Next Article in Journal
In Silico Identification of Lead Compounds for Pseudomonas Aeruginosa PqsA Enzyme: Computational Study to Block Biofilm Formation
Next Article in Special Issue
The Junctophilin-2 Mutation p.(Thr161Lys) Is Associated with Hypertrophic Cardiomyopathy Using Patient-Specific iPS Cardiomyocytes and Demonstrates Prolonged Action Potential and Increased Arrhythmogenicity
Previous Article in Journal
Co-Occurring X-Linked Agammaglobulinemia and X-Linked Chronic Granulomatous Disease: Two Isolated Pathogenic Variants in One Patient
Previous Article in Special Issue
Gq-Mediated Arrhythmogenic Signaling Promotes Atrial Fibrillation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nuclear Calcium in Cardiac (Patho)Physiology: Small Compartment, Big Impact

by
Mara Kiessling
1,*,
Nataša Djalinac
2,
Julia Voglhuber
1,3 and
Senka Ljubojevic-Holzer
1,3,4,*
1
Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
2
Department of Biology, University of Padua, 35122 Padova, Italy
3
BioTechMed Graz, 8010 Graz, Austria
4
Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
*
Authors to whom correspondence should be addressed.
Biomedicines 2023, 11(3), 960; https://doi.org/10.3390/biomedicines11030960
Submission received: 1 March 2023 / Accepted: 17 March 2023 / Published: 21 March 2023
(This article belongs to the Special Issue Regulation of Ca2+ Signals in Cardiovascular Biology)

Abstract

:
The nucleus of a cardiomyocyte has been increasingly recognized as a morphologically distinct and partially independent calcium (Ca2+) signaling microdomain, with its own Ca2+-regulatory mechanisms and important effects on cardiac gene expression. In this review, we (1) provide a comprehensive overview of the current state of research on the dynamics and regulation of nuclear Ca2+ signaling in cardiomyocytes, (2) address the role of nuclear Ca2+ in the development and progression of cardiac pathologies, such as heart failure and atrial fibrillation, and (3) discuss novel aspects of experimental methods to investigate nuclear Ca2+ handling and its downstream effects in the heart. Finally, we highlight current challenges and limitations and recommend future directions for addressing key open questions.

1. Introduction

Calcium (Ca2+) is a ubiquitous intracellular second messenger regulating a plethora of intricate cellular events and functions, e.g., electrical signaling, contraction, secretion, gene transcription and cell cycle [1]. In the heart, Ca2+ most prominently mediates the translation of electrical stimulation into the mechanical activity of cardiomyocytes—also known as excitation-contraction coupling (ECC)—thus inducing the contraction of cardiac muscle [2]. Each heartbeat is characterized by a precisely regulated transient rise in cytoplasmic free Ca2+ to systolic levels of about 1 µM at peak contraction and subsequent removal to diastolic levels of about 100 nM at full relaxation [3,4]. Beyond this well-described role in cardiomyocyte contractile function, Ca2+ has emerged as a key player in both physiological and pathophysiological cardiac signaling cascades and the regulation of gene transcription. It can thereby not only exert acute cellular effects but also contribute to long-term changes through so-called excitation-transcription coupling (ETC) [5,6,7].
Considering that the entire cardiomyocyte’s cytosol is flooded with Ca2+ on a beat-to-beat basis, there is an obvious need for the cell to spatio-temporally distinguish between “contractile” and “signaling” Ca2+ to properly elicit specific cellular functions [8]. This is in part achieved by the subcellular organization of cardiomyocytes in so-called microdomains that permit targeted Ca2+ release and thereby facilitate locally restricted Ca2+ signaling events [9,10]. The nucleus is considered as such distinct microdomain harboring its own nucleoplasmic Ca2+ transient cycling machinery and signaling properties with important implications for the regulation of gene transcription [7,10,11]. While alterations in cytoplasmic Ca2+ handling have been extensively studied and associated with the pathogenesis of cardiac remodeling, heart failure (HF) and atrial fibrillation (AF); nucleoplasmic Ca2+ has received considerably less attention. In this review, we will discuss the role of nuclear Ca2+ in cardiac physiology and pathophysiology and summarize recent findings on nuclear Ca2+ calibration methods and imaging tools. We will assess new developments in experimental approaches and highlight yet unanswered questions to stimulate further research.

2. Nuclear Ca2+ Dynamics

2.1. Structural Basis of Nuclear Ca2+ Handling

The nucleus is a subcellular compartment separated from the cytoplasm by the nuclear envelope (NE). Consisting of two phospholipid bilayers, the NE acts as a barrier and selective filter for the trafficking of substances and molecules in and out of the nucleus. Throughout the NE, nuclear pore complexes (NPCs) facilitate the diffusion of ions and small molecules (5 nm and 40–60 kDa) [12,13] and actively shuttle bigger proteins and RNAs (≤39 nm) via an associated transport system [14]. The inner and outer nuclear membranes show distinct characteristics determined by their orientation and the adjacent compartment. The outer nuclear membrane faces the cytoplasm and transitions directly into the membrane of the sarcoplasmic reticulum (SR), whereas the inner nuclear membrane is directed towards the nucleoplasm forming deep invaginations into the nuclear lumen, referred to as the nuclear reticulum [15,16]. Various Ca2+ release and re-uptake proteins and their regulators are embedded in the NE, such as inositol-1,4,5-triphosphate receptors (IP3Rs) [17], as well as the SR Ca2+ ATPase (SERCA) [18], phospholamban (PLB) [19] and the sodium-calcium exchanger (NCX) [20], highlighting the nucleus as an autonomous, fully-equipped Ca2+ handling compartment. The area between both membranes is similar in ion and protein composition to the SR and acts as a local Ca2+ storage [15]. Deep T-tubules are frequently located in very close proximity to the NE forming local Ca2+ signaling hubs, often accompanied by prominent mitochondrial accumulation [21,22,23].

2.2. Nuclear Ca2+ Transients

Every cytosolic Ca2+ transient (CaT) in a cardiomyocyte elicits a nuclear CaT (Figure 1, left). This also means that any change in cytoplasmic Ca2+ concentration, [Ca2+]cyto, e.g., following neurohormonal stimulation, will be accompanied by changes in nucleoplasmic Ca2+ concentration, [Ca2+]nuc [24]. Nuclear CaTs are characterized by a slower and delayed upstroke, lower peak and prolonged decay time in comparison to cytosolic CaTs. The delayed upstroke is likely caused by the insulation of the nucleus by the NE and the slower spread of Ca2+ released from the SR through a limited space within the NPCs, introducing a kinetic delay and simultaneously reducing the amplitude [25]. This idea is supported by the documented absence of major Ca2+ release channels from the NE, such as ryanodine receptors (RyRs), which are instead found in close proximity to the NE, forming a “cage” around the nucleus [26,27,28]. Similarly, [Ca2+] decline is mediated by passive diffusion out of the nucleus through NPCs and subsequent SR/NE SERCA reuptake or extrusion via NCX located in nearby T-tubules. As the large majority of SERCA is located on the outer nuclear membrane, Ca2+ first needs to diffuse out of the nucleus through NPCs to be taken up again into the lumen of the NE [24,29]. In a series of elegant experiments in which they modulated SERCA function with thapsigargin, Kiess and Kockskaemper indeed confirmed that—together with cytoplasmic Ca2+ levels—SERCA function is the single most important factor regulating diastolic [Ca2+]nuc [24]. Although the NPCs always remain open, their conductance and thereby total Ca2+ diffusion capacity can vary, depending on intracellular [Ca2+] and [ATP] [30], as well as distribution, density and position of NPCs on the NE [31]. Due to these structural and functional features, changes in stimulation frequency differentially affect nuclear and cytosolic diastolic CaTs. Whereas systolic [Ca2+]nuc increases to a similar extent as [Ca2+]cyto in response to faster pacing, diastolic [Ca2+]nuc rises to about twice the [Ca2+]cyto, suggesting that slower nucleoplasmic CaT kinetics provoke a build-up of Ca2+ when diastole is shortened [32]. If prolonged, such diastolic [Ca2+]nuc overload may lead to the activation of Ca2+-mediated hypertrophic pathways in the nucleus with important structural and functional consequences for the heart.
Recent work uncovered an additional regulatory mechanism of nuclear CaTs in cardiomyocytes, which prevents nucleoplasmic Ca2+ overload, especially in conditions of physiological stress. Namely, Voglhuber and Holzer et al. found that in healthy cardiomyocytes, densely packed mitochondria in perinuclear spaces—small areas of cytoplasm adjacent to the nucleus in the longitudinal direction—shape nucleoplasmic CaTs by taking up significant amounts of Ca2+ in close proximity to the nucleus. In contrast, pharmacological inhibition of mitochondrial Ca2+ uptake led to a significant increase in Ca2+ levels in and around the nucleus [21].

2.3. Nuclear Ca2+ Sparks and Puffs

As a result of the close proximity of NE and SR, the nucleus reacts to spontaneous, local Ca2+ release events from RyR2s and IP3Rs—so-called Ca2+ sparks and puffs—with a brief increase in [Ca2+]nuc independent of [Ca2+]cyto [33,34]. Similarly, the small distance between the NE and T-tubules and dyads regulates nuclear Ca2+ content without influencing global [Ca2+] [27]. PLB, the endogenous inhibitor of SERCA, is highly concentrated in the NE with a greater PLB to SERCA ratio than in the SR and plays a key role in the regulation of such localized nuclear Ca2+ dynamics [19,35]. Indeed, modulation of PLB activity affects the amount of Ca2+ taken up into lumen of the NE and regulates IP3R-RyR2 mediated spontaneous Ca2+ release [34]. As levels and activity of IP3R and PLB dramatically change during (patho)physiological cardiac stress [25,36], the altered dynamics of nuclear Ca2+ sparks and puffs are likely causally involved in the progression of adverse cardiac remodeling.

2.4. Receptor-Mediated Nuclear Ca2+ Handling

In addition to its regulation via passive diffusion of Ca2+ from the cytoplasm or specialized microdomains such as dyads, [Ca2+]nuc can be additionally and independently regulated via the direct release of Ca2+ from the NE [37]. Such active regulation of nuclear Ca2+ levels involves different classes of receptors located on and in close proximity to the NE.

2.4.1. IP3R

Key components of autonomous nuclear Ca2+ regulation processes are IP3Rs, with IP3R type 2 (IP3R2) expressed mainly in ventricular [26,36] and IP3R type 1 (IP3R1) in atrial cardiomyocytes [38], though recent evidence suggests the possibility of all IP3R subtypes (1–3) being present in both neonatal and adult cardiomyocytes [39]. IP3Rs are activated following G-protein coupled receptor (GPCR)-dependent activation of phospholipase C and generation of IP3 which then binds to the IP3Rs, instigating Ca2+ release from intracellular Ca2+ stores (Figure 1, right). Examples of GPCRs include endothelin-1 (ET-1), angiotensin-II (Ang II) and insulin-like growth factor 1 (IGF-1) receptors [40,41,42], which once coupled to their agonists are implicated in promoting hypertrophy. Cardiomyocytes show a unique pattern of IP3R distribution, with evidence that they are concentrated in nuclear and perinuclear compartments [26,28], thereby facilitating Ca2+-mediated transcriptional changes. The distribution of IP3Rs can, however, differ depending on the onset, intensity and duration of nuclear IP3-mediated Ca2+ signaling and finely tune activation of its downstream targets, such as Ca2+/calmodulin-dependent protein kinase II (CaMKII) and calcineurin (CaN) [26,28,40,43,44].
While IP3R-dependent regulation of [Ca2+]nuc is consistently shown to be mediated by Ca2+ release from perinuclear stores, precaution is needed when choosing an appropriate model to study IP3R signaling, since it is dependent on specific upstream stimuli, cardiomyocyte species and their maturation stage [32,40,45,46,47,48,49]. Ang II acts through IP3Rs to over-proportionally raise Ca2+ in the nucleus versus cytosol, both at rest [44] or when cells are paced at baseline frequencies [28]. This increase in [Ca2+]nuc is further sensed by CaN, which activates nuclear factor of activated T cells (NFAT)-dependent transcription [44]. Similarly, IGF-1 is also capable of eliciting an IP3R-mediated increase in nuclear Ca2+ levels. IGF-1 was shown to induce higher peak amplitudes and larger CaTs in the nucleus, which were clearly attributable to Ca2+ release via IP3Rs, as demonstrated using IP3R and RyR2 inhibitors [50]. This may be facilitated by the existence of a locally restricted signaling toolkit formed by deep T-tubular invaginations towards the NE, harboring the IGF-1 receptor and providing spatial insulation from whole cell Ca2+ oscillations [42]. Nakao et al. further suggested the involvement of neuronal calcium sensor-1 (NCS-1)—known for its pivotal role in various neuronal functions—in regulating nuclear and cytoplasmic Ca2+ signals mediated by IGF-1 in cardiomyocytes. In their study, NCS-1 co-localized with IP3Rs on the NE and in perinuclear regions [50]. This is in line with a previous report on the interaction between NCS-1 and IP3Rs as an important regulator of hypertrophy, engaging both CaMKII and CaN pathways [51].

2.4.2. Adrenergic Receptors

Both in vivo and in vitro studies have demonstrated nuclear and perinuclear localization of α1-adrenergic receptors and their signaling partners in cardiomyocytes [52,53,54,55,56]. This is in contrast to the conventional notion that GPCRs localize to and signal at the plasma membrane [57,58,59]. Mechanistically, evidence suggests a α1A-subtype/PKCδ/cTnI mediated “inside-out” (nuclear-to-cytoplasmic) signaling pathway, describing the transport of signals initiated at α1-ARs in the inner nuclear membrane to cytosolic (sarcomere) or membrane targets where the effects on cardiomyocyte contractile function are then elicited [52]. In line with this, α1-AR agonist, phenylephrine (PE), induced an increase in IP3R-mediated nuclear CaT frequency and triggered Ca2+-induced Ca2+ release in the cytosol of isolated neonatal rat cardiomyocytes [47]. However, insights into other contractile regulators controlled by nuclear α1-ARs are still sparse due to the limitations in techniques to detect α1-ARs [60]. Several studies including patient datasets have verified a cardioprotective role of α1-AR activation [52], thus emphasizing the therapeutic potential of modulating nuclear α-AR signaling.
Conversely, β-AR expression on cardiomyocyte nuclei is more controversially discussed. On the one hand, Boivin et al. demonstrated by immunological, ligand-binding and functional criteria that in rat and mouse adult ventricular cardiomyocytes β1AR- and β3AR-subtypes are located on the nuclear membrane [61]. Similarly, downstream effectors such as adenylyl cyclase (AC) and protein kinase A (PKA) were shown to be associated with the nucleus or the nuclear membrane [62], and β1AR stimulation with isoproterenol resulted in both increased AC activity and modulated gene expression levels in isolated nuclei from rat hearts [61,63]. Wang et al. further observed β1AR expression at the SR, associated with SERCA and PLB, to promote the phosphorylation of local targets via PKA activation [64]. On the other hand, Bedioune et al. showed that β1ARs and β2ARs located in the plasmalemma also differentially regulate nuclear PKA activity upon receptor stimulation, highlighting the formation of signaling routes from the plasmalemma into the nucleus with potential implications on the control of gene expression by βARs [65] without nuclear expression of the receptors. Furthermore, visualization of βARs in cardiomyocytes with highly sensitive microscopy methods failed to show a nuclear specific localization of βARs [66]. Recent evidence suggests the Golgi apparatus as a localized βAR-expression and signaling microdomain in close vicinity to the nucleus [67,68]. Overall, intracellular βAR-signaling emerges as an interesting concept in the regulation of cardiac contractility and may present a novel translational approach in optimizing the subcellular targeting of β-blockers in the future.
Together, the elaborate structural organization of the nucleus and its interplay with other subcellular compartments enable the formation of local, Ca2+ sensitive nuclear microdomains. In such a spatially and functionally restricted environment, Ca2+ signals can manifest immediate or long-term transcriptional regulatory effects and minutely respond to the current physiological demand of cardiomyocytes.

3. Role of Nuclear Ca2+ in Cardiomyocytes

Nuclear Ca2+ dynamics in cardiomyocytes are mainly associated with the process of ETC, where gene expression is regulated as part of the cellular response to physiological and pathological stress [69,70]. The best-studied signaling cascades activated by increased nuclear Ca2+ levels in cardiomyocytes are mediated by CaMKII and CaN. Downstream of CaMKII activation, phosphorylation of histone deacetylase 4 (HDAC4) was shown to promote nuclear HDAC4 export and thereby de-repress pro-hypertrophic transcription factor myocyte enhancer factor 2 (MEF2). MEF2 is involved in cardiac remodeling, fetal gene program re-expression and fibrosis [71,72]. Other transcription factors activated in a CaMKII-dependent manner have also been identified, e.g., the nuclear cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) [73] and nuclear factor-kappa B (NF-κB) [74]. Subedi et al. demonstrated that CaMKII mediates CREB activation in response to ET-1 and PE stimulation in rat ventricular cardiomyocytes [75], while Suetomi et al. identified CaMKII-mediated proinflammatory signaling involving NF-κB upon introduction of pressure overload in mice [74]. Similarly, CaMKII potently represses the expression of L-type Ca2+ channels (LTCC) by translocating the downstream regulatory element binding transcription factor DREAM, thus negatively regulating Ca2+ influx into the cytosol [76]. Still, it remains an open question whether activation of CaMKII upstream of CREB, NF-κB and DREAM is specifically mediated by nuclear Ca2+ accumulation or overall increases in [Ca2+] in cardiomyocytes. CaN, as CaMKII, is activated by Ca2+/calmodulin and thereby sensitive to elevated [Ca2+]. Several factors including differences in association rate with Ca2+/calmodulin, regional distribution of CaN, the presence of competing binding partners and endogenous inhibitors allow CaN to evade continuous activation following each contraction [77]. On the other hand, overactivation of CaN leads to pathological gene transcription by targeted phosphorylation of several transcription factors. For instance, CaN dephosphorylates cytosolic transcription factor NFAT and causes its translocation to the nucleus, where it interacts with GATA4 to mediate a hypertrophic response [78]. Importantly, recent work has demonstrated that CaN-NFAT signaling in cardiomyocytes is controlled by perinuclear signalosomes organized by the scaffold protein muscle A-Kinase anchoring protein β (mAKAPβ/AKAP6β) and is sensitive to (peri)nuclear Ca2+ [10]. Other major targets of CaN include CRTC, FOXO1 and TFEB [79], yet their specific activation by [Ca2+]nuc remains to be addressed.

4. Nuclear Ca2+ Dysregulation

While alterations in intracellular [Ca2+] have generally been accepted as drivers of pathological events in cardiomyocytes, studies reporting isolated changes in nucleoplasmic Ca2+ levels are gravely underrepresented in the literature. However, many arguments support the role of [Ca2+]nuc as an important regulator of pathological transcription. One strong argument is that functional and structural alterations of the nucleus precede the development of cardiac pathologies. These include changes in nuclear size and shape in the form of NE invaginations and blebbing, together with changes in expression of Ca2+ handling proteins, most importantly RyR2s, SERCA and IP3Rs [28,44,80,81].

4.1. Nuclear Ca2+ in Ventricular Remodeling: Hypertrophy to Heart Failure

Cardiac remodeling is characterized by changes in molecular, cellular and interstitial properties of the heart, manifesting as changes in size, shape and function [82]. This includes both physiological and pathological adaptations [83] where Ca2+-mediated signaling plays a crucial role. Pathological cardiac remodeling is recognized as an early feature of the diseased heart and most often transitions to HF with the critical involvement of Ca2+-dependent proteins, most notably CaMKII and CaN [84,85,86]. Hence, substantial efforts have been made in the last years to prevent or halt disease progression by restoring impaired cellular Ca2+ handling and Ca2+-dependent signaling pathways [7,87]. Common triggers of cardiac remodeling involve neurohumoral stimulation, pressure and volume overload and electrical abnormalities leading to arrhythmogenesis. The majority of conventional therapeutic strategies, e.g., β-blockers or angiotensin-converting enzyme (ACE) inhibitors, are directed towards alleviating the effects of neurohormonal overactivation. Newer attempts in treating cardiac remodeling and HF have focused on CaMKII and CaN as therapeutic targets [88,89,90,91], however with no clinical success, which is commonly attributed to the lack of specificity and selectivity of the drugs applied [92]. Similarly, gene therapy approaches to restore physiological SR Ca2+ reuptake, although promising in preclinical models [93], have failed to produce results in HF patients [94,95]. Encouraging results, however, were published very recently by Lebek et al. showing that CRISPR-Cas9 base editing to ablate the oxidative activation sites of CaMKIIδ was effective in protecting the heart from ischemia-reperfusion damage in mouse models [96].
Therefore, in order to identify novel and relevant therapeutic targets, strategies and/or candidate pharmacotherapeutics against cardiac remodeling, an improvement of the current knowledge base regarding spatio-temporal Ca2+ signaling regulation and interplay is imperative.

4.1.1. Early Cardiac Remodeling

Structural and functional cardiomyocyte remodeling in end-stage HF has been assessed extensively during the last decades, and disturbed Ca2+ homeostasis is now considered a hallmark of the terminal disease phenotype. Growing evidence, however, suggests that disturbed Ca2+ handling, especially in the cell nucleus, may be an early event in myocardial remodeling and that it may be causally involved in the development of hypertrophy and HF.
We provided the first direct evidence linking structural and functional NE changes to altered nucleoplasmic Ca2+ handling in early cardiac remodeling [28]. We observed progressive decline in NE invagination density, increases in nuclei sizes and changes in Ca2+-regulatory protein expression patterns on and around the NE following 1 week of transverse aortic constriction (TAC). Decreased NE invagination density harbors important consequences on nuclear Ca2+ cycling as it results in slower nuclear CaT propagation (partly due to fewer and less deeply located NPCs) and reduced expression of Ca2+ handling proteins such as SERCA. Most importantly, alterations in nuclear CaTs occurred well before cytoplasmic CaTs were measurably affected and they could activate the nuclear CaMKII-HDAC4 axis [28]. In agreement with these findings, Shimojima et al. reported an increase in nuclear area, decrease in number of nuclear invaginations and increase in half-decay time of the nuclear CaTs in neonatal rat ventricular cardiomyocytes in response to potent hypertrophic stimuli such as Ang II, ET-1 and PE [97].
Our more recent work provided further insights into the specifics of early remodeling with respect to nuclear CaTs and the localization of active CaMKIIδC [25]. At the very early phase of cardiomyocyte response to TAC—day 5 post-intervention—we observed enhanced cytoplasmic and nucleoplasmic CaT amplitudes and faster [Ca2+]cyto and [Ca2+]nuc kinetics. The changes in nucleoplasmic CaTs were more prominent than the cytoplasmic ones, and associated with accumulation of activated CaMKIIδC on the NE. This initial phenotype of preferentially sped up nuclear CaTs prevented the rise of diastolic [Ca2+]nuc and the activation of hypertrophic signaling pathways mediated by IL6R. However, such adaptative response was of limited duration only before transitioning into maladaptive changes, first in the nucleus and then in the cytoplasm as well [25,98].
Transient enhancement of CaTs, especially in the nucleus, was also observed in young spontaneously hypertensive rats, a well-characterized model of ventricular hypertrophy due to systemic hypertension. It was associated with highly increased SERCA activity and resulted in enhanced nuclear Ca2+ signaling via the CaMKIIδ-HDAC4 axis [80].
Along with changes in nuclear CaTs, changes in active receptor-mediated regulation of nuclear Ca2+ handling have also been documented [28,99]. In mice that developed hypertrophy due to chronic Ang II infusion, significant increases in IP3R2 expression were found at the NE and associated with increased [Ca2+]nuc [44]. Strikingly, elevated nuclear Ca2+ levels persisted even 3 weeks after removal of the Ang II stimulus and were sufficient to keep CaN active, thereby further increasing IP3R2 expression. The proposed hypothesis is that elevation of [Ca2+]nuc through newly expressed IP3Rs would continue promoting a vicious cycle of CaN-NFAT pro-hypertrophic transcription [44].

4.1.2. Late Cardiac Remodeling

When pathological changes in cellular Ca2+ handling, gene expression, structure and function persist, they gradually start manifesting as cardiac dysfunction at organ level and eventually progress to the full-blown HF phenotype [82]. Simultaneously, Ca2+-dependent signaling via the CaN-NFAT-GATA4 and CaMKII-HDAC-MEF2 axes remains critically activated, promoting pathological remodeling by altering the phosphorylation status of respective upstream regulators and initiating their nuclear translocation [7,28,80].
We specifically investigated consequences of long-term cardiac remodeling due to pressure overload on nuclear Ca2+ homeostasis and showed that 6–7 weeks post-TAC, diastolic [Ca2+] rose, CaT amplitude decreased and the rate of [Ca2+] decline slowed down in both cytosol and nucleus [25,28] (Figure 2A). This may be—at least in part—driven by impaired SERCA function and increased RyR2s open probability [100], as well as significant reduction in SERCA expression versus increases in IP3R2 levels in nuclear protein fractions from failing human myocytes [28]. At the same time, persistent CaMKII overactivation was visible and abolished by CaMKII inhibitor KN-93. Consistent with chronic CaMKII activation being a causal factor in the development of the disturbed Ca2+ phenotype, cardiomyocytes isolated from 11–13 week old CaMKIIδC overexpressing transgenic mice showed signs and symptoms of HF and were characterized by highly elevated diastolic [Ca2+] and slowed CaT kinetics in both subcellular compartments [25].
Importantly, we could determine distinct spatio-temporal profiles of CaMKII activation in early and late remodeling. Namely, in contrast to CaMKII accumulation on the NE observed in early remodeling which showed to be protective, in cardiomyocytes isolated from late TAC (6–7 weeks after surgery), CaMKIIδC translocated into the nucleus and promoted HDAC4 export [25]. Indeed, CaMKII mobility and its nuclear import are potentiated by increased intracellular Ca2+ levels and CaMKII autophosphorylation [101]. Translation of these findings into human myocardium revealed a large increase of CaMKIIδC expression in failing hearts versus non-failing controls with an especially strong rise in CaMKIIδC in the nuclear fraction. This is consistent with the idea that once CaMKIIδC starts accumulating inside the nucleus, it drives maladaptive transcriptional effects and eccentric hypertrophy [25]. Of note, prolonged CaMKIIδC activation is also observed in response to neurohormonal stimulation via ET-1, Ang II or α-/β-AR agonists (reviewed in [102]); however, the specific contribution of nuclear Ca2+ disbalance in these settings remains to be investigated.
In our latest work, we could demonstrate that defective mitochondrial Ca2+ uptake typically observed in cardiac remodeling and HF also contributes to increased diastolic [Ca2+]nuc in cardiomyocytes [21]. In particular, we demonstrated that perinuclear mitochondria from failing cardiomyocytes are more susceptible to depolarization of mitochondrial membrane potential, reactive oxygen species generation and impairment in Ca2+ uptake compared with intrafibrillar mitochondria at baseline and under physiological stress protocol. This may partially explain a disproportionate rise in [Ca2+]nuc compared to [Ca2+]cyto in failing cardiomyocytes at increased stimulation frequencies. On the other hand, upregulation of the mitochondrial calcium uniporter (MCU) under stress conditions elicits protective effects against cardiomyocyte death from Ca2+ overload [103]. MCU upregulation was capable of maintaining intracellular Ca2+ and energy homeostasis and thereby limited cardiac hypertrophy and dysfunction in response to chronic β-AR stimulation. Mechanistically, β-AR stimulation activates the nuclear CaMKIIδ variant, CaMKIIδB, which upregulates MCU gene transcription via the phosphorylation of CREB. Therefore, targeting the CaMKIIδB-CREB-MCU axis may present a novel approach in alleviating chronic stress-induced alterations in Ca2+ cycling and pathological cardiac remodeling [103]. Furthermore, Liu et al. recently showed in a guinea pig model of HF and sudden cardiac death that moderate MCU overexpression even has beneficial effects that persist weeks after the onset of HF and potently reverses the course of cardiac decompensation and arrhythmogenesis [104]. Ultimately, studying the interplay between (perinuclear) mitochondria, nuclear Ca2+ and downstream transcriptional regulation presents a promising and exciting field for future research and the development of new therapeutic options.
Finally, several reports describe NPC rearrangements and changes in nuclear trafficking in animal models and human failing cardiomyocytes and indicate their potential for therapeutic intervention [105,106]. By examining the properties of nuclear importins and exportins, such as expression and transportation magnitude and rate, Chanine et al. have found a reduction in nuclear import versus export, as well as changes in cell and nuclear size, reversible by leptomycin B [105]. We can speculate that changes in NE structure and cargo transport mechanisms may also translate into changes in nuclear Ca2+ handling and its downstream signaling; however, the experimental evidence is yet to be provided.
The complex dysregulation of nuclear Ca2+ homeostasis during all stages of cardiac remodeling, manifested through transcriptional and functional alterations, highlights the importance of [Ca2+]nuc as a clinically relevant parameter and potential therapeutic target for HF patients.
Figure 2. Averaged original recordings of nucleoplasmic (red) and cytoplasmic (black) Ca2+ transients from isolated cardiomyocytes. (A) Averaged nucleoplasmic (red) and cytoplasmic (black) Ca2+ transients of 1 Hz-stimulated isolated cardiomyocytes from sham-operated control mice (grey panel) and mice 7 weeks after transverse aortic constriction (TAC) intervention to induce pressure-overload mediated hypertrophy and heart failure (pink panel). TAC increased both cytoplasmic and nuclear diastolic Ca2+ concentration ([Ca2+]cyto and [Ca2+]nuc, respectively), decreased Ca2+ transient amplitude and prolonged time to peak and time from peak [Ca2+] to 50% decline. (B) Averaged nucleoplasmic (red) and cytoplasmic (black) Ca2+ transients of 1 Hz-stimulated isolated canine atrial cardiomyocytes from healthy controls (grey panel) and dogs subject to 1 week of atrial fibrillation pacing protocol (600 bpm) prior isolation (pink panel). [Ca2+]nuc averages 55% higher in AF vs. CTL with reduced nucleoplasmic Ca2+ transient amplitude and significantly increased time to peak and time from peak [Ca2+]nuc to 50% decline. Adapted with permission from Refs. [28,107]. 2023, Wolters Kluwer Health, Inc., Philadelphia, USA.
Figure 2. Averaged original recordings of nucleoplasmic (red) and cytoplasmic (black) Ca2+ transients from isolated cardiomyocytes. (A) Averaged nucleoplasmic (red) and cytoplasmic (black) Ca2+ transients of 1 Hz-stimulated isolated cardiomyocytes from sham-operated control mice (grey panel) and mice 7 weeks after transverse aortic constriction (TAC) intervention to induce pressure-overload mediated hypertrophy and heart failure (pink panel). TAC increased both cytoplasmic and nuclear diastolic Ca2+ concentration ([Ca2+]cyto and [Ca2+]nuc, respectively), decreased Ca2+ transient amplitude and prolonged time to peak and time from peak [Ca2+] to 50% decline. (B) Averaged nucleoplasmic (red) and cytoplasmic (black) Ca2+ transients of 1 Hz-stimulated isolated canine atrial cardiomyocytes from healthy controls (grey panel) and dogs subject to 1 week of atrial fibrillation pacing protocol (600 bpm) prior isolation (pink panel). [Ca2+]nuc averages 55% higher in AF vs. CTL with reduced nucleoplasmic Ca2+ transient amplitude and significantly increased time to peak and time from peak [Ca2+]nuc to 50% decline. Adapted with permission from Refs. [28,107]. 2023, Wolters Kluwer Health, Inc., Philadelphia, USA.
Biomedicines 11 00960 g002

4.2. Nuclear Ca2+ in Atrial Fibrillation

While ventricular and atrial cardiomyocytes share functional and structural commonalities in the form of ion handling and ECC, there are distinct tissue-specific differences in distribution of ion channel expression and thus electrophysiological properties. AF is the most common sustained cardiac arrhythmia with a lifetime risk of around 20% [108] and substantial health consequences. The propensity towards ectopic firing and re-entry, two key AF features, is aggravated by ion channel dysfunction, Ca2+ handling abnormalities, structural remodeling and autonomic neural dysfunction [109]. An increase in global average life expectancy, survival rate and risk factor exposure, coupled with the absence of optimal treatment strategies and continuous rise of AF incidence and prevalence, fundamentally underlie the high socio-economic burden of AF [110]. Similar to HF, most of the current research efforts are devoted to unraveling the mechanisms of global Ca2+ handling in AF and fail to consider [Ca2+]nuc and its downstream transcriptional targets as contributors to atrial arrhythmogenesis and contractile dysfunction.
The first study that specifically focused on nuclear Ca2+ alterations in atrial cardiomyocytes during AF was published only recently [107]. Isolated atrial cardiomyocytes from AF mongrel dogs showed nuclear enlargement and loss of NE invaginations, which was associated with reduced amplitude of nuclear CaTs and higher diastolic [Ca2+]nuc in AF samples, mediated by slower nuclear [Ca2+] decay kinetics (Figure 2B). Furthermore, even resting [Ca2+]nuc averaged 55% higher in AF vs. CTL cardiomyocytes, indicating a true accumulation of nuclear Ca2+ in AF. Expression patterns of Ca2+-regulating proteins were disturbed in AF, with the most striking increase in IP3R levels. Namely, IP3R1 expression increased upon AF on the NE and in non-nuclear compartments, while the smaller increase in IP3R2 expression was restricted to non-nuclear compartments. Importantly, IP3R-dependent Ca2+ mobilization from the NE was identified as an important driver of the observed changes in nuclear Ca2+ homeostasis, as IP3R inhibition resulted in a substantial return of nucleoplasmic CaTs to near control levels. Further, siRNA-mediated IP3R-knockdown in canine atrial cardiomyocytes largely diminished diastolic nuclear Ca2+ increases caused by in vitro tachypacing [107]. IP3R1 dysregulation emerged earlier as a key upstream factor decreasing LTCC current, a functional hallmark of AF [111]. Accumulation of [Ca2+]nuc correlated with increased nuclear CaMKII autophosphorylation and HDAC4 export, which was reversed upon IP3R-knockdown or CaMKII inhibition [107]. Finally, dysregulation of IP3R1 and thus nuclear Ca2+ handling in AF was accompanied with strong downregulation of micro-RNA 26, which may represent an exciting new target for future therapeutic interventions [107].
The observed changes in AF also harbor detrimental consequences for ventricular function and cardiomyocyte Ca2+ homeostasis, as evidenced in human left ventricular tissue from AF patients and induced pluripotent stem cell (iPSC)-derived cardiomyocytes under AF stimulation protocol [112]. In agreement with previously discussed studies, disturbed Ca2+ handling due to AF-simulating pacing protocol in iPSC-CMs induced CaMKII activation [112], which is likely to further promote a vicious cycle of Ca2+ dysregulation [25]. Thus, restoring (nuclear) Ca2+ handling in AF emerges as an exciting new area of research and it may offer strategies to prevent the potential development of both atrial and ventricular dysfunction in AF patients.

4.3. Nuclear Ca2+ in Familial Cardiomyopathy

Familial cardiomyopathy accounts for 30% to 50% of all dilated cardiomyopathy cases [113]. Among the genetic causes of familial cardiomyopathy numerous mutations in the structural proteins found in the nucleus, such as lamins and emerin have been identified [114,115,116]. These proteins are important for maintaining NE integrity, chromatin organization and regulation of gene transcription [116]. Mutations in these genes cause arrhythmogenic behavior, which was previously tied to disturbed Ca2+ handling and CaMKII [117,118]. Importantly, recent work showed that siRNA-mediated knockdown of emerin encoding gene emd—ultimately causing X-linked Emery-Dreifuss muscular dystrophy (X-EDMD)—was associated with nuclear Ca2+ dysregulation in neonatal rat ventricular cardiomyocytes. Likely provoked by the observed decrease in nuclear invagination incidence and thus disturbed Ca2+ pump back function into the NE, increased half-decay time of the nuclear CaT may contribute to the initiation of hypertrophic gene program activation and cardiac remodeling through prolonged high [Ca2+]nuc [97].
Disturbed cytosolic Ca2+ handling was studied more extensively and it was documented in cardiomyocytes with mutations in sarcomeric and ion handling genes such as TNNT (encoding troponin; [119]) and PLN (encoding PLB; [120]), while mutations in the splicing factor RNA binding motif 20 (RBM20), which controls alternate splicing of CaMKIIδ and titin, lead to an increase in cytoplasmic diastolic Ca2+, peak transient amplitude, increased SR load and increased frequency of spontaneous Ca2+ release in the cytosol [121]. Indeed, due to specific PLB accumulation around the nucleus [19,35] and CaMKIIδ translocation to the nuclear compartment over the course of cardiac remodeling [25,101], it is tempting to speculate that these mutations would have even stronger and/or earlier effects on nucleoplasmic Ca2+ levels and their regulation—yet experimental evidence needs to be provided in future.

5. Nuclear Ca2+: A Practical Approach

Reliable and accurate quantification of subcellular Ca2+ signals in cardiomyocytes is essential for assessing and understanding compartmentalized Ca2+ fluxes and their roles in ECC and ETC [122]. For successful experimental outcomes, an important aspect to consider is the choice of appropriate Ca2+ indicator and experimental model for acquiring signals in the subcellular compartment of interest.

5.1. Ca2+ Indicators

Detection of [Ca2+] changes in different cellular spaces is directly linked to Ca2+ binding affinities of fluorescent Ca2+ dyes, reflected by the dissociation constant (Kd). Kd allows an estimate of the detectable [Ca2+] range and should generally be near the midpoint of the expected [Ca2+] fluctuations [123]. Thus, low affinity Ca2+ indicators (e.g., Fluo-5N, Mag-fluo 4) are typically used for the visualization of [Ca2+] changes in the SR or NE and high affinity Ca2+ indicators (e.g., Fluo-3, Fluo-4, Fluo-8) for measuring changes in the cytosolic and nucleoplasmic free [Ca2+] [124,125,126,127]. Although Fluo-3 and Fluo-4 are the most commonly used Ca2+ indicators in cardiomyocytes, the emerging chemical indicator Calbryte-520 (or Cal-520) showed superior performance in intracellular retention, brightness and fluorescence intensity in CHO-K1 cell line [128]. Cal-520 was also found suitable for drug-screening purposes in iPSC-CM [129]; however, analyses were limited to cytosolic CaTs only.
Despite being an invaluable research tool since their discovery, chemical fluorescent Ca2+ indicators have several technical shortcomings: (1) their Ca2+ binding affinities and fluorescent properties vary between cytosol and nucleus, (2) they may be differentially sequestered into intracellular organelles or (3) they may leak from the cytoplasm to the extracellular medium via sarcolemmal anion transporters [32,130]. To overcome these challenges, substantial efforts have been made to transform raw fluorescence signals into calibrated [Ca2+] by determining in situ calibration curves in different types of cardiomyocytes isolated from different experimental animal models. Such curves take into account the effects of a particular cellular environment on indicator properties and allow for a direct comparison between [Ca2+] in different subcellular spaces [32,44,80,107,125]. An additional hurdle to consider is that cardiac remodeling processes may impact Kd, meaning that in some cases, signal calibration has to be performed independently for control and experimental groups [107]. Apart from the technical shortcomings one should keep in mind that Ca2+ fluorophores exhibit detrimental physiological effects on sodium-potassium ATPase (NKA) activity, cell viability and metabolic status [131], which can further complicate their use.
In contrast, genetically encoded fluorescent Ca2+ indicators (GECIs) interfere only minimally with NKA activity [131] and they have a big advantage with regard to their specific targeting to cellular compartments or organelles of interest via specific promoters and targeting sequences. GCaMP, one of the most successful and popular type of GECIs, has been continuously improved and updated to ultimately provide rapid response kinetics and powerful signal-to-noise ratios in subcellular Ca2+ imaging [132,133]. However, the CaM component in GCaMPs serving as the Ca2+ sensor element raised concerns regarding its use, as it was found to interfere with the gating of LTCCs in neurons leading to chronic nuclear Ca2+ accumulation and transcription dysregulation [134]. Similar effects on cardiac LTCCs have not been reported but may be prevented by modulating the CaM motif [134]. Another member of the GCaMP family, GCaMP6s, was evaluated as knockin in hPSC-CMs and successfully visualized isoprenaline-induced alterations in CaT kinetics [135]. Addition of a nuclear localization sequence, as in the case of jGCaMP7, allows highly efficient nuclear-specific targeting and thus real-time nuclear CaT acquisition as demonstrated in a mouse fibroblast line [136].
Promising progress has been recently made by combining newly designed MaPCa dyes—highly permeable rhodamine-based Ca2+ indicators of varying affinities and colors—with HaloTag fusion proteins targeted to specific subcellular localizations and compatible with both fluorescence and bioluminescence readouts [137]. In the first Ca2+ imaging experiments, AM esters of the MaPCa indicators were applied to co-cultures of 293 cells stably expressing a nuclear-localized HaloTag and nonexpressing 293 cells, demonstrating great power of the HaloTag expressing cells in detecting nuclear Ca2+ fluxes with excellent signal-to-background ratios [137]. Importantly, these indicators can be excited in the far-red range, thus enabling a more precise and detailed analysis of the Ca2+ dynamics in cardiomyocytes with special regard to the inter-compartmental crosstalk.

5.2. Emerging Models, Tools and Future Perspectives

Nuclear Ca2+ fluxes have been mostly monitored in cardiomyocytes from experimental animal models. While these models are necessary for in vivo and in vitro analyses of cardiac function in cardiac (patho)physiology, they differ fundamentally from humans. As such, results derived from preclinical studies, particularly in rodents, are not always transposable to humans. On the other hand, human cardiac tissue is either obtained from organ donors or as surgical biopsies [28,107,138,139], thus sample availability is relatively limited. With an increasing need for more “translatable” but at the same time readily accessible in vitro models, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have quickly proven invaluable. They reproduce many of the human cardiomyocyte characteristics such as Ca2+ cycling, contractility and response to exogenous stimuli, key sarcomeric proteins and Ca2+ signaling protein expression [140,141,142,143,144,145,146] and are—importantly—emerging as a tool for studying nuclear Ca2+ signals [147]. Furthermore, hiPSC-CMs can be derived from patients with preexisting genetic mutations influencing nuclear structure and consequently Ca2+ handling, such as lamin A/C cardiomyopathy (LMNA-DCM) [148], X-EDMD [149], but also mutations in CaMKIIδ splicing factor RBM20 [121].
The remaining downside of using hiPSC-CMs, however, is their incomplete maturation. To overcome this, advances in tissue engineering have enabled the development of novel two- or three-dimensional cardiovascular tissue models, namely cardiac organoids/microtissues and engineered heart tissue with superior cardiomyocyte maturation phenotypes [150,151]. However, detection of nuclear CaTs in such models is limited due to lack of standard digestion protocols that will consistently recover single cardiomyocytes from 3D structures with a high cellular yield and small penetration depth of confocal imaging systems. To surmount the challenges, Richards et al. developed a two-component imaging system able to track single cell CaTs from GCaMP6 labeled hiPSC-CMs within a microtissue model of cardiac toxicity. They took advantage of the increased scanning depth of two-photon excitation microscopy and the increased resolution of light-sheet microscopy systems equipped with a high-speed camera to record subcellular Ca2+ events [152]. Despite the current limitations, improving the isolation of single cardiomyocytes from cardiac organoids, as well as advancing technology for visualization of nuclear CaTs in intact microtissues holds a great promise for future work.
To our knowledge, two genetically encoded Ca2+ chelating proteins for selectively modulating (peri)nuclear [Ca2+] have been successfully used in isolated cardiomyocytes in the past [10,153]. In work by Higazi et al., nuclear Ca2+ was specifically buffered using a nuclear-targeted, red fluorescent protein-tagged form of the neuronal Ca2+ binding protein calbindin, demonstrating that increased nuclear Ca2+ levels are required for the induction of atrial natriuretic factor expression [153]. Turcotte et al. used mCherry-tagged parvalbumin β-nesprin fusion protein designed to buffer Ca2+ in the perinuclear space and showed that buffering perinuclear Ca2+ completely inhibits isoprenaline-induced perinuclear CaN activation in both neonatal and adult myocytes [10]. Further optimization and/or knockin of such nuclear Ca2+ regulatory proteins can be of great value for studying nucleus-restricted Ca2+ dynamics.

6. Conclusions

In this review, we demonstrate that nuclear Ca2+ handling—although not as extensively studied as an independent parameter of cardiac remodeling—significantly complements the knowledge on cytoplasmic Ca2+ regulation and alterations, and it augments the likelihood of mitigating pathological cardiac remodeling in the coming years. Future technological breakthroughs in the field of subcellular Ca2+ quantification, cardiac disease modeling and small molecule generation will surely help to better describe remodeling mechanisms and prevent or even halt cardiac disease progression. Understanding nuclear Ca2+ regulation will undoubtedly play an important role in these efforts, as Ca2+ cycling in the nucleus may be the “mastermind” behind the initiation and progression of various cardiac diseases [154].

Author Contributions

Conceptualization, M.K. and S.L.-H.; writing—original draft preparation, M.K., N.D. and S.L.-H.; writing—review and editing, M.K., N.D., J.V. and S.L.-H.; visualization, M.K. and S.L.-H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by BioTechMed-Graz, Young Researcher Groups (YRG) and the Austrian Science Fund (FWF), V-530 to S.L.-H. and NextGenerationEU (NGEU)—“MSCA Seal of Excellence @UNIPD” (Padova, IT) to N.D.; M.K. and J.V. received no external funding. M.K. and J.V. are currently trained as PhD candidates in the Program Molecular Medicine at the Medical University of Graz, Austria. For the purpose of open access, the author has applied a CC BY public copyright license to any Author Accepted Manuscript version arising from this submission.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Open Access Funding by the Austrian Science Fund (FWF).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Berridge, M.J.; Lipp, P.; Bootman, M.D. The versatility and universality of calcium signalling. Nat. Rev. Mol. Cell Biol. 2000, 1, 11–21. [Google Scholar] [CrossRef]
  2. Bers, D. Excitation-Contraction Coupling and Cardiac Contractile Force; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2001; Volume 237. [Google Scholar]
  3. Bers, D.M.; Guo, T. Calcium signaling in cardiac ventricular myocytes. Ann. N. Y. Acad. Sci. 2005, 1047, 86–98. [Google Scholar] [CrossRef]
  4. Kockskämper, J. Excitation–Contraction Coupling of Cardiomyocytes. In Cardiomyocytes–Active Players in Cardiac Disease; Schlüter, K.-D., Ed.; Springer International Publishing: Cham, Switzerland, 2016; pp. 67–96. [Google Scholar]
  5. Bers, D.M. Calcium cycling and signaling in cardiac myocytes. Annu. Rev. Physiol. 2008, 70, 23–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Frey, N.; McKinsey, T.A.; Olson, E.N. Decoding calcium signals involved in cardiac growth and function. Nat. Med. 2000, 6, 1221–1227. [Google Scholar] [CrossRef] [PubMed]
  7. Dewenter, M.; von der Lieth, A.; Katus, H.A.; Backs, J. Calcium Signaling and Transcriptional Regulation in Cardiomyocytes. Circ. Res. 2017, 121, 1000–1020. [Google Scholar] [CrossRef]
  8. Shaw, R.M.; Colecraft, H.M. L-type calcium channel targeting and local signalling in cardiac myocytes. Cardiovasc. Res. 2013, 98, 177–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Eisner, D.A.; Caldwell, J.L.; Kistamás, K.; Trafford, A.W. Calcium and Excitation-Contraction Coupling in the Heart. Circ. Res. 2017, 121, 181–195. [Google Scholar] [CrossRef]
  10. Turcotte, M.G.; Thakur, H.; Kapiloff, M.S.; Dodge-Kafka, K.L. A perinuclear calcium compartment regulates cardiac myocyte hypertrophy. J. Mol. Cell. Cardiol. 2022, 172, 26–40. [Google Scholar] [CrossRef]
  11. Tanaka, H.; Kawanishi, T.; Matsuda, T.; Takahashi, M.; Shigenobu, K. Intracellular free Ca2+ movements in cultured cardiac myocytes as shown by rapid scanning confocal microscopy. J. Cardiovasc. Pharmacol. 1996, 27, 761–769. [Google Scholar] [CrossRef]
  12. Winogradoff, D.; Chou, H.Y.; Maffeo, C.; Aksimentiev, A. Percolation transition prescribes protein size-specific barrier to passive transport through the nuclear pore complex. Nat. Commun. 2022, 13, 5138. [Google Scholar] [CrossRef]
  13. Mohr, D.; Frey, S.; Fischer, T.; Güttler, T.; Görlich, D. Characterisation of the passive permeability barrier of nuclear pore complexes. EMBO J. 2009, 28, 2541–2553. [Google Scholar] [CrossRef] [Green Version]
  14. Panté, N.; Kann, M. Nuclear pore complex is able to transport macromolecules with diameters of about 39 nm. Mol. Biol. Cell 2002, 13, 425–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wu, X.; Bers, D.M. Sarcoplasmic reticulum and nuclear envelope are one highly interconnected Ca2+ store throughout cardiac myocyte. Circ. Res. 2006, 99, 283–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Malhas, A.; Goulbourne, C.; Vaux, D.J. The nucleoplasmic reticulum: Form and function. Trends Cell Biol. 2011, 21, 362–373. [Google Scholar] [CrossRef] [PubMed]
  17. Guatimosim, S.; Amaya, M.J.; Guerra, M.T.; Aguiar, C.J.; Goes, A.M.; Gómez-Viquez, N.L.; Rodrigues, M.A.; Gomes, D.A.; Martins-Cruz, J.; Lederer, W.J.; et al. Nuclear Ca2+ regulates cardiomyocyte function. Cell Calcium 2008, 44, 230–242. [Google Scholar] [CrossRef]
  18. Gensburger, C.; Freyermuth, S.; Klein, C.; Malviya, A.N. In vivo nuclear Ca2+-ATPase phosphorylation triggers intermediate size molecular transport to the nucleus. Biochem. Biophys. Res. Commun. 2003, 303, 1225–1228. [Google Scholar] [CrossRef]
  19. Wu, A.Z.; Xu, D.; Yang, N.; Lin, S.F.; Chen, P.S.; Cala, S.E.; Chen, Z. Phospholamban is concentrated in the nuclear envelope of cardiomyocytes and involved in perinuclear/nuclear calcium handling. J. Mol. Cell. Cardiol. 2016, 100, 1–8. [Google Scholar] [CrossRef]
  20. Ledeen, R.W.; Wu, G. Sodium-calcium exchangers in the nucleus: An unexpected locus and an unusual regulatory mechanism. Ann. N. Y. Acad. Sci. 2007, 1099, 494–506. [Google Scholar] [CrossRef]
  21. Voglhuber, J.; Holzer, M.; Radulović, S.; Thai, P.N.; Djalinac, N.; Matzer, I.; Wallner, M.; Bugger, H.; Zirlik, A.; Leitinger, G.; et al. Functional remodelling of perinuclear mitochondria alters nucleoplasmic Ca2+ signalling in heart failure. Philos. Trans. R. Soc. Lond B Biol. Sci. 2022, 377, 20210320. [Google Scholar] [CrossRef]
  22. Lee, S.H.; Hadipour-Lakmehsari, S.; Miyake, T.; Gramolini, A.O. Three-dimensional imaging reveals endo(sarco)plasmic reticulum-containing invaginations within the nucleoplasm of muscle. Am. J. Physiol. Cell Physiol. 2018, 314, C257–C267. [Google Scholar] [CrossRef] [Green Version]
  23. Lu, X.; Thai, P.N.; Lu, S.; Pu, J.; Bers, D.M. Intrafibrillar and perinuclear mitochondrial heterogeneity in adult cardiac myocytes. J. Mol. Cell. Cardiol. 2019, 136, 72–84. [Google Scholar] [CrossRef] [PubMed]
  24. Kiess, T.O.; Kockskämper, J. SERCA Activity Controls the Systolic Calcium Increase in the Nucleus of Cardiac Myocytes. Front. Physiol. 2019, 10, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ljubojevic-Holzer, S.; Herren, A.W.; Djalinac, N.; Voglhuber, J.; Morotti, S.; Holzer, M.; Wood, B.M.; Abdellatif, M.; Matzer, I.; Sacherer, M.; et al. CaMKIIδC Drives Early Adaptive Ca2+ Change and Late Eccentric Cardiac Hypertrophy. Circ. Res. 2020, 127, 1159–1178. [Google Scholar] [CrossRef] [PubMed]
  26. Bare, D.J.; Kettlun, C.S.; Liang, M.; Bers, D.M.; Mignery, G.A. Cardiac type 2 inositol 1,4,5-trisphosphate receptor: Interaction and modulation by calcium/calmodulin-dependent protein kinase II. J. Biol. Chem. 2005, 280, 15912–15920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Escobar, M.; Cardenas, C.; Colavita, K.; Petrenko, N.B.; Franzini-Armstrong, C. Structural evidence for perinuclear calcium microdomains in cardiac myocytes. J. Mol. Cell. Cardiol. 2011, 50, 451–459. [Google Scholar] [CrossRef]
  28. Ljubojevic, S.; Radulovic, S.; Leitinger, G.; Sedej, S.; Sacherer, M.; Holzer, M.; Winkler, C.; Pritz, E.; Mittler, T.; Schmidt, A.; et al. Early remodeling of perinuclear Ca2+ stores and nucleoplasmic Ca2+ signaling during the development of hypertrophy and heart failure. Circulation 2014, 130, 244–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Ljubojevic, S.; Bers, D.M. Nuclear calcium in cardiac myocytes. J. Cardiovasc. Pharmacol. 2015, 65, 211–217. [Google Scholar] [CrossRef] [Green Version]
  30. Shahin, V.; Danker, T.; Enss, K.; Ossig, R.; Oberleithner, H. Evidence for Ca2+- and ATP-sensitive peripheral channels in nuclear pore complexes. Faseb. J. 2001, 15, 1895–1901. [Google Scholar] [CrossRef] [Green Version]
  31. Martins, T.V.; Evans, M.J.; Wysham, D.B.; Morris, R.J. Nuclear pores enable sustained perinuclear calcium oscillations. BMC Syst. Biol. 2016, 10, 55. [Google Scholar] [CrossRef] [Green Version]
  32. Ljubojević, S.; Walther, S.; Asgarzoei, M.; Sedej, S.; Pieske, B.; Kockskämper, J. In situ calibration of nucleoplasmic versus cytoplasmic Ca2+ concentration in adult cardiomyocytes. Biophys. J. 2011, 100, 2356–2366. [Google Scholar] [CrossRef] [Green Version]
  33. Lipp, P.; Thomas, D.; Berridge, M.J.; Bootman, M.D. Nuclear calcium signalling by individual cytoplasmic calcium puffs. EMBO J. 1997, 16, 7166–7173. [Google Scholar] [CrossRef] [Green Version]
  34. Chen, M.; Xu, D.; Wu, A.Z.; Kranias, E.; Lin, S.F.; Chen, P.S.; Chen, Z. Phospholamban regulates nuclear Ca2+ stores and inositol 1,4,5-trisphosphate mediated nuclear Ca2+ cycling in cardiomyocytes. J. Mol. Cell. Cardiol. 2018, 123, 185–197. [Google Scholar] [CrossRef] [PubMed]
  35. He, W.; Huang, D.; Guo, S.; Wang, D.; Guo, J.; Cala, S.E.; Chen, Z. Association with SERCA2a directs phospholamban trafficking to sarcoplasmic reticulum from a nuclear envelope pool. J. Mol. Cell. Cardiol. 2020, 143, 107–119. [Google Scholar] [CrossRef] [PubMed]
  36. Jin, X.; Amoni, M.; Gilbert, G.; Dries, E.; Doñate Puertas, R.; Tomar, A.; Nagaraju, C.K.; Pradhan, A.; Yule, D.I.; Martens, T.; et al. InsP(3)R-RyR Ca2+ channel crosstalk facilitates arrhythmias in the failing human ventricle. Basic Res. Cardiol. 2022, 117, 60. [Google Scholar] [CrossRef] [PubMed]
  37. Hohendanner, F.; Maxwell, J.T.; Blatter, L.A. Cytosolic and nuclear calcium signaling in atrial myocytes: IP3-mediated calcium release and the role of mitochondria. Channels 2015, 9, 129–138. [Google Scholar] [CrossRef] [Green Version]
  38. Kim, J.C.; Son, M.J.; Le, Q.A.; Woo, S.H. Role of inositol 1,4,5-trisphosphate receptor type 1 in ATP-induced nuclear Ca2+ signal and hypertrophy in atrial myocytes. Biochem. Biophys. Res. Commun. 2018, 503, 2998–3002. [Google Scholar] [CrossRef]
  39. Garcia, M.I.; Karlstaedt, A.; Chen, J.J.; Amione-Guerra, J.; Youker, K.A.; Taegtmeyer, H.; Boehning, D. Functionally redundant control of cardiac hypertrophic signaling by inositol 1,4,5-trisphosphate receptors. J. Mol. Cell. Cardiol. 2017, 112, 95–103. [Google Scholar] [CrossRef]
  40. Wu, X.; Zhang, T.; Bossuyt, J.; Li, X.; McKinsey, T.A.; Dedman, J.R.; Olson, E.N.; Chen, J.; Brown, J.H.; Bers, D.M. Local InsP3-dependent perinuclear Ca2+ signaling in cardiac myocyte excitation-transcription coupling. J. Clin. Investig. 2006, 116, 675–682. [Google Scholar] [CrossRef] [PubMed]
  41. Tadevosyan, A.; Maguy, A.; Villeneuve, L.R.; Babin, J.; Bonnefoy, A.; Allen, B.G.; Nattel, S. Nuclear-delimited angiotensin receptor-mediated signaling regulates cardiomyocyte gene expression. J. Biol. Chem. 2010, 285, 22338–22349. [Google Scholar] [CrossRef] [Green Version]
  42. Ibarra, C.; Vicencio, J.M.; Estrada, M.; Lin, Y.; Rocco, P.; Rebellato, P.; Munoz, J.P.; Garcia-Prieto, J.; Quest, A.F.; Chiong, M.; et al. Local control of nuclear calcium signaling in cardiac myocytes by perinuclear microdomains of sarcolemmal insulin-like growth factor 1 receptors. Circ. Res. 2013, 112, 236–245. [Google Scholar] [CrossRef] [Green Version]
  43. Arantes, L.A.; Aguiar, C.J.; Amaya, M.J.; Figueiró, N.C.; Andrade, L.M.; Rocha-Resende, C.; Resende, R.R.; Franchini, K.G.; Guatimosim, S.; Leite, M.F. Nuclear inositol 1,4,5-trisphosphate is a necessary and conserved signal for the induction of both pathological and physiological cardiomyocyte hypertrophy. J. Mol. Cell. Cardiol. 2012, 53, 475–486. [Google Scholar] [CrossRef]
  44. Olivares-Florez, S.; Czolbe, M.; Riediger, F.; Seidlmayer, L.; Williams, T.; Nordbeck, P.; Strasen, J.; Glocker, C.; Jänsch, M.; Eder-Negrin, P.; et al. Nuclear calcineurin is a sensor for detecting Ca2+ release from the nuclear envelope via IP(3)R. J. Mol. Med. 2018, 96, 1239–1249. [Google Scholar] [CrossRef]
  45. Ruiz-Hurtado, G.; Morel, E.; Domínguez-Rodríguez, A.; Llach, A.; Lezoualc’h, F.; Benitah, J.P.; Gomez, A.M. Epac in cardiac calcium signaling. J. Mol. Cell. Cardiol. 2013, 58, 162–171. [Google Scholar] [CrossRef] [PubMed]
  46. Zima, A.V.; Bare, D.J.; Mignery, G.A.; Blatter, L.A. IP3-dependent nuclear Ca2+ signalling in the mammalian heart. J. Physiol. 2007, 584, 601–611. [Google Scholar] [CrossRef] [PubMed]
  47. Luo, D.; Yang, D.; Lan, X.; Li, K.; Li, X.; Chen, J.; Zhang, Y.; Xiao, R.P.; Han, Q.; Cheng, H. Nuclear Ca2+ sparks and waves mediated by inositol 1,4,5-trisphosphate receptors in neonatal rat cardiomyocytes. Cell Calcium 2008, 43, 165–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Kockskämper, J.; Seidlmayer, L.; Walther, S.; Hellenkamp, K.; Maier, L.S.; Pieske, B. Endothelin-1 enhances nuclear Ca2+ transients in atrial myocytes through Ins(1,4,5)P3-dependent Ca2+ release from perinuclear Ca2+ stores. J. Cell Sci. 2008, 121, 186–195. [Google Scholar] [CrossRef] [Green Version]
  49. Pereira, L.; Ruiz-Hurtado, G.; Morel, E.; Laurent, A.C.; Métrich, M.; Domínguez-Rodríguez, A.; Lauton-Santos, S.; Lucas, A.; Benitah, J.P.; Bers, D.M.; et al. Epac enhances excitation-transcription coupling in cardiac myocytes. J. Mol. Cell. Cardiol. 2012, 52, 283–291. [Google Scholar] [CrossRef] [Green Version]
  50. Nakao, S.; Wakabayashi, S.; Nakamura, T.Y. Stimulus-dependent regulation of nuclear Ca2+ signaling in cardiomyocytes: A role of neuronal calcium sensor-1. PLoS ONE 2015, 10, e0125050. [Google Scholar] [CrossRef] [Green Version]
  51. Nakamura, T.Y.; Jeromin, A.; Mikoshiba, K.; Wakabayashi, S. Neuronal calcium sensor-1 promotes immature heart function and hypertrophy by enhancing Ca2+ signals. Circ. Res. 2011, 109, 512–523. [Google Scholar] [CrossRef] [Green Version]
  52. Wu, S.C.; Dahl, E.F.; Wright, C.D.; Cypher, A.L.; Healy, C.L.; O’Connell, T.D. Nuclear localization of a1A-adrenergic receptors is required for signaling in cardiac myocytes: An “inside-out” a1-AR signaling pathway. J. Am. Heart Assoc. 2014, 3, e000145. [Google Scholar] [CrossRef] [Green Version]
  53. Wright, C.D.; Chen, Q.; Baye, N.L.; Huang, Y.; Healy, C.L.; Kasinathan, S.; O’Connell, T.D. Nuclear alpha1-adrenergic receptors signal activated ERK localization to caveolae in adult cardiac myocytes. Circ. Res. 2008, 103, 992–1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Huang, Y.; Wright, C.D.; Merkwan, C.L.; Baye, N.L.; Liang, Q.; Simpson, P.C.; O’Connell, T.D. An alpha1A-adrenergic-extracellular signal-regulated kinase survival signaling pathway in cardiac myocytes. Circulation 2007, 115, 763–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wright, C.D.; Wu, S.C.; Dahl, E.F.; Sazama, A.J.; O’Connell, T.D. Nuclear localization drives α1-adrenergic receptor oligomerization and signaling in cardiac myocytes. Cell. Signal. 2012, 24, 794–802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Martin, R.D.; Sun, Y.; Bourque, K.; Audet, N.; Inoue, A.; Tanny, J.C.; Hébert, T.E. Receptor- and cellular compartment-specific activation of the cAMP/PKA pathway by α(1)-adrenergic and ETA endothelin receptors. Cell. Signal. 2018, 44, 43–50. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, J.; Gareri, C.; Rockman, H.A. G-Protein-Coupled Receptors in Heart Disease. Circ. Res. 2018, 123, 716–735. [Google Scholar] [CrossRef]
  58. Lohse, M.J.; Engelhardt, S.; Eschenhagen, T. What Is the Role of β-Adrenergic Signaling in Heart Failure? Circ. Res. 2003, 93, 896–906. [Google Scholar] [CrossRef] [Green Version]
  59. Drake, M.T.; Shenoy, S.K.; Lefkowitz, R.J. Trafficking of G protein-coupled receptors. Circ. Res. 2006, 99, 570–582. [Google Scholar] [CrossRef]
  60. O’Connell, T.D.; Jensen, B.C.; Baker, A.J.; Simpson, P.C. Cardiac alpha1-adrenergic receptors: Novel aspects of expression, signaling mechanisms, physiologic function, and clinical importance. Pharmacol. Rev. 2014, 66, 308–333. [Google Scholar] [CrossRef] [Green Version]
  61. Boivin, B.; Lavoie, C.; Vaniotis, G.; Baragli, A.; Villeneuve, L.R.; Ethier, N.; Trieu, P.; Allen, B.G.; Hébert, T.E. Functional beta-adrenergic receptor signalling on nuclear membranes in adult rat and mouse ventricular cardiomyocytes. Cardiovasc. Res. 2006, 71, 69–78. [Google Scholar] [CrossRef]
  62. Vaniotis, G.; Allen, B.G.; Hébert, T.E. Nuclear GPCRs in cardiomyocytes: An insider’s view of β-adrenergic receptor signaling. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H1754–H1764. [Google Scholar] [CrossRef] [Green Version]
  63. Vaniotis, G.; Del Duca, D.; Trieu, P.; Rohlicek, C.V.; Hébert, T.E.; Allen, B.G. Nuclear β-adrenergic receptors modulate gene expression in adult rat heart. Cell. Signal. 2011, 23, 89–98. [Google Scholar] [CrossRef] [Green Version]
  64. Wang, Y.; Shi, Q.; Li, M.; Zhao, M.; Reddy Gopireddy, R.; Teoh, J.P.; Xu, B.; Zhu, C.; Ireton, K.E.; Srinivasan, S.; et al. Intracellular β(1)-Adrenergic Receptors and Organic Cation Transporter 3 Mediate Phospholamban Phosphorylation to Enhance Cardiac Contractility. Circ. Res. 2021, 128, 246–261. [Google Scholar] [CrossRef]
  65. Bedioune, I.; Lefebvre, F.; Lechêne, P.; Varin, A.; Domergue, V.; Kapiloff, M.S.; Fischmeister, R.; Vandecasteele, G. PDE4 and mAKAPβ are nodal organizers of β2-ARs nuclear PKA signalling in cardiac myocytes. Cardiovasc. Res. 2018, 114, 1499–1511. [Google Scholar] [CrossRef] [Green Version]
  66. Bathe-Peters, M.; Gmach, P.; Boltz, H.H.; Einsiedel, J.; Gotthardt, M.; Hübner, H.; Gmeiner, P.; Lohse, M.J.; Annibale, P. Visualization of β-adrenergic receptor dynamics and differential localization in cardiomyocytes. Proc. Natl. Acad. Sci. USA 2021, 118, 818–823. [Google Scholar] [CrossRef]
  67. Nash, C.A.; Wei, W.; Irannejad, R.; Smrcka, A.V. Golgi localized β1-adrenergic receptors stimulate Golgi PI4P hydrolysis by PLCε to regulate cardiac hypertrophy. Elife 2019, 8, e48167. [Google Scholar] [CrossRef] [PubMed]
  68. Irannejad, R.; Pessino, V.; Mika, D.; Huang, B.; Wedegaertner, P.B.; Conti, M.; von Zastrow, M. Functional selectivity of GPCR-directed drug action through location bias. Nat. Chem. Biol. 2017, 13, 799–806. [Google Scholar] [CrossRef] [Green Version]
  69. Kehat, I.; Molkentin, J.D. Molecular pathways underlying cardiac remodeling during pathophysiological stimulation. Circulation 2010, 122, 2727–2735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Kreusser, M.M.; Lehmann, L.H.; Keranov, S.; Hoting, M.O.; Oehl, U.; Kohlhaas, M.; Reil, J.C.; Neumann, K.; Schneider, M.D.; Hill, J.A.; et al. Cardiac CaM Kinase II genes δ and γ contribute to adverse remodeling but redundantly inhibit calcineurin-induced myocardial hypertrophy. Circulation 2014, 130, 1262–1273. [Google Scholar] [CrossRef] [Green Version]
  71. Kim, Y.; Phan, D.; van Rooij, E.; Wang, D.Z.; McAnally, J.; Qi, X.; Richardson, J.A.; Hill, J.A.; Bassel-Duby, R.; Olson, E.N. The MEF2D transcription factor mediates stress-dependent cardiac remodeling in mice. J. Clin. Investig. 2008, 118, 124–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Helmstadter, K.G.; Ljubojevic-Holzer, S.; Wood, B.M.; Taheri, K.D.; Sedej, S.; Erickson, J.R.; Bossuyt, J.; Bers, D.M. CaMKII and PKA-dependent phosphorylation co-regulate nuclear localization of HDAC4 in adult cardiomyocytes. Basic Res. Cardiol. 2021, 116, 11. [Google Scholar] [CrossRef]
  73. Kreusser, M.M.; Backs, J. Integrated mechanisms of CaMKII-dependent ventricular remodeling. Front. Pharmacol. 2014, 5, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Suetomi, T.; Willeford, A.; Brand, C.S.; Cho, Y.; Ross, R.S.; Miyamoto, S.; Brown, J.H. Inflammation and NLRP3 Inflammasome Activation Initiated in Response to Pressure Overload by Ca2+/Calmodulin-Dependent Protein Kinase II δ Signaling in Cardiomyocytes Are Essential for Adverse Cardiac Remodeling. Circulation 2018, 138, 2530–2544. [Google Scholar] [CrossRef] [Green Version]
  75. Subedi, K.P.; Son, M.J.; Chidipi, B.; Kim, S.W.; Wang, J.; Kim, K.H.; Woo, S.H.; Kim, J.C. Signaling Pathway for Endothelin-1- and Phenylephrine-Induced cAMP Response Element Binding Protein Activation in Rat Ventricular Myocytes: Role of Inositol 1,4,5-Trisphosphate Receptors and CaMKII. Cell. Physiol. Biochem. 2017, 41, 399–412. [Google Scholar] [CrossRef] [PubMed]
  76. Ronkainen, J.J.; Hänninen, S.L.; Korhonen, T.; Koivumäki, J.T.; Skoumal, R.; Rautio, S.; Ronkainen, V.P.; Tavi, P. Ca2+-calmodulin-dependent protein kinase II represses cardiac transcription of the L-type calcium channel alpha(1C)-subunit gene (Cacna1c) by DREAM translocation. J. Physiol. 2011, 589, 2669–2686. [Google Scholar] [CrossRef] [PubMed]
  77. Mehta, S.; Aye-Han, N.N.; Ganesan, A.; Oldach, L.; Gorshkov, K.; Zhang, J. Calmodulin-controlled spatial decoding of oscillatory Ca2+ signals by calcineurin. Elife 2014, 3, e03765. [Google Scholar] [CrossRef]
  78. Chaklader, M.; Rothermel, B.A. Calcineurin in the heart: New horizons for an old friend. Cell. Signal. 2021, 87, 110134. [Google Scholar] [CrossRef]
  79. Parra, V.; Rothermel, B.A. Calcineurin signaling in the heart: The importance of time and place. J. Mol. Cell. Cardiol. 2017, 103, 121–136. [Google Scholar] [CrossRef] [Green Version]
  80. Plačkić, J.; Preissl, S.; Nikonova, Y.; Pluteanu, F.; Hein, L.; Kockskämper, J. Enhanced nucleoplasmic Ca2+ signaling in ventricular myocytes from young hypertensive rats. J. Mol. Cell. Cardiol. 2016, 101, 58–68. [Google Scholar] [CrossRef]
  81. Pérez-Hernández, M.; van Opbergen, C.J.M.; Bagwan, N.; Vissing, C.R.; Marrón-Liñares, G.M.; Zhang, M.; Torres Vega, E.; Sorrentino, A.; Drici, L.; Sulek, K.; et al. Loss of Nuclear Envelope Integrity and Increased Oxidant Production Cause DNA Damage in Adult Hearts Deficient in PKP2: A Molecular Substrate of ARVC. Circulation 2022, 146, 851–867. [Google Scholar] [CrossRef]
  82. Cohn, J.N.; Ferrari, R.; Sharpe, N. Cardiac remodeling--concepts and clinical implications: A consensus paper from an international forum on cardiac remodeling. Behalf of an International Forum on Cardiac Remodeling. J. Am. Coll. Cardiol. 2000, 35, 569–582. [Google Scholar] [CrossRef] [Green Version]
  83. Azevedo, P.S.; Polegato, B.F.; Minicucci, M.F.; Paiva, S.A.; Zornoff, L.A. Cardiac Remodeling: Concepts, Clinical Impact, Pathophysiological Mechanisms and Pharmacologic Treatment. Arq. Bras. Cardiol. 2016, 106, 62–69. [Google Scholar] [CrossRef] [PubMed]
  84. Ling, H.; Zhang, T.; Pereira, L.; Means, C.K.; Cheng, H.; Gu, Y.; Dalton, N.D.; Peterson, K.L.; Chen, J.; Bers, D.; et al. Requirement for Ca2+/calmodulin-dependent kinase II in the transition from pressure overload-induced cardiac hypertrophy to heart failure in mice. J. Clin. Investig. 2009, 119, 1230–1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sakata, Y.; Masuyama, T.; Yamamoto, K.; Nishikawa, N.; Yamamoto, H.; Kondo, H.; Ono, K.; Otsu, K.; Kuzuya, T.; Miwa, T.; et al. Calcineurin inhibitor attenuates left ventricular hypertrophy, leading to prevention of heart failure in hypertensive rats. Circulation 2000, 102, 2269–2275. [Google Scholar] [CrossRef] [Green Version]
  86. Fischer, T.H.; Eiringhaus, J.; Dybkova, N.; Förster, A.; Herting, J.; Kleinwächter, A.; Ljubojevic, S.; Schmitto, J.D.; Streckfuß-Bömeke, K.; Renner, A.; et al. Ca2+ /calmodulin-dependent protein kinase II equally induces sarcoplasmic reticulum Ca2+ leak in human ischaemic and dilated cardiomyopathy. Eur. J. Heart Fail. 2014, 16, 1292–1300. [Google Scholar] [CrossRef]
  87. Lou, Q.; Janardhan, A.; Efimov, I.R. Remodeling of calcium handling in human heart failure. Adv. Exp. Med. Biol. 2012, 740, 1145–1174. [Google Scholar] [CrossRef] [Green Version]
  88. Sossalla, S.; Fluschnik, N.; Schotola, H.; Ort, K.R.; Neef, S.; Schulte, T.; Wittköpper, K.; Renner, A.; Schmitto, J.D.; Gummert, J.; et al. Inhibition of elevated Ca2+/calmodulin-dependent protein kinase II improves contractility in human failing myocardium. Circ. Res. 2010, 107, 1150–1161. [Google Scholar] [CrossRef] [Green Version]
  89. Westenbrink, B.D.; Edwards, A.G.; McCulloch, A.D.; Brown, J.H. The promise of CaMKII inhibition for heart disease: Preventing heart failure and arrhythmias. Expert Opin. Ther. Targets 2013, 17, 889–903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Panther, F.; Williams, T.; Ritter, O. Inhibition of the calcineurin-NFAT signalling cascade in the treatment of heart failure. Recent Pat. Cardiovasc. Drug Discov. 2009, 4, 180–186. [Google Scholar] [CrossRef] [PubMed]
  91. Chen, C.H.; Lin, J.W.; Huang, C.Y.; Yeh, Y.L.; Shen, C.Y.; Badrealam, K.F.; Ho, T.J.; Padma, V.V.; Kuo, W.W.; Huang, C.Y. The combined inhibition of the CaMKIIδ and calcineurin signaling cascade attenuates IGF-IIR-induced cardiac hypertrophy. J. Cell. Physiol. 2020, 235, 3539–3547. [Google Scholar] [CrossRef]
  92. Pellicena, P.; Schulman, H. CaMKII inhibitors: From research tools to therapeutic agents. Front. Pharmacol. 2014, 5, 21. [Google Scholar] [CrossRef] [Green Version]
  93. Kawase, Y.; Ly, H.Q.; Prunier, F.; Lebeche, D.; Shi, Y.; Jin, H.; Hadri, L.; Yoneyama, R.; Hoshino, K.; Takewa, Y.; et al. Reversal of cardiac dysfunction after long-term expression of SERCA2a by gene transfer in a pre-clinical model of heart failure. J. Am. Coll. Cardiol. 2008, 51, 1112–1119. [Google Scholar] [CrossRef] [Green Version]
  94. Hulot, J.S.; Ishikawa, K.; Hajjar, R.J. Gene therapy for the treatment of heart failure: Promise postponed. Eur. Heart J. 2016, 37, 1651–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Greenberg, B.; Butler, J.; Felker, G.M.; Ponikowski, P.; Voors, A.A.; Desai, A.S.; Barnard, D.; Bouchard, A.; Jaski, B.; Lyon, A.R.; et al. Calcium upregulation by percutaneous administration of gene therapy in patients with cardiac disease (CUPID 2): A randomised, multinational, double-blind, placebo-controlled, phase 2b trial. Lancet 2016, 387, 1178–1186. [Google Scholar] [CrossRef] [PubMed]
  96. Lebek, S.; Chemello, F.; Caravia, X.M.; Tan, W.; Li, H.; Chen, K.; Xu, L.; Liu, N.; Bassel-Duby, R.; Olson, E.N. Ablation of CaMKIIδ oxidation by CRISPR-Cas9 base editing as a therapy for cardiac disease. Science 2023, 379, 179–185. [Google Scholar] [CrossRef]
  97. Shimojima, M.; Yuasa, S.; Motoda, C.; Yozu, G.; Nagai, T.; Ito, S.; Lachmann, M.; Kashimura, S.; Takei, M.; Kusumoto, D.; et al. Emerin plays a crucial role in nuclear invagination and in the nuclear calcium transient. Sci. Rep. 2017, 7, 44312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Matzer, I.; Voglhuber, J.; Kiessling, M.; Djalinac, N.; Trummer-Herbst, V.; Mabotuwana, N.; Rech, L.; Holzer, M.; Sossalla, S.; Rainer, P.P.; et al. β-Adrenergic Receptor Stimulation Maintains NCX-CaMKII Axis and Prevents Overactivation of IL6R-Signaling in Cardiomyocytes upon Increased Workload. Biomedicines 2022, 10, 1648. [Google Scholar] [CrossRef]
  99. Harzheim, D.; Talasila, A.; Movassagh, M.; Foo, R.S.; Figg, N.; Bootman, M.D.; Roderick, H.L. Elevated InsP3R expression underlies enhanced calcium fluxes and spontaneous extra-systolic calcium release events in hypertrophic cardiac myocytes. Channels 2010, 4, 67–71. [Google Scholar] [CrossRef] [Green Version]
  100. Bers, D.M. Cardiac ryanodine receptor phosphorylation: Target sites and functional consequences. Biochem. J. 2006, 396, e1–e3. [Google Scholar] [CrossRef] [Green Version]
  101. Wood, B.M.; Simon, M.; Galice, S.; Alim, C.C.; Ferrero, M.; Pinna, N.N.; Bers, D.M.; Bossuyt, J. Cardiac CaMKII activation promotes rapid translocation to its extra-dyadic targets. J. Mol. Cell. Cardiol. 2018, 125, 18–28. [Google Scholar] [CrossRef]
  102. Gómez, A.M.; Ruiz-Hurtado, G.; Benitah, J.P.; Domínguez-Rodríguez, A. Ca2+ fluxes involvement in gene expression during cardiac hypertrophy. Curr. Vasc. Pharmacol. 2013, 11, 497–506. [Google Scholar] [CrossRef]
  103. Wang, P.; Xu, S.; Xu, J.; Xin, Y.; Lu, Y.; Zhang, H.; Zhou, B.; Xu, H.; Sheu, S.S.; Tian, R.; et al. Elevated MCU Expression by CaMKIIδB Limits Pathological Cardiac Remodeling. Circulation 2022, 145, 1067–1083. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, T.; Yang, N.; Sidor, A.; O’Rourke, B. MCU Overexpression Rescues Inotropy and Reverses Heart Failure by Reducing SR Ca2+ Leak. Circ. Res. 2021, 128, 1191–1204. [Google Scholar] [CrossRef]
  105. Chahine, M.N.; Mioulane, M.; Sikkel, M.B.; O’Gara, P.; Dos Remedios, C.G.; Pierce, G.N.; Lyon, A.R.; Földes, G.; Harding, S.E. Nuclear pore rearrangements and nuclear trafficking in cardiomyocytes from rat and human failing hearts. Cardiovasc. Res. 2015, 105, 31–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Tarazón, E.; Rivera, M.; Roselló-Lletí, E.; Molina-Navarro, M.M.; Sánchez-Lázaro, I.J.; España, F.; Montero, J.A.; Lago, F.; González-Juanatey, J.R.; Portolés, M. Heart failure induces significant changes in nuclear pore complex of human cardiomyocytes. PLoS ONE 2012, 7, e48957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Qi, X.Y.; Vahdati Hassani, F.; Hoffmann, D.; Xiao, J.; Xiong, F.; Villeneuve, L.R.; Ljubojevic-Holzer, S.; Kamler, M.; Abu-Taha, I.; Heijman, J.; et al. Inositol Trisphosphate Receptors and Nuclear Calcium in Atrial Fibrillation. Circ. Res. 2021, 128, 619–635. [Google Scholar] [CrossRef]
  108. Staerk, L.; Wang, B.; Preis, S.R.; Larson, M.G.; Lubitz, S.A.; Ellinor, P.T.; McManus, D.D.; Ko, D.; Weng, L.C.; Lunetta, K.L.; et al. Lifetime risk of atrial fibrillation according to optimal, borderline, or elevated levels of risk factors: Cohort study based on longitudinal data from the Framingham Heart Study. BMJ 2018, 361, k1453. [Google Scholar] [CrossRef] [Green Version]
  109. Andrade, J.; Khairy, P.; Dobrev, D.; Nattel, S. The clinical profile and pathophysiology of atrial fibrillation: Relationships among clinical features, epidemiology, and mechanisms. Circ. Res. 2014, 114, 1453–1468. [Google Scholar] [CrossRef]
  110. Kornej, J.; Börschel, C.S.; Benjamin, E.J.; Schnabel, R.B. Epidemiology of Atrial Fibrillation in the 21st Century: Novel Methods and New Insights. Circ. Res. 2020, 127, 4–20. [Google Scholar] [CrossRef]
  111. Nattel, S.; Dobrev, D. The multidimensional role of calcium in atrial fibrillation pathophysiology: Mechanistic insights and therapeutic opportunities. Eur. Heart J. 2012, 33, 1870–1877. [Google Scholar] [CrossRef] [Green Version]
  112. Pabel, S.; Knierim, M.; Stehle, T.; Alebrand, F.; Paulus, M.; Sieme, M.; Herwig, M.; Barsch, F.; Körtl, T.; Pöppl, A.; et al. Effects of Atrial Fibrillation on the Human Ventricle. Circ. Res. 2022, 130, 994–1010. [Google Scholar] [CrossRef]
  113. Hershberger, R.E.; Siegfried, J.D. Update 2011: Clinical and genetic issues in familial dilated cardiomyopathy. J. Am. Coll. Cardiol. 2011, 57, 1641–1649. [Google Scholar] [CrossRef] [Green Version]
  114. Fatkin, D.; MacRae, C.; Sasaki, T.; Wolff, M.R.; Porcu, M.; Frenneaux, M.; Atherton, J.; Vidaillet, H.J., Jr.; Spudich, S.; De Girolami, U.; et al. Missense mutations in the rod domain of the lamin A/C gene as causes of dilated cardiomyopathy and conduction-system disease. N. Engl. J. Med. 1999, 341, 1715–1724. [Google Scholar] [CrossRef] [Green Version]
  115. Kimura, A. Molecular basis of hereditary cardiomyopathy: Abnormalities in calcium sensitivity, stretch response, stress response and beyond. J. Hum. Genet. 2010, 55, 81–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Holaska, J.M. Emerin and the nuclear lamina in muscle and cardiac disease. Circ. Res. 2008, 103, 16–23. [Google Scholar] [CrossRef]
  117. Nattel, S.; Heijman, J.; Zhou, L.; Dobrev, D. Molecular Basis of Atrial Fibrillation Pathophysiology and Therapy: A Translational Perspective. Circ. Res. 2020, 127, 51–72. [Google Scholar] [CrossRef] [PubMed]
  118. Hegyi, B.; Pölönen, R.P.; Hellgren, K.T.; Ko, C.Y.; Ginsburg, K.S.; Bossuyt, J.; Mercola, M.; Bers, D.M. Cardiomyocyte Na+ and Ca2+ mishandling drives vicious cycle involving CaMKII, ROS, and ryanodine receptors. Basic Res. Cardiol. 2021, 116, 58. [Google Scholar] [CrossRef] [PubMed]
  119. Kondo, T.; Higo, S.; Shiba, M.; Kohama, Y.; Kameda, S.; Tabata, T.; Inoue, H.; Okuno, S.; Ogawa, S.; Nakamura, S.; et al. Human-Induced Pluripotent Stem Cell-Derived Cardiomyocyte Model for TNNT2 Δ160E-Induced Cardiomyopathy. Circ. Genom. Precis. Med. 2022, 15, e003522. [Google Scholar] [CrossRef] [PubMed]
  120. Hughes, E.; Middleton, D.A. Comparison of the structure and function of phospholamban and the arginine-14 deficient mutant associated with dilated cardiomyopathy. PLoS ONE 2014, 9, e106746. [Google Scholar] [CrossRef] [Green Version]
  121. van den Hoogenhof, M.M.G.; Beqqali, A.; Amin, A.S.; van der Made, I.; Aufiero, S.; Khan, M.A.F.; Schumacher, C.A.; Jansweijer, J.A.; van Spaendonck-Zwarts, K.Y.; Remme, C.A.; et al. RBM20 Mutations Induce an Arrhythmogenic Dilated Cardiomyopathy Related to Disturbed Calcium Handling. Circulation 2018, 138, 1330–1342. [Google Scholar] [CrossRef] [PubMed]
  122. Dibb, K.M.; Graham, H.K.; Venetucci, L.A.; Eisner, D.A.; Trafford, A.W. Analysis of cellular calcium fluxes in cardiac muscle to understand calcium homeostasis in the heart. Cell Calcium 2007, 42, 503–512. [Google Scholar] [CrossRef]
  123. Bootman, M.D.; Rietdorf, K.; Collins, T.; Walker, S.; Sanderson, M. Ca2+-sensitive fluorescent dyes and intracellular Ca2+ imaging. Cold Spring Harb. Protoc. 2013, 2013, 83–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Takahashi, A.; Camacho, P.; Lechleiter, J.D.; Herman, B. Measurement of Intracellular Calcium. Physiol. Rev. 1999, 79, 1089–1125. [Google Scholar] [CrossRef] [PubMed]
  125. Ljubojević, S.; Bers, D.M. Measuring intranuclear and nuclear envelope [Ca2+] vs. cytosolic [Ca2+]. Methods Mol. Biol. 2015, 1234, 135–147. [Google Scholar] [CrossRef] [PubMed]
  126. Rossi, A.M.; Taylor, C.W. Reliable measurement of free Ca2+ concentrations in the ER lumen using Mag-Fluo-4. Cell Calcium 2020, 87, 102188. [Google Scholar] [CrossRef] [PubMed]
  127. Li, X.; Shen, L.; Zhao, F.; Zou, X.; He, Y.; Zhang, F.; Zhang, C.; Yu, B.; Cao, Z. Modification of distinct ion channels differentially modulates Ca2+ dynamics in primary cultured rat ventricular cardiomyocytes. Sci. Rep. 2017, 7, 40952. [Google Scholar] [CrossRef] [Green Version]
  128. Liao, J.; Patel, D.; Zhao, Q.; Peng, R.; Guo, H.; Diwu, Z. A novel Ca2+ indicator for long-term tracking of intracellular calcium flux. BioTechniques 2021, 70, 271–277. [Google Scholar] [CrossRef]
  129. Daily, N.J.; Santos, R.; Vecchi, J.; Kemanli, P.; Wakatsuki, T. Calcium Transient Assays for Compound Screening with Human iPSC-derived Cardiomyocytes: Evaluating New Tools. J. Evol. Stem Cell Res. 2017, 1, 1–11. [Google Scholar] [CrossRef] [Green Version]
  130. Thomas, D.; Tovey, S.C.; Collins, T.J.; Bootman, M.D.; Berridge, M.J.; Lipp, P. A comparison of fluorescent Ca2+ indicator properties and their use in measuring elementary and global Ca2+ signals. Cell Calcium 2000, 28, 213–223. [Google Scholar] [CrossRef]
  131. Smith, N.A.; Kress, B.T.; Lu, Y.; Chandler-Militello, D.; Benraiss, A.; Nedergaard, M. Fluorescent Ca2+ indicators directly inhibit the Na,K-ATPase and disrupt cellular functions. Sci. Signal. 2018, 11, eaal2039. [Google Scholar] [CrossRef] [Green Version]
  132. Nakai, J.; Ohkura, M.; Imoto, K. A high signal-to-noise Ca2+ probe composed of a single green fluorescent protein. Nat. Biotechnol. 2001, 19, 137–141. [Google Scholar] [CrossRef]
  133. Chen, T.W.; Wardill, T.J.; Sun, Y.; Pulver, S.R.; Renninger, S.L.; Baohan, A.; Schreiter, E.R.; Kerr, R.A.; Orger, M.B.; Jayaraman, V.; et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 2013, 499, 295–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Yang, Y.; Liu, N.; He, Y.; Liu, Y.; Ge, L.; Zou, L.; Song, S.; Xiong, W.; Liu, X. Improved calcium sensor GCaMP-X overcomes the calcium channel perturbations induced by the calmodulin in GCaMP. Nat. Commun. 2018, 9, 1504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Jiang, Y.; Zhou, Y.; Bao, X.; Chen, C.; Randolph, L.N.; Du, J.; Lian, X.L. An Ultrasensitive Calcium Reporter System via CRISPR-Cas9-Mediated Genome Editing in Human Pluripotent Stem Cells. iScience 2018, 9, 27–35. [Google Scholar] [CrossRef] [Green Version]
  136. Safaralizade, M.; Fuderer, R.; Grosse, R.; Zhao, B. Measuring nuclear calcium and actin assembly in living cells. J. Biochem. 2021, 169, 287–294. [Google Scholar] [CrossRef] [PubMed]
  137. Mertes, N.; Busch, M.; Huppertz, M.C.; Hacker, C.N.; Wilhelm, J.; Gürth, C.M.; Kühn, S.; Hiblot, J.; Koch, B.; Johnsson, K. Fluorescent and Bioluminescent Calcium Indicators with Tuneable Colors and Affinities. J. Am. Chem. Soc. 2022, 144, 6928–6935. [Google Scholar] [CrossRef] [PubMed]
  138. Voigt, N.; Heijman, J.; Wang, Q.; Chiang, D.Y.; Li, N.; Karck, M.; Wehrens, X.H.T.; Nattel, S.; Dobrev, D. Cellular and molecular mechanisms of atrial arrhythmogenesis in patients with paroxysmal atrial fibrillation. Circulation 2014, 129, 145–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ljubojević-Holzer, S.; Kraler, S.; Djalinac, N.; Abdellatif, M.; Voglhuber, J.; Schipke, J.; Schmidt, M.; Kling, K.M.; Franke, G.T.; Herbst, V.; et al. Loss of autophagy protein ATG5 impairs cardiac capacity in mice and humans through diminishing mitochondrial abundance and disrupting Ca2+ cycling. Cardiovasc. Res. 2022, 118, 1492–1505. [Google Scholar] [CrossRef]
  140. Liang, P.; Lan, F.; Lee, A.S.; Gong, T.; Sanchez-Freire, V.; Wang, Y.; Diecke, S.; Sallam, K.; Knowles, J.W.; Wang, P.J.; et al. Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation 2013, 127, 1677–1691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Itzhaki, I.; Rapoport, S.; Huber, I.; Mizrahi, I.; Zwi-Dantsis, L.; Arbel, G.; Schiller, J.; Gepstein, L. Calcium handling in human induced pluripotent stem cell derived cardiomyocytes. PLoS ONE 2011, 6, e18037. [Google Scholar] [CrossRef]
  142. Karakikes, I.; Ameen, M.; Termglinchan, V.; Wu, J.C. Human induced pluripotent stem cell-derived cardiomyocytes: Insights into molecular, cellular, and functional phenotypes. Circ. Res. 2015, 117, 80–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Hwang, H.S.; Kryshtal, D.O.; Feaster, T.K.; Sánchez-Freire, V.; Zhang, J.; Kamp, T.J.; Hong, C.C.; Wu, J.C.; Knollmann, B.C. Comparable calcium handling of human iPSC-derived cardiomyocytes generated by multiple laboratories. J. Mol. Cell. Cardiol. 2015, 85, 79–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Zhang, J.; Klos, M.; Wilson, G.F.; Herman, A.M.; Lian, X.; Raval, K.K.; Barron, M.R.; Hou, L.; Soerens, A.G.; Yu, J.; et al. Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: The matrix sandwich method. Circ. Res. 2012, 111, 1125–1136. [Google Scholar] [CrossRef] [PubMed]
  145. Lundy, S.D.; Zhu, W.Z.; Regnier, M.; Laflamme, M.A. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 2013, 22, 1991–2002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Ma, J.; Guo, L.; Fiene, S.J.; Anson, B.D.; Thomson, J.A.; Kamp, T.J.; Kolaja, K.L.; Swanson, B.J.; January, C.T. High purity human-induced pluripotent stem cell-derived cardiomyocytes: Electrophysiological properties of action potentials and ionic currents. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H2006–H2017. [Google Scholar] [CrossRef] [PubMed]
  147. Li, S.; Keung, W.; Cheng, H.; Li, R.A. Structural and Mechanistic Bases of Nuclear Calcium Signaling in Human Pluripotent Stem Cell-Derived Ventricular Cardiomyocytes. Stem Cells Int. 2019, 2019, 8765752. [Google Scholar] [CrossRef]
  148. Lee, J.; Termglinchan, V.; Diecke, S.; Itzhaki, I.; Lam, C.K.; Garg, P.; Lau, E.; Greenhaw, M.; Seeger, T.; Wu, H.; et al. Activation of PDGF pathway links LMNA mutation to dilated cardiomyopathy. Nature 2019, 572, 335–340. [Google Scholar] [CrossRef]
  149. Funakoshi, M.; Tsuchiya, Y.; Arahata, K. Emerin and cardiomyopathy in Emery-Dreifuss muscular dystrophy. Neuromuscul. Disord. 1999, 9, 108–114. [Google Scholar] [CrossRef]
  150. Thomas, D.; Choi, S.; Alamana, C.; Parker, K.K.; Wu, J.C. Cellular and Engineered Organoids for Cardiovascular Models. Circ. Res. 2022, 130, 1780–1802. [Google Scholar] [CrossRef]
  151. Campostrini, G.; Windt, L.M.; van Meer, B.J.; Bellin, M.; Mummery, C.L. Cardiac Tissues From Stem Cells: New Routes to Maturation and Cardiac Regeneration. Circ. Res. 2021, 128, 775–801. [Google Scholar] [CrossRef]
  152. Richards, D.J.; Li, Y.; Kerr, C.M.; Yao, J.; Beeson, G.C.; Coyle, R.C.; Chen, X.; Jia, J.; Damon, B.; Wilson, R.; et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat. Biomed. Eng. 2020, 4, 446–462. [Google Scholar] [CrossRef]
  153. Higazi, D.R.; Fearnley, C.J.; Drawnel, F.M.; Talasila, A.; Corps, E.M.; Ritter, O.; McDonald, F.; Mikoshiba, K.; Bootman, M.D.; Roderick, H.L. Endothelin-1-stimulated InsP3-induced Ca2+ release is a nexus for hypertrophic signaling in cardiac myocytes. Mol. Cell 2009, 33, 472–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Pfenniger, A.; Yoo, S.; Arora, R. Nucleoplasmic Ca2+: The ‘Mastermind’ Behind Pathological Atrial Remodeling? Circ. Res. 2021, 128, 636–638. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Nuclear Ca2+ handling in cardiomyocytes. (Left) Cardiac action potential (AP) causes myocyte membrane depolarization (1) and creates an inward Ca2+ current through opening of L-type Ca2+ channels (LTCC) located at the plasma membrane, including T-tubules (2). Increased local Ca2+ concentration ([Ca2+]) triggers the release of Ca2+ stored in the sarcoplasmic reticulum (SR) via ryanodine receptors (RyR) (3). Nuclear pore complexes (NPC) facilitate passive diffusion of free cytoplasmic Ca2+ into the nucleus (4), where the transient rise in [Ca2+]nuc can mediate transcriptional effects. For [Ca2+]nuc to decline, Ca2+ has to diffuse back out of the nucleus through NPCs to be taken up into the nuclear envelope by SR Ca2+ ATPase (SERCA) expressed on the outer nuclear membrane (5). (Right) G-protein coupled receptors (GPCRs) are stimulated by endothelin-1 (ET-1), angiotensin II (Ang II) or noradrenaline (NA) and activate phospholipase C (PLC) (1), which synthesizes diacylglycerol (DAG) and inositol-1,4,5-triphospate (IP3) (2). IP3 then passively diffuses through NPCs to initiate Ca2+ release from the nuclear envelope via IP3 receptors (IP3R) and thus elicits a nuclear Ca2+ release (3). Increased [Ca2+]nuc can again exert effects on transcription factors before diffusing back out of the nucleoplasm via NPCs and being taken up by SERCA expressed on the outer nuclear membrane (4).
Figure 1. Nuclear Ca2+ handling in cardiomyocytes. (Left) Cardiac action potential (AP) causes myocyte membrane depolarization (1) and creates an inward Ca2+ current through opening of L-type Ca2+ channels (LTCC) located at the plasma membrane, including T-tubules (2). Increased local Ca2+ concentration ([Ca2+]) triggers the release of Ca2+ stored in the sarcoplasmic reticulum (SR) via ryanodine receptors (RyR) (3). Nuclear pore complexes (NPC) facilitate passive diffusion of free cytoplasmic Ca2+ into the nucleus (4), where the transient rise in [Ca2+]nuc can mediate transcriptional effects. For [Ca2+]nuc to decline, Ca2+ has to diffuse back out of the nucleus through NPCs to be taken up into the nuclear envelope by SR Ca2+ ATPase (SERCA) expressed on the outer nuclear membrane (5). (Right) G-protein coupled receptors (GPCRs) are stimulated by endothelin-1 (ET-1), angiotensin II (Ang II) or noradrenaline (NA) and activate phospholipase C (PLC) (1), which synthesizes diacylglycerol (DAG) and inositol-1,4,5-triphospate (IP3) (2). IP3 then passively diffuses through NPCs to initiate Ca2+ release from the nuclear envelope via IP3 receptors (IP3R) and thus elicits a nuclear Ca2+ release (3). Increased [Ca2+]nuc can again exert effects on transcription factors before diffusing back out of the nucleoplasm via NPCs and being taken up by SERCA expressed on the outer nuclear membrane (4).
Biomedicines 11 00960 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kiessling, M.; Djalinac, N.; Voglhuber, J.; Ljubojevic-Holzer, S. Nuclear Calcium in Cardiac (Patho)Physiology: Small Compartment, Big Impact. Biomedicines 2023, 11, 960. https://doi.org/10.3390/biomedicines11030960

AMA Style

Kiessling M, Djalinac N, Voglhuber J, Ljubojevic-Holzer S. Nuclear Calcium in Cardiac (Patho)Physiology: Small Compartment, Big Impact. Biomedicines. 2023; 11(3):960. https://doi.org/10.3390/biomedicines11030960

Chicago/Turabian Style

Kiessling, Mara, Nataša Djalinac, Julia Voglhuber, and Senka Ljubojevic-Holzer. 2023. "Nuclear Calcium in Cardiac (Patho)Physiology: Small Compartment, Big Impact" Biomedicines 11, no. 3: 960. https://doi.org/10.3390/biomedicines11030960

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop