Aging is a natural biological process influenced by endogenous and exogenous factors such as genetics, environment, and individual lifestyle. The aging-dependent decline in resting and maximum heart rate is a conserved feature across multiple species, including humans. Such changes in heart rhythm control underscore fundamental alterations in the primary cardiac pacemaker, the sinoatrial node (SAN). Older individuals often present symptoms of SAN dysfunction (SND), including sinus bradycardia, sinus arrest, and bradycardia-tachycardia syndrome. These can lead to a broad range of symptoms from palpitations, dizziness to recurrent syncope. The sharp rise in the incidence of SND among individuals over 65 years old, coupled with projected longevity over the next decades, highlights the urgent need for a deeper mechanistic understanding of aging-related SND to develop novel and effective therapeutic alternatives. In this review, we will revisit current knowledge on the ionic and structural remodeling underlying age-related decline in SAN function, and a particular emphasis will be made on new directions for future research.

The sinoatrial node (SAN) is the primary pacemaker of the heart, responsible for the initiation of rhythmic electrical impulses that lead to synchronized cardiac contractions. Located in the right atrium at the junction of the crista terminalis and the superior vena cava [1], the SAN consists of a small cluster of highly specialized pacemaker cells that generate spontaneous action potentials. The automaticity of these pacemaker cells is a key feature that distinguishes them from other cardiac myocytes. This unique electrogenesis allows the human heart to beat over two billion times throughout an average lifetime. However, when the SAN fails to self-generate electrical impulses, it compromises the systemic circulation leading to dizziness and syncope. Early-stage SAN dysfunction (SND) may be latent and asymptomatic, while more severe cases demand the implantation of an electronic pacemaker.

SND can arise from a variety of conditions, broadly categorized into genetic and acquired causes. Genetic factors include inherited conditions such as sick sinus syndrome, where specific gene mutations result in structural and/or functional abnormalities in the SAN tissue [2-5]. Mutations in several genes, including those encoding ion channels such as hyperpolarization-activated cyclic nucleotide-gated 2 and 4 (Hcn2 and Hcn4) and sodium voltage-gated channel alpha subunit 5 (Scn5a), as well as cytoskeletal proteins and transcription factors involved in heart development, have been linked to SND [3,6,7]. For a comprehensive review of the genetic mechanisms underlying SND, we recommend readers see references [2,3,6]. However, most SND cases are due to non-genetic factors including, aging [8-10], heart failure (HF with reduced and preserved ejection fraction) [11-16], amyloidosis [17], systemic sclerosis [18,19], rheumatic fever [20], Chagas disease [21,22], and secondary to chemotherapies including immune checkpoint inhibitors [23-25].

Aging-related SND is a common cardiac condition characterized by a progressive decline of SAN function (Figure 1A), impairing its ability to accurately regulate heart rhythm. SND affects approximately 1 in every 600 individuals over the age of 65 [7,26] (Figure 1B). According to the World Population Prospects 2024 from the United Nations, the current global population of people aged ≥80 years is 137 million, a number projected to triple by 2050 [27]. As human lifespans continue to increase, SND has become an increasingly relevant clinical problem, especially since current treatment options are limited to the costly and not risk-free implantation of electronic pacemakers. Therefore, a better understanding of the cellular and molecular mechanisms involved in aging-related SND could potentially generate new alternative therapeutic approaches for aging and other diseases that manifest with SND. While previous reviews have discussed important aspects of aging-related SND [28,29], the next section will provide a brief overview of the major ionic and structural remodeling that occur in SAN during aging.

Age-dependent SND.

Figure 1:
Age-dependent SND.

(A) Decline in intrinsic and maximum heart rates with age. (B) Estimative of overall number of sick sinus syndrome (SSS) cases per year and stratified by age (United States, 2012–2060; reproduced with permission from [7]). Part of panel A was created with BioRender.com. SAN, sinoatrial node; SND, SAN dysfunction.

Figure 1:
Age-dependent SND.

(A) Decline in intrinsic and maximum heart rates with age. (B) Estimative of overall number of sick sinus syndrome (SSS) cases per year and stratified by age (United States, 2012–2060; reproduced with permission from [7]). Part of panel A was created with BioRender.com. SAN, sinoatrial node; SND, SAN dysfunction.

Close modal

In the mammalian heart, three major structures are endowed with automaticity and are capable of driving the heartbeat: the SAN, which serves as the primary pacemaker, the atrioventricular node (AVN), and the Purkinje fibers network. In 1882, Walter Gaskell demonstrated in the tortoise heart that cardiac electrical impulses are generated in the sinus auricle and then conducted to the atrium and ventricles [30,31]. A more detailed anatomical localization of the SAN pacemaker was later identified by Arthur Keith and Martin Flack in 1907 [32].

SAN pacemaker cells are specialized cell types that spontaneously oscillate their membrane potential, generating rhythmic electrical impulses that travel through the cardiac conduction system (CCS) to stimulate the entire heart (Figure 2). These electrical impulses quickly spread to the right and left atria, slowing down when reaching the AVN. This delay, along with the decremental property of the AVN, allows the atria to contract, completing the filling of ventricles with blood in preparation for their contraction at different heart rates (HRs). Subsequently, electrical impulses travel through the left and right bundle branches, which rapidly propagate throughout the ventricles via the Purkinje fibers to produce a synchronized biventricular contraction. Although pacemaker cells are distributed throughout the CCS, SAN cells exhibit a higher rate of spontaneous depolarizations compared with those in the AVN, His bundle, bundle branches, and Purkinje fibers, establishing the SAN as the primary pacemaker of the heart (Figure 2B).

Cardiac conduction system.

Figure 2:
Cardiac conduction system.

(A) Cardiac conduction system visualized using contactin2-eGFP mice (reproduced with permission from [33]). (B) Schematic representative anatomy of the cardiac conduction system. The cardiac electric impulse originates in the sinoatrial node (SAN) and travels across the atrioventricular node (AVN), the His bundle, left and right bundle branches, and Purkinje fibers (PFs) (reproduced with permission from [34]).

Figure 2:
Cardiac conduction system.

(A) Cardiac conduction system visualized using contactin2-eGFP mice (reproduced with permission from [33]). (B) Schematic representative anatomy of the cardiac conduction system. The cardiac electric impulse originates in the sinoatrial node (SAN) and travels across the atrioventricular node (AVN), the His bundle, left and right bundle branches, and Purkinje fibers (PFs) (reproduced with permission from [34]).

Close modal

The automaticity of SAN pacemaker cells relies on the synchronized activity of two oscillators [34-37], referred to as the ‘membrane clock’ and the ‘calcium (Ca2+) clock’. Although SAN pacemaker cells are morphologically diverse [38,39] (Figure 3A), they are capable of generating rhythmic depolarizations with a unique action potential profile. Briefly, the membrane clock operates within the plasma membrane and relies on the hyperpolarization-activated funny current (If), which provides an inward current during diastolic depolarization through the Hcn1, Hcn2 and Hcn4 channels [37,40-44]. As the membrane potential depolarizes, activation of T-type calcium channels (mainly Cav3.1 and Cav3.2) and l-type calcium channels (primarily Cav1.2 and Cav1.3) takes place, contributing to inward calcium currents (ICa) [37,45-48]. The influx of ions from If and ICa synchronizes the rhythmic Ca2+ release from the sarcoplasmic reticulum via ryanodine receptors (RyR) [49-51], facilitating diastolic depolarization through the electrogenic sodium–calcium exchanger (NCX) [52,53]. The repolarization of the membrane potential is achieved through rapid and slow delayed rectifier potassium currents (IKr and IKs, respectively), with additional contributions from IK,ACh, IK,ATP, IK,Ado, and IK,Ca [37,54-59]. Subsequently, Ca2+ reuptake into the sarcoplasmic reticulum by the sarcoplasmic reticulum (SR) Ca2+ ATPase (SERCA) takes place, which, in combination with repolarizing currents, reestablish the maximum diastolic membrane potential to facilitate the initiation of a new heartbeat (Figure 3B and C). For more details on the numerous ionic currents involved in cardiac pacemaking, we encourage readers to consult the following reviews [37,60]. Although not discussed in detail here, alterations in calsequestrin 2 [61], junctophilin 2 [62], desmosomal proteins (e.g., desmoplakin and desmin) [63,64], natriuretic peptides receptors (NPR-A, NPR-B, and NPR-C) [65-68], Hippo-Yap signaling [69], glucagon-like peptide-1 receptor [70], p21-activated kinase 1 [71], phosphodiesterases [72-76], cyclic guanosine monophosphate, and protein kinase D [77] have been linked with the control of SAN function in healthy and diseased hearts.

Depressed automaticity of aged SAN pacemaker cells.

Figure 3:
Depressed automaticity of aged SAN pacemaker cells.

(A) Diverse morphology of pacemaker cells within the canine SAN tissue (center: spindle cell; right, spider cell; Reproduced with permission from [39]). (B) The coupled-clock system generates spontaneous diastolic depolarizations by functional interplay between ion channels at the plasma membrane (membrane clock) and local diastolic Ca2+ release (calcium clock). (C) SAN pacemaker cell action potential and ionic fluxes responsible for automaticity (B and C, adapted and reproduced with permission from [34]). (D) Reduced intrinsic spontaneous action potential (AP) firing rate and impaired β-adrenergic receptor (β-AR) response in aged SAN cells compared to young. AC, adenylyl cyclases; CamKII, Ca2+/calmodulin-dependent protein kinase II; PDE, phosphodiesterases; PKA, protein kinase A; SR, sarcoplasmic reticulum.

Figure 3:
Depressed automaticity of aged SAN pacemaker cells.

(A) Diverse morphology of pacemaker cells within the canine SAN tissue (center: spindle cell; right, spider cell; Reproduced with permission from [39]). (B) The coupled-clock system generates spontaneous diastolic depolarizations by functional interplay between ion channels at the plasma membrane (membrane clock) and local diastolic Ca2+ release (calcium clock). (C) SAN pacemaker cell action potential and ionic fluxes responsible for automaticity (B and C, adapted and reproduced with permission from [34]). (D) Reduced intrinsic spontaneous action potential (AP) firing rate and impaired β-adrenergic receptor (β-AR) response in aged SAN cells compared to young. AC, adenylyl cyclases; CamKII, Ca2+/calmodulin-dependent protein kinase II; PDE, phosphodiesterases; PKA, protein kinase A; SR, sarcoplasmic reticulum.

Close modal

The gradual decline in HR with aging reflects a slowing of the intrinsic SAN activity (Figure 3D), resulting from both electrical remodeling of individual pacemaker cells and structural changes within the SAN tissue. Aging-dependent ionic remodeling of SAN is considered a key mechanism that explains the reduced automaticity of pacemaker cells. A decrease in the densities of If, INa, ICa,L, and ICa,T occurs in an age-dependent manner [78-81], thereby contributing to lower action potential firing rates in aged SAN. Despite the clear role of INCX and IK in pacemaker function, there is currently no functional evidence regarding their contributions in aged SAN cells. Changes in several Ca2+-handling proteins have been reported in aged SAN, suggesting an impairment of the Ca2+ clock. The expression levels of RyR2, SERCA2a, and NCX1 are decreased in aged SAN cells [82,83], which partially justifies the reduced Ca2+ transient amplitude, prolonged SR Ca2+ reuptake, and consequently, SR Ca2+ load [82]. Although the cellular components of membrane and Ca2+ clocks have been considered as spatially distinct [84], there is compelling evidence highlighting the requisite of a coupled-clock system to maintain the automaticity of SAN pacemaker cells [35]. Thus, further studies are still required to further dissect the precise mechanisms driving the ‘uncoupled’-clock system in aged SAN cells.

Chronotropic incompetence is defined as the inability of the heart to accelerate its rate to adequately match cardiac output to the increased body’s demand [85,86]. This condition is considered a major limiting factor in exercise capacity, which limits the quality of life and serves as an independent predictor of major adverse cardiovascular events and overall mortality [85,86]. Although the exact mechanisms of chronotropic incompetence have yet to be elucidated, among other factors [85,86], SND is considered an important contributor. During strenuous exertion, the HR can be increased by up to 300%; however, a blunted maximum HR in response to exercise and sympathetic stimulation is observed in older individuals [85-87]. This acute chronotropic adaptation is a mechanism modulated by the autonomic nervous system, which releases catecholamines by sympathetic neurons. These neurotransmitters activate the β-adrenergic receptor (βAR), a Gs protein-coupled receptor located at the membrane of SAN pacemaker cells, with subsequent augmented intracellular levels of cyclic adenosine monophosphate (cAMP) via adenylyl cyclase [88,89]. cAMP directly binds to HCN channels increasing If [90] and stimulates protein kinase A (PKA), which phosphorylates numerous proteins that increase the firing rate of SAN cells [82,91-95]. The impaired response of aged SAN to βAR stimulation is partly justified by the altered expression of PKA-sensitive proteins. So far, no age-dependent changes in βARs transcription have been found in SAN [83,96]. Most previous research in aged SAN has focused on dissecting the cellular responses of components downstream of βAR signaling, revealing that stimulating adenylyl cyclase or inhibiting phosphodiesterases increases firing rates in both young and aged SAN cells [78,79]. While the relative increase in firing rates in young and aged SAN was similar, the absolute number of action potentials generated by aged SAN cells was significantly lower [78,79] (Figure 3D). Interestingly, the replenishment of intracellular concentrations of cAMP restored the firing rate in aged SAN to young levels [79]. These findings suggest that the impaired fight-or-flight response in aged SAN cells is more closely associated with reduced intracellular cAMP production than changes in βAR expression.

The SAN tissue is unique in its cellular and extracellular matrix composition (ECM) [97,98]. Pacemaker cells within the SAN are connected to each other via gap junctions, primarily connexin 45 (Cx45) and connexin 30.2 (Cx30.2), unlike the working myocardium where connexin 43 (Cx43) is more commonly expressed. Along with the particular ECM microstructure of SAN tissue discussed below, the pattern of gap junctions contributes to the functional connection between the SAN and atria myocardium through SAN conduction pathways (SACPs) [99-101]. Age-related slowing of SAN conduction has been observed [102,103], with studies reporting a progressive reduction in the expression of Cx43 and Cx30.2 without significant changes in Cx40 or Cx45 [83,102]. Given the naturally low levels of Cx43 in the SAN, the link between slowing SAN conduction and Cx43 expression requires further exploration.

The ECM of the SAN is notably distinct from other compartments of the heart, containing a high content of collagen, elastin, and fibroblasts [97,99-101,104,105]. This rich network of connective tissue not only supports the structural integrity of the SAN but also functions as an electrical insulator, regulating the velocity and directionality of electrical impulse propagation and ensuring that the SAN remains the dominant pacemaker of the heart [97,106]. Increased fibrosis within the SAN has been widely observed across various species and is strongly associated with reduced pacemaker function [8,107-110]. This architectural remodeling disrupts electrical conduction, with more pronounced effects seen in older individuals with SND [10,111]. However, while fibrosis is commonly linked to pacemaker abnormalities, its precise role in the age-related slowing of intrinsic HR remains uncertain. Interestingly, fibrotic remodeling is not always correlated with SND, as some aged hearts with significant fibrosis maintain normal sinus rhythm [107]. Therefore, the extent to which fibrosis contributes to the decline of SAN function with age has yet to be determined.

The management of symptomatic patients with SND focuses on alleviating symptoms and preventing complications (Figure 4A), such as syncope, dizziness, or HF. Acute and reversible SND cases are commonly managed with medications or temporary transvenous or transesophageal pacing [2,112,113]. However, most cases of aging-related SND are chronic and irreversible [26,112]. Unfortunately, there are currently limited options for treating chronic SND, with the implantation of a permanent electronic pacemaker remaining the primary treatment option. The Dutch registry data from 96,900 patients demonstrated that 32.6% of individuals aged 80 or older necessitate pacemaker implantation, with SND the most common cause of indication (42.3%) [114]. In a larger cohort, the analysis of 178,000 pacemakers implanted in the United States between 1997 and 2004 indicated that 64% of all implantations were in patients aged 75 years or older [115]. Although benefits on the quality of life were observed in elderly patients treated with a permanent pacemaker, particularly using dual-chamber rate-modulated pacing [116], there are conflicting findings on survival benefits [26,117-121]. For a detailed discussion on electronic devices and biological pacemakers, we recommend reading the following review [122].

Postulated mechanisms of aging-related SND.

Figure 4:
Postulated mechanisms of aging-related SND.

(A) Classifications of SND (reproduced with permission from [2]). (B) Underlying mechanisms associated with aging-dependent SND. SAN, sinoatrial node; SND, SAN dysfunction.

Figure 4:
Postulated mechanisms of aging-related SND.

(A) Classifications of SND (reproduced with permission from [2]). (B) Underlying mechanisms associated with aging-dependent SND. SAN, sinoatrial node; SND, SAN dysfunction.

Close modal

Multiple pharmacological agents have been shown to affect SAN physiology. Table 1 briefly summarizes the most clinically relevant drugs that affect the chronotropism of the heart and their mechanisms of action. Despite the therapeutic benefits, drug intoxication has been reported with consequent effects on the sinus rhythm, as previously discussed [113]. However, molecules without anticipated effects on SAN chronotropy such as glucagon-like peptide-1 receptor agonist were recently shown to increase the firing rate of pacemaker cells via phosphorylation of Ca2+ cycling proteins [70]. While the repurposing of drugs for SND remains largely unexplored, the development of synthetic molecules, biological and gene therapeutic approaches are areas that warrant further studies.the

Table 1:
Summary of the most clinically relevant drugs that affect the heart rate.
Class of drugMechanism of actionHR effect
β-AR blocker
(e.g., atenolol, propranolol) 
β-AR antagonism ↓ 
Cholinergic blockers
(e.g., atropine) 
muscarinic receptor antagonism ↑ 
Methylxanthine
(e.g., theophylline) 
Phosphodiesterase inhibitor and adenosine receptors blocker ↑ 
Benzazepine
(e.g., ivabradine) 
HCN blocker ↓ 
Digitalis glycoside
(e.g., digoxin) 
Na+/K+ ATPase inhibitor ↓ 
Ca2+ channel blockers
(e.g., verapamil) 
Ca2+ channel blockers ↓ 
K+ channel blockers
(e.g., amiodarone) 
K+ channel blockers ↓ 
Class of drugMechanism of actionHR effect
β-AR blocker
(e.g., atenolol, propranolol) 
β-AR antagonism ↓ 
Cholinergic blockers
(e.g., atropine) 
muscarinic receptor antagonism ↑ 
Methylxanthine
(e.g., theophylline) 
Phosphodiesterase inhibitor and adenosine receptors blocker ↑ 
Benzazepine
(e.g., ivabradine) 
HCN blocker ↓ 
Digitalis glycoside
(e.g., digoxin) 
Na+/K+ ATPase inhibitor ↓ 
Ca2+ channel blockers
(e.g., verapamil) 
Ca2+ channel blockers ↓ 
K+ channel blockers
(e.g., amiodarone) 
K+ channel blockers ↓ 

Additionally, bioinformatic pipelines that analyze transcriptional signatures alongside predicted drug effects—using databases of bioactive molecules with drug-like properties [98,123]—could accelerate drug repurposing for SND. While these predictions require experimental validations, advances in data mining are necessary to enhance their reliability. For instance, integrating multiple bioactive molecule databases may empower the discovery of new classes of drugs or analogs. Furthermore, incorporating disease-associated transcriptional signatures and the introduction of artificial intelligence methods such as machine learning in combination with proteomics and metabolomics studies may further increase the accuracy of predictions and expand the therapeutic options for SND.

Understanding the molecular mechanisms and pathways involved in the development of SND holds the potential to uncover novel therapeutic targets (Figure 4B), paving the way for more effective and patient-specific therapies. Much of our current knowledge about the molecular mechanisms governing the SAN function has been derived from animal models, mainly due to practical reasons. Despite numerous structural and functional commonalities across multiple species, differences exist and should be interpreted with caution.

To develop more effective and alternative therapeutic strategies for SND, future research must prioritize a deeper exploration of the cellular and molecular mechanisms that underlie the condition. In this section, we provide our perspective on key topics that deserve further investigation in the context of aging-related SND (Figure 4B). Given the numerous underexplored areas, we emphasize that the mechanisms discussed below are just a few among many that require further studies.

Mechanosensitivity of SAN pacemaker cells

As previously mentioned, voltage-gated ion channels are crucial for SAN automaticity. In addition, pacemaker cells are equipped with a variety of mechanosensitive ion channels that influence their automaticity [124]. A landmark study by Cooper et al. [125] demonstrated that moderate stretching (5–10% of resting cell length) of isolated SAN cells led to an increased beating rate, suggesting that pacemaker cells functionally encode mechanosensitive mechanisms that modulate their chronotropic response.

The positive chronotropic response induced by sustained stretch seems to be linked to the activation of quiescent nodal cells [126]. Interestingly, prolonged stretching (up to 5 min) of SAN elicits a biphasic response, characterized by an immediate acceleration of beating rate followed by a gradual return to pre-stretch levels [127]. This reversible stretch-induced behavior may arise from the activation of slow mechanosensitive channels and mechanochemical signaling pathways [124,128-130]. These ion channels can be classified into two categories: fast (within seconds), directly mechanoactivated (Piezo1-2, TREK-1, TRAAK, and BK) and slow (within minutes), indirectly mechanoresponsive (TRP channels, LRRC8, and ClC). In murine SAN, transcripts for Piezo1, LRRC8a, ANO1, TASK-1, and TRPM7 were found to be more abundant than HCN4 [131]. Although the precise mechanisms by which mechanical forces alter the chronotropic response are not fully understood, it is likely that the activation of mechanochemical pathways, which generate intracellular second messengers like reactive oxygen species, cAMP, and inositol trisphosphate, modifies Ca2+ handling and ion channel activity within the SAN.

Increased and sustained intra-atrial pressures are known contributors to the development of ectopic foci and atrial arrhythmogenesis [132-134]. However, there is limited information regarding the causal role of mechanosensitive pathways in SND. Transcripts for TREK1 and TASK1 were found to be upregulated in aged compared to young [83]. A recent deep RNA sequencing performed on aged and young SAN tissues provides a valuable resource for further exploration into whether other mechanoactivated channels are also altered by aging [135]. While the transcriptional landscape offers insights, the mechanisms driving these transcriptional changes—and, more importantly, the functional implications of mechanoactivated channels in aging-related SND—remain largely unexplored. Given that mechanical forces can be influenced by fibrotic remodeling of aged SAN tissue, elucidating the precise mechanisms involved poses significant experimental challenges. In summary, the interplay between mechanoelectrical and mechanochemical signals within SAN tissue introduces a layer of complexity to the regulation of pacemaker automaticity, forming a novel concept of a ‘mechano clock’ that may affect the dual voltage-Ca2+ clocks. Therefore, a deeper understanding of the regulation, localization, and function of these pathways is essential for considering them as potential therapeutic targets for SND.

Heterogeneity of SAN pacemaker cells

The SAN is a heterogeneous tissue composed of a variety of cell types [98,136]. Sub-specialized pacemaker cells have been classified into three morphologically distinct cells: elongated spindle, spindle, and spider cells [38]. Pacemaker cells are densely packed within the core region of the SAN, initiating the spontaneous depolarizations that set the rhythm of the heart. Characteristically smaller and spindle-shaped, these cells contain fewer myofibrils than neighboring atrial myocytes. Interestingly, a gradual transition in action potential properties from the central SAN area toward its periphery suggests the presence of transitional cells [137]. While not fully characterized, these transitional cells display intermediate characteristics between pacemaker cells and atrial myocytes. Unlike SAN cells from the core region, transitional cells may be interconnected by gap junctions [100]. Although a deep molecular and electrical understanding of each morphologically distinct pacemaker cells warrants investigation, the extent to which they individually contribute to SND in aging has yet to be determined.

A coordinated depolarization of a cluster of SAN pacemaker cells generates a relatively weak current flow, acting as a ‘source’ that diffuses into the adjacent atrial myocardium, which serves as the ‘sink’. This capacity of SAN currents to drive the overwhelming hyperpolarized potentials of well-coupled atrial myocardium highlights the intricate tissue architecture that is composed of specialized branching myofibers from SACPs [100,101,138]. Histological analyses have shown that the volume of SAN cells declines with age [139,140]. Additionally, cellular hypertrophy and increased membrane capacitance have been observed in aged SAN [78,139,140]. Thus, the numerical reduction of SAN pacemaker cells as a result of aging may limit the cluster of electrically coupled cells needed to effectively excite the surrounding atria myocardium, leading to a source-sink mismatch. Although aging is linked with the activation of multiple signaling pathways that promote cell death [141], determining the mechanism mediating the aging-related pacemaker cell loss may open avenues for new therapeutic targets. Moreover, although structural remodeling also occurs in aged SAN, further investigation is needed to determine whether the formation of discontinuous myofiber tracts in specific regions of SACPs is causally related to conduction blocks in aged SAN tissue.

Non-canonical roles of macrophages in SAN function

Many arrhythmic disorders are associated with an inflammatory component. We and others have demonstrated that excessive production of inflammatory cytokines and chemokines contributes to the development of arrhythmogenic substrates [142-147]. Recent discoveries suggest that immune cells can also influence the cardiac rhythm by non-canonical electrotonic coupling with cardiomyocytes [142,148]. Tissue-resident macrophages are the most abundant immune cell type in rodent and human hearts (≈5% total cells), playing a wide range of physiological and pathological roles [149-151]. Single-cell RNA sequencing of mice hearts has identified three transcriptionally distinct macrophage subsets: TLF+ macrophages (expressing Timd4, Lyve1, and Folr2, but lacking Ccr2 and low MHC-II expression), MHC-IIhi macrophages (lacking Timd4, Lyve1, Folr2, and Ccr2), and CCR2+ macrophages (lacking Timd4, Lyve1, and Folr2, but high MHC-II expression) [149-151]. This diversity clarifies the biological distinctions between self-renewing resident macrophages (CCR2) and monocyte-dependent macrophages (CCR2+), shedding light on their specific roles during cardiac injury [149,152,153].

The role of macrophages in the CCS remains largely unexplored. Macrophages were previously shown to directly couple with pacemaker cells in the AVN facilitating the electrical conduction via a Cx43-dependent mechanism [148]. However, to our knowledge, no previous research has specifically addressed the role of resident macrophages in regulating SAN function. In this context, a recent study combined single-nuclei RNA sequencing and spatial transcriptomics to analyze human SAN tissue from individuals without a history of cardiac disease or arrhythmia [98]. Three distinct populations of tissue-resident macrophages were identified, including Lyve1+Igf1+, Lyve1+Timd4+, and Lyve1+ cycling. However, their functional role has yet to be established. Intriguingly, Cx43 or other connexins were minimally expressed in macrophages present in SAN and AVN [98]. Therefore, the mechanisms by which the depletion of resident macrophages, using Cd11b-expressing diphtheria toxin receptor mice, leads to third-degree AV block remain to be further investigated.

In isolated cardiac resident macrophages, four patterns of outward and two patterns of inward-rectifier potassium currents have been described [154]. Kv1.3, Kv1.5, and Kir2.1 were abundantly expressed in macrophages [154], likely contributing to the observed ion currents and potentially influencing cardiac excitability. Given the presence of distinct macrophage subsets in the heart, further research is needed to determine if all these identified subsets similarly affect cardiac electrophysiology. In a mouse model of HF with SND, significant remodeling of the inflammatory proteome was observed [155]. A notable increase in CCR2-expressing macrophages was observed in the SAN of failing hearts, while systemic inhibition of the pro-inflammatory galectin-3 improved the SND [155]. However, this area of research remains largely unexplored in the context of aging-related SND. This knowledge gap highlights the need for future studies to investigate whether resident and/or recruited macrophages contribute to aging-related SND, and to uncover the mechanisms through which they may influence pacemaker function.

Regulation of SAN gene expression

The understanding of the early development of the SAN may reveal molecular insights into a potential loss of gene expression programming in aged pacemaker cells. The first spontaneous Ca2+ oscillations occur in the mouse embryo at around E7.75 [156,157], initiating the uninterrupted automaticity that sustains heart function throughout life. This critical event is the result of a finely orchestrated regulation of transcription factors that guide the differentiation and maturation of pacemaker cells [158-160]. During early cardiogenesis, T-box transcription factors Tbx5, Tbx18, and insulin gene enhancer protein ISL1 give rise to the sinus venosus myocardium. Tbx5 activates short stature homeobox protein 2 (Shox2) and Tbx3, which along with Isl1, regulate the SAN pacemaker gene program [158-160]. Shox2 also inhibits the expression of the homeobox protein NKX2.5, which along with Tbx3 and pituitary homeobox 2 protein (Pitx2) determine the formation of the SAN region and prevents its ‘atrialization’ [6,158].

Transcriptomic analysis of aged SAN tissue revealed an up-regulation of Tbx3 and Tbx5 compared with young counterparts, consistent with increased expression of pacemaker-related genes HCN1 and HCN4 [135]. Despite this, a reduction in If has been consistently reported in aged SAN cells, pointing to complex gene expression regulatory mechanisms in aged pacemaker cells that require further investigation. One limitation of many current studies using next-generation RNA sequencing is the lack of transcriptional changes at single-cell resolution. Pacemaker cells are considered to be large in size for conventional droplet-based platforms, with most of the single-cell RNA sequencing data obtained from SAN cells during embryonic or neonatal stages [161,162]. However, single-nuclei RNA sequencing and newer technologies such as plate-based platforms and spatial transcriptomics can circumvent cell size limitations [98,136]. Given the heterogeneity of pacemaker cells in the SAN [38], embracing the patch-sequencing technology may connect distinct morphology and electrophysiology of pacemaker cells with unique transcriptomic signatures [163].

In recent decades, research on non-coding RNA molecules, particularly microRNAs (miRNAs), has highlighted their critical role as regulators of mRNA and protein expression [164]. Approximately 30% of protein-coding genes are influenced by miRNAs [165], with their expression and function undergoing dynamic changes during development and various pathological conditions [166,167]. miRNAs also regulate genes essential for cardiac pacemaking. Profiling of miRNAs in human SAN tissue has revealed a distinct expression pattern compared to the right atrium, with many SAN-enriched miRNAs predicted to target key pacemaking genes including, Hcn4, Cacna1c, Cacna1d, Kcnj3, Kcnj5, and Ryr2 [168,169]. A summary of the miRNA interaction with ion channels in the SAN is provided in Table 2. While this list focuses on findings from SAN tissue, a broader list of ion channels modulated by miRNAs in atrial and ventricular cells can be found elsewhere [173,174].

Table 2:
miRNAs involved in the regulation of genes related to pacemaking function.
microRNA in the SANGenesRef.
INa miR 153–3 p, miR-486–3p, miR-92a-3p, miR-652–5p, miR-3200–3p, let-7g-3p, mir-1180–3p Scn8a, Scn5a [168,169
If miR-1, mir-30c-5p, miR-133–3p, miR-187–3p, miR-211–5p, miR-370–3p, miR-423–5p, miR-486–3p, miR-652–5p, miR-3200–3p Hcn1, Hcn2, Hcn4 [168-171
IK mir-1247–5p, mir-30c-5p, mir-486–3p, mir-423–5p, mir-1260-a, mir-744–5p, mir-193a-3p, mir-574–5p, mir-133a-3p, mir-215 Kcnj3, Kcnj5, Kcnj11, Kcne1, Kcnq1, Kcna4 [168,169
ICa,L miR 30 c‐5 p, miR-1976, mir-198, mir-153–3p, mir-204–5p, mir-371–3p, mir-938 Cacna1c, Cacna1d [168,169,172
Ryr mir-198, mir-153–3p Ryr2 [168
INCX miR-1–3p Slc8a1 [169
microRNA in the SANGenesRef.
INa miR 153–3 p, miR-486–3p, miR-92a-3p, miR-652–5p, miR-3200–3p, let-7g-3p, mir-1180–3p Scn8a, Scn5a [168,169
If miR-1, mir-30c-5p, miR-133–3p, miR-187–3p, miR-211–5p, miR-370–3p, miR-423–5p, miR-486–3p, miR-652–5p, miR-3200–3p Hcn1, Hcn2, Hcn4 [168-171
IK mir-1247–5p, mir-30c-5p, mir-486–3p, mir-423–5p, mir-1260-a, mir-744–5p, mir-193a-3p, mir-574–5p, mir-133a-3p, mir-215 Kcnj3, Kcnj5, Kcnj11, Kcne1, Kcnq1, Kcna4 [168,169
ICa,L miR 30 c‐5 p, miR-1976, mir-198, mir-153–3p, mir-204–5p, mir-371–3p, mir-938 Cacna1c, Cacna1d [168,169,172
Ryr mir-198, mir-153–3p Ryr2 [168
INCX miR-1–3p Slc8a1 [169

There is compelling evidence that miRNA-based therapeutics may offer promising avenues for the treatment of SND. For instance, the down-regulation of miR-423–5p has been shown to reverse exercise training-induced remodeling of the Hcn4 channel, alleviating sinus bradycardia [175]. Additionally, local delivery of anti-miR-370–3p successfully restored If in a mouse model of HF [170]. Furthermore, our recent research revealed that chemically modified human Tbx18 mRNA is suppressed by endogenous miRNAs. By employing counterstrategies to inhibit these suppressive miRNAs, we were able to achieve robust expression of Tbx18 protein, resulting in increased automaticity in a rat model of AV block [176].

While miRNAs have been implicated in regulating age-related processes across various mammalian tissues, their specific role in aging-related SND has yet to be investigated. Given that a single miRNA can influence hundreds of target genes involved in diverse signaling pathways, miRNA-based therapies could potentially lead to significant therapeutic benefits [166,167,177]. However, the possibility of off-target effects and adverse effects must be carefully considered. Thus, further research is needed to assess the viability and safety of miRNA-based therapies for cardiac rhythm disorders.

Metabolic control of SAN pacemaker cells

Pacemaker cells from SAN tissue maintain a high basal cAMP-PKA signaling driven by Ca2+-dependent activation of adenylyl cyclase [51]. Despite the relatively low density of contractile myofilaments in SAN myocytes compared to ventricular myocytes [97], SAN cells consume more oxygen than ventricular myocytes paced at 3 Hz [178]. However, such elevated demand comes with a high energetic cost, requiring an efficient energy supply and consumption. The SAN is indeed equipped with a dense mitochondrial network with a high basal respiratory rate [178,179]. It was previously shown that decreased production of adenosine triphosphate (ATP) in SAN cells suppressed the automaticity by interfering with basal Ca2+-cAMP/PKA [178] or Ca2+-calmodulin-dependent protein kinase II CaMKII [180] signaling. In contrast, when the HR increases, such as during β-AR stimulation, ATP production must rise to meet the higher energy demand needed to sustain the elevated firing rate. In this context, the mitochondrial calcium uniporter revealed important for the acceleration of HR by facilitating Ca2+ entry into the mitochondrial matrix to enhance energy production via oxidative phosphorylation [181]. A significant remodeling of mitochondria with disruption of mitochondria–SR microdomains was recently described in the SAN of HF mice [179]. Down-regulation of mitofusin-2 was mechanistically linked with impairing mitochondrial function, energy production, and, consequently, SND [179]. AMP-activated protein kinase (AMPK) is a key cellular fuel sensor, detecting energy imbalances during metabolic stress [182], being the AMPK γ2 subunit the most abundant in the SAN [182]. Mice with gain-of-function mutation on R302Q γ2 exhibit sinus bradycardia due to reduced If and local SR Ca2+release in SAN cells [182]. Together, these findings reveal that energy homeostasis regulation in pacemaker cells is critical for preserving SAN automaticity.

The primary pacemaking site within the SAN region shifts in response to physiological stimuli, including activation by the autonomic nervous system [12,84]. Two spatially distinct pacemaker sites coexist within the SAN, identified in the superior and inferior anatomic regions of SAN tissue and preferentially regulating fast and slow HRs, respectively [84]. A recent investigation added an important layer of complexity by demonstrating the influence of anatomical variations of the local vasculature on SAN physiology [183]. High-resolution 3D reconstructions from intact SAN revealed that the superior site is highly vascularized and Hcn4-expressing pacemaker cells have more near contact with blood vessels compared with the inferior site [183]. These findings suggest that high vascular density in the superior SAN region may locally regulate nutrient supply to meet the SAN’s electrical demands. However, further studies are needed to determine whether microvascular rarefaction of SAN tissue occurs with aging. Additionally, the potential mechanistic link between aging-related SND and energy mismatch or metabolic inflexibility remains to be elucidated.

Aging is a natural and unavoidable process that is often associated with SND. Despite significant progress made in understanding aging-related SND, electronic pacemaker implantation remains the only treatment option for these patients, regardless of the underlying mechanism. Extensive research has uncovered key age-related structural and functional changes in the SAN, which correlate with the age-dependent slowing of HR and SAN cells depolarizations. However, correlation does not imply causation, and further mechanistic studies are needed to clarify these connections. While we have highlighted some important gaps in knowledge, it is important to note that much of this research was conducted using preclinical animal models, such as rodents, pigs, and dogs due to practical and logistic reasons. While these models are valuable for mechanistic studies, they do not fully capture the complexity of human physiology, requiring cautious interpretation and conclusions. Additionally, there is a surprising lack of research on potential sex-dependent changes in the aging SAN, an area that warrants further investigation. Thus, expanding our understanding of these key identified areas, and others beyond the scope of this review, is crucial for ‘decoding the clock’ of aging hearts, reducing the reliance on electronic pacemakers, and developing innovative and disease-specific therapies for sinus node dysfunction in the elderly and related diseases.

The authors declare that there are no competing interests associated with the manuscript.

This work was supported by the National Institutes of Health (grants R01 HL135866 and R01 HL147570 to Dr. Cingolani), the American Heart Association (Career Development Award: 940033 and Innovative Project Award: 24IPA1275047 to Dr. Mesquita), California Institute for Regenerative Medicine (CLIN1-14874 to Dr. Cingolani and EDUC4-12751 to Dr. Miguel-dos-Santos), and the Cedars-Sinai Board of Governors.

T.M., R.M.S., and E.C.: Conceptualization, Supervision, Writing—original draft, Writing—review & editing.

AMPK

AMP-activated protein kinase

AVN

atrioventricular node

CCS

cardiac conduction system

CaMKII

Ca2+-calmodulin-dependent protein kinase II

Cav

voltage-gated calcium channel

ECM

extracellular matrix

HCN

hyperpolarization-activated cyclic nucleotide-gated channel

HF

heart failure

HRs

heart rates

Kv

Voltage-gated potassium channels

NCX

sodium-calcium exchanger

PKA

protein kinase A

SAN

sinoatrial node

SERCA

sarcoplasmic reticulum calcium ATPase

SND

SAN dysfunction

SR

sarcoplasmic reticulum

miRNA

microRNA

βAR

beta-adrenergic receptor

1
Opthof
,
T
. (
1988
)
The mammalian sinoatrial node
.
Cardiovasc. Drugs Ther.
1
,
573
597
https://doi.org/10.1007/BF02125744
2
Manoj
,
P.
,
Kim
,
J.A.
,
Kim
,
S.
,
Li
,
T.
,
Sewani
,
M.
,
Chelu
,
M.G.
et al.
(
2023
)
Sinus node dysfunction: current understanding and future directions
.
Am. J. Physiol. Heart Circ. Physiol.
324
,
H259
H278
https://doi.org/10.1152/ajpheart.00618.2022
3
Wallace
,
M.J.
,
El Refaey
,
M.
,
Mesirca
,
P.
,
Hund
,
T.J.
,
Mangoni
,
M.E.
and
Mohler
,
P.J
. (
2021
)
Genetic complexity of sinoatrial node dysfunction
.
Front. Genet.
12
, 654925 https://doi.org/10.3389/fgene.2021.654925
4
Neco
,
P.
,
Torrente
,
A.G.
,
Mesirca
,
P.
,
Zorio
,
E.
,
Liu
,
N.
,
Priori
,
S.G.
et al.
(
2012
)
Paradoxical effect of increased diastolic Ca(2+) release and decreased sinoatrial node activity in a mouse model of catecholaminergic polymorphic ventricular tachycardia
.
Circulation
126
,
392
401
https://doi.org/10.1161/CIRCULATIONAHA.111.075382
5
Wang
,
Y.Y.
,
Mesirca
,
P.
,
Marqués-Sulé
,
E.
,
Zahradnikova
,
A.
Jr
,
Villejoubert
,
O.
,
D’Ocon
,
P.
et al.
(
2017
)
RyR2R420Q catecholaminergic polymorphic ventricular tachycardia mutation induces bradycardia by disturbing the coupled clock pacemaker mechanism
.
JCI Insight
2
,
91872
https://doi.org/10.1172/jci.insight.91872
6
Maarel
,
L.E.
,
Postma
,
A.V.
and
Christoffels
,
V.M
. (
2023
)
Genetics of sinoatrial node function and heart rate disorders
.
Dis. Model. Mech.
16
, dmm050101 https://doi.org/10.1242/dmm.050101
7
Jensen
,
P.N.
,
Gronroos
,
N.N.
,
Chen
,
L.Y.
,
Folsom
,
A.R.
,
deFilippi
,
C.
,
Heckbert
,
S.R.
et al.
(
2014
)
Incidence of and risk factors for sick sinus syndrome in the general population
.
J. Am. Coll. Cardiol.
64
,
531
538
https://doi.org/10.1016/j.jacc.2014.03.056
8
Shiraishi
,
I.
,
Takamatsu
,
T.
,
Minamikawa
,
T.
,
Onouchi
,
Z.
and
Fujita
,
S
. (
1992
)
Quantitative histological analysis of the human sinoatrial node during growth and aging
.
Circulation
85
,
2176
2184
https://doi.org/10.1161/01.cir.85.6.2176
9
Mesquita
,
T.R.R.
,
Zhang
,
R.
,
Couto
,
G.
,
Valle
,
J.
,
Sanchez
,
L.
,
Rogers
,
R.G.
et al.
(
2020
)
Mechanisms of atrial fibrillation in aged rats with heart failure with preserved ejection fraction
.
Heart Rhythm
17
,
1025
1033
https://doi.org/10.1016/j.hrthm.2020.02.007
10
Monfredi
,
O.
and
Boyett
,
M.R
. (
2015
)
Sick sinus syndrome and atrial fibrillation in older persons - a view from the sinoatrial nodal myocyte
.
J. Mol. Cell. Cardiol.
83
,
88
100
https://doi.org/10.1016/j.yjmcc.2015.02.003
11
Sanders
,
P.
,
Kistler
,
P.M.
,
Morton
,
J.B.
,
Spence
,
S.J.
and
Kalman
,
J.M
. (
2004
)
Remodeling of sinus node function in patients with congestive heart failure: reduction in sinus node reserve
.
Circulation
110
,
897
903
https://doi.org/10.1161/01.CIR.0000139336.69955.AB
12
Mesquita
,
T.
,
Zhang
,
R.
,
Cho
,
J.H.
,
Zhang
,
R.
,
Lin
,
Y.N.
,
Sanchez
,
L.
et al.
(
2022
)
Mechanisms of sinoatrial node dysfunction in heart failure with preserved ejection fraction
.
Circulation
145
,
45
60
https://doi.org/10.1161/CIRCULATIONAHA.121.054976
13
Xue
,
J.B.
,
Val-Blasco
,
A.
,
Davoodi
,
M.
,
Gómez
,
S.
,
Yaniv
,
Y.
,
Benitah
,
J.P.
et al.
(
2022
)
Heart failure in mice induces a dysfunction of the sinus node associated with reduced CaMKII signaling
.
J. Gen. Physiol.
154
, e202112895 https://doi.org/10.1085/jgp.202112895
14
Verkerk
,
A.O.
,
Wilders
,
R.
,
Coronel
,
R.
,
Ravesloot
,
J.H.
and
Verheijck
,
E.E
. (
2003
)
Ionic remodeling of sinoatrial node cells by heart failure
.
Circulation
108
,
760
766
https://doi.org/10.1161/01.CIR.0000083719.51661.B9
15
Swaminathan
,
P.D.
,
Purohit
,
A.
,
Soni
,
S.
,
Voigt
,
N.
,
Singh
,
M.V.
,
Glukhov
,
A.V.
et al.
(
2011
)
Oxidized CaMKII causes cardiac sinus node dysfunction in mice
.
J. Clin. Invest.
121
,
3277
3288
https://doi.org/10.1172/JCI57833
16
Gams
,
A.
,
Brennan
,
J.A.
,
Goldrick
,
K.
and
Efimov
,
I.R
. (
2022
)
Molecular and functional remodeling of superior and inferior SAN in a rat model of HCM
.
JACC Clin. Electrophysiol.
8
,
1341
1353
https://doi.org/10.1016/j.jacep.2022.08.003
17
Pattanshetty
,
D.J.
,
Bhat
,
P.K.
,
Chamberlain
,
W.A.
and
Lyons
,
M.R
. (
2013
)
Isolated cardiac involvement in primary amyloidosis: presenting as sick sinus syndrome and heart failure
.
Tex. Heart Inst. J.
40
,
615
618
18
Faraj
,
R.
,
Laktib
,
N.
,
Hilal
,
S.
,
Hassan
,
F.
,
Krimech
,
A.
,
Bouanani
,
A.
et al.
(
2022
)
Systemic sclerosis and tachycardia-bradycardia syndrome: a case report
.
J. Med. Case Rep.
16
, 258 https://doi.org/10.1186/s13256-022-03462-z
19
Inazumi
,
T.
,
Tajima
,
S.
,
Ando
,
T.
and
Shimada
,
M
. (
1995
)
A case of CREST syndrome associated with sick sinus syndrome
.
J. Dermatol.
22
,
801
803
https://doi.org/10.1111/j.1346-8138.1995.tb03925.x
20
Maisch
,
B.
,
Lotze
,
U.
,
Schneider
,
J.
and
Kochsiek
,
K
. (
1986
)
Antibodies to human sinus node in sick sinus syndrome
.
Pacing Clin. Electrophysiol.
9
,
1101
1109
https://doi.org/10.1111/j.1540-8159.1986.tb06677.x
21
Roman-Campos
,
D.
,
Marin-Neto
,
J.A.
,
Santos-Miranda
,
A.
,
Kong
,
N.
,
D’Avila
,
A.
and
Rassi
,
A.
Jr
. (
2024
)
Arrhythmogenic manifestations of chagas disease: perspectives from the bench to bedside
.
Circ. Res.
134
,
1379
1397
https://doi.org/10.1161/CIRCRESAHA.124.324507
22
Elizari
,
M.V.
and
Chiale
,
P.A
. (
1993
)
Cardiac arrhythmias in Chagas’ heart disease
.
J. Cardiovasc. Electrophysiol.
4
,
596
608
https://doi.org/10.1111/j.1540-8167.1993.tb01247.x
23
Buza
,
V.
,
Rajagopalan
,
B.
and
Curtis
,
A.B
. (
2017
)
Cancer treatment-induced Arrhythmias: focus on chemotherapy and targeted therapies
.
Circ. Arrhythm. Electrophysiol.
10
, e005443 https://doi.org/10.1161/CIRCEP.117.005443
24
Nishikawa
,
T.
,
Kunimasa
,
K.
,
Ohta-Ogo
,
K.
,
Ikeda
,
Y.
,
Yasui
,
T.
,
Shioyama
,
W.
et al.
(
2022
)
Sinus node dysfunction co-occurring with immune checkpoint inhibitor-associated Myocarditis
.
Intern. Med.
61
,
2161
2165
https://doi.org/10.2169/internalmedicine.8575-21
25
Wei
,
S.C.
,
Meijers
,
W.C.
,
Axelrod
,
M.L.
,
Anang
,
N.A.A.S.
,
Screever
,
E.M.
,
Wescott
,
E.C.
et al.
(
2021
)
A genetic mouse model recapitulates immune checkpoint inhibitor-associated Myocarditis and supports a mechanism-based therapeutic intervention
.
Cancer Discov.
11
,
614
625
https://doi.org/10.1158/2159-8290.CD-20-0856
26
Curtis
,
A.B.
,
Karki
,
R.
,
Hattoum
,
A.
and
Sharma
,
U.C
. (
2018
)
Arrhythmias in patients ≥80 years of age: pathophysiology, management, and outcomes
.
J. Am. Coll. Cardiol.
71
,
2041
2057
https://doi.org/10.1016/j.jacc.2018.03.019
27
World population prospects
. https://population.un.org/wpp/
28
Choi
,
S.
,
Baudot
,
M.
,
Vivas
,
O.
and
Moreno
,
C.M
. (
2022
)
Slowing down as we age: aging of the cardiac pacemaker’s neural control
.
Geroscience
44
,
1
17
https://doi.org/10.1007/s11357-021-00420-3
29
Peters
,
C.H.
,
Sharpe
,
E.J.
and
Proenza
,
C
. (
2020
)
Cardiac pacemaker activity and aging
.
Annu. Rev. Physiol.
82
,
21
43
https://doi.org/10.1146/annurev-physiol-021119-034453
30
Gaskell
,
W.H
. (
1883
)
On the innervation of the heart, with especial reference to the heart of the tortoise
.
J. Physiol. (Lond.)
4
,
43
230
. https://doi.org/10.1113/jphysiol.1883.sp000121
31
Gaskell
,
W.H
. (
1882
)
Preliminary observations on the innervation of the heart of the tortoise
.
J. Physiol. (Lond.)
3
,
369
379
https://doi.org/10.1113/jphysiol.1882.sp000110
32
Keith
,
A.
and
Flack
,
M
. (
1907
)
The form and nature of the muscular connections between the primary divisions of the vertebrate heart
.
J. Anat. Physiol.
41
,
172
189
33
Shekhar
,
A.
,
Lin
,
X.
,
Liu
,
F.Y.
,
Zhang
,
J.
,
Mo
,
H.
,
Bastarache
,
L.
et al.
(
2016
)
Transcription factor ETV1 is essential for rapid conduction in the heart
.
J. Clin. Invest.
126
,
4444
4459
https://doi.org/10.1172/JCI87968
34
Mesquita
,
T.
,
Miguel-Dos-Santos
,
R.
and
Cingolani
,
E
. (
2024
)
Biological pacemakers: present and future
.
Circ. Res.
134
,
837
841
https://doi.org/10.1161/CIRCRESAHA.123.323180
35
Tsutsui
,
K.
,
Monfredi
,
O.J.
,
Sirenko-Tagirova
,
S.G.
,
Maltseva
,
L.A.
,
Bychkov
,
R.
,
Kim
,
M.S.
et al.
(
2018
)
A coupled-clock system drives the automaticity of human sinoatrial nodal pacemaker cells
.
Sci. Signal.
11
, eaap7608 https://doi.org/10.1126/scisignal.aap7608
36
Hennis
,
K.
,
Biel
,
M.
,
Wahl-Schott
,
C.
and
Fenske
,
S
. (
2021
)
Beyond pacemaking: HCN channels in sinoatrial node function
.
Prog. Biophys. Mol. Biol.
166
,
51
60
https://doi.org/10.1016/j.pbiomolbio.2021.03.004
37
Mangoni
,
M.E.
and
Nargeot
,
J
. (
2008
)
Genesis and regulation of the heart automaticity
.
Physiol. Rev.
88
,
919
982
https://doi.org/10.1152/physrev.00018.2007
38
Verheijck
,
E.E.
,
Wessels
,
A.
,
Ginneken
,
A.C.
,
Bourier
,
J.
,
Markman
,
M.W.
,
Vermeulen
,
J.L.
et al.
(
1998
)
Distribution of atrial and nodal cells within the rabbit sinoatrial node: models of sinoatrial transition
.
Circulation
97
,
1623
1631
https://doi.org/10.1161/01.cir.97.16.1623
39
Gao
,
Z.
,
Chen
,
B.
,
Joiner
,
M.L.A.
,
Wu
,
Y.
,
Guan
,
X.
,
Koval
,
O.M.
et al.
(
2010
)
I(f) and SR Ca(2+) release both contribute to pacemaker activity in canine sinoatrial node cells
.
J. Mol. Cell. Cardiol.
49
,
33
40
https://doi.org/10.1016/j.yjmcc.2010.03.019
40
Peters
,
C.H.
,
Liu
,
P.W.
,
Morotti
,
S.
,
Gantz
,
S.C.
,
Grandi
,
E.
,
Bean
,
B.P.
et al.
(
2021
)
Bidirectional flow of the funny current (If) during the pacemaking cycle in murine sinoatrial node myocytes
.
Proc. Natl. Acad. Sci. U.S.A.
118
, e2104668118 https://doi.org/10.1073/pnas.2104668118
41
Mangoni
,
M.E.
and
Nargeot
,
J
. (
2001
)
Properties of the hyperpolarization-activated current (I(f)) in isolated mouse sino-atrial cells
.
Cardiovasc. Res.
52
,
51
64
https://doi.org/10.1016/s0008-6363(01)00370-4
42
DiFrancesco
,
D.
and
Mangoni
,
M
. (
1994
)
Modulation of single hyperpolarization-activated channels (i(f)) by cAMP in the rabbit sino-atrial node
.
J. Physiol. (Lond.)
474
,
473
482
https://doi.org/10.1113/jphysiol.1994.sp020038
43
Fenske
,
S.
,
Krause
,
S.C.
,
Hassan
,
S.I.H.
,
Becirovic
,
E.
,
Auer
,
F.
,
Bernard
,
R.
et al.
(
2013
)
Sick sinus syndrome in HCN1-deficient mice
.
Circulation
128
,
2585
2594
https://doi.org/10.1161/CIRCULATIONAHA.113.003712
44
Herrmann
,
S.
,
Stieber
,
J.
,
Stöckl
,
G.
,
Hofmann
,
F.
and
Ludwig
,
A
. (
2007
)
HCN4 provides a “depolarization reserve” and is not required for heart rate acceleration in mice
.
EMBO J.
26
,
4423
4432
https://doi.org/10.1038/sj.emboj.7601868
45
Mesirca
,
P.
,
Chemin
,
J.
,
Barrère
,
C.
,
Torre
,
E.
,
Gallot
,
L.
,
Monteil
,
A.
et al.
(
2024
)
Selective blockade of Cav1.2 (α1C) versus Cav1.3 (α1D) L-type calcium channels by the black mamba toxin calciseptine
.
Nat. Commun.
15
,
54
https://doi.org/10.1038/s41467-023-43502-w
46
Christel
,
C.J.
,
Cardona
,
N.
,
Mesirca
,
P.
,
Herrmann
,
S.
,
Hofmann
,
F.
,
Striessnig
,
J.
et al.
(
2012
)
Distinct localization and modulation of Cav1.2 and Cav1.3 L-type Ca2+ channels in mouse sinoatrial node
.
J. Physiol. (Lond.)
590
,
6327
6342
https://doi.org/10.1113/jphysiol.2012.239954
47
Mangoni
,
M.E.
,
Couette
,
B.
,
Bourinet
,
E.
,
Platzer
,
J.
,
Reimer
,
D.
,
Striessnig
,
J.
et al.
(
2003
)
Functional role of L-type Cav1.3 Ca2+ channels in cardiac pacemaker activity
.
Proc. Natl. Acad. Sci. U.S.A.
100
,
5543
5548
https://doi.org/10.1073/pnas.0935295100
48
Torrente
,
A.G.
,
Mesirca
,
P.
,
Neco
,
P.
,
Rizzetto
,
R.
,
Dubel
,
S.
,
Barrere
,
C.
et al.
(
2016
)
L-type Cav1.3 channels regulate ryanodine receptor-dependent Ca2+ release during sino-atrial node pacemaker activity
.
Cardiovasc. Res.
109
,
451
461
https://doi.org/10.1093/cvr/cvw006
49
Lakatta
,
E.G.
,
Maltsev
,
V.A.
and
Vinogradova
,
T.M
. (
2010
)
A coupled system of intracellular Ca2+ clocks and surface membrane voltage clocks controls the timekeeping mechanism of the heart’s pacemaker
.
Circ. Res.
106
,
659
673
https://doi.org/10.1161/CIRCRESAHA.109.206078
50
Vinogradova
,
T.M.
,
Zhou
,
Y.Y.
,
Maltsev
,
V.
,
Lyashkov
,
A.
,
Stern
,
M.
and
Lakatta
,
E.G
. (
2004
)
Rhythmic ryanodine receptor Ca2+ releases during diastolic depolarization of sinoatrial pacemaker cells do not require membrane depolarization
.
Circ. Res.
94
,
802
809
https://doi.org/10.1161/01.RES.0000122045.55331.0F
51
Vinogradova
,
T.M.
,
Lyashkov
,
A.E.
,
Zhu
,
W.
,
Ruknudin
,
A.M.
,
Sirenko
,
S.
,
Yang
,
D.
et al.
(
2006
)
High basal protein kinase a-dependent phosphorylation drives rhythmic internal Ca2+ store oscillations and spontaneous beating of cardiac pacemaker cells
.
Circ. Res.
98
,
505
514
https://doi.org/10.1161/01.RES.0000204575.94040.d1
52
Torrente
,
A.G.
,
Zhang
,
R.
,
Zaini
,
A.
,
Giani
,
J.F.
,
Kang
,
J.
,
Lamp
,
S.T.
et al.
(
2015
)
Burst pacemaker activity of the sinoatrial node in sodium-calcium exchanger knockout mice
.
Proc. Natl. Acad. Sci. U.S.A.
112
,
9769
9774
https://doi.org/10.1073/pnas.1505670112
53
Groenke
,
S.
,
Larson
,
E.D.
,
Alber
,
S.
,
Zhang
,
R.
,
Lamp
,
S.T.
,
Ren
,
X.
et al.
(
2013
)
Complete atrial-specific knockout of sodium-calcium exchange eliminates sinoatrial node pacemaker activity
.
Plos One
8
, e81633 https://doi.org/10.1371/journal.pone.0081633
54
Bidaud
,
I.
,
D’Souza
,
A.
,
Forte
,
G.
,
Torre
,
E.
,
Greuet
,
D.
,
Thirard
,
S.
et al.
(
2020
)
Genetic ablation of G protein-gated inwardly rectifying K+ channels prevents training-induced sinus bradycardia
.
Front. Physiol.
11
, 519382 https://doi.org/10.3389/fphys.2020.519382
55
Clark
,
R.B.
,
Mangoni
,
M.E.
,
Lueger
,
A.
,
Couette
,
B.
,
Nargeot
,
J.
and
Giles
,
W.R
. (
2004
)
A rapidly activating delayed rectifier K+ current regulates pacemaker activity in adult mouse sinoatrial node cells
.
Am. J. Physiol. Heart Circ. Physiol.
286
,
H1757
H1766
https://doi.org/10.1152/ajpheart.00753.2003
56
Mesirca
,
P.
,
Marger
,
L.
,
Toyoda
,
F.
,
Rizzetto
,
R.
,
Audoubert
,
M.
,
Dubel
,
S.
et al.
(
2013
)
The G-protein-gated K+ channel, IKACh, is required for regulation of pacemaker activity and recovery of resting heart rate after sympathetic stimulation
.
J. Gen. Physiol.
142
,
113
126
https://doi.org/10.1085/jgp.201310996
57
Torrente
,
A.G.
,
Zhang
,
R.
,
Wang
,
H.
,
Zaini
,
A.
,
Kim
,
B.
,
Yue
,
X.
et al.
(
2017
)
Contribution of small conductance K+ channels to sinoatrial node pacemaker activity: insights from atrial-specific Na+ /Ca2+ exchange knockout mice
.
J Physiol
595
,
3847
3865
https://doi.org/10.1113/JP274249
58
Aziz
,
Q.
,
Li
,
Y.
and
Tinker
,
A
. (
2018
)
Potassium channels in the sinoatrial node and their role in heart rate control
.
Channels (Austin).
12
,
356
366
https://doi.org/10.1080/19336950.2018.1532255
59
Lai
,
M.H.
,
Wu
,
Y.
,
Gao
,
Z.
,
Anderson
,
M.E.
,
Dalziel
,
J.E.
and
Meredith
,
A.L
. (
2014
)
BK channels regulate sinoatrial node firing rate and cardiac pacing in vivo
.
Am. J. Physiol. Heart Circ. Physiol.
307
,
H1327
H1338
https://doi.org/10.1152/ajpheart.00354.2014
60
Monfredi
,
O.
,
Dobrzynski
,
H.
,
Mondal
,
T.
,
Boyett
,
M.R.
and
Morris
,
G.M
. (
2010
)
The anatomy and physiology of the sinoatrial node-a contemporary review
.
Pacing Clin. Electrophysiol.
33
,
1392
1406
https://doi.org/10.1111/j.1540-8159.2010.02838.x
61
Glukhov
,
A.V.
,
Kalyanasundaram
,
A.
,
Lou
,
Q.
,
Hage
,
L.T.
,
Hansen
,
B.J.
,
Belevych
,
A.E.
et al.
(
2015
)
Calsequestrin 2 deletion causes sinoatrial node dysfunction and atrial arrhythmias associated with altered sarcoplasmic reticulum calcium cycling and degenerative fibrosis within the mouse atrial pacemaker complex1
.
Eur. Heart J.
36
,
686
697
https://doi.org/10.1093/eurheartj/eht452
62
Landstrom
,
A.P.
,
Yang
,
Q.
,
Sun
,
B.
,
Perelli
,
R.M.
,
Bidzimou
,
M.T.
,
Zhang
,
Z.
et al.
(
2023
)
Reduction in junctophilin 2 expression in cardiac nodal tissue results in intracellular calcium-driven increase in nodal cell automaticity
.
Circ. Arrhythm. Electrophysiol.
16
, e010858 https://doi.org/10.1161/CIRCEP.122.010858
63
Mavroidis
,
M.
,
Athanasiadis
,
N.C.
,
Rigas
,
P.
,
Kostavasili
,
I.
,
Kloukina
,
I.
,
Te Rijdt
,
W.P.
et al.
(
2020
)
Desmin is essential for the structure and function of the sinoatrial node: implications for increased arrhythmogenesis
.
Am. J. Physiol. Heart Circ. Physiol.
319
,
H557
H570
https://doi.org/10.1152/ajpheart.00594.2019
64
Mezzano
,
V.
,
Liang
,
Y.
,
Wright
,
A.T.
,
Lyon
,
R.C.
,
Pfeiffer
,
E.
,
Song
,
M.Y.
et al.
(
2016
)
Desmosomal junctions are necessary for adult sinus node function
.
Cardiovasc. Res.
111
,
274
286
https://doi.org/10.1093/cvr/cvw083
65
Egom
,
E.E.
,
Vella
,
K.
,
Hua
,
R.
,
Jansen
,
H.J.
,
Moghtadaei
,
M.
,
Polina
,
I.
et al.
(
2015
)
Impaired sinoatrial node function and increased susceptibility to atrial fibrillation in mice lacking natriuretic peptide receptor C
.
J. Physiol. (Lond.)
593
,
1127
1146
https://doi.org/10.1113/jphysiol.2014.283135
66
Dorey
,
T.W.
,
Mackasey
,
M.
,
Jansen
,
H.J.
,
McRae
,
M.D.
,
Bohne
,
L.J.
,
Liu
,
Y.
et al.
(
2022
)
Natriuretic peptide receptor B maintains heart rate and sinoatrial node function via cyclic GMP-mediated signalling
.
Cardiovasc. Res.
118
,
1917
1931
https://doi.org/10.1093/cvr/cvab245
67
Azer
,
J.
,
Hua
,
R.
,
Vella
,
K.
and
Rose
,
R.A
. (
2012
)
Natriuretic peptides regulate heart rate and sinoatrial node function by activating multiple natriuretic peptide receptors
.
J. Mol. Cell. Cardiol.
53
,
715
724
https://doi.org/10.1016/j.yjmcc.2012.08.020
68
Mackasey
,
M.
,
Egom
,
E.E.
,
Jansen
,
H.J.
,
Hua
,
R.
,
Moghtadaei
,
M.
,
Liu
,
Y.
et al.
(
2018
)
Natriuretic peptide receptor-C protects against angiotensin ii-mediated sinoatrial node disease in mice
.
JACC Basic Transl. Sci.
3
,
824
843
https://doi.org/10.1016/j.jacbts.2018.08.004
69
Zheng
,
M.
,
Li
,
R.G.
,
Song
,
J.
,
Zhao
,
X.
,
Tang
,
L.
,
Erhardt
,
S.
et al.
(
2022
)
Hippo-yap signaling maintains sinoatrial node homeostasis
.
Circulation
146
,
1694
1711
https://doi.org/10.1161/CIRCULATIONAHA.121.058777
70
Lubberding
,
A.F.
,
Veedfald
,
S.
,
Achter
,
J.S.
,
Nissen
,
S.D.
,
Soattin
,
L.
,
Sorrentino
,
A.
et al.
(
2024
)
Glucagon-like peptide-1 increases heart rate by a direct action on the sinus node
.
Cardiovasc. Res.
120
,
1427
1441
https://doi.org/10.1093/cvr/cvae120
71
Pereira
,
C.H.
,
Bare
,
D.J.
,
Rosas
,
P.C.
,
Dias
,
F.A.L.
and
Banach
,
K
. (
2023
)
The role of P21-activated kinase (Pak1) in sinus node function
.
J. Mol. Cell. Cardiol.
179
,
90
101
https://doi.org/10.1016/j.yjmcc.2023.04.004
72
Dorey
,
T.W.
,
McRae
,
M.D.
,
Belke
,
D.D.
and
Rose
,
R.A
. (
2023
)
PDE4D mediates impaired β-adrenergic receptor signalling in the sinoatrial node in mice with hypertensive heart disease
.
Cardiovasc. Res.
119
,
2697
2711
https://doi.org/10.1093/cvr/cvad138
73
Hua
,
R.
,
Adamczyk
,
A.
,
Robbins
,
C.
,
Ray
,
G.
and
Rose
,
R.A
. (
2012
)
Distinct patterns of constitutive phosphodiesterase activity in mouse sinoatrial node and atrial Myocardium
.
Plos One
7
, e47652 https://doi.org/10.1371/journal.pone.0047652
74
Vinogradova
,
T.M.
,
Kobrinsky
,
E.
and
Lakatta
,
E.G
. (
2018
)
Dual activation of phosphodiesterases 3 and 4 regulates basal spontaneous beating rate of cardiac pacemaker cells: role of compartmentalization?
Front. Physiol.
9
, 1301 https://doi.org/10.3389/fphys.2018.01301
75
Vinogradova
,
T.M.
,
Sirenko
,
S.
,
Lukyanenko
,
Y.O.
,
Yang
,
D.
,
Tarasov
,
K.V.
,
Lyashkov
,
A.E.
et al.
(
2018
)
Basal spontaneous firing of rabbit sinoatrial node cells is regulated by dual activation of PDEs (phosphodiesterases) 3 and 4
.
Circ. Arrhythm. Electrophysiol.
11
, e005896 https://doi.org/10.1161/CIRCEP.117.005896
76
Jansen
,
H.J.
,
Moghtadaei
,
M.
,
Rafferty
,
S.A.
,
Belke
,
D.D.
and
Rose
,
R.A
. (
2022
)
Loss of natriuretic peptide receptor C enhances sinoatrial node dysfunction in aging and frail mice
.
J. Gerontol. A Biol. Sci. Med. Sci.
77
,
902
908
https://doi.org/10.1093/gerona/glab357
77
Reddy
,
G.R.
,
Ren
,
L.
,
Thai
,
P.N.
,
Caldwell
,
J.L.
,
Zaccolo
,
M.
,
Bossuyt
,
J.
et al.
(
2022
)
Deciphering cellular signals in adult mouse sinoatrial node cells
.
iScience
25
, 103693 https://doi.org/10.1016/j.isci.2021.103693
78
Larson
,
E.D.
,
St Clair
,
J.R.
,
Sumner
,
W.A.
,
Bannister
,
R.A.
and
Proenza
,
C
. (
2013
)
Depressed pacemaker activity of sinoatrial node myocytes contributes to the age-dependent decline in maximum heart rate
.
Proc. Natl. Acad. Sci. U.S.A.
110
,
18011
18016
https://doi.org/10.1073/pnas.1308477110
79
Sharpe
,
E.J.
,
Larson
,
E.D.
and
Proenza
,
C
. (
2017
)
Cyclic AMP reverses the effects of aging on pacemaker activity and if in sinoatrial node myocytes
.
J. Gen. Physiol.
149
,
237
247
https://doi.org/10.1085/jgp.201611674
80
Protas
,
L.
,
Oren
,
R.V.
,
Clancy
,
C.E.
and
Robinson
,
R.B
. (
2010
)
Age-dependent changes in Na current magnitude and TTX-sensitivity in the canine sinoatrial node
.
J. Mol. Cell. Cardiol.
48
,
172
180
https://doi.org/10.1016/j.yjmcc.2009.07.028
81
Choi
,
S.
,
Vivas
,
O.
,
Baudot
,
M.
and
Moreno
,
C.M
. (
2022
)
Aging alters the formation and functionality of signaling microdomains between L-type calcium channels and β2-adrenergic receptors in cardiac pacemaker cells
.
Front. Physiol.
13
, 805909 https://doi.org/10.3389/fphys.2022.805909
82
Liu
,
J.
,
Sirenko
,
S.
,
Juhaszova
,
M.
,
Sollott
,
S.J.
,
Shukla
,
S.
,
Yaniv
,
Y.
et al.
(
2014
)
Age-associated abnormalities of intrinsic automaticity of sinoatrial nodal cells are linked to deficient cAMP-PKA-Ca(2+) signaling
.
Am. J. Physiol. Heart Circ. Physiol.
306
,
H1385
H1397
https://doi.org/10.1152/ajpheart.00088.2014
83
Tellez
,
J.O.
,
Mczewski
,
M.
,
Yanni
,
J.
,
Sutyagin
,
P.
,
Mackiewicz
,
U.
,
Atkinson
,
A.
et al.
(
2011
)
Ageing-dependent remodelling of ion channel and Ca2+ clock genes underlying sino-atrial node pacemaking
.
Exp. Physiol.
96
,
1163
1178
https://doi.org/10.1113/expphysiol.2011.057752
84
Brennan
,
J.A.
,
Chen
,
Q.
,
Gams
,
A.
,
Dyavanapalli
,
J.
,
Mendelowitz
,
D.
,
Peng
,
W.
et al.
(
2020
)
Evidence of superior and inferior sinoatrial nodes in the mammalian heart
.
JACC Clin. Electrophysiol.
6
,
1827
1840
https://doi.org/10.1016/j.jacep.2020.09.012
85
Witte
,
K.K.A.
,
Cleland
,
J.G.F.
and
Clark
,
A.L
. (
2006
)
Chronic heart failure, chronotropic incompetence, and the effects of beta blockade
.
Heart
92
,
481
486
https://doi.org/10.1136/hrt.2004.058073
86
Brubaker
,
P.H.
and
Kitzman
,
D.W
. (
2011
)
Chronotropic incompetence: causes, consequences, and management
.
Circulation
123
,
1010
1020
https://doi.org/10.1161/CIRCULATIONAHA.110.940577
87
Stratton
,
J.R.
,
Cerqueira
,
M.D.
,
Schwartz
,
R.S.
,
Levy
,
W.C.
,
Veith
,
R.C.
,
Kahn
,
S.E.
et al.
(
1992
)
Differences in cardiovascular responses to isoproterenol in relation to age and exercise training in healthy men
.
Circulation
86
,
504
512
https://doi.org/10.1161/01.cir.86.2.504
88
Moen
,
J.M.
,
Matt
,
M.G.
,
Ramirez
,
C.
,
Tarasov
,
K.V.
,
Chakir
,
K.
,
Tarasova
,
Y.S.
et al.
(
2019
)
Overexpression of a neuronal type adenylyl cyclase (type 8) in sinoatrial node markedly impacts heart rate and rhythm
.
Front. Neurosci.
13
, 615 https://doi.org/10.3389/fnins.2019.00615
89
Ren
,
L.
,
Thai
,
P.N.
,
Gopireddy
,
R.R.
,
Timofeyev
,
V.
,
Ledford
,
H.A.
,
Woltz
,
R.L.
et al.
(
2022
)
Adenylyl cyclase isoform 1 contributes to sinoatrial node automaticity via functional microdomains
.
JCI Insight
7
, e162602 https://doi.org/10.1172/jci.insight.162602
90
St Clair
,
J.R.
,
Liao
,
Z.
,
Larson
,
E.D.
and
Proenza
,
C
. (
2013
)
PKA-independent activation of I(f) by cAMP in mouse sinoatrial myocytes
.
Channels (Austin).
7
,
318
321
https://doi.org/10.4161/chan.25293
91
Vinogradova
,
T.M.
,
Bogdanov
,
K.Y.
and
Lakatta
,
E.G
. (
2002
)
Beta-adrenergic stimulation modulates ryanodine receptor Ca(2+) release during diastolic depolarization to accelerate pacemaker activity in rabbit sinoatrial nodal cells
.
Circ. Res.
90
,
73
79
https://doi.org/10.1161/hh0102.102271
92
Liao
,
Z.
,
Lockhead
,
D.
,
Larson
,
E.D.
and
Proenza
,
C
. (
2010
)
Phosphorylation and modulation of hyperpolarization-activated HCN4 channels by protein kinase A in the mouse sinoatrial node
.
J. Gen. Physiol.
136
,
247
258
https://doi.org/10.1085/jgp.201010488
93
Vinogradova
,
T.M.
,
Brochet
,
D.X.P.
,
Sirenko
,
S.
,
Li
,
Y.
,
Spurgeon
,
H.
and
Lakatta
,
E.G
. (
2010
)
Sarcoplasmic reticulum Ca2+ pumping kinetics regulates timing of local Ca2+ releases and spontaneous beating rate of rabbit sinoatrial node pacemaker cells
.
Circ. Res.
107
,
767
775
https://doi.org/10.1161/CIRCRESAHA.110.220517
94
Wu
,
Y.
,
Valdivia
,
H.H.
,
Wehrens
,
X.H.T.
and
Anderson
,
M.E
. (
2016
)
A single protein kinase a or calmodulin kinase II site does not control the cardiac pacemaker Ca2+ clock
.
Circ. Arrhythm. Electrophysiol.
9
, e003180 https://doi.org/10.1161/CIRCEP.115.003180
95
Hennis
,
K.
,
Piantoni
,
C.
,
Biel
,
M.
,
Fenske
,
S.
and
Wahl-Schott
,
C
. (
2024
)
Pacemaker channels and the chronotropic response in health and disease
.
Circ. Res.
134
,
1348
1378
https://doi.org/10.1161/CIRCRESAHA.123.323250
96
Hardouin
,
S.
,
Bourgeois
,
F.
,
Toraasson
,
M.
,
Oubenaissa
,
A.
,
Elalouf
,
J.M.
,
Fellmann
,
D.
et al.
(
1998
)
Beta-adrenergic and muscarinic receptor mRNA accumulation in the sinoatrial node area of adult and senescent rat hearts
.
Mech. Ageing Dev.
100
,
277
297
https://doi.org/10.1016/s0047-6374(97)00142-5
97
Boyett
,
M.R.
,
Honjo
,
H.
and
Kodama
,
I
. (
2000
)
The sinoatrial node, a heterogeneous pacemaker structure
.
Cardiovasc. Res.
47
,
658
687
https://doi.org/10.1016/s0008-6363(00)00135-8
98
Kanemaru
,
K.
,
Cranley
,
J.
,
Muraro
,
D.
,
Miranda
,
A.M.A.
,
Ho
,
S.Y.
,
Wilbrey-Clark
,
A.
et al.
(
2023
)
Spatially resolved multiomics of human cardiac niches
.
Nature
619
,
801
810
https://doi.org/10.1038/s41586-023-06311-1
99
Fedorov
,
V.V.
,
Schuessler
,
R.B.
,
Hemphill
,
M.
,
Ambrosi
,
C.M.
,
Chang
,
R.
,
Voloshina
,
A.S.
et al.
(
2009
)
Structural and functional evidence for discrete exit pathways that connect the canine sinoatrial node and atria
.
Circ. Res.
104
,
915
923
https://doi.org/10.1161/CIRCRESAHA.108.193193
100
Csepe
,
T.A.
,
Zhao
,
J.
,
Hansen
,
B.J.
,
Li
,
N.
,
Sul
,
L.V.
,
Lim
,
P.
et al.
(
2016
)
Human sinoatrial node structure: 3D microanatomy of sinoatrial conduction pathways
.
Prog. Biophys. Mol. Biol.
120
,
164
178
https://doi.org/10.1016/j.pbiomolbio.2015.12.011
101
Li
,
N.
,
Hansen
,
B.J.
,
Csepe
,
T.A.
,
Zhao
,
J.
,
Ignozzi
,
A.J.
,
Sul
,
L.V.
et al.
(
2017
)
Redundant and diverse intranodal pacemakers and conduction pathways protect the human sinoatrial node from failure
.
Sci. Transl. Med.
9
, eaam5607 https://doi.org/10.1126/scitranslmed.aam5607
102
Jones
,
S.A.
,
Lancaster
,
M.K.
and
Boyett
,
M.R
. (
2004
)
Ageing‐related changes of connexins and conduction within the sinoatrial node
.
J. Physiol. (Lond.)
560
,
429
437
https://doi.org/10.1113/jphysiol.2004.072108
103
Moghtadaei
,
M.
,
Jansen
,
H.J.
,
Mackasey
,
M.
,
Rafferty
,
S.A.
,
Bogachev
,
O.
,
Sapp
,
J.L.
et al.
(
2016
)
The impacts of age and frailty on heart rate and sinoatrial node function
.
J. Physiol. (Lond.)
594
,
7105
7126
https://doi.org/10.1113/JP272979
104
Kalyanasundaram
,
A.
,
Li
,
N.
,
Gardner
,
M.L.
,
Artiga
,
E.J.
,
Hansen
,
B.J.
,
Webb
,
A.
et al.
(
2021
)
Fibroblast-specific proteotranscriptomes reveal distinct fibrotic signatures of human sinoatrial node in nonfailing and failing hearts
.
Circulation
144
,
126
143
https://doi.org/10.1161/CIRCULATIONAHA.120.051583
105
Smithers
,
R.L.
,
Kao
,
H.K.J.
,
Zeigler
,
S.
,
Yechikov
,
S.
,
Nolta
,
J.A.
,
Chan
,
J.W.
et al.
(
2021
)
Making heads or tails of the large mammalian sinoatrial node micro-organization
.
Circ. Arrhythm. Electrophysiol.
14
, e010465 https://doi.org/10.1161/CIRCEP.121.010465
106
Csepe
,
T.A.
,
Kalyanasundaram
,
A.
,
Hansen
,
B.J.
,
Zhao
,
J.
and
Fedorov
,
V.V
. (
2015
)
Fibrosis: a structural modulator of sinoatrial node physiology and dysfunction
.
Front. Physiol.
6
, 37 https://doi.org/10.3389/fphys.2015.00037
107
Thery
,
C.
,
Gosselin
,
B.
,
Lekieffre
,
J.
and
Warembourg
,
H
. (
1977
)
Pathology of sinoatrial node. correlations with electrocardiographic findings in 111 patients
.
Am. Heart J.
93
,
735
740
https://doi.org/10.1016/s0002-8703(77)80070-7
108
Inoue
,
S.
,
Shinohara
,
F.
,
Niitani
,
H.
and
Gotoh
,
K
. (
1986
)
A new method for the histological study of aging changes in the sinoatrial node
.
Jpn. Heart J.
27
,
653
660
https://doi.org/10.1536/ihj.27.653
109
Alings
,
A.M.
,
Abbas
,
R.F.
and
Bouman
,
L.N
. (
1995
)
Age-related changes in structure and relative collagen content of the human and feline sinoatrial node. a comparative study
.
Eur. Heart J.
16
,
1655
1667
https://doi.org/10.1093/oxfordjournals.eurheartj.a060792
110
DiFrancesco
,
M.L.
,
Marrot
,
M.
,
Torre
,
E.
,
Mesirca
,
P.
,
Davaze
,
R.
,
Lautier
,
C.
et al.
(
2023
)
Characterization of sinoatrial automaticity in microcebus murinus to study the effect of aging on cardiac activity and the correlation with longevity
.
Sci. Rep.
13
, 3054 https://doi.org/10.1038/s41598-023-29723-5
111
Sanders
,
P.
,
Morton
,
J.B.
,
Kistler
,
P.M.
,
Spence
,
S.J.
,
Davidson
,
N.C.
,
Hussin
,
A.
et al.
(
2004
)
Electrophysiological and electroanatomic characterization of the atria in sinus node disease: evidence of diffuse atrial remodeling
.
Circulation
109
,
1514
1522
https://doi.org/10.1161/01.CIR.0000121734.47409.AA
112
Kusumoto
,
F.M.
,
Schoenfeld
,
M.H.
,
Barrett
,
C.
,
Edgerton
,
J.R.
,
Ellenbogen
,
K.A.
,
Gold
,
M.R.
et al.
(
2019
)
2018 ACC/AHA/HRS guideline on the evaluation and management of patients with bradycardia and cardiac conduction delay: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines, and the Heart Rhythm Society
.
J. Am. Coll. Cardiol.
74
,
932
987
https://doi.org/10.1016/j.jacc.2018.10.043
113
Mesirca
,
P.
,
Fedorov
,
V.V.
,
Hund
,
T.J.
,
Torrente
,
A.G.
,
Bidaud
,
I.
,
Mohler
,
P.J.
et al.
(
2021
)
Pharmacologic approach to sinoatrial node dysfunction
.
Annu. Rev. Pharmacol. Toxicol.
61
,
757
778
https://doi.org/10.1146/annurev-pharmtox-031120-115815
114
Vries
,
L.M.
,
Dijk
,
W.A.
,
Hooijschuur
,
C.A.M.
,
Leening
,
M.J.G.
,
Stricker
,
B.H.C.
and
Hemel
,
N.M
. (
2017
)
Utilisation of cardiac pacemakers over a 20-year period: results from a nationwide pacemaker registry
.
Neth. Heart J.
25
,
47
55
https://doi.org/10.1007/s12471-016-0880-0
115
Zhan
,
C.
,
Baine
,
W.B.
,
Sedrakyan
,
A.
and
Steiner
,
C
. (
2008
)
Cardiac device implantation in the United States from 1997 through 2004: a population-based analysis
.
J. Gen. Intern. Med.
23
,
13
19
https://doi.org/10.1007/s11606-007-0392-0
116
Lamas
,
G.A.
,
Orav
,
E.J.
,
Stambler
,
B.S.
,
Ellenbogen
,
K.A.
,
Sgarbossa
,
E.B.
,
Huang
,
S.K.
et al.
(
1998
)
Quality of life and clinical outcomes in elderly patients treated with ventricular pacing as compared with dual-chamber pacing. Pacemaker selection in the elderly investigators
.
N. Engl. J. Med.
338
,
1097
1104
https://doi.org/10.1056/NEJM199804163381602
117
Shen
,
W.K.
,
Hayes
,
D.L.
,
Hammill
,
S.C.
,
Bailey
,
K.R.
,
Ballard
,
D.J.
and
Gersh
,
B.J
. (
1996
)
Survival and functional independence after implantation of a permanent pacemaker in octogenarians and nonagenarians. A population-based study
.
Ann. Intern. Med.
125
,
476
480
https://doi.org/10.7326/0003-4819-125-6-199609150-00008
118
Strauss
,
H.D.
and
Berman
,
N.D
. (
1978
)
Permanent pacing in the elderly
.
Pacing Clin. Electrophysiol.
1
,
458
464
https://doi.org/10.1111/j.1540-8159.1978.tb03507.x
119
Pyatt
,
J.R.
,
Somauroo
,
J.D.
,
Jackson
,
M.
,
Grayson
,
A.D.
,
Osula
,
S.
,
Aggarwal
,
R.K.
et al.
(
2002
)
Long-term survival after permanent pacemaker implantation: analysis of predictors for increased mortality
.
Europace
4
,
113
119
https://doi.org/10.1053/eupc.2002.0233
120
Brunner
,
M.
,
Olschewski
,
M.
,
Geibel
,
A.
,
Bode
,
C.
and
Zehender
,
M
. (
2004
)
Long-term survival after pacemaker implantation. Prognostic importance of gender and baseline patient characteristics
.
Eur. Heart J.
25
,
88
95
https://doi.org/10.1016/j.ehj.2003.10.022
121
Udo
,
E.O.
,
Hemel
,
N.M.
,
Zuithoff
,
N.P.A.
,
Kelder
,
J.C.
,
Crommentuijn
,
H.A.
,
Koopman-Verhagen
,
A.M.
et al.
(
2012
)
Long-term outcome of cardiac pacing in octogenarians and nonagenarians
.
Europace
14
,
502
508
https://doi.org/10.1093/europace/eur329
122
Cingolani
,
E.
,
Goldhaber
,
J.I.
and
Marbán
,
E
. (
2018
)
Next-generation pacemakers: from small devices to biological pacemakers
.
Nat. Rev. Cardiol.
15
,
139
150
https://doi.org/10.1038/nrcardio.2017.165
123
Wang
,
Y.Y.
,
Kang
,
H.
,
Xu
,
T.
,
Hao
,
L.
,
Bao
,
Y.
and
Jia
,
P
. (
2022
)
CeDR Atlas: a knowledgebase of cellular drug response
.
Nucleic Acids Res.
50
,
D1164
D1171
https://doi.org/10.1093/nar/gkab897
124
Quinn
,
T.A.
and
Kohl
,
P
. (
2021
)
Cardiac mechano-electric coupling: acute effects of mechanical stimulation on heart rate and rhythm
.
Physiol. Rev.
101
,
37
92
https://doi.org/10.1152/physrev.00036.2019
125
Cooper
,
P.J.
,
Lei
,
M.
,
Cheng
,
L.X.
and
Kohl
,
P
. (
2000
)
Selected contribution: axial stretch increases spontaneous pacemaker activity in rabbit isolated sinoatrial node cells
.
J. Appl. Physiol. (1985)
89
,
2099
2104
https://doi.org/10.1152/jappl.2000.89.5.2099
126
Lange
,
G.
,
Lu
,
H.H.
,
Chang
,
A.
and
Brooks
,
C.M
. (
1966
)
Effect of stretch on the isolated cat sinoatrial node
.
Am. J. Physiol.
211
,
1192
1196
https://doi.org/10.1152/ajplegacy.1966.211.5.1192
127
Brooks
,
C.M.
,
Lu
,
H.H.
,
Lange
,
G.
,
Mangi
,
R.
,
Shaw
,
R.B.
and
Geoly
,
K
. (
1966
)
Effects of localized stretch of the sinoatrial node region of the dog heart
.
Am. J. Physiol.
211
,
1197
1202
https://doi.org/10.1152/ajplegacy.1966.211.5.1197
128
Huang
,
Z.M.
,
Prasad
,
C.
,
Britton
,
F.C.
,
Ye
,
L.L.
,
Hatton
,
W.J.
and
Duan
,
D
. (
2009
)
Functional role of CLC-2 chloride inward rectifier channels in cardiac sinoatrial nodal pacemaker cells
.
J. Mol. Cell. Cardiol.
47
,
121
132
https://doi.org/10.1016/j.yjmcc.2009.04.008
129
Seol
,
C.A.
,
Kim
,
W.T.
,
Ha
,
J.M.
,
Choe
,
H.
,
Jang
,
Y.J.
,
Youm
,
J.B.
et al.
(
2008
)
Stretch-activated currents in cardiomyocytes isolated from rabbit pulmonary veins
.
Prog. Biophys. Mol. Biol.
97
,
217
231
https://doi.org/10.1016/j.pbiomolbio.2008.02.008
130
Petroff
,
M.G.
,
Kim
,
S.H.
,
Pepe
,
S.
,
Dessy
,
C.
,
Marbán
,
E.
,
Balligand
,
J.L.
et al.
(
2001
)
Endogenous nitric oxide mechanisms mediate the stretch dependence of Ca2+ release in cardiomyocytes
.
Nat. Cell Biol.
3
,
867
873
https://doi.org/10.1038/ncb1001-867
131
Turner
,
D.
,
Kang
,
C.
,
Mesirca
,
P.
,
Hong
,
J.
,
Mangoni
,
M.E.
,
Glukhov
,
A.V.
et al.
(
2021
)
Electrophysiological and molecular mechanisms of sinoatrial node mechanosensitivity
.
Front. Cardiovasc. Med.
8
, 662410 https://doi.org/10.3389/fcvm.2021.662410
132
Egorov
,
Y.V.
,
Lang
,
D.
,
Tyan
,
L.
,
Turner
,
D.
,
Lim
,
E.
,
Piro
,
Z.D.
et al.
(
2019
)
Caveolae-mediated activation of mechanosensitive chloride channels in pulmonary veins triggers atrial arrhythmogenesis
.
J. Am. Heart Assoc.
8
, e012748 https://doi.org/10.1161/JAHA.119.012748
133
Bode
,
F.
,
Katchman
,
A.
,
Woosley
,
R.L.
and
Franz
,
M.R
. (
2000
)
Gadolinium decreases stretch-induced vulnerability to atrial fibrillation
.
Circulation
101
,
2200
2205
https://doi.org/10.1161/01.cir.101.18.2200
134
Chang
,
S.L.
,
Chen
,
Y.C.
,
Chen
,
Y.J.
,
Wangcharoen
,
W.
,
Lee
,
S.H.
,
Lin
,
C.I.
et al.
(
2007
)
Mechanoelectrical feedback regulates the arrhythmogenic activity of pulmonary veins
.
Heart
93
,
82
88
https://doi.org/10.1136/hrt.2006.089359
135
Qu
,
J.H.
,
Tarasov
,
K.V.
,
Tarasova
,
Y.S.
,
Chakir
,
K.
and
Lakatta
,
E.G
. (
2023
)
Transcriptome of left ventricle and sinoatrial node in young and old C57 mice
.
Fortune J. Health Sci.
6
,
332
356
https://doi.org/10.26502/fjhs.134
136
Linscheid
,
N.
,
Logantha
,
S.J.R.J.
,
Poulsen
,
P.C.
,
Zhang
,
S.
,
Schrölkamp
,
M.
,
Egerod
,
K.L.
et al.
(
2019
)
Quantitative proteomics and single-nucleus transcriptomics of the sinus node elucidates the foundation of cardiac pacemaking
.
Nat. Commun.
10
, 2889 https://doi.org/10.1038/s41467-019-10709-9
137
Kodama
,
I.
and
Boyett
,
M.R
. (
1985
)
Regional differences in the electrical activity of the rabbit sinus node
.
Pflugers Arch.
404
,
214
226
https://doi.org/10.1007/BF00581242
138
Fedorov
,
V.V.
,
Glukhov
,
A.V.
,
Chang
,
R.
,
Kostecki
,
G.
,
Aferol
,
H.
,
Hucker
,
W.J.
et al.
(
2010
)
Optical mapping of the isolated coronary-perfused human sinus node
.
J. Am. Coll. Cardiol.
56
,
1386
1394
https://doi.org/10.1016/j.jacc.2010.03.098
139
Yanni
,
J.
,
Tellez
,
J.O.
,
Sutyagin
,
P.V.
,
Boyett
,
M.R.
and
Dobrzynski
,
H
. (
2010
)
Structural remodelling of the sinoatrial node in obese old rats
.
J. Mol. Cell. Cardiol.
48
,
653
662
https://doi.org/10.1016/j.yjmcc.2009.08.023
140
Melo
,
S.R.
,
Souza
,
R.R.
and
Mandarim-de-Lacerda
,
C.A
. (
2002
)
Stereologic study of the sinoatrial node of rats - age related changes
.
Biogerontology
3
,
383
390
https://doi.org/10.1023/a:1021376002896
141
Tower
,
J
. (
2015
)
Programmed cell death in aging
.
Ageing Res. Rev.
23
,
90
https://doi.org/10.1016/j.arr.2015.04.002
142
Grune
,
J.
,
Yamazoe
,
M.
and
Nahrendorf
,
M
. (
2021
)
Electroimmunology and cardiac arrhythmia
.
Nat. Rev. Cardiol.
18
,
547
564
https://doi.org/10.1038/s41569-021-00520-9
143
Mesquita
,
T.
,
Lin
,
Y.N.
,
Chen
,
S.
,
Lee
,
Y.
,
Miguel-Dos-Santos
,
R.
,
Atici
,
A.E.
et al.
(
2024
)
Inhibition of IL-1 ameliorates cardiac dysfunction and Arrhythmias in a murine model of kawasaki disease
.
Arterioscler. Thromb. Vasc. Biol.
44
,
e117
e130
https://doi.org/10.1161/ATVBAHA.123.320382
144
Mesquita
,
T.
and
Cingolani
,
E
. (
2024
)
Targeting arrhythmogenic macrophages: lessons learned from arrhythmogenic cardiomyopathy
.
J. Clin. Invest.
134
, e180482 https://doi.org/10.1172/JCI180482
145
Zhang
,
R.
,
Mesquita
,
T.
,
Cho
,
J.H.
,
Li
,
C.
,
Sanchez
,
L.
,
Holm
,
K.
et al.
(
2023
)
Systemic delivery of extracellular vesicles attenuates atrial fibrillation in heart failure with preserved ejection fraction
.
JACC Clin. Electrophysiol.
9
,
147
158
https://doi.org/10.1016/j.jacep.2022.09.012
146
Lin
,
Y.N.
,
Mesquita
,
T.
,
Sanchez
,
L.
,
Chen
,
Y.H.
,
Liu
,
W.
,
Li
,
C.
et al.
(
2021
)
Extracellular vesicles from immortalized cardiosphere-derived cells attenuate arrhythmogenic cardiomyopathy in desmoglein-2 mutant mice
.
Eur. Heart J.
42
,
3558
3571
https://doi.org/10.1093/eurheartj/ehab419
147
Chelko
,
S.P.
,
Penna
,
V.R.
,
Engel
,
M.
,
Shiel
,
E.A.
,
Centner
,
A.M.
,
Farra
,
W.
et al.
(
2024
)
NFĸB signaling drives myocardial injury via CCR2+ macrophages in a preclinical model of arrhythmogenic cardiomyopathy
.
J. Clin. Invest.
134
, e172014 https://doi.org/10.1172/JCI172014
148
Hulsmans
,
M.
,
Clauss
,
S.
,
Xiao
,
L.
,
Aguirre
,
A.D.
,
King
,
K.R.
,
Hanley
,
A.
et al.
(
2017
)
Macrophages facilitate electrical conduction in the heart
.
Cell
169
,
510
522
https://doi.org/10.1016/j.cell.2017.03.050
149
Dick
,
S.A.
,
Macklin
,
J.A.
,
Nejat
,
S.
,
Momen
,
A.
,
Clemente-Casares
,
X.
,
Althagafi
,
M.G.
et al.
(
2019
)
Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction
.
Nat. Immunol.
20
,
29
39
https://doi.org/10.1038/s41590-018-0272-2
150
Dick
,
S.A.
,
Wong
,
A.
,
Hamidzada
,
H.
,
Nejat
,
S.
,
Nechanitzky
,
R.
,
Vohra
,
S.
et al.
(
2022
)
Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles
.
Sci. Immunol.
7
, eabf7777 https://doi.org/10.1126/sciimmunol.abf7777
151
Holt
,
M.
,
Lin
,
J.
,
Cicka
,
M.
,
Wong
,
A.
,
Epelman
,
S.
and
Lavine
,
K.J
. (
2024
)
Dissecting and visualizing the functional diversity of cardiac macrophages
.
Circ. Res.
134
,
1791
1807
https://doi.org/10.1161/CIRCRESAHA.124.323817
152
Bajpai
,
G.
,
Bredemeyer
,
A.
,
Li
,
W.
,
Zaitsev
,
K.
,
Koenig
,
A.L.
,
Lokshina
,
I.
et al.
(
2019
)
Tissue resident CCR2- and CCR2+ cardiac macrophages differentially orchestrate monocyte recruitment and fate specification following myocardial injury
.
Circ. Res.
124
,
263
278
https://doi.org/10.1161/CIRCRESAHA.118.314028
153
Bajpai
,
G.
,
Schneider
,
C.
,
Wong
,
N.
,
Bredemeyer
,
A.
,
Hulsmans
,
M.
,
Nahrendorf
,
M.
et al.
(
2018
)
The human heart contains distinct macrophage subsets with divergent origins and functions
.
Nat. Med.
24
,
1234
1245
https://doi.org/10.1038/s41591-018-0059-x
154
Simon-Chica
,
A.
,
Fernández
,
M.C.
,
Wülfers
,
E.M.
,
Lother
,
A.
,
Hilgendorf
,
I.
,
Seemann
,
G.
et al.
(
2022
)
Novel insights into the electrophysiology of murine cardiac macrophages: relevance of voltage-gated potassium channels
.
Cardiovasc. Res.
118
,
798
813
https://doi.org/10.1093/cvr/cvab126
155
Kahnert
,
K.
,
Soattin
,
L.
,
Mills
,
R.W.
,
Wilson
,
C.
,
Maurya
,
S.
,
Sorrentino
,
A.
et al.
(
2024
)
Proteomics couples electrical remodelling to inflammation in a murine model of heart failure with sinus node dysfunction
.
Cardiovasc. Res.
120
,
927
942
https://doi.org/10.1093/cvr/cvae054
156
Tyser
,
R.C.
,
Miranda
,
A.M.
,
Chen
,
C.M.
,
Davidson
,
S.M.
,
Srinivas
,
S.
and
Riley
,
P.R
. (
2016
)
Calcium handling precedes cardiac differentiation to initiate the first heartbeat
.
Elife
5
, e17113 https://doi.org/10.7554/eLife.17113
157
Jia
,
B.Z.
,
Qi
,
Y.
,
Wong-Campos
,
J.D.
,
Megason
,
S.G.
and
Cohen
,
A.E
. (
2023
)
A bioelectrical phase transition patterns the first vertebrate heartbeats
.
Nature
622
,
149
155
https://doi.org/10.1038/s41586-023-06561-z
158
Eif
,
V.W.W.
,
Devalla
,
H.D.
,
Boink
,
G.J.J.
and
Christoffels
,
V.M
. (
2018
)
Transcriptional regulation of the cardiac conduction system
.
Nat. Rev. Cardiol.
15
,
617
630
https://doi.org/10.1038/s41569-018-0031-y
159
Bhattacharyya
,
S.
and
Munshi
,
N.V
. (
2020
)
Development of the cardiac conduction system
.
Cold Spring Harb. Perspect. Biol.
12
, a037408 https://doi.org/10.1101/cshperspect.a037408
160
Mandla
,
R.
,
Jung
,
C.
and
Vedantham
,
V
. (
2021
)
Transcriptional and epigenetic landscape of cardiac pacemaker cells: insights into cellular specialization in the sinoatrial node
.
Front. Physiol.
12
, 712666 https://doi.org/10.3389/fphys.2021.712666
161
Goodyer
,
W.R.
,
Beyersdorf
,
B.M.
,
Paik
,
D.T.
,
Tian
,
L.
,
Li
,
G.
,
Buikema
,
J.W.
et al.
(
2019
)
Transcriptomic profiling of the developing cardiac conduction system at single-cell resolution
.
Circ. Res.
125
,
379
397
https://doi.org/10.1161/CIRCRESAHA.118.314578
162
Hill
,
M.C.
,
Kadow
,
Z.A.
,
Li
,
L.
,
Tran
,
T.T.
,
Wythe
,
J.D.
and
Martin
,
J.F
. (
2019
)
A cellular atlas of Pitx2-dependent cardiac development
.
Development
146
, dev180398 https://doi.org/10.1242/dev.180398
163
Lipovsek
,
M.
,
Bardy
,
C.
,
Cadwell
,
C.R.
,
Hadley
,
K.
,
Kobak
,
D.
and
Tripathy
,
S.J
. (
2021
)
Patch-seq: past, present, and future
.
J. Neurosci.
41
,
937
946
https://doi.org/10.1523/JNEUROSCI.1653-20.2020
164
Ha
,
M.
and
Kim
,
V.N
. (
2014
)
Regulation of microRNA biogenesis
.
Nat. Rev. Mol. Cell Biol.
15
,
509
524
https://doi.org/10.1038/nrm3838
165
Lewis
,
B.P.
,
Burge
,
C.B.
and
Bartel
,
D.P
. (
2005
)
Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets
.
Cell
120
,
15
20
https://doi.org/10.1016/j.cell.2004.12.035
166
Rooij
,
E.
and
Olson
,
E.N
. (
2012
)
MicroRNA therapeutics for cardiovascular disease: opportunities and obstacles
.
Nat. Rev. Drug Discov.
11
,
860
872
https://doi.org/10.1038/nrd3864
167
Santovito
,
D.
and
Weber
,
C
. (
2022
)
Non-canonical features of microRNAs: paradigms emerging from cardiovascular disease
.
Nat. Rev. Cardiol.
19
,
620
638
https://doi.org/10.1038/s41569-022-00680-2
168
Petkova
,
M.
,
Atkinson
,
A.J.
,
Yanni
,
J.
,
Stuart
,
L.
,
Aminu
,
A.J.
,
Ivanova
,
A.D.
et al.
(
2020
)
Identification of key small non-coding microRNAs controlling pacemaker mechanisms in the human sinus node
.
J. Am. Heart Assoc.
9
, e016590 https://doi.org/10.1161/JAHA.120.016590
169
Li
,
N.
,
Artiga
,
E.
,
Kalyanasundaram
,
A.
,
Hansen
,
B.J.
,
Webb
,
A.
,
Pietrzak
,
M.
et al.
(
2021
)
Altered microRNA and mRNA profiles during heart failure in the human sinoatrial node
.
Sci. Rep.
11
,
19328
https://doi.org/10.1038/s41598-021-98580-x
170
Yanni
,
J.
,
D’Souza
,
A.
,
Wang
,
Y.
,
Li
,
N.
,
Hansen
,
B.J.
,
Zakharkin
,
S.O.
et al.
(
2020
)
Silencing miR-370-3p rescues funny current and sinus node function in heart failure
.
Sci. Rep.
10
, 11279 https://doi.org/10.1038/s41598-020-67790-0
171
D’Souza
,
A.
,
Bucchi
,
A.
,
Johnsen
,
A.B.
,
Logantha
,
S.J.R.J.
,
Monfredi
,
O.
,
Yanni
,
J.
et al.
(
2014
)
Exercise training reduces resting heart rate via downregulation of the funny channel HCN4
.
Nat. Commun.
5
, 3775 https://doi.org/10.1038/ncomms4775
172
Zhang
,
J.
,
Wei
,
F.
,
Ding
,
L.
,
Wang
,
L.
,
Zhang
,
X.
,
Yu
,
L.
et al.
(
2019
)
MicroRNA-1976 regulates degeneration of the sinoatrial node by targeting Cav1.2 and Cav1.3 ion channels
.
J. Mol. Cell. Cardiol.
134
,
74
85
https://doi.org/10.1016/j.yjmcc.2019.06.018
173
Fan
,
W.
,
Sun
,
X.
,
Yang
,
C.
,
Wan
,
J.
,
Luo
,
H.
and
Liao
,
B
. (
2023
)
Pacemaker activity and ion channels in the sinoatrial node cells: microRNAs and Arrhythmia
.
Prog. Biophys. Mol. Biol.
177
,
151
167
https://doi.org/10.1016/j.pbiomolbio.2022.11.005
174
Lozano-Velasco
,
E.
,
Aranega
,
A.
and
Franco
,
D
. (
2021
)
Non-coding RNAs in the cardiac action potential and their impact on Arrhythmogenic cardiac diseases
.
Hearts
2
,
307
330
https://doi.org/10.3390/hearts2030026
175
D’Souza
,
A.
,
Pearman
,
C.M.
,
Wang
,
Y.
,
Nakao
,
S.
,
Logantha
,
S.J.R.J.
,
Cox
,
C.
et al.
(
2017
)
Targeting miR-423-5p reverses exercise training-induced HCN4 channel remodeling and sinus bradycardia
.
Circ. Res.
121
,
1058
1068
https://doi.org/10.1161/CIRCRESAHA.117.311607
176
Sanchez
,
L.
,
Mesquita
,
T.
,
Zhang
,
R.
,
Liao
,
K.
,
Rogers
,
R.
,
Lin
,
Y.N.
et al.
(
2022
)
MicroRNA-dependent suppression of biological pacemaker activity induced by TBX18
.
Cell Rep. Med.
3
, 100871 https://doi.org/10.1016/j.xcrm.2022.100871
177
Anglicheau
,
D.
,
Muthukumar
,
T.
and
Suthanthiran
,
M
. (
2010
)
MicroRNAs: small RNAs with big effects
.
Transplantation
90
,
105
112
https://doi.org/10.1097/TP.0b013e3181e913c2
178
Yaniv
,
Y.
,
Juhaszova
,
M.
,
Lyashkov
,
A.E.
,
Spurgeon
,
H.A.
,
Sollott
,
S.J.
and
Lakatta
,
E.G
. (
2011
)
Ca2+-regulated-cAMP/PKA signaling in cardiac pacemaker cells links ATP supply to demand
.
J. Mol. Cell. Cardiol.
51
,
740
748
https://doi.org/10.1016/j.yjmcc.2011.07.018
179
Ren
,
L.
,
Gopireddy
,
R.R.
,
Perkins
,
G.
,
Zhang
,
H.
,
Timofeyev
,
V.
,
Lyu
,
Y.
et al.
(
2022
)
Disruption of mitochondria-sarcoplasmic reticulum microdomain connectomics contributes to sinus node dysfunction in heart failure
.
Proc. Natl. Acad. Sci. U.S.A.
119
, e2206708119 https://doi.org/10.1073/pnas.2206708119
180
Yaniv
,
Y.
,
Spurgeon
,
H.A.
,
Ziman
,
B.D.
and
Lakatta
,
E.G
. (
2013
)
Ca2Ca
.
Plos One
8
, e57079 https://doi.org/10.1371/journal.pone.0057079
181
Wu
,
Y.
,
Rasmussen
,
T.P.
,
Koval
,
O.M.
,
Joiner
,
M.A.
,
Hall
,
D.D.
,
Chen
,
B.
et al.
(
2015
)
The mitochondrial uniporter controls fight or flight heart rate increases
.
Nat. Commun.
6
,
6081
https://doi.org/10.1038/ncomms7081
182
Yavari
,
A.
,
Bellahcene
,
M.
,
Bucchi
,
A.
,
Sirenko
,
S.
,
Pinter
,
K.
,
Herring
,
N.
et al.
(
2017
)
Mammalian γ2 AMPK regulates intrinsic heart rate
.
Nat. Commun.
8
, 1258 https://doi.org/10.1038/s41467-017-01342-5
183
Grainger
,
N.
,
Guarina
,
L.
,
Cudmore
,
R.H.
and
Santana
,
L.F
. (
2021
)
The organization of the sinoatrial node microvasculature varies regionally to match local myocyte excitability
.
Function (Oxf)
2
, zqab031 https://doi.org/10.1093/function/zqab031
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).