulation of phosphatase inhibitor-1 in cardiac myocytes. Biochem Biophys
lase, HDAC4. Implications in cardiac muscle gene regulation during Res Commun. 2006;346:700–706. doi: 10.1016/j.bbrc.2006.05.182.
hypertrophy. J Biol Chem. 2003;278:20047–20058. doi: 10.1074/jbc. 127. Bradshaw JM, Kubota Y, Meyer T, Schulman H. An ultrasensitive Ca2+/
M209998200. calmodulin-dependent protein kinase II-protein phosphatase 1 switch fa-
107. Backs J, Worst BC, Lehmann LH, Patrick DM, Jebessa Z, Kreusser MM, cilitates specificity in postsynaptic calcium signaling. Proc Natl Acad Sci
Sun Q, Chen L, Heft C, Katus HA, Olson EN. Selective repression of USA. 2003;100:10512–10517. doi: 10.1073/pnas.1932759100.
MEF2 activity by PKA-dependent proteolysis of HDAC4. J Cell Biol. 128. Wang K, Long B, Zhou J, Li PF. miR-9 and NFATc3 regulate myocar-
2011;195:403–415. doi: 10.1083/jcb.201105063. din in cardiac hypertrophy. J Biol Chem. 2010;285:11903–11912. doi:
108. Backs J, Backs T, Bezprozvannaya S, McKinsey TA, Olson EN. Histone 10.1074/jbc.M109.098004.
deacetylase 5 acquires calcium/calmodulin-dependent kinase II respon- 129. Li M, He HP, Gong HQ, Zhang J, Ma WJ, Zhou H, Cao DS, Wang N,
siveness by oligomerization with histone deacetylase 4. Mol Cell Biol. Zhang TC. NFATc4 and myocardin synergistically up-regulate the ex-
2008;28:3437–3445. doi: 10.1128/MCB.01611-07. pression of LTCC α1C in ET-1-induced cardiomyocyte hypertrophy. Life
109. Zhang CL, McKinsey TA, Olson EN. Association of class II histone Sci. 2016;155:11–20. doi: 10.1016/j.lfs.2016.05.007.
deacetylases with heterochromatin protein 1: potential role for his- 130. Ceribelli M, Dolfini D, Merico D, Gatta R, Viganò AM, Pavesi G,
tone methylation in control of muscle differentiation. Mol Cell Biol. Mantovani R. The histone-like NF-Y is a bifunctional transcription fac-
2002;22:7302–7312. tor. Mol Cell Biol. 2008;28:2047–2058. doi: 10.1128/MCB.01861-07.
110. Hohl M, Wagner M, Reil JC, Müller SA, Tauchnitz M, Zimmer AM, 131. Cha-Molstad H, Xu G, Chen J, Jing G, Young ME, Chatham JC, Shalev
Lehmann LH, Thiel G, Böhm M, Backs J, Maack C. HDAC4 controls A. Calcium channel blockers act through nuclear factor Y to control tran-
histone methylation in response to elevated cardiac load. J Clin Invest. scription of key cardiac genes. Mol Pharmacol. 2012;82:541–549. doi:
2013;123:1359–1370. doi: 10.1172/JCI61084. 10.1124/mol.112.078253.
111. Rusnak F, Mertz P. Calcineurin: form and function. Physiol Rev.
132. Yoshioka J, Chutkow WA, Lee S, Kim JB, Yan J, Tian R, Lindsey ML,
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

2000;80:1483–1521. Feener EP, Seidman CE, Seidman JG, Lee RT. Deletion of thioredox-
112. Molkentin JD. Calcineurin and beyond: cardiac hypertrophic signaling. in-interacting protein in mice impairs mitochondrial function but pro-
Circ Res. 2000;87:731–738. tects the myocardium from ischemia-reperfusion injury. J Clin Invest.
113. Molkentin JD, Lu JR, Antos CL, Markham B, Richardson J, Robbins J, 2012;122:267–279. doi: 10.1172/JCI44927.
Grant SR, Olson EN. A calcineurin-dependent transcriptional pathway 133. Yoshioka J, Imahashi K, Gabel SA, Chutkow WA, Burds AA, Gannon J,
for cardiac hypertrophy. Cell. 1998;93:215–228. Schulze PC, MacGillivray C, London RE, Murphy E, Lee RT. Targeted
114. Hallhuber M, Burkard N, Wu R, Buch MH, Engelhardt S, Hein L, Neyses deletion of thioredoxin-interacting protein regulates cardiac dysfunction
L, Schuh K, Ritter O. Inhibition of nuclear import of calcineurin prevents in response to pressure overload. Circ Res. 2007;101:1328–1338. doi:
myocardial hypertrophy. Circ Res. 2006;99:626–635. doi: 10.1161/01. 10.1161/CIRCRESAHA.106.160515.
RES.0000243208.59795.d8. 134. Matsumura Y, Saeki E, Otsu K, Morita T, Takeda H, Kuzuya T, Hori M,
115. Zhu J, McKeon F. NF-AT activation requires suppression of Crm1- Kusuoka H. Intracellular calcium level required for calpain activation in
dependent export by calcineurin. Nature. 1999;398:256–260. doi: a single myocardial cell. J Mol Cell Cardiol. 2001;33:1133–1142. doi:
10.1038/18473. 10.1006/jmcc.2001.1373.
116. Wu H, Naya FJ, McKinsey TA, Mercer B, Shelton JM, Chin ER, Simard 135. Neuhof C, Neuhof H. Calpain system and its involvement in myocardial
AR, Michel RN, Bassel-Duby R, Olson EN, Williams RS. MEF2 re- ischemia and reperfusion injury. World J Cardiol. 2014;6:638–652. doi:
sponds to multiple calcium-regulated signals in the control of skel- 10.4330/wjc.v6.i7.638.
etal muscle fiber type. EMBO J. 2000;19:1963–1973. doi: 10.1093/ 136. Zatz M, Starling A. Calpains and disease. N Engl J Med. 2005;352:2413–
emboj/19.9.1963. 2423. doi: 10.1056/NEJMra043361.
117. Mao Z, Wiedmann M. Calcineurin enhances MEF2 DNA binding activ- 137. Kang MY, Zhang Y, Matkovich SJ, Diwan A, Chishti AH, Dorn

ity in calcium-dependent survival of cerebellar granule neurons. J Biol GW II. Receptor-independent cardiac protein kinase Calpha activa-
Chem. 1999;274:31102–31107. tion by calpain-mediated truncation of regulatory domains. Circ Res.
118. Blaeser F, Ho N, Prywes R, Chatila TA. Ca(2+)-dependent gene ex- 2010;107:903–912. doi: 10.1161/CIRCRESAHA.110.220772.
pression mediated by MEF2 transcription factors. J Biol Chem. 138. Rohde S, Sabri A, Kamasamudran R, Steinberg SF. The alpha(1)-

2000;275:197–209. adrenoceptor subtype- and protein kinase C isoform-dependence
119. Grégoire S, Tremblay AM, Xiao L, Yang Q, Ma K, Nie J, Mao Z, Wu Z, of norepinephrine’s actions in cardiomyocytes. J Mol Cell Cardiol.
Giguère V, Yang XJ. Control of MEF2 transcriptional activity by coor- 2000;32:1193–1209. doi: 10.1006/jmcc.2000.1153.
dinated phosphorylation and sumoylation. J Biol Chem. 2006;281:4423– 139. Zhang Y, Matkovich SJ, Duan X, Diwan A, Kang MY, Dorn GW II.
4433. doi: 10.1074/jbc.M509471200. Receptor-independent protein kinase C alpha (PKCalpha) signaling by
120. Li J, Vargas MA, Kapiloff MS, Dodge-Kafka KL. Regulation of MEF2 calpain-generated free catalytic domains induces HDAC5 nuclear ex-
transcriptional activity by calcineurin/mAKAP complexes. Exp Cell Res. port and regulates cardiac transcription. J Biol Chem. 2011;286:26943–
2013;319:447–454. doi: 10.1016/j.yexcr.2012.12.016. 26951. doi: 10.1074/jbc.M111.234757.
121. van Oort RJ, van Rooij E, Bourajjaj M, Schimmel J, Jansen MA, van 140. Burkard N, Becher J, Heindl C, Neyses L, Schuh K, Ritter O. Targeted
der Nagel R, Doevendans PA, Schneider MD, van Echteld CJ, De Windt proteolysis sustains calcineurin activation. Circulation. 2005;111:1045–
LJ. MEF2 activates a genetic program promoting chamber dilation and 1053. doi: 10.1161/01.CIR.0000156458.80515.F7.
contractile dysfunction in calcineurin-induced heart failure. Circulation. 141. Wang JC, Zhao Y, Li XD, Zhou NN, Sun H, Sun YY. Proteolysis by en-
2006;114:298–308. doi: 10.1161/CIRCULATIONAHA.105.608968. dogenous calpain I leads to the activation of calcineurin in human heart.
122. Dai YS, Xu J, Molkentin JD. The DnaJ-related factor Mrj interacts with Clin Lab. 2012;58:1145–1152.
nuclear factor of activated T cells c3 and mediates transcriptional repres- 142. Gomez-Ospina N, Tsuruta F, Barreto-Chang O, Hu L, Dolmetsch R.
sion through class II histone deacetylase recruitment. Mol Cell Biol. The C terminus of the L-type voltage-gated calcium channel Ca(V)1.2
2005;25:9936–9948. doi: 10.1128/MCB.25.22.9936-9948.2005. encodes a transcription factor. Cell. 2006;127:591–606. doi: 10.1016/j.
123. MacDonnell SM, Weisser-Thomas J, Kubo H, Hanscome M, Liu Q, cell.2006.10.017.
Jaleel N, Berretta R, Chen X, Brown JH, Sabri AK, Molkentin JD, 143. Satin J, Schroder EA, Crump SM. L-type calcium channel auto-regu-
Houser SR. CaMKII negatively regulates calcineurin-NFAT signal- lation of transcription. Cell Calcium. 2011;49:306–313. doi: 10.1016/j.
ing in cardiac myocytes. Circ Res. 2009;105:316–325. doi: 10.1161/ ceca.2011.01.001.
CIRCRESAHA.109.194035. 144. Braunlin WH, Drakenberg T, Nordenskiöld L. Ca2+ binding environ-
124. Kreusser MM, Lehmann LH, Keranov S, et al. Cardiac CaM Kinase ments on natural and synthetic polymeric DNA’s. J Biomol Struct Dyn.
II genes δ and γ contribute to adverse remodeling but redundantly 1992;10:333–343. doi: 10.1080/07391102.1992.10508651.
inhibit calcineurin-induced myocardial hypertrophy. Circulation. 145. Lyon AR, MacLeod KT, Zhang Y, Garcia E, Kanda GK, Lab MJ,

2014;130:1262–1273. doi: 10.1161/CIRCULATIONAHA.114.006185. Korchev YE, Harding SE, Gorelik J. Loss of T-tubules and other changes
125. Kubokawa M, Nakamura K, Komagiri Y. Interaction between Calcineurin to surface topography in ventricular myocytes from failing human and
and Ca/Calmodulin kinase-II in modulating cellular functions. Enzyme rat heart. Proc Natl Acad Sci USA. 2009;106:6854–6859. doi: 10.1073/
Res. 2011;2011:587359. doi: 10.4061/2011/587359. pnas.0809777106.
Dewenter et al   Calcium Signaling and Transcription   1017

146. Yan SM, Finato N, Di Loreto C, Beltrami CA. Nuclear size of myo- 164. Grimm M, Ling H, Willeford A, Pereira L, Gray CB, Erickson JR, Sarma
cardial cells in end-stage cardiomyopathies. Anal Quant Cytol Histol. S, Respress JL, Wehrens XH, Bers DM, Brown JH. CaMKIIδ mediates
1999;21:174–180. β-adrenergic effects on RyR2 phosphorylation and SR Ca(2+) leak and
147. Cortés R, Roselló-Lletí E, Rivera M, Martínez-Dolz L, Salvador A, the pathophysiological response to chronic β-adrenergic stimulation. J
Azorín I, Portolés M. Influence of heart failure on nucleocytoplasmic Mol Cell Cardiol. 2015;85:282–291. doi: 10.1016/j.yjmcc.2015.06.007.
transport in human cardiomyocytes. Cardiovasc Res. 2010;85:464–472. 165. Kreusser MM, Lehmann LH, Wolf N, Keranov S, Jungmann A, Gröne HJ,
doi: 10.1093/cvr/cvp336. Müller OJ, Katus HA, Backs J. Inducible cardiomyocyte-specific dele-
148. Chahine MN, Mioulane M, Sikkel MB, O’Gara P, Dos Remedios CG, tion of CaM kinase II protects from pressure overload-induced heart fail-
Pierce GN, Lyon AR, Földes G, Harding SE. Nuclear pore rearrange- ure. Basic Res Cardiol. 2016;111:65. doi: 10.1007/s00395-016-0581-2.
ments and nuclear trafficking in cardiomyocytes from rat and human fail- 166. Zhang M, Hagenmueller M, Riffel JH, Kreusser MM, Bernhold E, Fan
ing hearts. Cardiovasc Res. 2015;105:31–43. doi: 10.1093/cvr/cvu218. J, Katus HA, Backs J, Hardt SE. Calcium/calmodulin-dependent protein
149. Bers DM. Altered cardiac myocyte Ca regulation in heart failure.
kinase II couples Wnt signaling with histone deacetylase 4 and mediates
Physiology. 2006;21:380–387. doi: 10.1152/physiol.00019.2006. dishevelled-induced cardiomyopathy. Hypertension. 2015;65:335–344.
150. Lehnart SE, Maier LS, Hasenfuss G. Abnormalities of calcium metabo- doi: 10.1161/HYPERTENSIONAHA.114.04467.
lism and myocardial contractility depression in the failing heart. Heart 167. Zhang R, Khoo MS, Wu Y, et al. Calmodulin kinase II inhibition pro-
Fail Rev. 2009;14:213–224. doi: 10.1007/s10741-009-9146-x. tects against structural heart disease. Nat Med. 2005;11:409–417. doi:
151. Studer R, Reinecke H, Bilger J, Eschenhagen T, Böhm M, Hasenfuss G, 10.1038/nm1215.
Just H, Holtz J, Drexler H. Gene expression of the cardiac Na(+)-Ca2+ 168. Singh MV, Kapoun A, Higgins L, et al. Ca2+/calmodulin-dependent ki-
exchanger in end-stage human heart failure. Circ Res. 1994;75:443–453. nase II triggers cell membrane injury by inducing complement factor B
152. Kuwahara K, Wang Y, McAnally J, Richardson JA, Bassel-Duby R, gene expression in the mouse heart. J Clin Invest. 2009;119:986–996.
Hill JA, Olson EN. TRPC6 fulfills a calcineurin signaling circuit during doi: 10.1172/JCI35814.
pathologic cardiac remodeling. J Clin Invest. 2006;116:3114–3126. doi: 169. Khoo MS, Li J, Singh MV, et al. Death, cardiac dysfunction, and ar-
10.1172/JCI27702. rhythmias are increased by calmodulin kinase II in calcineurin car-
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

153. Bush EW, Hood DB, Papst PJ, Chapo JA, Minobe W, Bristow MR, diomyopathy. Circulation. 2006;114:1352–1359. doi: 10.1161/
Olson EN, McKinsey TA. Canonical transient receptor potential chan- CIRCULATIONAHA.106.644583.
nels promote cardiomyocyte hypertrophy through activation of calcineu- 170. Chang S, McKinsey TA, Zhang CL, Richardson JA, Hill JA, Olson
rin signaling. J Biol Chem. 2006;281:33487–33496. doi: 10.1074/jbc. EN. Histone deacetylases 5 and 9 govern responsiveness of the
M605536200. heart to a subset of stress signals and play redundant roles in heart
154. Ai X, Curran JW, Shannon TR, Bers DM, Pogwizd SM. Ca2+/calmodu- development. Mol Cell Biol. 2004;24:8467–8476. doi: 10.1128/
lin-dependent protein kinase modulates cardiac ryanodine receptor phos- MCB.24.19.8467-8476.2004.
phorylation and sarcoplasmic reticulum Ca2+ leak in heart failure. Circ 171. Zhang CL, McKinsey TA, Chang S, Antos CL, Hill JA, Olson EN. Class
Res. 2005;97:1314–1322. doi: 10.1161/01.RES.0000194329.41863.89. II histone deacetylases act as signal-responsive repressors of cardiac hy-
155. Go LO, Moschella MC, Watras J, Handa KK, Fyfe BS, Marks AR. pertrophy. Cell. 2002;110:479–488.
Differential regulation of two types of intracellular calcium release chan- 172. Xu J, Gong NL, Bodi I, Aronow BJ, Backx PH, Molkentin JD. Myocyte
nels during end-stage heart failure. J Clin Invest. 1995;95:888–894. doi: enhancer factors 2A and 2C induce dilated cardiomyopathy in transgenic
10.1172/JCI117739. mice. J Biol Chem. 2006;281:9152–9162. doi: 10.1074/jbc.M510217200.
156. Zhang T, Maier LS, Dalton ND, Miyamoto S, Ross J Jr, Bers DM, 173. Wilkins BJ, De Windt LJ, Bueno OF, Braz JC, Glascock BJ, Kimball TF,
Brown JH. The deltaC isoform of CaMKII is activated in cardiac hyper- Molkentin JD. Targeted disruption of NFATc3, but not NFATc4, reveals
trophy and induces dilated cardiomyopathy and heart failure. Circ Res. an intrinsic defect in calcineurin-mediated cardiac hypertrophic growth.
2003;92:912–919. doi: 10.1161/01.RES.0000069686.31472.C5. Mol Cell Biol. 2002;22:7603–7613.
157. Zhang T, Guo T, Mishra S, Dalton ND, Kranias EG, Peterson KL, 174. Rothermel BA, McKinsey TA, Vega RB, Nicol RL, Mammen P, Yang
Bers DM, Brown JH. Phospholamban ablation rescues sarcoplas- J, Antos CL, Shelton JM, Bassel-Duby R, Olson EN, Williams RS.
mic reticulum Ca(2+) handling but exacerbates cardiac dysfunction Myocyte-enriched calcineurin-interacting protein, MCIP1, inhibits car-
in CaMKIIdelta© transgenic mice. Circ Res. 2010;106:354–362. doi: diac hypertrophy in vivo. Proc Natl Acad Sci USA. 2001;98:3328–3333.
10.1161/CIRCRESAHA.109.207423. doi: 10.1073/pnas.041614798.
158. Huke S, Desantiago J, Kaetzel MA, Mishra S, Brown JH, Dedman JR, 175. Heineke J, Wollert KC, Osinska H, Sargent MA, York AJ, Robbins J,
Bers DM. SR-targeted CaMKII inhibition improves SR Ca²+ handling, Molkentin JD. Calcineurin protects the heart in a murine model of di-
but accelerates cardiac remodeling in mice overexpressing CaMKIIδC. J lated cardiomyopathy. J Mol Cell Cardiol. 2010;48:1080–1087. doi:
Mol Cell Cardiol. 2011;50:230–238. doi: 10.1016/j.yjmcc.2010.10.014. 10.1016/j.yjmcc.2009.10.012.
159. Dewenter M, Neef S, Vettel C, et al. Calcium/calmodulin-dependent pro- 176. Antos CL, McKinsey TA, Frey N, Kutschke W, McAnally J, Shelton
tein kinase II activity persists during chronic β-adrenoceptor blockade in JM, Richardson JA, Hill JA, Olson EN. Activated glycogen synthase-3
experimental and human heart failure. Circ Heart Fail. 2017;10:e003840. beta suppresses cardiac hypertrophy in vivo. Proc Natl Acad Sci USA.
doi: 10.1161/CIRCHEARTFAILURE.117.003840. 2002;99:907–912. doi: 10.1073/pnas.231619298.
160. Dybkova N, Sedej S, Napolitano C, Neef S, Rokita AG, Hünlich
177. Lim HW, De Windt LJ, Mante J, Kimball TR, Witt SA, Sussman MA,
M, Brown JH, Kockskämper J, Priori SG, Pieske B, Maier LS. Molkentin JD. Reversal of cardiac hypertrophy in transgenic disease
Overexpression of CaMKIIδc in RyR2R4496C+/- knock-in mice leads models by calcineurin inhibition. J Mol Cell Cardiol. 2000;32:697–709.
to altered intracellular Ca2+ handling and increased mortality. J Am Coll doi: 10.1006/jmcc.2000.1113.
Cardiol. 2011;57:469–479. doi: 10.1016/j.jacc.2010.08.639. 178. Berry JM, Le V, Rotter D, Battiprolu PK, Grinsfelder B, Tannous P,
161. Zhang T, Johnson EN, Gu Y, Morissette MR, Sah VP, Gigena MS, Belke Burchfield JS, Czubryt M, Backs J, Olson EN, Rothermel BA, Hill JA.
DD, Dillmann WH, Rogers TB, Schulman H, Ross J Jr, Brown JH. The Reversibility of adverse, calcineurin-dependent cardiac remodeling. Circ
cardiac-specific nuclear delta(B) isoform of Ca2+/calmodulin-dependent Res. 2011;109:407–417. doi: 10.1161/CIRCRESAHA.110.228452.
protein kinase II induces hypertrophy and dilated cardiomyopathy as- 179. Zhao G, Jeoung NH, Burgess SC, Rosaaen-Stowe KA, Inagaki T, Latif
sociated with increased protein phosphatase 2A activity. J Biol Chem. S, Shelton JM, McAnally J, Bassel-Duby R, Harris RA, Richardson JA,
2002;277:1261–1267. doi: 10.1074/jbc.M108525200. Kliewer SA. Overexpression of pyruvate dehydrogenase kinase 4 in
162. Ling H, Zhang T, Pereira L, Means CK, Cheng H, Gu Y, Dalton ND, heart perturbs metabolism and exacerbates calcineurin-induced cardio-
Peterson KL, Chen J, Bers D, Brown JH, Heller Brown J. Requirement myopathy. Am J Physiol Heart Circ Physiol. 2008;294:H936–H943. doi:
for Ca2+/calmodulin-dependent kinase II in the transition from pressure 10.1152/ajpheart.00870.2007.
overload-induced cardiac hypertrophy to heart failure in mice. J Clin 180. Vega RB, Rothermel BA, Weinheimer CJ, Kovacs A, Naseem RH,

Invest. 2009;119:1230–1240. doi: 10.1172/JCI38022. Bassel-Duby R, Williams RS, Olson EN. Dual roles of modulatory cal-
163. Backs J, Backs T, Neef S, Kreusser MM, Lehmann LH, Patrick DM, cineurin-interacting protein 1 in cardiac hypertrophy. Proc Natl Acad Sci
Grueter CE, Qi X, Richardson JA, Hill JA, Katus HA, Bassel-Duby R, USA. 2003;100:669–674. doi: 10.1073/pnas.0237225100.
Maier LS, Olson EN. The delta isoform of CaM kinase II is required 181. Bueno OF, Wilkins BJ, Tymitz KM, Glascock BJ, Kimball TF, Lorenz
for pathological cardiac hypertrophy and remodeling after pressure JN, Molkentin JD. Impaired cardiac hypertrophic response in Calcineurin
overload. Proc Natl Acad Sci USA. 2009;106:2342–2347. doi: 10.1073/ Abeta -deficient mice. Proc Natl Acad Sci USA. 2002;99:4586–4591.
pnas.0813013106. doi: 10.1073/pnas.072647999.
1018  Circulation Research  September 29, 2017

182. Zou Y, Hiroi Y, Uozumi H, Takimoto E, Toko H, Zhu W, Kudoh S, mice with chronic pressure overload. Am J Physiol Heart Circ Physiol.
Mizukami M, Shimoyama M, Shibasaki F, Nagai R, Yazaki Y, Komuro I. 2009;297:H1814–H1819. doi: 10.1152/ajpheart.00449.2009.
Calcineurin plays a critical role in the development of pressure overload- 202. Kirchhefer U, Schmitz W, Scholz H, Neumann J. Activity of cAMP-de-
induced cardiac hypertrophy. Circulation. 2001;104:97–101. pendent protein kinase and Ca2+/calmodulin-dependent protein kinase in
183. Müller JG, Nemoto S, Laser M, Carabello BA, Menick DR. Calcineurin failing and nonfailing human hearts. Cardiovasc Res. 1999;42:254–261.
inhibition and cardiac hypertrophy. Science. 1998;282:1007. 203. Palomeque J, Rueda OV, Sapia L, Valverde CA, Salas M, Petroff MV,
184. Luo Z, Shyu KG, Gualberto A, Walsh K. Calcineurin inhibitors and car- Mattiazzi A. Angiotensin II-induced oxidative stress resets the Ca2+ de-
diac hypertrophy. Nat Med. 1998;4:1092–1093. doi: 10.1038/2578. pendence of Ca2+-calmodulin protein kinase II and promotes a death
185. Zhang W, Kowal RC, Rusnak F, Sikkink RA, Olson EN, Victor RG. pathway conserved across different species. Circ Res. 2009;105:1204–
Failure of calcineurin inhibitors to prevent pressure-overload left ven- 1212. doi: 10.1161/CIRCRESAHA.109.204172.
tricular hypertrophy in rats. Circ Res. 1999;84:722–728. 204. Zhu W, Zou Y, Shiojima I, Kudoh S, Aikawa R, Hayashi D, Mizukami
186. Ding B, Price RL, Borg TK, Weinberg EO, Halloran PF, Lorell BH. M, Toko H, Shibasaki F, Yazaki Y, Nagai R, Komuro I. Ca2+/calmodulin-
Pressure overload induces severe hypertrophy in mice treated with cyclo- dependent kinase II and calcineurin play critical roles in endothelin-1-in-
sporine, an inhibitor of calcineurin. Circ Res. 1999;84:729–734. duced cardiomyocyte hypertrophy. J Biol Chem. 2000;275:15239–15245.
187. Hill JA, Karimi M, Kutschke W, Davisson RL, Zimmerman K, Wang 205. O-Uchi J, Komukai K, Kusakari Y, Obata T, Hongo K, Sasaki H, Kurihara
Z, Kerber RE, Weiss RM. Cardiac hypertrophy is not a required com- S. alpha1-adrenoceptor stimulation potentiates L-type Ca2+ current
pensatory response to short-term pressure overload. Circulation. through Ca2+/calmodulin-dependent PK II (CaMKII) activation in rat
2000;101:2863–2869. ventricular myocytes. Proc Natl Acad Sci USA. 2005;102:9400–9405.
188. Yang G, Meguro T, Hong C, Asai K, Takagi G, Karoor VL, Sadoshima J, doi: 10.1073/pnas.0503569102.
Vatner DE, Bishop SP, Vatner SF. Cyclosporine reduces left ventricular 206. Mangmool S, Shukla AK, Rockman HA. beta-Arrestin-dependent acti-
mass with chronic aortic banding in mice, which could be due to apopto- vation of Ca(2+)/calmodulin kinase II after beta(1)-adrenergic receptor
sis and fibrosis. J Mol Cell Cardiol. 2001;33:1505–1514. doi: 10.1006/ stimulation. J Cell Biol. 2010;189:573–587. doi: 10.1083/jcb.200911047.
jmcc.2001.1413. 207. Curran J, Hinton MJ, Ríos E, Bers DM, Shannon TR. Beta-adrenergic
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

189. Lim HW, De Windt LJ, Steinberg L, Taigen T, Witt SA, Kimball TR, enhancement of sarcoplasmic reticulum calcium leak in cardiac myo-
Molkentin JD. Calcineurin expression, activation, and function in cardiac cytes is mediated by calcium/calmodulin-dependent protein kinase. Circ
pressure-overload hypertrophy. Circulation. 2000;101:2431–2437. Res. 2007;100:391–398. doi: 10.1161/01.RES.0000258172.74570.e6.
190. Meguro T, Hong C, Asai K, Takagi G, McKinsey TA, Olson EN, Vatner 208. Wang W, Zhu W, Wang S, Yang D, Crow MT, Xiao RP, Cheng H.
SF. Cyclosporine attenuates pressure-overload hypertrophy in mice Sustained beta1-adrenergic stimulation modulates cardiac contractility
while enhancing susceptibility to decompensation and heart failure. Circ by Ca2+/calmodulin kinase signaling pathway. Circ Res. 2004;95:798–
Res. 1999;84:735–740. 806. doi: 10.1161/01.RES.0000145361.50017.aa.
209. Said M, Mundiña-Weilenmann C, Vittone L, Mattiazzi A. The relative
191. Mackiewicz U, Maczewski M, Klemenska E, Brudek M, Konior A,
relevance of phosphorylation of the Thr(17) residue of phospholamban is
Czarnowska E, Lewartowski B. Brief postinfarction calcineurin blockade
different at different levels of beta-adrenergic stimulation. Pflugers Arch.
affects left ventricular remodeling and Ca2+ handling in the rat. J Mol
2002;444:801–809. doi: 10.1007/s00424-002-0885-y.
Cell Cardiol. 2010;48:1307–1315. doi: 10.1016/j.yjmcc.2009.12.016.
210. Ramirez MT, Zhao XL, Schulman H, Brown JH. The nuclear deltaB
192. Oie E, Bjørnerheim R, Clausen OP, Attramadal H. Cyclosporin A

isoform of Ca2+/calmodulin-dependent protein kinase II regulates atrial
inhibits cardiac hypertrophy and enhances cardiac dysfunction dur-
natriuretic factor gene expression in ventricular myocytes. J Biol Chem.
ing postinfarction failure in rats. Am J Physiol Heart Circ Physiol.
1997;272:31203–31208.
2000;278:H2115–H2123.
211. Sei CA, Irons CE, Sprenkle AB, McDonough PM, Brown JH,

193. Deng L, Huang B, Qin D, Ganguly K, El-Sherif N. Calcineurin inhi-
Glembotski CC. The alpha-adrenergic stimulation of atrial natri-
bition ameliorates structural, contractile, and electrophysiologic con-
uretic factor expression in cardiac myocytes requires calcium influx,
sequences of postinfarction remodeling. J Cardiovasc Electrophysiol.
protein kinase C, and calmodulin-regulated pathways. J Biol Chem.
2001;12:1055–1061.
1991;266:15910–15916.
194. Youn TJ, Piao H, Kwon JS, Choi SY, Kim HS, Park DG, Kim DW, Kim
212. Sucharov CC, Mariner PD, Nunley KR, Long C, Leinwand L, Bristow
YG, Cho MC. Effects of the calcineurin dependent signaling pathway MR. A beta1-adrenergic receptor CaM kinase II-dependent pathway me-
inhibition by cyclosporin A on early and late cardiac remodeling follow- diates cardiac myocyte fetal gene induction. Am J Physiol Heart Circ
ing myocardial infarction. Eur J Heart Fail. 2002;4:713–718. Physiol. 2006;291:H1299–H1308. doi: 10.1152/ajpheart.00017.2006.
195. Sussman MA, Lim HW, Gude N, Taigen T, Olson EN, Robbins J, 213. Lim HW, Molkentin JD. Calcineurin and human heart failure. Nat Med.
Colbert MC, Gualberto A, Wieczorek DF, Molkentin JD. Prevention 1999;5:246–247. doi: 10.1038/6430.
of cardiac hypertrophy in mice by calcineurin inhibition. Science. 214. Haq S, Choukroun G, Lim H, Tymitz KM, del Monte F, Gwathmey J,
1998;281:1690–1693. Grazette L, Michael A, Hajjar R, Force T, Molkentin JD. Differential
196. Goldspink PH, McKinney RD, Kimball VA, Geenen DL, Buttrick PM. activation of signal transduction pathways in human hearts with hyper-
Angiotensin II induced cardiac hypertrophy in vivo is inhibited by cyclo- trophy versus advanced heart failure. Circulation. 2001;103:670–677.
sporin A in adult rats. Mol Cell Biochem. 2001;226:83–88. 215. Jiang DS, Wei X, Zhang XF, et al. IRF8 suppresses pathological car-
197. Shimoyama M, Hayashi D, Takimoto E, Zou Y, Oka T, Uozumi H, Kudoh diac remodelling by inhibiting calcineurin signalling. Nat Commun.
S, Shibasaki F, Yazaki Y, Nagai R, Komuro I. Calcineurin plays a criti- 2014;5:3303. doi: 10.1038/ncomms4303.
cal role in pressure overload-induced cardiac hypertrophy. Circulation. 216. Taigen T, De Windt LJ, Lim HW, Molkentin JD. Targeted inhibition of
1999;100:2449–2454. calcineurin prevents agonist-induced cardiomyocyte hypertrophy. Proc
198. De Windt LJ, Lim HW, Bueno OF, Liang Q, Delling U, Braz JC, Glascock Natl Acad Sci USA. 2000;97:1196–1201.
BJ, Kimball TF, del Monte F, Hajjar RJ, Molkentin JD. Targeted inhi- 217. Chu G, Carr AN, Young KB, Lester JW, Yatani A, Sanbe A, Colbert MC,
bition of calcineurin attenuates cardiac hypertrophy in vivo. Proc Natl Schwartz SM, Frank KF, Lampe PD, Robbins J, Molkentin JD, Kranias
Acad Sci USA. 2001;98:3322–3327. doi: 10.1073/pnas.031371998. EG. Enhanced myocyte contractility and Ca2+ handling in a calcineurin
199. Hill JA, Rothermel B, Yoo KD, Cabuay B, Demetroulis E, Weiss RM, transgenic model of heart failure. Cardiovasc Res. 2002;54:105–116.
Kutschke W, Bassel-Duby R, Williams RS. Targeted inhibition of cal- 218. Eto Y, Yonekura K, Sonoda M, Arai N, Sata M, Sugiura S, Takenaka K,
cineurin in pressure-overload cardiac hypertrophy. Preservation of sys- Gualberto A, Hixon ML, Wagner MW, Aoyagi T. Calcineurin is activated
tolic function. J Biol Chem. 2002;277:10251–10255. doi: 10.1074/jbc. in rat hearts with physiological left ventricular hypertrophy induced by
M110722200. voluntary exercise training. Circulation. 2000;101:2134–2137.
200. van Rooij E, Doevendans PA, Crijns HJ, Heeneman S, Lips DJ, van 219. Chung E, Yeung F, Leinwand LA. Calcineurin activity is required for
Bilsen M, Williams RS, Olson EN, Bassel-Duby R, Rothermel BA, De cardiac remodelling in pregnancy. Cardiovasc Res. 2013;100:402–410.
Windt LJ. MCIP1 overexpression suppresses left ventricular remodel- doi: 10.1093/cvr/cvt208.
ing and sustains cardiac function after myocardial infarction. Circ Res. 220. Hainsey T, Csiszar A, Sun S, Edwards JG. Cyclosporin A does not
2004;94:e18–e26. doi: 10.1161/01.RES.0000118597.54416.00. block exercise-induced cardiac hypertrophy. Med Sci Sports Exerc.
201. Gelpi RJ, Gao S, Zhai P, Yan L, Hong C, Danridge LM, Ge H, Maejima 2002;34:1249–1254.
Y, Donato M, Yokota M, Molkentin JD, Vatner DE, Vatner SF, Sadoshima 221. Oliveira RS, Ferreira JC, Gomes ER, Paixão NA, Rolim NP, Medeiros
J. Genetic inhibition of calcineurin induces diastolic dysfunction in A, Guatimosim S, Brum PC. Cardiac anti-remodelling effect of aerobic
Dewenter et al   Calcium Signaling and Transcription   1019

training is associated with a reduction in the calcineurin/NFAT signal- targets attenuating this effect. J Cell Physiol. 2008;217:242–249. doi:
ling pathway in heart failure mice. J Physiol. 2009;587:3899–3910. doi: 10.1002/jcp.21501.
10.1113/jphysiol.2009.173948. 240. Satoh N, Suter TM, Liao R, Colucci WS. Chronic alpha-adrenergic re-
222. Wilkins BJ, Dai YS, Bueno OF, Parsons SA, Xu J, Plank DM, Jones ceptor stimulation modulates the contractile phenotype of cardiac myo-
F, Kimball TR, Molkentin JD. Calcineurin/NFAT coupling participates cytes in vitro. Circulation. 2000;102:2249–2254.
in pathological, but not physiological, cardiac hypertrophy. Circ Res. 241. Osadchii OE, Norton GR, McKechnie R, Deftereos D, Woodiwiss AJ.
2004;94:110–118. doi: 10.1161/01.RES.0000109415.17511.18. Cardiac dilatation and pump dysfunction without intrinsic myocardial
223. Cooley N, Ouyang K, McMullen JR, Kiriazis H, Sheikh F, Wu W, Mu systolic failure following chronic beta-adrenoreceptor activation. Am
Y, Du XJ, Chen J, Woodcock EA. No contribution of IP3-R(2) to dis- J Physiol Heart Circ Physiol. 2007;292:H1898–H1905. doi: 10.1152/
ease phenotype in models of dilated cardiomyopathy or pressure over- ajpheart.00740.2006.
load hypertrophy. Circ Heart Fail. 2013;6:318–325. doi: 10.1161/ 242. Lechat P, Packer M, Chalon S, Cucherat M, Arab T, Boissel JP. Clinical
CIRCHEARTFAILURE.112.972158. effects of beta-adrenergic blockade in chronic heart failure: a meta-anal-
224. Seth M, Zhang ZS, Mao L, Graham V, Burch J, Stiber J, Tsiokas L, Winn ysis of double-blind, placebo-controlled, randomized trials. Circulation.
M, Abramowitz J, Rockman HA, Birnbaumer L, Rosenberg P. TRPC1 1998;98:1184–1191.
channels are critical for hypertrophic signaling in the heart. Circ Res. 243. Flather MD, Yusuf S, Køber L, Pfeffer M, Hall A, Murray G, Torp-
2009;105:1023–1030. doi: 10.1161/CIRCRESAHA.109.206581. Pedersen C, Ball S, Pogue J, Moyé L, Braunwald E. Long-term
225. Nakayama H, Wilkin BJ, Bodi I, Molkentin JD. Calcineurin-dependent ACE-inhibitor therapy in patients with heart failure or left-ventricular
cardiomyopathy is activated by TRPC in the adult mouse heart. FASEB J. dysfunction: a systematic overview of data from individual patients.
2006;20:1660–1670. doi: 10.1096/fj.05-5560com. ACE-Inhibitor Myocardial Infarction Collaborative Group. Lancet.
226. Seo K, Rainer PP, Shalkey Hahn V, Lee DI, Jo SH, Andersen A, Liu T, Xu 2000;355:1575–1581.
X, Willette RN, Lepore JJ, Marino JP Jr, Birnbaumer L, Schnackenberg 244. Satoh S, Ueda Y, Suematsu N, Oyama J, Kadokami T, Sugano M,

CG, Kass DA. Combined TRPC3 and TRPC6 blockade by selective Yoshikawa Y, Makino N. Beneficial effects of angiotensin-converting
small-molecule or genetic deletion inhibits pathological cardiac hyper- enzyme inhibition on sarcoplasmic reticulum function in the failing heart
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

trophy. Proc Natl Acad Sci USA. 2014;111:1551–1556. doi: 10.1073/ of the Dahl rat. Circ J. 2003;67:705–711.
pnas.1308963111. 245. Reiken S, Wehrens XH, Vest JA, Barbone A, Klotz S, Mancini D, Burkhoff
227. Camacho Londoño JE, Tian Q, Hammer K, et al. A background Ca2+ D, Marks AR. Beta-blockers restore calcium release channel function and
entry pathway mediated by TRPC1/TRPC4 is critical for development of improve cardiac muscle performance in human heart failure. Circulation.
pathological cardiac remodelling. Eur Heart J. 2015;36:2257–2266. doi: 2003;107:2459–2466. doi: 10.1161/01.CIR.0000068316.53218.49.
10.1093/eurheartj/ehv250. 246. Shannon TR, Pogwizd SM, Bers DM. Elevated sarcoplasmic reticulum
228. Wu X, Eder P, Chang B, Molkentin JD. TRPC channels are necessary Ca2+ leak in intact ventricular myocytes from rabbits in heart failure.
mediators of pathologic cardiac hypertrophy. Proc Natl Acad Sci USA. Circ Res. 2003;93:592–594. doi: 10.1161/01.RES.0000093399.11734.
2010;107:7000–7005. doi: 10.1073/pnas.1001825107. B3.
229. Correll RN, Goonasekera SA, van Berlo JH, Burr AR, Accornero F, 247. Marx SO, Reiken S, Hisamatsu Y, Jayaraman T, Burkhoff D, Rosemblit
Zhang H, Makarewich CA, York AJ, Sargent MA, Chen X, Houser SR, N, Marks AR. PKA phosphorylation dissociates FKBP12.6 from the cal-
Molkentin JD. STIM1 elevation in the heart results in aberrant Ca²⁺ cium release channel (ryanodine receptor): defective regulation in failing
handling and cardiomyopathy. J Mol Cell Cardiol. 2015;87:38–47. doi: hearts. Cell. 2000;101:365–376.
10.1016/j.yjmcc.2015.07.032. 248. Wehrens XH, Lehnart SE, Reiken S, Vest JA, Wronska A, Marks AR.
230. Collins HE, He L, Zou L, Qu J, Zhou L, Litovsky SH, Yang Q, Young ME, Ryanodine receptor/calcium release channel PKA phosphorylation: a
Marchase RB, Chatham JC. Stromal interaction molecule 1 is essential critical mediator of heart failure progression. Proc Natl Acad Sci USA.
for normal cardiac homeostasis through modulation of ER and mitochon- 2006;103:511–518. doi: 10.1073/pnas.0510113103.
drial function. Am J Physiol Heart Circ Physiol. 2014;306:H1231– 249. Benkusky NA, Weber CS, Scherman JA, Farrell EF, Hacker TA, John
H1239. doi: 10.1152/ajpheart.00075.2014. MC, Powers PA, Valdivia HH. Intact beta-adrenergic response and
231. Wu X, Zhang T, Bossuyt J, Li X, McKinsey TA, Dedman JR, Olson EN, unmodified progression toward heart failure in mice with genetic
Chen J, Brown JH, Bers DM. Local InsP3-dependent perinuclear Ca2+ ablation of a major protein kinase A phosphorylation site in the car-
signaling in cardiac myocyte excitation-transcription coupling. J Clin diac ryanodine receptor. Circ Res. 2007;101:819–829. doi: 10.1161/
Invest. 2006;116:675–682. doi: 10.1172/JCI27374. CIRCRESAHA.107.153007.
232. Voelkers M, Salz M, Herzog N, Frank D, Dolatabadi N, Frey N, Gude 250. Zhang H, Makarewich CA, Kubo H, Wang W, Duran JM, Li Y,

N, Friedrich O, Koch WJ, Katus HA, Sussman MA, Most P. Orai1 and Berretta RM, Koch WJ, Chen X, Gao E, Valdivia HH, Houser SR.
Stim1 regulate normal and hypertrophic growth in cardiomyocytes. J Mol Hyperphosphorylation of the cardiac ryanodine receptor at serine 2808 is
Cell Cardiol. 2010;48:1329–1334. doi: 10.1016/j.yjmcc.2010.01.020. not involved in cardiac dysfunction after myocardial infarction. Circ Res.
233. Troupes CD, Wallner M, Borghetti G, Zhang C, Mohsin S, von
2012;110:831–840. doi: 10.1161/CIRCRESAHA.111.255158.
Lewinski D, Berretta RM, Kubo H, Chen X, Soboloff J, Houser S. 251. Guo T, Zhang T, Mestril R, Bers DM. Ca2+/calmodulin-dependent pro-
Role of STIM1 (stromal interaction molecule 1) in hypertrophy-relat- tein kinase II phosphorylation of ryanodine receptor does affect calcium
ed contractile dysfunction. Circ Res. 2017;121:125–136. doi: 10.1161/ sparks in mouse ventricular myocytes. Circ Res. 2006;99:398–406. doi:
CIRCRESAHA.117.311094. 10.1161/01.RES.0000236756.06252.13.
234. Hulot JS, Fauconnier J, Ramanujam D, et al. Critical role for stromal in- 252. Sossalla S, Fluschnik N, Schotola H, Ort KR, Neef S, Schulte T,

teraction molecule 1 in cardiac hypertrophy. Circulation. 2011;124:796– Wittköpper K, Renner A, Schmitto JD, Gummert J, El-Armouche A,
805. doi: 10.1161/CIRCULATIONAHA.111.031229. Hasenfuss G, Maier LS. Inhibition of elevated Ca2+/calmodulin-
235. Pieske B, Beyermann B, Breu V, Löffler BM, Schlotthauer K, Maier LS, dependent protein kinase II improves contractility in human fail-
Schmidt-Schweda S, Just H, Hasenfuss G. Functional effects of endothe- ing myocardium. Circ Res. 2010;107:1150–1161. doi: 10.1161/
lin and regulation of endothelin receptors in isolated human nonfailing CIRCRESAHA.110.220418.
and failing myocardium. Circulation. 1999;99:1802–1809. 253. Respress JL, van Oort RJ, Li N, et al. Role of RyR2 phosphorylation at
236. Serneri GG, Boddi M, Cecioni I, Vanni S, Coppo M, Papa ML,
S2814 during heart failure progression. Circ Res. 2012;110:1474–1483.
Bandinelli B, Bertolozzi I, Polidori G, Toscano T, Maccherini M, doi: 10.1161/CIRCRESAHA.112.268094.
Modesti PA. Cardiac angiotensin II formation in the clinical course of 254. van Oort RJ, McCauley MD, Dixit SS, Pereira L, Yang Y, Respress
heart failure and its relationship with left ventricular function. Circ Res. JL, Wang Q, De Almeida AC, Skapura DG, Anderson ME, Bers DM,
2001;88:961–968. Wehrens XH. Ryanodine receptor phosphorylation by calcium/calmodu-
237. Thomas JA, Marks BH. Plasma norepinephrine in congestive heart fail- lin-dependent protein kinase II promotes life-threatening ventricular ar-
ure. Am J Cardiol. 1978;41:233–243. rhythmias in mice with heart failure. Circulation. 2010;122:2669–2679.
238. Benedict CR, Shelton B, Johnstone DE, Francis G, Greenberg B,
doi: 10.1161/CIRCULATIONAHA.110.982298.
Konstam M, Probstfield JL, Yusuf S. Prognostic significance of plasma 255. Pleger ST, Most P, Boucher M, Soltys S, Chuprun JK, Pleger W, Gao E,
norepinephrine in patients with asymptomatic left ventricular dysfunc- Dasgupta A, Rengo G, Remppis A, Katus HA, Eckhart AD, Rabinowitz
tion. SOLVD Investigators. Circulation. 1996;94:690–697. JE, Koch WJ. Stable myocardial-specific AAV6-S100A1 gene ther-
239. Mufti S, Wenzel S, Euler G, Piper HM, Schlüter KD. Angiotensin II- apy results in chronic functional heart failure rescue. Circulation.
dependent loss of cardiac function: mechanisms and pharmacological 2007;115:2506–2515. doi: 10.1161/CIRCULATIONAHA.106.671701.
1020  Circulation Research  September 29, 2017

256. Kho C, Lee A, Jeong D, Oh JG, Chaanine AH, Kizana E, Park WJ, Hajjar 260. Lisman J, Yasuda R, Raghavachari S. Mechanisms of CaMKII action
RJ. SUMO1-dependent modulation of SERCA2a in heart failure. Nature. in long-term potentiation. Nat Rev Neurosci. 2012;13:169–182. doi:
2011;477:601–605. doi: 10.1038/nature10407. 10.1038/nrn3192.
257. Kawase Y, Ly HQ, Prunier F, et al. Reversal of cardiac dysfunction af- 261. Backs J, Stein P, Backs T, Duncan FE, Grueter CE, McAnally J, Qi X,
ter long-term expression of SERCA2a by gene transfer in a pre-clinical Schultz RM, Olson EN. The gamma isoform of CaM kinase II controls
model of heart failure. J Am Coll Cardiol. 2008;51:1112–1119. doi: mouse egg activation by regulating cell cycle resumption. Proc Natl Acad
10.1016/j.jacc.2007.12.014. Sci USA. 2010;107:81–86. doi: 10.1073/pnas.0912658106.
258. Greenberg B, Butler J, Felker GM, Ponikowski P, Voors AA, Desai 262. Bui JD, Calbo S, Hayden-Martinez K, Kane LP, Gardner P, Hedrick SM.
AS, Barnard D, Bouchard A, Jaski B, Lyon AR, Pogoda JM, Rudy JJ, A role for CaMKII in T cell memory. Cell. 2000;100:457–467.
Zsebo KM. Calcium upregulation by percutaneous administration of 263. Pellicena P, Schulman H. CaMKII inhibitors: from research tools

gene therapy in patients with cardiac disease (CUPID 2): a randomised, to therapeutic agents. Front Pharmacol. 2014;5:21. doi: 10.3389/
multinational, double-blind, placebo-controlled, phase 2b trial. Lancet. fphar.2014.00021.
2016;387:1178–1186. doi: 10.1016/S0140-6736(16)00082-9. 264. Ottani F, Latini R, Staszewsky L, et al; CYCLE Investigators.

259. Hulot JS, Ishikawa K, Hajjar RJ. Gene therapy for the treatment of Cyclosporine A in reperfused myocardial infarction: the multicenter,
heart failure: promise postponed. Eur Heart J. 2016;37:1651–1658. doi: controlled, open-label CYCLE trial. J Am Coll Cardiol. 2016;67:365–
10.1093/eurheartj/ehw019. 374. doi: 10.1016/j.jacc.2015.10.081.
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017
Calcium Signaling and Transcriptional Regulation in Cardiomyocytes
Matthias Dewenter, Albert von der Lieth, Hugo A. Katus and Johannes Backs
Downloaded from http://circres.ahajournals.org/ by guest on October 8, 2017

Circ Res. 2017;121:1000-1020


doi: 10.1161/CIRCRESAHA.117.310355
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2017 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/121/8/1000

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the
Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information
about this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation Research is online at:


http://circres.ahajournals.org//subscriptions/

You might also like