* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download The cardiac pacemaker current Journal of Molecular and Cellular
Survey
Document related concepts
Transcript
Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 Contents lists available at ScienceDirect Journal of Molecular and Cellular Cardiology j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / y j m c c Review article The cardiac pacemaker current Mirko Baruscotti ⁎, Andrea Barbuti, Annalisa Bucchi Department of Biomolecular Sciences and Biotechnology, Laboratory of Molecular Physiology and Neurobiology, Università degli Studi di Milano; Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata (CIMMBA), via Celoria 26, 20133 Milano, Italy a r t i c l e i n f o Article history: Received 29 April 2009 Received in revised form 15 June 2009 Accepted 26 June 2009 Available online 8 July 2009 Keywords: Pacemaker current SAN HCN clones HCN knockout mice HCN-associated pathologies a b s t r a c t In mammals cardiac rate is determined by the duration of the diastolic depolarization of sinoatrial node (SAN) cells which is mainly determined by the pacemaker If current. f-channels are encoded by four members of the hyperpolarization-activated cyclic nucleotide-gated gene (HCN1–4) family. HCN4 is the most abundant isoform in the SAN, and its relevance to pacemaking has been further supported by the discovery of four loss-of-function mutations in patients with mild or severe forms of cardiac rate disturbances. Due to its selective contribution to pacemaking, the If current is also the pharmacological target of a selective heart rate-reducing agent (ivabradine) currently used in the clinical practice. Albeit to a minor extent, the If current is also present in other spontaneously active myocytes of the cardiac conduction system (atrioventricular node and Purkinje fibres). In working atrial and ventricular myocytes f-channels are expressed at a very low level and do not play any physiological role; however in certain pathological conditions over-expression of HCN proteins may represent an arrhythmogenic mechanism. In this review some of the most recent findings on f/HCN channels contribution to pacemaking are described. © 2009 Elsevier Inc. All rights reserved. Contents The mechanism of cardiac pacemaking and the If current . Basic biophysical properties of the pacemaker current . . Molecular structure of pacemaker channels . . . . . . . Pacemaker channels during embryonic development . . . Pacemaker channels in the adult heart . . . . . . . . . 5.1. SAN . . . . . . . . . . . . . . . . . . . . . . 5.2. Atrioventricular node (AVN) . . . . . . . . . . . 5.3. Purkinje fibres (PFs) . . . . . . . . . . . . . . . 5.4. Working myocardium . . . . . . . . . . . . . . 6. Basis of functional heterogeneity of pacemaker currents . 6.1. MiRP1 . . . . . . . . . . . . . . . . . . . . . 6.2. PI(4,5)P2 . . . . . . . . . . . . . . . . . . . . 6.3. Caveolin 3 . . . . . . . . . . . . . . . . . . . 7. Genetics of HCN: HCN knockout mice and HCN-associated 8. Biological pacemaker . . . . . . . . . . . . . . . . . . 9. f-channels blockers. . . . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . 1. 2. 3. 4. 5. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . pathologies in . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1. The mechanism of cardiac pacemaking and the If current Cardiac pacemaking originates in the sinoatrial node (SAN) as a consequence of spontaneous firing of rhythmic action potentials ⁎ Corresponding author. Tel.: +39 02 5031 4931; fax: +39 02 5031 4932. E-mail address: [email protected] (M. Baruscotti). 0022-2828/$ – see front matter © 2009 Elsevier Inc. All rights reserved. doi:10.1016/j.yjmcc.2009.06.019 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . humans . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55 56 56 57 57 58 58 58 59 59 59 59 59 59 61 61 61 61 generated by specialized myocytes. Although the electrical behavior of a typical SAN cell differs in several aspects from that of a working myocyte, the functional hallmark can be precisely identified in the events that take place during the diastolic interval. During this phase atrial and ventricular myocytes rest in a standby-like condition at a stable voltage (∼−80 mV); a quite different situation characterizes SAN cells, where the cell potential slowly creeps up from the M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 56 maximum diastolic potential of about −60 mV to the threshold for the ignition of a new action potential. Since this time interval sets the pace of the heart, this phase is named “pacemaker depolarization”. Given the large spectrum of heart rates observed in mammals the duration of this phase can vary substantially, however the voltage range encompassed is extremely constant and roughly extends from −60 to −40 mV [1–3]. To sustain this phase several ionic currents and pumps enter in action at variable times and voltages [4–6], and this complexity allows for a highly flexible system since the chronotropic fine tuning operated by neuro-hormonal regulators can target different effectors. In this review we will focus on the If current which is responsible for initiating the diastolic depolarization of SAN cells. Due to its fundamental role and its unusual characteristics of being activated in hyperpolarization, this current was named “pacemaker current” or “funny” (If) current [7–9]. The unique property of a reverse voltage dependence, together with the inward nature of the current at diastolic potentials, makes this current apt to initiate and support the diastolic depolarization. In addition, the direct modulation of the current operated by the second messenger cAMP, represents one of the main pathways by which the autonomic nervous system controls cardiac chronotropism [10]. Two recent clinical findings further confirm the role of f-channels in setting the cardiac rate: one is the evidence of a causative link between the presence of loss-of-function mutations found in these channels and the arrhythmic state of individuals carrying the mutations, and the other is the specific heart rate reduction observed in patients treated with ivabradine, a drug that at therapeutic doses selectively reduces the If current (see specific sections in this review). Although originally discovered in the heart, the If current is also abundantly present in a large fraction of neuronal elements, where it contributes to rhythmic firing, synaptic integration, and dendritic integration [11]. 2. Basic biophysical properties of the pacemaker current The If current is carried by Na+ and K+ ions and its reversal potential is between − 10 and −20 mV, and permeation occurs according to a multiion, single-file mechanism [9,12,13]. Interestingly, recent single channel experiments carried out in rat and human working myocytes have also reported a weak permeability to Ca2+ ions [14]. Voltage-clamp studies have shown that activation is a complex event determined initially by an intrinsic rearrangement of the closed structure of the channel, which originates a “shoulder-like” delay, and followed by the proper close-to-open transition which proceeds according to an exponential relaxation [15,16]. The quantitative aspects of this activation process, i.e. the time-course of the current activation, and the steady-state current level reached at the end of each pulse, are finely controlled both by the membrane voltage and by a direct interaction with the second messenger cAMP [17,18]. Although variable data have been reported, the voltage threshold for sinoatrial If activation is compatible with a functional presence of the current at diastolic potentials (Table 1 in [19]); for example, recent experiments carried out on human SAN cells indicate that threshold for If activation occurs between −50 and − 60 mV, a voltage range well comprised in the diastolic range [3]. 3. Molecular structure of pacemaker channels The molecular constituents of pacemaker channels belong to the hyperpolarization-activated cyclic nucleotide-gated (HCN) channel family which is part of the superfamily of K+ channels. In mammals four HCN isoforms are known (HCN1–4), and heterologous expression and in vivo investigations have shown that they can assemble both as homotetramers and heterotetramers (with the exceptions of HCN2– HCN3 heteromers) to yield functional channels. Currents resulting from homomeric and heteromeric assembly have biophysical and modulatory properties qualitatively similar to the native If [20]; but when quantitative aspects are taken into account noticeable differences are evident in terms of kinetics, voltage dependence, and cAMP sensitivity [21–26]. Each HCN isoform is composed of three large macrodomains: a cytoplasmic N-terminus, a 6 transmembranespanning “core” region, and a cytoplasmic C-terminus. Each of these regions mediates important functional aspects. The N-terminus contains a conserved stretch of residues which appears to be important in mediating channel trafficking [27]. The core transmembrane region is the more conserved region among isoforms and contains both the voltage sensor (S4 segment) and residues involved in pore formation (S5-P-S6 regions), and therefore constitutes the proper gating module of the channel. All HCN isoforms exhibit within the permeation pathway the GYG triplet which, as in pure K+ channels, constitutes the selectivity filter. It is not immediately apparent why HCN channels are also permeable to Na+ ions, but it is now believed that the inner pore of HCN channels is somewhat less rigid than that of K + channels, and for this reason it also accommodates partially hydrated Na+ ions [28]. Further information Table 1 Effects of “heart rate-lowering” agents on electrical properties of cardiac pacemaker cells. If reduction IK reduction ICa reduction DDS APD50 Rate reduction Refs Alinidine Zatebradine (UL-FS49) Cilobradine ZD7288 Ivabradine ∼80% 30 μM rbSAN cells 30% 30 μM rbSAN cells 11% 30 μM rbSAN cells ↓ 30 μM rbSAN cells + 23% 30 μM rbSAN cells 22% 30 μM rbSAN cells [146] ∼65% 1 μM rbSAN cells ∼20% 1 μM rbSAN cells No effect 1 μM rbSAN cells − 42% 3 μM rbSAN tissue + 29% 3 μM rbSAN tissue 28% 3 μM rbSAN tissue [147–149] ∼60% 1 μM mSAN cells ∼22% 5 μM mSAN cells Not tested 78% 1 μM gpSAN cells No effect 1 μM gpSAN cells 14% 1 μM gpSAN cells − 54% 1 μM gpSAN tissue + 8% 1 μM gpSAN tissue ∼35% 1 μM gp right atrium [150,151] ∼60% 3 μM rbSAN cells No effect 3 μM rbSAN cells No effect 3 μM rbSAN cells −67% 3 μM rbSAN tissue + 9% 3 μM rbSAN tissue ∼24% 3 μM rbSAN tissue [10,13,148,149] − 70% 1 μM mSAN cells + 60%⁎ 1 μM mSAN cells ∼55% 1 μM mSAN cells [139] DDS: diastolic depolarization slope; APD50: action potential duration measured at 50% of repolarization (⁎APD measured from the threshold to the following maximum diastolic potential); rb, rabbit; m, mouse; gp, guinea-pig. M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 on the structural organization of the pore comes from the observation that the pore blocker ZD7288 directly interacts with residues of the S6 segment, and that closing of the channel entraps the drug into the inner vestibule of the HCN1 channel [29]. Taken together, these data and the homology model based on the KcsA K+ channel crystal [30], suggest that the four S6 segments line the inner wall of HCN pore, and that access to the S6 residues involved in ZD7288 binding is guarded by a gating structure likely to comprise part of the S6 segment. The Cterminus region comprises three separate structural elements: the Clinker, which is organized in 6 α-helices, the cyclic nucleotide binding domain (CNBD), and the proper C-terminus. Collectively, the CNBD and the C-linker act as a functional unit that modulates the open probability of a pacemaker channel. Detailed information on several aspects of the HCN channel structure–function relation is now available [21,31,32] and here we will only provide some general concepts. It is now clear that voltage-dependent ion channels do not behave like rigid structures and therefore the rotational/translational movement of the S4 segments imposed by changes in the membrane electrical field does not induce a general rearrangement of the protein leading to the opening of the pore [33]. Due to its direct interaction with the S4 segment a particular focus was placed on the role of the S4–S5 linker, which indeed was shown to undergo a spatial reorganization upon S4 movement [33,34]. Single alanine substitution of aminoacids E324, Y331, and R339 of the HCN2 S4–S5 linker was able to disrupt channel closing thus indicating that this linker plays a crucial role in gating [34]. There is now evidence that the S4–S5 linker functionally interacts with the C-linker region; for example in HCN2 channels the electrostatic interaction between R339 of the S4–S5 linker and D443 of C-linker stabilizes the closed state of the channel [35]. These data indicate that the interaction between the S4–S5 linker and the C-linker mediates the coupling of voltage sensing and channel opening and closing. In addition to the voltage, the HCN channel open probability is also controlled by the second messenger cAMP through a direct binding to the channel [17]. Crystallization data indicate that in the presence of cAMP the A′ and B′ helices of the C-linker of each subunit interact with the C′ and D′ helices of the neighboring subunit, and this tetrameric arrangement favors the open state of the channel. In the absence of cAMP, the tetrameric assembly is lost and this favors the closed channel conformation [21,36,37]. This mechanism thus represents the final molecular event in the modulation of the If current exerted by the autonomic nervous system. 4. Pacemaker channels during embryonic development Pacemaker activity characterizes the developing heart at a very early stage; for example, in chick embryos electrical pacemaker activity is present even before the onset of regular contractions of the primitive heart tube [38]. In mammals, the linear heart tube starts to beat around embryonic day 8.5 (ED8.5), a stage at which all cardiomyocytes are autorhythmic and express the If current [39]. With further development (ED18) only 33% of myocytes present spontaneous but irregular action potentials and the If current is decreased by 82% [39]. In agreement with functional data the same authors also reported a significant down-regulation of HCN1 and HCN4 mRNAs and a moderate increase of HCN2; HCN3 was not detected [39]. A detailed analysis of HCN4 localization in the developing heart, by in situ hybridization, indicates that the HCN4 mRNA is already detectable in the cardiac crescent at ED7.5, and its expression remains confined to the venous pole, the cardiac region from which develops the mature SAN [40]. Specific expression of HCN4 mRNA in the SAN and conduction system has also been demonstrated in a series of studies addressing the molecular pathway leading to the formation of these regions. Tbx3 is a transcriptional repressor whose expression during heart development specifically delineates the SAN/conduction system and match the HCN4 expression [41,42]. Interestingly, ectopic expression of Tbx3 in the atria 57 generates foci of autorhythmic cells in which HCN4 results upregulated [43]. Also, constitutive deletion of the transcription factors Shox2 causes embryonic lethality at mid-gestation (ED11.5–ED12.5), and this genetic alteration prevents the expression of both HCN4 and Tbx3 and causes formation of an underdeveloped SAN and significant bradycardia [44]. Taken together these data indicate that HCN4 is a major component of the f-channel in the developing SAN, but they leave some uncertainty regarding its presence in the embryonic working myocytes. The embryological lethality of HCN4 knockout mice further indicates that the presence of the If current is necessary for proper cardiac development [45]. Despite the observation that Tbx3 and Shox2 are important in the modulation of HCN4 expression during normal heart development, they do not account for basal HCN gene transcription. Indeed, the lack of Tbx3 does not influence HCN4 expression [43], and the lack of Shox2 does not modify HCN4 expression until ED10 [44]. Two conserved sequences accounting for HCN4 gene transcription have recently been found in the non-coding region of HCN4 [46]. The first, an 846 bp long sequence is located at the 5′UTR, the second sequence is a neuron-restrictive silencer element (NRSE) located in the intronic region between exons 1 and 2, which specifically represses the promoter activity of the first sequence [46]. A different approach in studying early events of cardiac development consists of the in vitro differentiation of embryonic stem cells (ESC) through the formation of embryoid bodies (EBs), which are three-dimensional cell aggregates able to recapitulate early embryonic developmental events. 7–8 days after cell aggregation mouse EBs start to display foci of spontaneous contraction generated by early cardiomyocytes [47–49]. The If current has been found both in human and in mouse ESC-derived cardiomyocytes and its direct involvement in the generation and modulation of rate has been assessed by applying specific If inhibitors [48,50–54]. In mouse ESC-derived cardiomyocytes, ivabradine (3 μM) slowed beating rate by 25% and reduced the If current by 50%, while ZD7288 (0.3 μM) slowed beating rate by approximately 50% and reduced the If current by 15% [52,53]. Similarly, zatebradine, another If blocker, slowed spontaneous rate of human ESC-derived cardiomyocytes [50]. Patch-clamp investigations have shown that the If current is already present in cells isolated from 10 day-old mouse EBs, and that its density increases significantly with the progression of differentiation [52–54]. There is some variability concerning the mRNA and protein expression of HCN isoforms in mouse ESC-derived myocytes. While two studies reported the expression of mRNAs of all four isoforms [52,55], another study showed that mouse ESC-derived myocytes only express HCN1 and HCN4 [56]. At the protein level, western blot experiments revealed the expression of HCN2 and HCN3 isoforms [53], while immunofluorescence experiments reported that HCN1 and HCN4 are the only isoforms expressed on the membrane of pacemaker cells [52,57]. Investigations of human ESC-derived cardiomyocytes revealed a high and stable expression of HCN2 mRNA while HCN1 and HCN4 appear to be present in undifferentiated human ESC and their expression decreases with differentiation [50,51]. Despite this variability, likely originating from differences in ES clones, species and in the techniques employed, it is clear that the If current and the underlying HCN channels are expressed very early during cardiac development and significantly contribute to the pacemaking mechanism. 5. Pacemaker channels in the adult heart The If current has been described in all tissues of the heart, however its functional contribution in non pathological conditions is limited to the cardiac conduction system. Here we provide an overview of the characteristics of the If current in various heart regions. 58 M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 Fig. 1. Molecular and functional properties of SAN myocytes. (A) Spontaneous action potentials (left) and If current traces (right) recorded from typical rabbit SAN myocytes; currents were elicited by hyperpolarizing voltage steps in the range − 45 to − 75 mV. (B) Immunofluorescence analysis of rabbit SAN tissue slice labelled with anti-connexin 43 (Cx43, red) and anti-HCN4 (green) antibodies. HCN4 is strongly expressed in the central region of the SAN, while the opposite staining is observed for Cx43; crista terminalis (CT), interatrial septum (IS). (C) HCN4 labelling of single myocytes isolated from CT, SAN and IS (top), and representative current traces recorded at − 125 mV from myocytes isolated from the same regions (bottom). Both If current density and HCN4 labelling are more abundant in the central nodal area. (Panels B and C from [61] with permission). 5.1. SAN In agreement with the leading role of the SAN in pacing, this tissue presents the highest level of If current density and HCN expression [58–62]. As previously mentioned, the presence of the If current has been documented in adult SAN myocytes of many species including humans [3,19], and its activation threshold (∼− 50.8 mV, n = 9, from Table 1 in [19]) falls within the voltage excursion encompassed by SAN cells during the diastolic depolarization. In SAN tissues of lower mammals and humans, the predominant molecular constituent of fchannel is the HCN4 isoform (Fig. 1); HCN1 and HCN2 have also been detected, but at low to moderate levels depending on the species [58– 66]. HCN3 is absent from the SAN [58,67]. 5.2. Atrioventricular node (AVN) The AVN is able to pace the heart in the absence of proper sinus rhythm, and the If current has been recorded in the majority of cells isolated from the AVN of rabbit, mouse and guinea-pig [68–70]. A more detailed investigation on ovoid- and rod-shaped cells isolated from rabbit AVN indicates that in ovoid cells the pacemaker current is abundantly present (− 5.18 pA/pF at − 100 mV) and its activation threshold is about −60 mV, while in rod-shaped cells the current is nearly absent [68]. Studies on mRNA distribution indicate that in the AVN, as well as in the SAN, the HCN4 isoform is the most abundant isoform [71], and, at least in mice, the overall HCN4 protein expression is about one third of that found in the SA node [72]. 5.3. Purkinje fibres (PFs) Isolated PFs are able to beat spontaneously, and for this reason they were largely used in early electrophysiological experiments investigating the nature of the pacemaker currents responsible for the diastolic depolarization [8,9,73,74]. The diastolic depolarization of these fibres develops between −90 mV and − 70 mV, thus in a range of potential more negative than that of a typical SAN cell. Interestingly, the threshold for If current activation in these cells is also negatively shifted (∼ −80 mV), and this suggests an active role of the current in the generation of the spontaneous activity [8,9]. The molecular composition of f-channels in PFs is extremely dependent on the species investigated. For example, canine PFs, which exhibit a significant automaticity and a large If current, have high levels of HCN mRNA (∼ 35% of the HCN signal recorded in the SAN), with ∼90% of the transcripts constituted by HCN4 and the remaining by HCN2 M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 [75,76]; interestingly, at the protein level HCN2 is the major isoform expressed [76]. In contrast, rabbit PFs, which tend not to be automatic and exhibit little If current, show minimal levels of HCN messenger (∼4% of SAN) with an equivalent presence of HCN4 and HCN1, and a minor contribution of HCN2 (∼10% [65]). In human PFs HCN4 is the predominant isoform expressed [77]. 5.4. Working myocardium The presence of the If current in the working myocardium is well documented but its range of activation is much more negative than in the SAN. Reported values of thresholds are extremely variable and range from −60 to − 120 mV [78–83]. Furthermore, even when assessed at −130 mV the current densities are extremely low; for example in human atrial and ventricular myocytes the reported values are −0.8 pA/pF and −0.47 pA/pF, respectively [79,81]. For comparison at − 130 mV the If density measured in human SAN cells is − 8 pA/pF [3]. Taken together these data indicate that under physiological conditions the If current is not expected to play a functional role in atria and ventricles. The molecular composition of fchannels in working myocytes is extremely complex due to interspecies variability and sometimes due to contrasting results between transcriptional and protein data. In general, it can be stated that the major protein isoform both in atria and ventricles is HCN2. HCN4 is only occasionally reported, while HCN1 does not appear to be present [62,71,76,84,85]. Despite the fact that pacemaker currents do not play a role in the healthy atria and ventricles, growing evidence in humans and animals demonstrates that the over-expression of the If current in these tissues is often associated to some cardiac diseases, and this over-expression may represent an important arrhythmogenic source [82,83,85–90]. For example, in human ventricular myocytes isolated from failing hearts of patients with ischemic cardiomyopathy, the If current is over-expressed by approximately two-fold and its threshold of activation is shifted positively by 9.6 mV [82]. In agreement with these observations up-regulation of HCN2 and HCN4 mRNAs/proteins was found in both atria and ventricle myocytes obtained from failing hearts explanted from patients with end-stage ischemic cardiomyopathy [85]. A regulatory mechanism controlling the expression of HCN isoforms may consist of post-translational events. One such mechanism, known to control the functional expression of both HCN2 and HCN4 is operated by two muscle-specific micro RNAs, miR-133 and miR-1 [91,92]. For example, during cardiac development, up-regulation of miR-133 and miR-1 decreases the ventricular If current by reducing HCN2 and HCN4 proteins without significantly affecting mRNA levels. The opposite event occurs in hypertrophied hearts where miR-133 and miR-1 are decreased and this down-regulation causes the re-expression of HCN2 and HCN4 channels [91,92]. This same mechanism could also explain the variability between mRNA and protein expression data found in different systems. 6. Basis of functional heterogeneity of pacemaker currents The identification of the exact molecular composition of native pacemaker channels was initially challenged by the attempt to reproduce the native kinetic and modulatory properties of the native f through heterologous expressions of HCN isoforms. Unfortunately, this approach proved unsuccessful. Although HCN4 and HCN1 are the main isoforms of the SAN, their expression either alone or in combination failed to reproduce the sinoatrial If current. For example, even though homomeric HCN4 channels retain a cAMP modulation similar to that observed in native channels, both the activation and deactivation kinetics and the voltage dependence are much different. On the contrary HCN4–HCN1 heteromers generate currents with kinetics approaching those of native SAN f-channels, but they do not reproduce the same voltage dependence of activation [25]. Furthermore, it has been shown that even when the same isoform is 59 expressed in different cells, the resulting currents do not display identical properties. To this regard Qu et al. [93] have shown that currents resulting from HCN2 expression in neonatal and in adult ventricle myocytes have different biophysical properties such as a different position of the voltage-dependent curve (V1/2, − 76 mV and −96 mV, respectively). These data clearly suggest that, in addition to the heterogeneity provided by different HCN isoform and cAMP modulation, the large phenotypic variation of If properties observed both in the healthy heart and in pathological conditions likely reflects the presence of additional modulatory factors. Recent evidence indicates that protein–protein and protein–phospholipid interactions, and modulatory cytoplasmic factors play a relevant role in modulating the pacemaker current. Taken together all these regulatory elements are defined as “context dependence”. Some of the most relevant modulatory factors are MiRP1, phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), and caveolin 3. 6.1. MiRP1 Mink-related protein 1 (MiRP1 or KCNE2) is a single transmembrane-spanning protein that acts as a β-subunit of HCN channels. Although its modulatory actions depend on the HCN isoform and on the expression system used, the most relevant effect is to increase current density [94–97]. 6.2. PI(4,5)P2 Recent data indicate that the voltage dependence of native sinoatrial f- and HCN1/2 channels is also regulated by local pools of PI(4,5)P2 [98,99]. Further experiments suggested that stimulation of receptors coupled to phospholipase C (PLC), such as bradykinin BK2 receptor and the muscarinic M1 receptor, can modulate the gating of both recombinant and native HCN channels via an increase of PI(4,5) P2 [100]. Specifically, activation of the BK2 receptor induces a positive shift of the voltage dependence of HCN2 (∼ 20 mV), HCN1 (∼ 6 mV), and sinoatrial f-channels (∼8 mV), and also affects time constants of activation and deactivation. Noteworthy is the fact that basal levels of PI(4,5)P2 may change with development, stress, and pathological conditions [101–103]. 6.3. Caveolin 3 It has also been shown that in rabbit SAN cells HCN4 localizes in caveolin-rich membrane microdomains (caveolae) and interacts with caveolin 3 [104,105]. Disorganization of caveolae strongly affects fchannel kinetics, shifting the activation curve toward more positive potentials and slowing the deactivation kinetics. Similar effects were observed for HCN4 current after lipid raft disruption in HEK cells [106]. Post translational modification events may also contribute to modulate HCN properties: both native and HCN channels can indeed be modulated by phosphorylation processes and particularly by tyrosine-kinase phosphorylation, but a clear understanding of the functional role of this regulatory mechanism remains in part elusive [107–115]. 7. Genetics of HCN: HCN knockout mice and HCN-associated pathologies in humans Genetically manipulated knockout mice and genetic analysis in human patients affected by severe or mild cardiac rhythm alterations have brought new information on the role and relevance of HCN4 channels to cardiac pacemaking. In recent years four HCN4 knockout mouse models have been developed to evaluate the functional contribution of this isoform to pacemaking in vivo [45,116,117]. Both global and cardiac specific constitutive knockouts determine the 60 M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 premature death of mice at mid-gestation (ED9.5–ED11.5), period in which a regular contractile activity of the developing heart normally appears [45]. Hearts isolated from knockout embryos at day ED9.5 exhibited a decreased spontaneous rate (−36.7%) and If current (−75/−90%). Interestingly, the knockout process also led to the complete loss of cAMP-mediated β-adrenergic chronotropic modulation in both intact hearts and single cells; this finding is consistent with the observation that the residual If current could be carried by the HCN1 and HCN3 isoforms which are known to be mildly or not affected by cAMP modulation [67,118,119]. Taken together these data indicate that expression of HCN4 is a necessary element for proper embryonic heart development, but unfortunately they do not provide any hint on the role of the HCN4 current in the adult animal. In order to overcome this limitation, temporally-controlled global [116], and cardiac specific [117] HCN4 knockout models were developed. ECG recordings in freely moving knockout animals showed the presence of sinus pauses (mean duration = 321 ms, mean frequency = 8.1/min) in otherwise normal rhythmic activity. Interestingly, β-modulation of rate was not lost in the adult knockout animals, and any acceleration in heart rate led to a reduction of sinus pauses. When the pacemaking activity was investigated at the single cell level, it was found that 90% of sinoatrial node myocytes of the global knockout and 50% of the cardiac specific knockout were quiescent [116,117], and that the If current was reduced by about 75%–80%. Interestingly, β-adrenergic stimulation was able to rescue spontaneous activity in knockout cells [116]. As pointed out by the same authors, the presence in vivo of a sympathetic tone and of additional network properties may explain the persistence of spontaneous activity in vivo, however it does not fully explain the presence of sinus pauses [116]. The members of the HCN channel family were among the last ion channel genes to be cloned; for this reason research aiming at identifying a link between alterations in the HCN genes and cardiac pathologies has only recently begun. During the course of independent screenings of patients affected by various forms of rhythm disturbances, four mutations/deletions of the HCN4 open reading frame have been identified and directly correlated to the phenotypes [120–123]. Although all the identified mutations determine a reduced contribution of the pacemaker If current to the diastolic depolarization (loss-of-function), the functional mechanisms by which this reduction is achieved are quite different (Fig. 2). The first evidence of a mutation of the HCN4 in humans was found by Schulze-Bahr et al. [120] who reported the presence of the heterozygous 1-bp deletion (1631delC) in a single patient affected by idiopathic sinus node dysfunction (SND) with severe bradycardia (41 beats per minute, bpm), intermittent atrial fibrillation, and chronotropic incompetence. The resulting HCN4 protein (573X) had a shorter C-terminus, lacking the CNBD, caused by the presence of an earlier stop codon. In vitro heterologous expression of the 573X channels revealed that the major alteration in the current was a dominant negative loss of cAMP modulation, which could explain the chronotropic incompetence of the patient both at rest and during maximal workload [120]. Although these data are extremely intriguing, the causative relation between the altered function of the channel and the pathological phenotype of the patient is not conclusive since the study is based on a single-case report. In another study Ueda et al. [121] reported the case of arrhythmic patients affected by SND with recurrent syncope, severe bradycardia, QT prolongation, polymorphic ventricular tachycardia. The study, based on a two generation family, suggested a possible linkage between the presence of the D553N mutation in the C-linker region and the disease since members carrying the mutation (n = 3) also presented the clinical phenotype. Patch-clamp and immunofluorescence analysis showed that this mutation exerts a dominant negative effect and severely impairs channel trafficking to the membrane leading to an almost complete loss of the pacemaker current. Despite the strong suggestion of a link between the mutation and the disease, the Fig. 2. hHCN4 mutations associate to cardiac rhythm alterations. Schematic topology of a single HCN4 subunit with the 6 transmembrane segments, and the N- and C-termini. The C-terminal region is composed by the C-linker (A′ to F′ helices), the CNBD (A helix, β-roll, B and C helices) and the proper C-terminus. Orange dots indicate the position of the point mutations (G480R, D553N, and S672R), while the orange X indicates the site where the mutant protein (573X), originated by the 1631delC, is truncated. Insets show the main effects on If properties induced by the mutations. Top insets show bar-graphs indicating the reduction in current densities observed for mutant G480R (at −100 mV) and D553N (at − 120 mV) proteins (measured by eye from [121,123]). Bottom left inset shows that cAMP does not modulate the position of the voltage-dependent curve of the mutant 573X (reproduced from [120]), while bottom right inset shows that S672R mutant channels display a negative shift of the voltage-dependent curve (reproduced from [122]). For clarity data relative to homomeric channel expressions have been used. F.A., fractional activation. complexity of the clinical manifestations cannot be easily explained by in vitro results. A direct correlation between a heterozygous mutation and cardiac rhythm alteration has been demonstrated for the S672R mutation which induces a form of familial asymptomatic bradycardia [122]. An extensive familial analysis carried out on 27 members allowed the conclusion that the mutation co-segregated with the bradycardic phenotype according to an autosomal dominant pattern (LOD score N5). The mean heart rate of affected individuals (52.2 bpm) was reduced by 29% when compared to unaffected members (73.2 bpm) of the same family. In vitro heterologous expression of mutant and wild type channels demonstrated that the half-activation voltage (V1/2) of homomeric mutant channels was 8.4 mV (heteromeric channels: 4.9 mV) more negative than that of wild type channels; a slower kinetics of deactivation was also observed. Despite the fact that this mutation is localized in the CNBD region, cAMP retained its normal M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 modulatory action. Interestingly, the quantitative and qualitative effects of the mutation (4.9 mV shift and 29% heart rate reduction) completely resemble those of a low dose (30 nM) of acetylcholine. This study not only identifies a tight linkage between the mutation and bradycardia but it is also the first one that fully describes the underlying molecular mechanism. A form of familial sinus bradycardia was also found to co-segregate with the mutation G480R which is located in the pore region of the HCN4 channel [123]. In vitro studies seem to indicate both an extreme negative shift (∼ 40 mV) of the voltage dependence and trafficking defects. Taken together studies on HCN4 channelopathies seem to suggest a tight relation between the amount of current suppressed by the presence of the mutation and the severity of the associated disease. Indeed, while the limited reduction of the If current caused by the S672R mutation only determines asymptomatic bradycardia, progressively more severe clinical manifestations appear with the deletion 1631delC which eliminates the cAMP modulation, and with the D553N mutation which suppresses the presence of functional channels. 8. Biological pacemaker The search of new therapeutic tools consisting of gene- and/or cell-based intervention aimed to restore compromised cardiac functions has prompted researchers to exploit the use of HCN channels to alter cellular electrical activity in order to generate, in normally quiescent substrates, stable rhythmic activity similar to that of native pacemaker myocytes. The specific features of pacemaker channels and in particular the fact that they are activated only at diastolic potentials and do not contribute to other phases of the action potentials, make them particularly suitable for such purpose. Early in vitro studies demonstrated that virus-mediated over-expression of HCN2 channels induced a significant increase in the rate of spontaneously beating neonatal ventricular myocytes by causing an If-mediated increase of the diastolic depolarization slope [93]. This approach was later confirmed in vivo by showing that direct injection of the HCN2-adenovirus in the left atrium or into the ventricular conduction system of dogs, was able to induce ectopic regular spontaneous activity after AV block [93,124–126]. Similarly, adenovirus-mediated over-expression of HCN1 or HCN4 was sufficient to induce a regular rhythm in quiescent cardiomyocytes [127,128]. Alternative cell-based strategies, aimed to avoid the use of viruses, have been developed by engineering cells in order to express high levels of HCN channels. Engineered human mesenchymal stem cells (hMSCs) expressing either HCN2 or HCN4 have been shown in vitro to properly connect to neonatal cardiomyocytes and to increase their intrinsic spontaneous rhythm [129,130]. HCN2-expressing hMSCs have also been successfully transplanted in canine left ventricular wall where they were able to induce stable ectopic beats [129]. Furthermore, spontaneously beating heterokarion cells generated by the fusion of HCN1 expressing lung fibroblast and ventricular myocytes were shown to induce in vivo ectopic beats at the site of injection [131]. A different approach to generate a biological pacemaker would be to use a cellular substrate as close as possible to native pacemaker myocytes. Both murine and human embryonic stem cells differentiate into spontaneously beating cell aggregates (EBs) which contain myocytes with functional and molecular properties typical of pacemaker cells [50,52–54,132–135]. Two separate studies have demonstrated that spontaneously beating portions of human EBs are able to pace either cultures of neonatal rat cardiac myocytes in vitro or the whole heart in vivo [136,137]. Although neither the If current nor HCN expression was directly addressed in these studies, the increase in rate by β-adrenergic stimulation and, more importantly, the decrease by the f-channel blocker ZD7288 [136,137] strongly suggest a role of the pacemaker current in the generation of the rhythmic activity. 61 9. f-channels blockers The specific and restricted contribution of If to the generation and modulation of the sinoatrial diastolic depolarization phase has long made this current a crucial target for pharmacological applications. In principle, a selective reduction of the If current should cause a slowing of heart rate devoid of undesired side effects. For this reason, drugs able to block f-channels are expected to have the potential for treatment of heart diseases characterized by a deficiency of oxygen supply to the working myocardium such as angina pectoris, heart failure and ischemic heart disease. Several f-channel blockers, called specific “heart ratelowering” agents, have been developed and extensively characterized in in vitro and in vivo studies. This family includes alinidine (ST567), zatebradine (UL-FS49), cilobradine (DK-AH26), ZD-7288, and ivabradine (S16257) [19]. The effects of these agents on the action potential parameters and membrane currents of pacemaker cells/tissue are shown in Table 1. The only If blocker extremely selective at therapeutic doses and with mild side effects is ivabradine [19]. The action of ivabradine on native f-channels displays both a marked use-dependence, since the drug can only access its binding site when channels are in the open state, and current-dependence since the direction of current through the channel affects the stability of the binding [13]. The “usedependence” is the mechanism responsible for block accumulation during repetitive channel opening–closing cycles with the possible consequence that the higher the initial cardiac rate the more effective is the ivabradine block [66]. Experiments investigating the affinity of ivabradine for homomeric HCN channels do not reveal any substantial isoform-specificity although the drug was shown to act with a higher degree of co-operativity in binding to HCN1 than to HCN4 and native fchannels [138,139]. A striking difference however exists between HCN4 and HCN1 in the state-dependence of channel block: while ivabradine behaves as an open-channel blocker of HCN4 and native channels, it can only block HCN1 channels when they are in the closed state [138]. Currently, ivabradine is marketed for treatment of chronic stable angina in patients with normal sinus rhythm who have a contraindication or intolerance to β-blockers; clinical studies of patients with chronic stable angina have shown that ivabradine acts as a pure heart rate-reducing agent and has anti-ischemic and anti-anginal properties equivalent to βblockers and Ca2+ channel blockers and presents a good safety and tolerability profile even during long-term treatment [140–143]. Mild visual symptoms (phosphenes) were occasionally reported, but were generally well tolerated [140,143]. Additional information comes from results from a recent large clinical trial (BEAUTIFUL) which indicate that ivabradine treatment of patients with stable coronary artery disease (CAD) and heart rate ≥70 bpm can reduce the incidence of some CAD outcomes such as hospitalization for myocardial infarction and coronary revascularization [144,145]. Acknowledgments This work was supported by grants from the Ministero dell' Istruzione dell'Università e della Ricerca (Cofin 2007WB35CW) to MB and by the European Union (Normacor) grant. We would like to thank Prof. DiFrancesco for his helpful suggestions and discussions. References [1] Mangoni ME, Nargeot J. Properties of the hyperpolarization-activated current (I (f)) in isolated mouse sino-atrial cells. Cardiovasc Res 2001;52:51–64. [2] DiFrancesco D, Ferroni A, Mazzanti M, Tromba C. Properties of the hyperpolarizing-activated current (if) in cells isolated from the rabbit sino-atrial node. J Physiol 1986;377:61–88. [3] Verkerk AO, Wilders R, van Borren MM, Peters RJ, Broekhuis E, Lam K, et al. Pacemaker current (I(f)) in the human sinoatrial node. Eur Heart J 2007;28: 2472–8. [4] Mangoni ME, Nargeot J. Genesis and regulation of the heart automaticity. Physiol Rev 2008;88:919–82. [5] Lakatta EG, DiFrancesco D. What keeps us thicking, a funny current, a calcium clock, or both? J Mol Cell Cardiol 2009;47:157–70. 62 M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 [6] DiFrancesco D. Pacemaker mechanisms in cardiac tissue. Annu Rev Physiol 1993;55:455–72. [7] Brown HF, DiFrancesco D, Noble SJ. How does adrenaline accelerate the heart? Nature 1979;280:235–6. [8] DiFrancesco D. A new interpretation of the pace-maker current in calf Purkinje fibres. J Physiol 1981;314:359–76. [9] DiFrancesco D. A study of the ionic nature of the pace-maker current in calf Purkinje fibres. J Physiol 1981;314:377–93. [10] Bucchi A, Baruscotti M, Robinson RB, DiFrancesco D. Modulation of rate by autonomic agonists in SAN cells involves changes in diastolic depolarization and the pacemaker current. J Mol Cell Cardiol 2007;43:39–48. [11] Wahl-Schott C, Biel M. HCN channels: structure, cellular regulation and physiological function. Cell Mol Life Sci 2009;66:470–94. [12] Frace AM, Maruoka F, Noma A. External K+ increases Na+ conductance of the hyperpolarization-activated current in rabbit cardiac pacemaker cells. Pflugers Arch 1992;421:97–9. [13] Bucchi A, Baruscotti M, DiFrancesco D. Current-dependent block of rabbit sinoatrial node I(f) channels by ivabradine. J Gen Physiol 2002;120:1–13. [14] Michels G, Brandt MC, Zagidullin N, Khan IF, Larbig R, van Aaken S, et al. Direct evidence for calcium conductance of hyperpolarization-activated cyclic nucleotide-gated channels and human native If at physiological calcium concentrations. Cardiovasc Res 2008;78:466–75. [15] DiFrancesco D, Ferroni A. Delayed activation of the cardiac pacemaker current and its dependence on conditioning pre-hyperpolarizations. Pflugers Arch 1983;396:265–7. [16] DiFrancesco D. Characterization of the pace-maker current kinetics in calf Purkinje fibres. J Physiol 1984;348:341–67. [17] DiFrancesco D, Tortora P. Direct activation of cardiac pacemaker channels by intracellular cyclic AMP. Nature 1991;351:145–7. [18] DiFrancesco D, Mangoni M. Modulation of single hyperpolarization-activated channels (i(f)) by cAMP in the rabbit sino-atrial node. J Physiol 1994;474:473–82. [19] Baruscotti M, Bucchi A, DiFrancesco D. Physiology and pharmacology of the cardiac pacemaker (“funny”) current. Pharmacol Ther 2005;107:59–79. [20] Accili EA, Proenza C, Baruscotti M, DiFrancesco D. From funny current to HCN channels: 20 years of excitation. News Physiol Sci 2002;17:32–7. [21] Zagotta WN, Olivier NB, Black KD, Young EC, Olson R, Gouaux E. Structural basis for modulation and agonist specificity of HCN pacemaker channels. Nature 2003;425:200–5. [22] Chen S, Wang J, Siegelbaum SA. Properties of hyperpolarization-activated pacemaker current defined by coassembly of HCN1 and HCN2 subunits and basal modulation by cyclic nucleotide. J Gen Physiol 2001;117:491–504. [23] Ulens C, Tytgat J. Functional heteromerization of HCN1 and HCN2 pacemaker channels. J Biol Chem 2001;276:6069–72. [24] Much B, Wahl-Schott C, Zong X, Schneider A, Baumann L, Moosmang S, et al. Role of subunit heteromerization and N-linked glycosylation in the formation of functional hyperpolarization-activated cyclic nucleotide-gated channels. J Biol Chem 2003;278:43781–6. [25] Altomare C, Terragni B, Brioschi C, Milanesi R, Pagliuca C, Viscomi C, et al. Heteromeric HCN1–HCN4 channels: a comparison with native pacemaker channels from the rabbit sinoatrial node. J Physiol 2003;549:347–59. [26] Moroni A, Barbuti A, Altomare C, Viscomi C, Morgan J, Baruscotti M, et al. Kinetic and ionic properties of the human HCN2 pacemaker channel. Pflugers Arch 2000;439:618–26. [27] Tran N, Proenza C, Macri V, Petigara F, Sloan E, Samler S, et al. A conserved domain in the NH2 terminus important for assembly and functional expression of pacemaker channels. J Biol Chem 2002;277:43588–92. [28] Giorgetti A, Carloni P, Mistrik P, Torre V. A homology model of the pore region of HCN channels. Biophys J 2005;89:932–44. [29] Shin KS, Rothberg BS, Yellen G. Blocker state dependence and trapping in hyperpolarization-activated cation channels: evidence for an intracellular activation gate. J Gen Physiol 2001;117:91–101. [30] Doyle DA, Morais CJ, Pfuetzner RA, Kuo A, Gulbis JM, Cohen SL, et al. The structure of the potassium channel: molecular basis of K+ conduction and selectivity. Science 1998;280:69–77. [31] Craven KB, Zagotta WN. CNG and HCN channels: two peas, one pod. Annu Rev Physiol 2006;68:375–401. [32] Zhou L, Siegelbaum SA. Gating of HCN channels by cyclic nucleotides: residue contacts that underlie ligand binding, selectivity, and efficacy. Structure 2007;15: 655–70. [33] Long SB, Campbell EB, MacKinnon R. Voltage sensor of Kv1.2: structural basis of electromechanical coupling. Science 2005;309:903–8. [34] Chen J, Mitcheson JS, Tristani-Firouzi M, Lin M, Sanguinetti MC. The S4–S5 linker couples voltage sensing and activation of pacemaker channels. Proc Natl Acad Sci U S A 2001;98:11277–82. [35] Decher N, Chen J, Sanguinetti MC. Voltage-dependent gating of hyperpolarization-activated, cyclic nucleotide-gated pacemaker channels: molecular coupling between the S4–S5 and C-linkers. J Biol Chem 2004;279:13859–65. [36] Wainger BJ, DeGennaro M, Santoro B, Siegelbaum SA, Tibbs GR. Molecular mechanism of cAMP modulation of HCN pacemaker channels. Nature 2001;411:805–10. [37] Craven KB, Zagotta WN. Salt bridges and gating in the COOH-terminal region of HCN2 and CNGA1 channels. J Gen Physiol 2004;124:663–77. [38] Van Mierop LH. Location of pacemaker in chick embryo heart at the time of initiation of heartbeat. Am J Physiol 1967;212:407–15. [39] Yasui K, Liu W, Opthof T, Kada K, Lee JK, Kamiya K, et al. I(f) current and spontaneous activity in mouse embryonic ventricular myocytes. Circ Res 2001;88:536–42. [40] Garcia-Frigola C, Shi Y, Evans SM. Expression of the hyperpolarization-activated cyclic nucleotide-gated cation channel HCN4 during mouse heart development. Gene Expr Patterns 2003;3:777–83. [41] Hoogaars WM, Tessari A, Moorman AF, de Boer PA, Hagoort J, Soufan AT, et al. The transcriptional repressor Tbx3 delineates the developing central conduction system of the heart. Cardiovasc Res 2004;62:489–99. [42] Remme CA, Verkerk AO, Hoogaars WM, Aanhaanen WT, Scicluna BP, Annink C, et al. The cardiac sodium channel displays differential distribution in the conduction system and transmural heterogeneity in the murine ventricular myocardium. Basic Res Cardiol 2009 PMID: 19255801 [Electronic publication ahead of print]. [43] Hoogaars WM, Engel A, Brons JF, Verkerk AO, de Lange FJ, Wong LY, et al. Tbx3 controls the sinoatrial node gene program and imposes pacemaker function on the atria. Genes Dev 2007;21:1098–112. [44] Espinoza-Lewis RA, Yu L, He F, Liu H, Tang R, Shi J, et al. Shox2 is essential for the differentiation of cardiac pacemaker cells by repressing Nkx2-5. Dev Biol 2009;327:376–85. [45] Stieber J, Herrmann S, Feil S, Loster J, Feil R, Biel M, et al. The hyperpolarizationactivated channel HCN4 is required for the generation of pacemaker action potentials in the embryonic heart. Proc Natl Acad Sci U S A 2003;100:15235–40. [46] Kuratomi S, Kuratomi A, Kuwahara K, Ishii TM, Nakao K, Saito Y, et al. NRSF regulates the developmental and hypertrophic changes of HCN4 transcription in rat cardiac myocytes. Biochem Biophys Res Commun 2007;353:67–73. [47] Maltsev VA, Rohwedel J, Hescheler J, Wobus AM. Embryonic stem cells differentiate in vitro into cardiomyocytes representing sinusnodal, atrial and ventricular cell types. Mech Dev 1993;44:41–50. [48] Maltsev VA, Wobus AM, Rohwedel J, Bader M, Hescheler J. Cardiomyocytes differentiated in vitro from embryonic stem cells developmentally express cardiac-specific genes and ionic currents. Circ Res 1994;75:233–44. [49] Hescheler J, Fleischmann BK, Lentini S, Maltsev VA, Rohwedel J, Wobus AM, et al. Embryonic stem cells: a model to study structural and functional properties in cardiomyogenesis. Cardiovasc Res 1997;36:149–62. [50] Sartiani L, Bettiol E, Stillitano F, Mugelli A, Cerbai E, Jaconi ME. Developmental changes in cardiomyocytes differentiated from human embryonic stem cells: a molecular and electrophysiological approach. Stem Cells 2007;25:1136–44. [51] Satin J, Kehat I, Caspi O, Huber I, Arbel G, Itzhaki I, et al. Mechanism of spontaneous excitability in human embryonic stem cell derived cardiomyocytes. J Physiol 2004;559:479–96. [52] Barbuti A, Crespi A, Capilupo D, Mazzocchi N, Baruscotti M, DiFrancesco D. Molecular composition and functional properties of f-channels in murine embryonic stem cell-derived pacemaker cells. J Mol Cell Cardiol 2009;46:343–51. [53] Qu Y, Whitaker GM, Hove-Madsen L, Tibbits GF, Accili EA. Hyperpolarizationactivated cyclic nucleotide-modulated ‘HCN’ channels confer regular and faster rhythmicity to beating mouse embryonic stem cells. J Physiol 2008;586:701–16. [54] Abi-Gerges N, Ji GJ, Lu ZJ, Fischmeister R, Hescheler J, Fleischmann BK. Functional expression and regulation of the hyperpolarization activated non-selective cation current in embryonic stem cell-derived cardiomyocytes. J Physiol 2000;523: 377–89. [55] White SM, Claycomb WC. Embryonic stem cells form an organized, functional cardiac conduction system in vitro. Am J Physiol, Heart Circ Physiol 2005;288: H670–9. [56] van Kempen M, van Ginneken A, de Grijs I, Mutsaers N, Opthof T, Jongsma H, et al. Expression of the electrophysiological system during murine embryonic stem cell cardiac differentiation. Cell Physiol Biochem 2003;13:263–70. [57] Yanagi K, Takano M, Narazaki G, Uosaki H, Hoshino T, Ishii T, et al. Hyperpolarization-activated cyclic nucleotide-gated channels and T-type calcium channels confer automaticity of embryonic stem cell-derived cardiomyocytes. Stem Cells 2007;25:2712–9. [58] Liu J, Dobrzynski H, Yanni J, Boyett MR, Lei M. Organisation of the mouse sinoatrial node: structure and expression of HCN channels. Cardiovasc Res 2007;73:729–38. [59] Marionneau C, Couette B, Liu J, Li H, Mangoni ME, Nargeot J, et al. Specific pattern of ionic channel gene expression associated with pacemaker activity in the mouse heart. J Physiol 2005;562:223–34. [60] Tellez JO, Dobrzynski H, Greener ID, Graham GM, Laing E, Honjo H, et al. Differential expression of ion channel transcripts in atrial muscle and sinoatrial node in rabbit. Circ Res 2006;99:1384–93. [61] Brioschi C, Micheloni S, Tellez JO, Pisoni G, Longhi R, Moroni P, et al. Distribution of the pacemaker HCN4 channel mRNA and protein in the rabbit sinoatrial node. J Mol Cell Cardiol 2009;47:221–7. [62] Chandler NJ, Greener ID, Tellez JO, Inada S, Musa H, Molenaar P, et al. Molecular architecture of the human sinus node: insights into the function of the cardiac pacemaker. Circulation 2009;119:1562–75. [63] Moosmang S, Stieber J, Zong X, Biel M, Hofmann F, Ludwig A. Cellular expression and functional characterization of four hyperpolarization-activated pacemaker channels in cardiac and neuronal tissues. Eur J Biochem 2001;268:1646–52. [64] Ishii TM, Takano M, Xie LH, Noma A, Ohmori H. Molecular characterization of the hyperpolarization-activated cation channel in rabbit heart sinoatrial node. J Biol Chem 1999;274:12835–9. [65] Shi W, Wymore R, Yu H, Wu J, Wymore RT, Pan Z, et al. Distribution and prevalence of hyperpolarization-activated cation channel (HCN) mRNA expression in cardiac tissues. Circ Res 1999;85:e1–6. [66] Thollon C, Bedut S, Villeneuve N, Coge F, Piffard L, Guillaumin JP, et al. Usedependent inhibition of hHCN4 by ivabradine and relationship with reduction in pacemaker activity. Br J Pharmacol 2007;150:37–46. [67] Stieber J, Stockl G, Herrmann S, Hassfurth B, Hofmann F. Functional expression of the human HCN3 channel. J Biol Chem 2005;280:34635–43. M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 [68] Munk AA, Adjemian RA, Zhao J, Ogbaghebriel A, Shrier A. Electrophysiological properties of morphologically distinct cells isolated from the rabbit atrioventricular node. J Physiol 1996;493:801–18. [69] Hancox JC, Levi AJ, Lee CO, Heap P. A method for isolating rabbit atrioventricular node myocytes which retain normal morphology and function. Am J Physiol 1993;265:H755–66. [70] Yuill KH, Hancox JC. Characteristics of single cells isolated from the atrioventricular node of the adult guinea-pig heart. Pflugers Arch 2002;445:311–20. [71] Dobrzynski H, Nikolski VP, Sambelashvili AT, Greener ID, Yamamoto M, Boyett MR, et al. Site of origin and molecular substrate of atrioventricular junctional rhythm in the rabbit heart. Circ Res 2003;93:1102–10. [72] Liu J, Noble PJ, Xiao G, Abdelrahman M, Dobrzynski H, Boyett MR, Lei M, Noble D. Role of pacemaking current in cardiac nodes: insights from a comparative study of sinoatrial node and atrioventricular node. Prog Biophys Mol Biol 2008;96: 294–304. [73] Weidmann S. Effect of current flow on the membrane potential of cardiac muscle. J Physiol 1951;115:227–36. [74] Vassalle M. Analysis of cardiac pacemaker potential using a “voltage clamp” technique. Am J Physiol 1966;210:1335–41. [75] Shi W, Yu H, Wu J, Zuckerman J, Wymore R. The distribution and prevalence of HCN isoforms in the canine heart and their relation to the voltage dependence of If. Biophys J 2000;78:353A (Abstract). [76] Han W, Bao W, Wang Z, Nattel S. Comparison of ion-channel subunit expression in canine cardiac Purkinje fibers and ventricular muscle. Circ Res 2002;91:790–7. [77] Gaborit N, Le Bouter S, Szuts V, Varro A, Escande D, Nattel S, et al. Regional and tissue specific transcript signatures of ion channel genes in the non-diseased human heart. J Physiol 2007;582:675–93. [78] Porciatti F, Pelzmann B, Cerbai E, Schaffer P, Pino R, Bernhart E, et al. The pacemaker current I(f) in single human atrial myocytes and the effect of β-adrenoceptor and A1-adenosine receptor stimulation. Br J Pharmacol 1997;122:963–9. [79] Hoppe UC, Beuckelmann DJ. Characterization of the hyperpolarization-activated inward current in isolated human atrial myocytes. Cardiovasc Res 1998;38: 788–801. [80] Yu H, Chang F, Cohen IS. Pacemaker current i(f) in adult canine cardiac ventricular myocytes. J Physiol 1995;485:469–83. [81] Hoppe UC, Jansen E, Sudkamp M, Beuckelmann DJ. Hyperpolarization-activated inward current in ventricular myocytes from normal and failing human hearts. Circulation 1998;97:55–65. [82] Cerbai E, Sartiani L, DePaoli P, Pino R, Maccherini M, Bizzarri F, et al. The properties of the pacemaker current I(F)in human ventricular myocytes are modulated by cardiac disease. J Mol Cell Cardiol 2001;33:441–8. [83] Fernandez-Velasco M, Goren N, Benito G, Blanco-Rivero J, Bosca L, Delgado C. Regional distribution of hyperpolarization-activated current (If) and hyperpolarization-activated cyclic nucleotide-gated channel mRNA expression in ventricular cells from control and hypertrophied rat hearts. J Physiol 2003;553: 395–405. [84] Zicha S, Fernandez-Velasco M, Lonardo G, L'Heureux N, Nattel S. Sinus node dysfunction and hyperpolarization-activated (HCN) channel subunit remodeling in a canine heart failure model. Cardiovasc Res 2005;66:472–81. [85] Stillitano F, Lonardo G, Zicha S, Varro A, Cerbai E, Mugelli A, et al. Molecular basis of funny current (If) in normal and failing human heart. J Mol Cell Cardiol 2008;45:289–99. [86] Opthof T. The membrane current (I(f)) in human atrial cells: implications for atrial arrhythmias. Cardiovasc Res 1998;38:537–40. [87] Lai LP, Su MJ, Lin JL, Tsai CH, Lin FY, Chen YS, et al. Measurement of funny current (I(f)) channel mRNA in human atrial tissue: correlation with left atrial filling pressure and atrial fibrillation. J Cardiovasc Electrophysiol 1999;10: 947–53. [88] Workman AJ, Rankin AC. Serotonin, I(f) and human atrial arrhythmia. Cardiovasc Res 1998;40:436–7. [89] Cerbai E, Barbieri M, Mugelli A. Occurrence and properties of the hyperpolarization-activated current If in ventricular myocytes from normotensive and hypertensive rats during aging. Circulation 1996;94:1674–81. [90] Cerbai E, Pino R, Porciatti F, Sani G, Toscano M, Maccherini M, et al. Characterization of the hyperpolarization-activated current, I(f), in ventricular myocytes from human failing heart. Circulation 1997;95:568–71. [91] Xiao J, Yang B, Lin H, Lu Y, Luo X, Wang Z. Novel approaches for gene-specific interference via manipulating actions of microRNAs: examination on the pacemaker channel genes HCN2 and HCN4. J Cell Physiol 2007;212:285–92. [92] Luo X, Lin H, Pan Z, Xiao J, Zhang Y, Lu Y, et al. Down-regulation of miR-1/miR-133 contributes to re-expression of pacemaker channel genes HCN2 and HCN4 in hypertrophic heart. J Biol Chem 2008;283:20045–52. [93] Qu J, Barbuti A, Protas L, Santoro B, Cohen IS, Robinson RB. HCN2 overexpression in newborn and adult ventricular myocytes: distinct effects on gating and excitability. Circ Res 2001;89:E8–E14. [94] Yu H, Wu J, Potapova I, Wymore RT, Holmes B, Zuckerman J, et al. MinK-related peptide 1: a β subunit for the HCN ion channel subunit family enhances expression and speeds activation. Circ Res 2001;88:E84–7. [95] Decher N, Bundis F, Vajna R, Steinmeyer K. KCNE2 modulates current amplitudes and activation kinetics of HCN4: influence of KCNE family members on HCN4 currents. Pflugers Arch 2003;446:633–40. [96] Qu J, Kryukova Y, Potapova IA, Doronin SV, Larsen M, Krishnamurthy G, et al. MiRP1 modulates HCN2 channel expression and gating in cardiac myocytes. J Biol Chem 2004;279:43497–502. [97] Proenza C, Angoli D, Agranovich E, Macri V, Accili EA. Pacemaker channels produce an instantaneous current. J Biol Chem 2002;277:5101–9. 63 [98] Pian P, Bucchi A, Robinson RB, Siegelbaum SA. Regulation of gating and rundown of HCN hyperpolarization-activated channels by exogenous and endogenous PIP2. J Gen Physiol 2006;128:593–604. [99] Zolles G, Klocker N, Wenzel D, Weisser-Thomas J, Fleischmann BK, Roeper J, et al. Pacemaking by HCN channels requires interaction with phosphoinositides. Neuron 2006;52:1027–36. [100] Pian P, Bucchi A, Decostanzo A, Robinson RB, Siegelbaum SA. Modulation of cyclic nucleotide-regulated HCN channels by PIP(2) and receptors coupled to phospholipase C. Pflugers Arch 2007;455:125–45. [101] Suchy SF, Olivos-Glander IM, Nussabaum RL. Lowe syndrome, a deficiency of phosphatidylinositol 4,5-bisphosphate 5-phosphatase in the Golgi apparatus. Hum Mol Genet 1995;4:2245–50. [102] Pasquare SJ, Salvador GA, Giusto NM. Phospholipase D and phosphatidate phosphohydrolase activities in rat cerebellum during aging. Lipids 2004;39:553–60. [103] Suh BC, Hille B. Regulation of ion channels by phosphatidylinositol 4,5bisphosphate. Curr Opin Neurobiol 2005;15:370–8. [104] Barbuti A, Terragni B, Brioschi C, DiFrancesco D. Localization of f-channels to caveolae mediates specific β2-adrenergic receptor modulation of rate in sinoatrial myocytes. J Mol Cell Cardiol 2007;42:71–8. [105] Ye B, Balijepalli RC, Foell JD, Kroboth S, Ye Q, Luo YH, et al. Caveolin-3 associates with and affects the function of hyperpolarization-activated cyclic nucleotidegated channel 4. Biochemistry 2008;47:12312–8. [106] Barbuti A, Gravante B, Riolfo M, Milanesi R, Terragni B, DiFrancesco D. Localization of pacemaker channels in lipid rafts regulates channel kinetics. Circ Res 2004;94:1325–31. [107] Accili EA, Redaelli G, DiFrancesco D. Differential control of the hyperpolarizationactivated current (i(f)) by cAMP gating and phosphatase inhibition in rabbit sinoatrial node myocytes. J Physiol 1997;500:643–51. [108] Wu JY, Cohen IS. Tyrosine kinase inhibition reduces i(f) in rabbit sinoatrial node myocytes. Pflugers Arch 1997;434:509–14. [109] Altomare C, Tognati A, Bescond J, Ferroni A, Baruscotti M. Direct inhibition of the pacemaker (If) current in rabbit sinoatrial node cells by genistein. Br J Pharmacol 2006;147:36–44. [110] Yu H, Chang F, Cohen IS. Phosphatase inhibition by calyculin A increases i(f) in canine Purkinje fibers and myocytes. Pflugers Arch 1993;422:614–6. [111] Zong X, Eckert C, Yuan H, Wahl-Schott C, Abicht H, Fang L, et al. A novel mechanism of modulation of hyperpolarization-activated cyclic nucleotide-gated channels by Src kinase. J Biol Chem 2005;280:34224–32. [112] Arinsburg SS, Cohen IS, Yu HG. Constitutively active Src tyrosine kinase changes gating of HCN4 channels through direct binding to the channel proteins. J Cardiovasc Pharmacol 2006;47:578–86. [113] Li CH, Zhang Q, Teng B, Mustafa SJ, Huang JY, Yu HG. Src tyrosine kinase alters gating of hyperpolarization-activated HCN4 pacemaker channel through Tyr531. Am J Physiol, Cell Physiol 2008;294:C355–62. [114] Huang J, Huang A, Zhang Q, Lin YC, Yu HG. Novel mechanism for suppression of hyperpolarization-activated cyclic nucleotide-gated pacemaker channels by receptor-like tyrosine phosphatase-alpha. J Biol Chem 2008;283:29912–9. [115] Kryukova Y, Rybin VO, Qu J, Steinberg SF, Robinson RB. Age-dependent differences in the inhibition of HCN2 current in rat ventricular myocytes by the tyrosine kinase inhibitor erbstatin. Pflugers Arch 2009;457:821–30. [116] Herrmann S, Stieber J, Stockl G, Hofmann F, Ludwig A. HCN4 provides a ‘depolarization reserve’ and is not required for heart rate acceleration in mice. EMBO J 2007;26:4423–32. [117] Hoesl E, Stieber J, Herrmann S, Feil S, Tybl E, Hofmann F, et al. Tamoxifeninducible gene deletion in the cardiac conduction system. J Mol Cell Cardiol 2008;45:62–9. [118] Mistrik P, Mader R, Michalakis S, Weidinger M, Pfeifer A, Biel M. The murine HCN3 gene encodes a hyperpolarization-activated cation channel with slow kinetics and unique response to cyclic nucleotides. J Biol Chem 2005;280:27056–61. [119] Moroni A, Gorza L, Beltrame M, Gravante B, Vaccari T, Bianchi ME, et al. Hyperpolarization-activated cyclic nucleotide-gated channel 1 is a molecular determinant of the cardiac pacemaker current I(f). J Biol Chem 2001;276: 29233–41. [120] Schulze-Bahr E, Neu A, Friederich P, Kaupp UB, Breithardt G, Pongs O, et al. Pacemaker channel dysfunction in a patient with sinus node disease. J Clin Invest 2003;111:1537–45. [121] Ueda K, Nakamura K, Hayashi T, Inagaki N, Takahashi M, Arimura T, et al. Functional characterization of a trafficking-defective HCN4 mutation, D553N, associated with cardiac arrhythmia. J Biol Chem 2004;279:27194–8. [122] Milanesi R, Baruscotti M, Gnecchi-Ruscone T, DiFrancesco D. Familial sinus bradycardia associated with a mutation in the cardiac pacemaker channel. N Engl J Med 2006;354:151–7. [123] Nof E, Luria D, Brass D, Marek D, Lahat H, Reznik-Wolf H, et al. Point mutation in the HCN4 cardiac ion channel pore affecting synthesis, trafficking, and functional expression is associated with familial asymptomatic sinus bradycardia. Circulation 2007;116:463–70. [124] Qu J, Plotnikov AN, Danilo Jr P, Shlapakova I, Cohen IS, Robinson RB, et al. Expression and function of a biological pacemaker in canine heart. Circulation 2003;107:1106–9. [125] Plotnikov AN, Sosunov EA, Qu J, Shlapakova IN, Anyukhovsky EP, Liu L, et al. Biological pacemaker implanted in canine left bundle branch provides ventricular escape rhythms that have physiologically acceptable rates. Circulation 2004;109:506–12. [126] Bucchi A, Plotnikov AN, Shlapakova I, Danilo Jr P, Kryukova Y, Qu J, et al. Wild-type and mutant HCN channels in a tandem biological-electronic cardiac pacemaker. Circulation 2006;114:992–9. 64 M. Baruscotti et al. / Journal of Molecular and Cellular Cardiology 48 (2010) 55–64 [127] Tse HF, Xue T, Lau CP, Siu CW, Wang K, Zhang QY, et al. Bioartificial sinus node constructed via in vivo gene transfer of an engineered pacemaker HCN Channel reduces the dependence on electronic pacemaker in a sick-sinus syndrome model. Circulation 2006;114:1000–1. [128] Cai J, Yi FF, Li YH, Yang XC, Song J, Jiang XJ, et al. Adenoviral gene transfer of HCN4 creates a genetic pacemaker in pigs with complete atrioventricular block. Life Sci 2007;80:1746–53. [129] Potapova I, Plotnikov A, Lu Z, Danilo Jr P, Valiunas V, Qu J, et al. Human mesenchymal stem cells as a gene delivery system to create cardiac pacemakers. Circ Res 2004;94:952–9. [130] Yang XJ, Zhou YF, Li HX, Han LH, Jiang WP. Mesenchymal stem cells as a gene delivery system to create biological pacemaker cells in vitro. J Int Med Res 2008;36:1049–55. [131] Cho HC, Kashiwakura Y, Marban E. Creation of a biological pacemaker by cell fusion. Circ Res 2007;100:1112–5. [132] Wobus AM, Kleppisch T, Maltsev V, Hescheler J. Cardiomyocyte-like cells differentiated in vitro from embryonic carcinoma cells P19 are characterized by functional expression of adrenoceptors and Ca2+ channels. In Vitro Cell Dev Biol, Anim 1994;30A:425–34. [133] Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest 2001;108:407–14. [134] He JQ, Ma Y, Lee Y, Thomson JA, Kamp TJ. Human embryonic stem cells develop into multiple types of cardiac myocytes: action potential characterization. Circ Res 2003;93:32–9. [135] Mummery C, Ward-van Oostwaard D, Doevendans P, Spijker R, van den Brink S, Hassink R, et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation 2003;107: 2733–40. [136] Kehat I, Khimovich L, Caspi O, Gepstein A, Shofti R, Arbel G, et al. Electromechanical integration of cardiomyocytes derived from human embryonic stem cells. Nat Biotechnol 2004;22:1282–9. [137] Xue T, Cho HC, Akar FG, Tsang SY, Jones SP, Marban E, et al. Functional integration of electrically active cardiac derivatives from genetically engineered human embryonic stem cells with quiescent recipient ventricular cardiomyocytes: insights into the development of cell-based pacemakers. Circulation 2005;111:11–20. [138] Bucchi A, Tognati A, Milanesi R, Baruscotti M, DiFrancesco D. Properties of ivabradine-induced block of HCN1 and HCN4 pacemaker channels. J Physiol 2006;572:335–46. [139] Stieber J, Wieland K, Stockl G, Ludwig A, Hofmann F. Bradycardic and proarrhythmic properties of sinus node inhibitors. Mol Pharmacol 2006;69:1328–37. [140] Borer JS, Fox K, Jaillon P, Lerebours G. Antianginal and antiischemic effects of ivabradine, an I(f) inhibitor, in stable angina: a randomized, double-blind, multicentered, placebo-controlled trial. Circulation 2003;107:817–23. [141] Tardif JC, Ford I, Tendera M, Bourassa MG, Fox K. Efficacy of ivabradine, a new selective I(f) inhibitor, compared with atenolol in patients with chronic stable angina. Eur Heart J 2005;26:2529–36. [142] Ruzyllo W, Tendera M, Ford I, Fox KM. Antianginal efficacy and safety of ivabradine compared with amlodipine in patients with stable effort angina pectoris: a 3-month randomised, double-blind, multicentre, noninferiority trial. Drugs 2007;67:05–393. [143] Lopez-Bescos L, Filipova S, Martos R. Long-term safety and efficacy of ivabradine in patients with chronic stable angina. Cardiology 2007;108:387–96. [144] Fox K, Ford I, Steg PG, Tendera M, Ferrari R. Ivabradine for patients with stable coronary artery disease and left-ventricular systolic dysfunction (BEAUTIFUL): a randomised, double-blind, placebo-controlled trial. Lancet 2008;372:807–16. [145] Fox K, Ford I, Steg PG, Tendera M, Robertson M, Ferrari R. Heart rate as a prognostic risk factor in patients with coronary artery disease and leftventricular systolic dysfunction (BEAUTIFUL): a subgroup analysis of a randomised controlled trial. Lancet 2008;372:817–21. [146] Satoh H, Hashimoto K. Electrophysiological study of alinidine in voltage clamped rabbit sino-atrial node cells. Eur J Pharmacol 1986;121:211–9. [147] Goethals M, Raes A, Van Bogaert PP. Use-dependent block of the pacemaker current I(f) in rabbit sinoatrial node cells by zatebradine (UL-FS 49). On the mode of action of sinus node inhibitors. Circulation 1993;88:2389–401. [148] Thollon C, Cambarrat C, Vian J, Prost JF, Peglion JL, Vilaine JP. Electrophysiological effects of S 16257, a novel sino-atrial node modulator, on rabbit and guinea-pig cardiac preparations: comparison with UL-FS 49. Br J Pharmacol 1994;112:37–42. [149] Bois P, Bescond J, Renaudon B, Lenfant J. Mode of action of bradycardic agent, S 16257, on ionic currents of rabbit sinoatrial node cells. Br J Pharmacol 1996;118: 1051–7. [150] BoSmith RE, Briggs I, Sturgess NC. Inhibitory actions of ZENECA ZD7288 on whole-cell hyperpolarization activated inward current (If) in guinea-pig dissociated sinoatrial node cells. Br J Pharmacol 1993;110:343–9. [151] Marshall PW, Rouse W, Briggs I, Hargreaves RB, Mills SD, McLoughlin BJ. ICI D7288, a novel sinoatrial node modulator. J Cardiovasc Pharmacol 1993;21:902–6.