You are on page 1of 12

REVIEWS

Exploring HCN channels as novel


drug targets
Otilia Postea* and Martin Biel‡*
Abstract | Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have a key
role in the control of heart rate and neuronal excitability. Ivabradine is the first compound
acting on HCN channels to be clinically approved for the treatment of angina pectoris. HCN
channels may offer excellent opportunities for the development of novel anticonvulsant,
anaesthetic and analgesic drugs. In support of this idea, some well-established drugs that act
on the central nervous system — including lamotrigine, gabapentin and propofol — have
been found to modulate HCN channel function. This Review gives an up-to-date summary of
compounds acting on HCN channels, and discusses strategies to further explore the potential
of these channels for therapeutic intervention.
Hyperpolarization-activated cyclic nucleotide-gated HCN channels are members of the pore loop cation
Pacemaker cells
Cardiac cells that display
(HCN) channels were discovered over ten years ago, yet channel superfamily 10. In mammals the HCN channel
automaticity. The primary many aspects of their activation mechanism, their modu- family comprises four homologous members (HCN1–
pacemaker cells of the heart lation in vivo, their cellular and subcellular distribution, HCN4) that share about 60% sequence identity with
are localized in the sinoatrial and their interaction with agonists or antagonists remain each other and 25% sequence identity with their closest
node region.
unclear 1–3. HCN channels allow the passage of a depolar- relatives — the cyclic nucleotide-gated channels (FIG. 1a).
Sinoatrial node izing mixed Na+ and K+ current — the hyperpolarization- HCN channels are present in all vertebrates and also in
Tissue located in the right activated current (termed Ih) or the funny current (termed several invertebrates11–20.
atrium of the heart that If) — which has a key role in controlling the rhythmic HCN channels are tetrameric complexes consisting of
generates cardiac sinus
activity (also known as the pacemaker current) in cardiac either identical (homomeric) or different (heteromeric)
rhythm.
pacemaker cells and spontaneously firing neurons. Ih is also types of subunits. The topology of a HCN channel sub­
involved in several non-pacemaking functions, including unit is shown in FIG. 1. The transmembrane core includes
determination of resting membrane potential, dendritic six α-helices (S1–S6) and a loop domain between the
integration, synaptic transmission and learning. Thus, S5 and S6 helix that forms the ion selectivity filter.
HCN channels are interesting targets not only for the Like other voltage-operated cation channels, HCN chan-
development of drugs to lower heart rate but also for the nels contain a positively charged S4 helix that functions
treatment of diseases related to impaired neuronal activity. as a voltage sensor 21. HCN channels contain a cyclic
In this Review we briefly outline the current knowl- nucleotide-binding domain (CNBD) in the carboxyl
*Center for Drug Research, edge on the structure–function relationships, expression terminus that is connected to the channel core via an
Department of Pharmacy, patterns and basic biophysical profiles of HCN channels. 80‑amino-acid peptide called the C‑linker. The struc-
Ludwig-Maximilians- This is followed by a summary of the role of HCN chan- ture of the CNBD–C-linker has been determined at
Universität München,
Butenandtstr. 7, 81377
nels in disease. A more detailed overview of these topics atomic resolution22–23. Following the binding of cyclic
Munich, Germany. is provided in several recent publications1–6. This Review AMP or cyclic GMP to the channel, the CNBD–C-linker

Center for Integrated Protein focuses on the potential of HCN channels as targets for facilitates HCN channel activation by inducing a right-
Science Munich, Ludwig- bradycardic, anticonvulsant, analgesic and anaesthetic ward shift in the voltage-dependence of activation14,24,25
Maximilians-Universität
compounds, and discusses the challenges and future (FIG. 2a). Elucidation of the crystal structure of the HCN
München, Butenandtstr.
5-13, 81377 Munich, directions in the development of novel drugs targeting channel core should shed further light on the gating
Germany. HCN channels. machinery that couples the movement of the S4 helix
Correspondence to M.B. in the electrical field with the conformational change in
e-mail: martin.biel@cup. The HCN channel family the CNBD–C-linker domain following cAMP binding.
uni-muenchen.de
doi:10.1038/nrd3576
HCN channels represent the molecular correlate of Ih When they are expressed in heterologous systems the
Published online or If, which was identified in the late 1970s in sinoatrial HCN channel subunits give rise to currents that display
18 November 2011 node cells and neurons1,7–9. the typical hallmarks of native Ih: activation with sigmoidal

NATURE REVIEWS | DRUG DISCOVERY VOLUME 10 | DECEMBER 2011 | 903

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

a TRP b Pore the individual channel isoforms, is provided in TABLE 1.


TPCN Expression of HCN channels in the central nervous
Extracellular
Kir system (CNS) has been extensively studied at both the
protein and transcript level28–32 (reviewed in REF. 1).
KCa
Kv1–9
Among the four subtypes, HCN2 is expressed most
extensively and is found in most parts of the brain. By
K2P Intracellular contrast, expression of HCN1 and HCN4 is spatially more
Kv10 Ca2+
restricted. HCN1 is enriched in the neocortex, hippocam-
Kv11 K+ pus, cerebellar cortex and brainstem, whereas HCN4 is
Kv12 mainly present in thalamic nuclei, basal ganglia and the
Na+
CNGB olfactory bulb. HCN3 is weakly expressed in heart
CNGA + muscle33 and most parts of the CNS, except for the olfac-
S1 S2 S3 S4 S5 S6
+ tory bulb and some hypothalamic nuclei, which display
HCN2 +
HCN4 higher HCN3 expression levels26,31. HCN channels are also
Ih(If)
HCN1 C-linker found in retinal cells32,34 and in the peripheral nervous
HCN3 system, including dorsal root ganglion neurons32,35,36.
N C
Nav CNBD In all mammals studied so far, including humans
Cav cAMP
and mice, HCN4 represents the principal HCN channel
Figure 1 | HCN channels: phylogenetic tree and transmembrane topology. isoform present in the sinoatrial node12,20,32,37,38. The expres-
a | A schematic phylogenetic tree representing the superfamily of pore loop cation
sion of other isoforms in the sinoatrial node is much
channels is shown, including the hyperpolarization-activated cyclic nucleotide-gated
Nature Reviews
(HCN) channels. b | The diagram illustrates the transmembrane | Drug
topology Discovery
of HCN weaker and also seems to vary between species20,32. The key
channels. The channels are tetramers (as shown in the upper panel). Each subunit importance of HCN4 for the generation of sinoatrial If is
consists of six transmembrane α-helices (numbered S1–S6 on the figure). The fourth corroborated by electrophysiological studies that revealed
α-helix is positively charged and forms the putative voltage sensor of the channel. a 70–80% reduction in the total If in HCN4‑deficient
In the carboxyl terminus the subunits contain a cyclic nucleotide-binding domain sinoatrial node cells compared with wild-type cells39,40.
(CNBD) that is connected with the sixth transmembrane α-helix via the C‑linker. By contrast, deletion of HCN2 diminished total If only
HCN channels allow the passage of Na+ and K+. There is also evidence of minor by about 20% in murine sinoatrial node cells41. HCN
permeation of Ca2+. Cav, voltage-gated calcium channel; CNGA, cyclic nucleotide-gated
channels are also present in non-pacemaking cardio-
cation channel A subunit; CNGB, cyclic nucleotide-gated channel B subunit; If, funny
current; Ih, hyperpolarization-activated current; K2P, two-pore potassium channel;
myocytes of the atrium and ventricle14–15,20. The func-
KCa, calcium-activated potassium channel; Kir, inwardly rectifying potassium channel; tion of HCN channels in these cell types remains to be
Kv1, voltage-gated potassium channel subunit Kv1; Nav, voltage-gated sodium determined. HCN channel transcripts have also been
channel; TRP, transient receptor potential channel; TPCN, two-pore calcium channel. identified in other organs including the testis, smooth
muscle and kidney 1. However, in most of these organs
the presence of the HCN channel protein has not yet been
kinetics following membrane hyperpolarization (FIG. 2b); detected. Similarly, the functional roles of HCN channels
lack of voltage-dependent inactivation; conduction of Na+ in these organs are unclear. It should also be noted that
and K+; a shift in the activation curve as a result of direct the expression pattern of HCN channels displays an age-
interaction with cAMP; and inhibition by millimolar con- dependence and may change during development42–44.
centrations of external Cs+ (REFS 1,2). Currents mediated
by the four HCN channels differ from each other with Regulation of HCN channel function
regard to their activation kinetics, which are fast in HCN1, Like most ion channels, HCN channels form macro­
intermediate in HCN2, and slow in HCN3 and HCN4 mol­ec­ular complexes that consist of the principal ion-
(REFS 12,15,17,26). The speed of channel activation is also conducting channel core and auxiliary subunits (for
strongly voltage-dependent, and increases as activation example, scaffolding proteins and modulator proteins)
voltages become more hyperpolarized. HCN channels that are either permanently assembled with the channel
also differ from each other in their response to cAMP. core or that can bind and unbind in a regulated fashion.
The activation curves of HCN2 and HCN4 are shifted Affinity purification, combined with high-resolution
by about +15 mV, whereas HCN1 and HCN3 are only mass spectrometry, has revealed that HCN channels
weakly modulated by cAMP, if at all. As mentioned in the brain are tightly co-assembled with peroxisomal
above, HCN channels can be assembled into hetero­tetra­ biogenesis factor 5‑like protein (PEX5L; also known
mers. The biophysical properties of these heteromeric as TRIP8b) in a 1:1 ratio45. Functional studies suggest that
channels are a mixture of those determined for pure TRIP8b has an antagonistic effect on the cyclic nucleotide
homomeric channels1,27. It is likely that the formation of gating of HCN channels45–47. TRIP8b is also an important
heteromeric channels is an important process underlying modulator of cell-surface expression and intraneuronal
the diversity of the native Ih observed in different types trafficking of HCN channels48. There are more than ten
of neurons1,9. alternative amino‑terminal splice forms of TRIP8b in the
brain. Interestingly, some of these variants enhance HCN
Expression pattern of HCN channel subtypes. HCN channel densities, whereas others strongly downregulate
channels are predominantly expressed in the brain and the levels of HCN channels. These findings suggest that
heart. An overview of the different expression sites, as TRIP8b acts as a gatekeeper of neuronal HCN channel
well as the implicated functions and/or phenotypes of activity (reviewed in REF. 49).

904 | DECEMBER 2011 | VOLUME 10 www.nature.com/reviews/drugdisc

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

sO8
C 
D sO8

 sO8
UGEQPFU


++OCZ






s s s


8O O8 P#
sE#/2 E#/2 UGEQPFU

Figure 2 | Functional properties of HCN channel currents. a | The graph shows the voltage-dependence of
hyperpolarization-activated cyclic nucleotide-gated channel 2 (HCN2) activation in the absence or presence of 10 μM
cyclic AMP. cAMP induces a positive shift in the activation curve. The inset shows the accelerating effect of cAMP on
0CVWTG4GXKGYU^&TWI&KUEQXGT[
HCN2 channel activation. In this experiment, currents were evoked in an inside-out patch in the absence or presence of
10 μM cAMP by stepping from a holding potential of –40 mV to –120 mV (data were compiled from REF. 15). b | A family
of HCN2 current traces are shown that are activated following hyperpolarizing steps from a holding potential of
–40 mV to various test potentials, ranging between –140 mV and –50 mV. The current is activated with sigmoidal kinetics
and does not display voltage-dependent inactivation. I/Imax, ratio between current at a given membrane potential (I) and
the maximal current (Imax); nA, nanoampere; Vm, membrane potential.

The functional significance of several other proteins HCN channel currents are upregulated in hippocampal
that have been proposed to be assembled with HCN pyramidal neurons by p38 mitogen-activated protein
channels is less clear. Such proteins include: potassium kinase65, whereas the phosphatase calcineurin inhibits
voltage-gated channel, Isk-related family member 2 HCN channels in hippocampal pyramidal neurons by
(also known as MIRP1)50; potassium channel regulator 1 inducing a hyperpolarizing shift in the activation curve66.
(REF. 51) ; filamin A 52; the complex formed by GRP1 It remains to be determined whether the bidirectional reg-
(general receptor for phosphoinositides 1)-associated ulation exerted by p38 MAPK and calcineurin is caused
scaffold protein (also known as tamalin) and amyloid-β by direct phosphorylation and dephosphorylation of the
A4 precursor protein-binding family A member 2 (also channel or by a mechanism involving other proteins.
known as MINT2)53; as well as caveolin 3 (REF. 54). Further cGMP-dependent protein kinase 2 has been identified as
studies will be required to validate the specific roles of another regulator of HCN channels in the brain; it binds
these proteins with regard to HCN channel function. to the C terminus of HCN2 and phosphorylates a specific
In addition to auxiliary subunits, HCN channels are serine residue in the CNBD67. This modification induces a
regulated by several low-molecular-weight factors. The hyperpolarizing shift in the activation curve. Thus, cGMP
best characterized of these factors is phosphatidylinositol regulates HCN channel activity in two ways: it enhances
4,5‑bisphosphate, which was shown to shift the activation HCN currents by directly binding to the CNBD and it
curve of HCN channels by about +20 mV55,56. This regu- inhibits gating by phosphorylating the CNBD.
lation is of crucial importance because it is needed to
activate HCN channels in the physiological voltage range. HCN channels in human disease
In addition to phosphatidylinositol 4,5‑bisphosphate, HCN channels have an important role in the generation
other acidic lipids — like phosphatidic acid and arachi- of cardiac pacemaker activity and the control of neuronal
donic acid, which are products of diacylglycerol kinase– excitability. Impairment of HCN channel function (for
phospholipase A2 signalling pathways — directly facilitate example, resulting from inadequate expression levels or
HCN channel gating57. HCN channel activity is also regu- mutations in the primary sequence) has been implicated
lated by extracellular concentrations of K+ (REF. 58), extra- in the pathology of diseases of the heart and the nervous
and intracellular concentrations of H+ (REFS 59,60), and system. Here, we give a brief overview of pathologies
external concentrations of Cl– (REFS 58,62). involving HCN channels (TABLE 1; for further details see
A third group of HCN channel regulators comprises REFS 1,68–72).
Diastolic depolarization protein kinases and phosphatases. SRC kinase directly
Slow membrane interacts with HCN1, HCN2 and HCN4, and regulates HCN channels in arrhythmogenic diseases. Cardiac pace­
depolarization that occurs Ih by phosphorylating tyrosine residues. The regulation maker cells generate a specialized kind of action potential
between action potentials
(that is, in the diastole of
of HCN channels by SRC kinases has been demonstrated (known as a pacemaker potential) that is characterized
the heart) in pacemaker under physiological conditions in sinoatrial pacemaker by the presence of a progressive diastolic depolarization in
cells of the cardiac system. cells in murine and rat hearts as well as in neurons19,63,64. the voltage range between –65 mV and –45 mV (FIG. 3).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 10 | DECEMBER 2011 | 905

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Although the diastolic depolarization results from the are heterozygous for the respective HCN4 mutations.
Sinus bradycardia
Heart rhythm that originates
concerted action of several currents, If is generally con- A possible explanation for this finding might be that,
from the sinus node with a sidered to have a key role because it is activated at nega- like in mice, homozygous deletion of HCN4 function39
resting heart rate of 60 beats tive potentials and can therefore potentially serve as a or complete loss of cAMP-dependent modulation79 may
per minute or less. primary initiator of diastolic depolarization. Importantly, result in embryonic lethality.
up- and downregulation of If by an increase or decrease The first HCN4 channelopathy was identified in a
Syncope
Partial or complete loss
in sinoatrial cAMP levels, respectively, is believed to single patient who developed an idiopathic sinus node
of consciousness, with have a pivotal role in the autonomic control of heart rate. dysfunction that was characterized by a marked sinus
interruption of awareness of As described above, HCN4 is the predominant compo- bradycardia, episodes of syncope, intermittent atrial
oneself and one’s surroundings. nent of sinoatrial If in all mammals. The intensive search fibrillation and chronotropic incompetence even under
Syncope often occurs as a
result of an irregular heartbeat.
for genetically determined cardiac channelopathies has maximal workload73. Molecular genetic analysis revealed
resulted in the identification of six different mutations the presence of a single base deletion in exon 5 of the
Chronotropic incompetence in the HCN4 gene. All six mutations are associated HCN4 gene, which leads to the generation of a truncated
The inability to increase heart with the induction of sinus bradycardia73–78. In addition, protein lacking the CNBD and subsequent downstream
rate commensurately with one mutation (D553N) is associated with a complex sequence (referred to as L573X). In heterologous expres-
increased activity or demand.
arrhythmia syndrome74. All patients identified so far sion systems the truncated HCN channels are normally
targeted to the plasma membrane but — as expected —
they do not respond to cAMP.
In 2004 a missense mutation (D553N) in the HCN4
Table 1 | Expression pattern and involvement of HCN channels in disease gene was identified in a Japanese family 74. The patients
Isoform Expression sites Phenotypes had severe bradycardia combined with a complex pat-
tern of rhythmic disturbances that included recurrent
Nervous system
syncopes, QT prolongation, polymorphic ventricular tachy­
HCN1 Neocortex, • Mouse knockout: impaired motor learning, cardia and torsade de pointes. In vitro studies indicate that
hippocampus, enhanced hippocampal-dependent learning
brain stem19,28,30,31 and memory, less cold allodynia, epilepsy and
the D553N mutation is associated with impairment of
altered vision116,126,141 the cell-surface expression of HCN4. Downregulation
of HCN4 expression could explain the bradycardia but
HCN2 Ubiquitous28,30,31 • Mouse knockout: ataxia, absence
epilepsy41, as well as reduced inflammatory it does not readily explain the other complex symptoms
and neuro­pathic pain127 of these patients.
HCN3 Olfactory bulb, • Mouse knockout: not reported Another point mutation (S672R) in the HCN4 gene,
hypothalamic localized at a highly conserved position in the CNBD
nuclei, retinal cone domain, was identified in an Italian family with asymp-
pedicles2,26,28,31,34 tomatic sinus bradycardia75. Heterologous co-expression
HCN4 Thalamic nuclei, • Mouse knockout: not reported of mutant and wild-type HCN4 channels to mimic
basal ganglia, heterozygosis as present in patients revealed that the
olfactory bulb28,30,31 S672R mutation does not impair the cAMP-dependent
Heart modulation of the channel; rather, it induces a 5 mV
HCN1 Sinoatrial and • Not reported hyperpolarizing shift in the If activation curve. This
atrioventricular shift would explain the basal bradycardia combined with
node20, 142,143 normal autonomic control of heart rate that is observed
HCN2 Ubiquitous20,32 • Mouse knockout: sinus arrhythmia41 in patients with this mutation. Unexpectedly, autonomic
up- and downregulation of heart rate is completely
HCN3 Heart muscle 33
• Mouse knockout: increased T wave amplitude
in the electrocardiogram at basal heart rate33 preserved in patients with bradycardia who express an
HCN4 variant with a truncated and hence non-functional
HCN4 Sinoatrial and • Mouse knockout: embryonic lethal phenotype,
atrioventricular node, bradycardia, nonfunctional pacemaker cells39 CNBD77 (HCN4‑695X). This finding indicates that
Purkinje fibres20,32,39,145 • Conditional mouse knockout: repetitive sinus HCN4 is crucial for determining the basal heart rate but
pauses80,81, deep bradycardia, heart block40 challenges the notion that it is required for autonomic
(a disease in the electrical system of the heart) control of cardiac pacemaking.
• Human mutation: bradycardia, QT prolongation,
Although all four HCN4 mutations described above
syncope and conduction disturbances in
the sinoatrial and atrioventricular node73–76 occur in the C terminus of HCN4, a fifth mutation
(G480R) associated with asymptomatic bradycardia
Other tissues
was discovered in the pore region of this channel76. This
HCN1 Not reported • Not reported mutation involves the GYG signature that forms the
HCN2 Not reported • Not reported ion selectivity filter of potassium and HCN channels.
HCN3 Mouse liver, lung • Not reported Surprisingly, heterologous expression of this channel in
and kidney18,144 Xenopus laevis oocytes and human embryonic kidney
HCN4 Lung, skeletal • Not reported
293 (HEK293) cells showed that the ion selectivity of
muscle18, human HCN4 is not affected by the G480R mutation. Rather,
testis17 (but not the mutation shifts the activation curve to values that
mouse testis18) are more hyperpolarized and leads to a pronounced
HCN, hyperpolarization-activated cyclic nucleotide-gated channel. decrease in the cell-surface expression of HCN4 (REF. 76).

906 | DECEMBER 2011 | VOLUME 10 www.nature.com/reviews/drugdisc

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Similarly, another mutation in the HCN4 gene (A485V) a


Iv
downstream of the GYG signature has been identified
in three Moroccan families with sinus bradycardia, and 0
this mutation profoundly impairs HCN4 function
and synthesis78. –20

mV
The key role of HCN4 in cardiac pacemaking is Diastolic
depolarization
corroborated by genetic mouse models. Constitutive
cardiac deletion of HCN4 results in embryonic death –60
If
because of the failure to generate mature sinoatrial pace-
maker cells39. There is, however, an ongoing controversy
concerning the relevance of HCN4 in the adult murine b
heart. Herrmann et al.80 reported that the conditional
Iv Na+ Extracellular
deletion of HCN4 in mice does not induce bradycardia + + – –
at rest — rather, it leads to arrhythmia characterized by
recurrent sinus pauses81. By contrast, using a slightly dif- + +
+ Iv +
ferent strategy to conditionally delete HCN4 in sinoatrial + +
+ +
node cells, Baruscotti and colleagues40 observed a pro- + Iv +
+ +
gressive development of severe bradycardia and atrio-
ventricular block in mice, which led to cardiac arrest – – + +
Iv K+
and death. A cardiac phenotype was also observed in Intracellular
HCN2‑knockout mice. HCN2 makes up about 20% of Figure 3 | Mechanism of action of ivabradine on
total sinoatrial If 41. Deletion of the HCN2‑mediated cur- sinoatrial HCN channels. a | Hyperpolarization-
rent fraction does not impair basal heart rate nor does activated cyclic nucleotide-gated (HCN) channels
Nature Reviews allow
| Drug Discovery
it interfere with autonomic regulation of the heartbeat. the passage of the funny current (If), which is a major
Rather, HCN2‑deficient mice display a mild sinus dys- current producing slow diastolic depolarization of the
sinoatrial pacemaker potential. For clarity, other currents
rhythmia at rest. So far, HCN2 channelopathies have not
contributing to diastolic depolarization are omitted.
been reported in humans, and no naturally occurring Ivabradine (shown on the figure as ‘Iv’) reduces the
deletions or mutations in HCN1 have been described. frequency of sinoatrial pacemaker potentials by blocking
Besides the pathology resulting from inherited HCN HCN channels and hence reducing the slope of diastolic
channel mutations, there is evidence that upregulation depolarization. b | Current-dependent block of HCN
of the levels of HCN channel expression may have a role channels by ivabradine. Ivabradine enters the pore of
in the induction of ventricular and atrial arrhythmia in HCN channels from the intracellular side when the
heart failure (reviewed in REFS 82,83). Basal levels of channels are in the open state (left side of panel). When
HCN channels are very low in healthy heart muscle cells the membrane potentials are more positive than about
compared with sinoatrial pacemaker cells. However, –20 mV, HCN-mediated currents are mainly carried by K+
and are outwardly directed. The outward current drives
elevated mRNA and protein levels of HCN2 and HCN4
ivabradine into the pore of the channel, where it tightly
have been found in ventricular and atrial samples from binds to its binding site (right side of panel). The polarity
failing hearts explanted from patients with end-stage of the plasma membrane is indicated by ‘+’ or ‘–’ signs.
ischaemic cardiomyopathy 84. It is therefore tempting to The movement of the positively charged S4 helix of HCN
assume that increased If densities that may result from channels in the electrical field is also indicated.
HCN channel upregulation could endow cardiomyo-
cytes with pacemaker-like activity, thereby promoting
ectopic pacemaker activity.
on the pathophysiological importance of HCN channels
HCN channels in neurological diseases. HCN channels is by and large based on studies in animals (mostly mice
are widely expressed in the CNS and peripheral nervous and rats) and has yet to be confirmed in humans.
QT prolongation system. At the cellular level several basic functions have HCN channels are known to be involved in two com-
The QT interval represents been attributed to these channels, including neuronal plex groups of diseases: epilepsies and neuropathic pain
the time for electrical pacemaking, control of the membrane resting potential disorders1,4,71,72,85,86. Both groups of diseases are charac-
activation and inactivation
of the ventricles — the lower
and dendritic integration. It was therefore hypothesized terized by enhanced firing of neurons and/or aberrant
chambers of the heart. that the dysfunction and/or inadequate expression of neuronal firing patterns. HCN channels are key determi-
Prolongation of the QT interval HCN channels may be a disease-causing factor. This nants of neuronal network activity. Even subtle changes
can result in the potentially assumption has been supported by the analysis of in their expression levels, localization to subcellular
lethal arrythmia known
HCN1- and HCN2‑deficient mice that has revealed compartments, subunit composition or their cellular
as torsades de pointes.
several phenotypes resembling human neurological dis­ regulation can have profound consequences on neu-
Torsades de pointes orders, including epilepsy, ataxia and impaired learning. ronal network activity. Given the multifaceted functions
A form of polymorphous Conversely, channelopathies for the two major neu- of HCN channels, effects resulting from HCN channel
ventricular tachycardia ronal HCN channels, HCN1 and HCN2, have not been dysfunction are intrinsically complex and may strongly
that is associated with
prolongation of the cardiac
reported so far in humans, and patients with mutations depend on the pathophysiological context. For exam-
QT interval that can lead to in HCN4 do not have obvious defects other than cardiac ple, both upregulation87,88 and downregulation89 of HCN
sudden cardiac death. bradycardia or arrhythmia. Thus, our current knowledge channels have been associated with increased firing of

NATURE REVIEWS | DRUG DISCOVERY VOLUME 10 | DECEMBER 2011 | 907

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

peripheral sensory neurons following nerve injury. The Use-dependency is a very useful clinical property,
involvement of HCN channels in the development of as it specifically increases the efficiency of ivabradine
epilepsies is even more complex. As in pain disorders, during tachycardia. Interestingly, the mode of action of
there is evidence that both up- and downregulation of ivabradine seems to depend on the particular isoform
HCN channel subunits, particularly subunits of HCN1 of HCN channels. Ivabradine displays pronounced
and HCN2, are associated with epilepsies 90. There is current-dependency in HCN4 and in native sinoatrial
growing evidence that several mechanisms, including If 98; by contrast, it is a closed-channel blocker of HCN1
transcriptional control, trafficking and channel modu- channels. Zatebradine — which is structurally closely
lation (for example, alterations in subunit composition related to ivabradine — blocked HCN channels in a
that affect the biophysical properties of Ih), act at dif- purely voltage-dependent fashion and did not display
ferent temporal and spatial scales to modulate Ih in the current-dependency 97. Together, these findings suggest
brain of patients with epilepsy. For a detailed overview of that zatebradine and ivabradine, although structurally
the role of HCN channels in the development of different closely related to each other, have slightly different bind-
types of epilepsies, see REFS 4,72,86. ing sites on HCN channels. The interaction of both com-
pounds with HCN channels may be affected by residues
HCN channels as drug targets that are either distinct or spatially oriented in a different
HCN channel blockers as heart rate-reducing agents. way in the respective HCN channel isoforms.
Increased heart rate is an important risk factor in vari- Ivabradine was approved by the European Medicines
ous cardiac diseases, including heart failure, arterial Agency in 2005, and is the first clinically approved
hypertension and coronary heart disease 91–93. The compound that specifically targets HCN channels. The
reduction of an increased basal heart rate is beneficial therapeutic indication of ivabradine is the symptomatic
to patients and has been shown to prolong lifespan94. treatment of chronic stable angina pectoris in patients
Currently used bradycardic drugs (such as β‑adrenergic with coronary artery disease with a normal sinus
receptor antagonists) and blockers of the cardiac L‑type rhythm, who have contraindication or intolerance for
calcium channel efficiently reduce heart rate, but their beta blockers. Ivabradine may also be effective in various
use is limited by adverse reactions (for example, negative other cardiovascular conditions in which an exclusive
inotropic effects and interactions with vascular and lowering of heart rate is beneficial (for example, chronic
pulmonary smooth muscle)93,95. Inhibitors of HCN chan- heart failure, atherosclerosis and heart transplantation)99.
nels — particularly inhibitors of the major sinoatrial In particular, ivabradine and other If blockers may sup-
isoform, HCN4 — are well suited for therapeutically press the generation of ectopic action potentials that
lowering heart rate. Importantly, as HCN channels are are thought to be triggered by the upregulation of HCN
specifically expressed in the heart and the CNS, drugs channels in congestive heart failure100,101. Importantly, in
interfering with HCN channel activity will not be associ- contrast to β‑adrenergic receptor antagonists, ivabradine
ated with adverse effects on airway and vascular smooth does not induce depression of left ventricular function in
muscle tone. patients with left ventricular dysfunction102.
Drugs identified as selective bradycardic agents A large-scale multicenter clinical trial — BEAUTIFUL
include alinidine (ST567), which is a derivative of cloni- (morbidity-mortality evaluation of the If inhibitor
dine, ZD7288 and the ‘bradines’, which are a group of Procoralan in patients with coronary disease and left ven-
compounds derived from the calcium channel blocker tricular dysfunction) — enrolling almost 11,000 patients
verapamil96. The group of bradines comprises zatebradine evaluated the efficacy of ivabradine in reducing morbidity
(UL‑FS49), cilobradine (DK‑AH269) and ivabradine and/or mortality in patients with coronary artery dis-
(Procoralan; Servier); chemical structures of these and ease with left ventricular dysfunction103,104. This study
other drugs discussed in this article are provided in FIG. 4. revealed a clear benefit for the subgroup of patients with
These substances block sinoatrial If and hence reduce the a basal heart rate ≥70 beats per minute with respect to the
slope of the slow diastolic depolarization phase as well as occurrence of fatal and non-fatal myocardial infarction
the frequency of pacemaker potentials (FIG. 3a). and revascularization. In another clinical study — SHIFT
The mechanism of action of ivabradine has been (systolic heart failure treatment with the I f inhibitor
studied in detail in isolated rabbit sinoatrial node cells97 ivabradine trial) — that included about 6,500 patients
(FIG. 3b). Ivabradine can access HCN channels only from with chronic heart failure, ivabradine lowered major
the intracellular side when the channel pore is opened by risks associated with heart failure when it was adminis-
hyperpolarization and the current is inwardly directed. tered in addition to guideline-based and evidence-based
During the following depolarization phase in the action treatment 105 (most patients received beta blockers and
potential cycle, which is characterized by an outward ion antagonists of the renin–angiotensin–aldosterone sys-
flow, ivabradine is driven into the pore where it binds tem). In agreement with the results of the BEAUTIFUL
(to a site that has not yet been identified) and blocks study, the therapeutic benefit associated with ivabradine
the HCN channels (FIG. 3b). This mechanism of action directly correlated with pre-treatment heart rate (it was
is consistent with the observed use-dependency of significant in patients with a median baseline heart rate
ivabradine: the more frequently the direction of ion flow that was higher than 77 beats per minute).
Inotropic effects
Effects pertaining to the force
through the HCN channel changes, the more efficiently Given their promising therapeutic value, it is clearly
of muscular contractions, the ivabradine molecule can access its binding site in the desirable to further optimize bradycardic drugs acting
particularly those of the heart. inner pore. on HCN channels. Ivabradine and the other compounds

908 | DECEMBER 2011 | VOLUME 10 www.nature.com/reviews/drugdisc

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

O
O
N O
O
O
N N
O O
O N
O
O  

O
O
N HN HN
H O
N HN N N N
N Cl Cl Cl Cl
N

N N

   
O O

Cl

Cl OH
N
H2N Cl
H2N F
N O
Br
N N F H
F
N
NH2
   

Cl F OH
NH
F
O
O F
F N Cl Cl
F
N O OH

   
Figure 4 | Structures of compounds interacting with hyperpolarization-activated cyclic nucleotide-gated
channels. Compound 1, ivabradine (Procoralan; Servier); compound 2, zatebradine; compound 3, cilobradine;
0CVWTG4GXKGYU^&TWI&KUEQXGT[
compound 4, ZD7288; compound 5, clonidine; compound 6, alinidine; compound 7, nicotine; compound 8, lamotrigine
(Lamictal; GlaxoSmithKline); compound 9, gabapentin (Gabapen; Pfizer); compound 10, halothane; compound 11,
isofluorane; compound 12, loperamide; compound 13, propofol; compound 14, ketamine.

currently available have only a modest affinity for HCN and bipolar cells34,110 probably underlies the transient
channels. The IC50 value (the half-maximal inhibitory changes in visual sensation that are observed in about
concentration) of ivabradine is 1.5–4.5 μM under physio­ 15% of patients following initial treatment with ivabra-
logical conditions97,106. Off-target effects of ivabradine dine111. A recent study demonstrated that subtype speci-
have not yet been reported 96,107,108. Nevertheless, to ficity can be achieved by modifying the substituent on
further increase the specificity and long-term safety of the nitrogen atom in the benzazepin-4‑one backbone
HCN channel-based therapy (as patients will have to of ivabradine106,112 (FIG. 5). Ivabradine does not cross the
receive the treatment for many years), the development blood–brain barrier — a property that is crucial for its
of compounds acting in the nanomolar concentration clinical use as a bradycardic agent113. Future development
range would be a desirable goal. Achieving subtype of specific HCN channel blockers should also aim for
specificity among the four HCN channel subtypes is compounds that — unlike ivabradine — do not cross
another important goal. Ivabradine does not selectively the blood–retinal barrier.
target specific HCN channel subtypes106,109. Another strategy for developing cardiac-specific
As HCN4 is the principal HCN channel isoform in HCN channel blockers could rely on using clonidine as
the sinoatrial node (responsible for over 80% of total If), a a lead structure. Clonidine is a well-known α2-adrenergic
drug that specifically blocks this particular channel with- receptor agonist, and is chemically related to the brady-
out interfering with HCN1–HCN3 would be superior cardic agent alinidine. Studies in mice in which the
to the currently available compounds. Interaction with α2-adrenergic receptor was knocked out indicated that
the HCN1 isoform expressed in retinal photoreceptors inhibition of sinoatrial If contributes substantially to

NATURE REVIEWS | DRUG DISCOVERY VOLUME 10 | DECEMBER 2011 | 909

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

the bradycardic action of clonidine114. Clonidine blocks HCN channel blockers in the treatment of neuropathic
HCN4 with an IC50 of about 10 μM, but in contrast pain. There is growing evidence that HCN channels are
to ivabradine it acts from the extracellular side of the involved in the pathology of peripheral neuropathic pain
plasma membrane (M.B. and X. Zong, unpublished and in pain processing (reviewed in REFS 5,71). A hallmark
observations) and shifts the voltage-dependence of the of neuropathic pain disorders is the generation of ectopic
channel by 10–20 mV to voltages that are more hyper- discharges in primary sensory neurons and/or enhanced
polarized114. As clonidine has several neuronal effects nociceptive synaptic transmission in the spinal dorsal horn
that are mediated by central α2-adrenergic receptors that may be accompanied by changes in the descending
(for example, analgesia and sedation), it cannot be used regulation of the supraspinal CNS. Several studies have
as a bradycardic drug. However, compounds could be reported the upregulation of Ih densities following periph-
designed — based on the clonidine backbone — that eral nerve injury, particularly in large and medium-sized
do not act on α2-adrenergic receptors but preserve the neurons in which most ectopic discharges are produced87,88.
HCN-blocking effect. Increased Ih densities might lead to a shift in the resting
membrane potential of neurons towards more positive
HCN channels as targets for anticonvulsant drugs. values and hence lower the threshold for the generation
As discussed previously, dysregulation of HCN chan- of action potentials. By contrast, a study by Doan et al.89
nel expression and aberrant HCN channel function suggests that the suppression of Ih by increasing neuronal
have been implicated in various types of idiopathic and input resistance may also lead to enhanced peripheral
acquired epilepsies. HCN2-deficient mice have the typical nerve firing. All four HCN channel subtypes have been
clinical hallmarks of absence epilepsy41. Similarly, deletion identified in peripheral sensory neurons, with HCN1 and
of HCN1 in mice is associated with increased seizure HCN2 being the most widely expressed isoforms32,124.
severity and risk of seizure-related death in different There is experimental evidence for increased HCN1 and
limbic seizure induction models115,116. The results from HCN2 expression in peripheral pain conditions35.
genetic mouse models suggest that suppression of Ih is A recent study 125 showed that the anticancer agent
involved in the generation of neuronal hyperexcitability. oxaliplatin induces the upregulation of HCN1 in cold-
In support of this hypothesis, downregulation of HCN1 sensitive primary afferent neurons. This process is of
expression levels has been reported in experimental clinical relevance because it may underlie the generation of
temporal lobe epilepsy, which is the most common and hypersensitivity to cold in patients receiving oxaliplatin-
severe form of epilepsy in adults66,117,118. Surprisingly, based chemotherapy. The relevance of HCN1 in the
however, there is also growing evidence that not only pathology of cold hyperalgesia is supported by the
downregulation but also enhancement of HCN channel- findings of Momin and colleagues126. These authors
mediated currents may contribute to the pathology of demonstrated that HCN1‑knockout mice display sub-
seizures. For example, an HCN2 variant (in which resi- stantially less cold allodynia than their wild-type litter-
dues 719–721 are deleted) was identified in patients suf- mates, but their responsiveness to inflammatory pain
fering from febrile seizure syndromes; when heterologously stimuli is unaffected. There is evidence that HCN2 has
expressed, this variant leads to 35% larger currents than a central role in this particular pain modality 127. Genetic
Absence epilepsy controls119. Finally, in addition to the absolute expression deletion of HCN2 in a subfraction of nociceptive neurons
A form of childhood epilepsy levels, the stoichiometry and subunit composition of het- that are characterized by the expression of the voltage-
that typically results from eromeric channels (for example, the HCN1:HCN2 ratio)120 dependent sodium channel isoform Nav1.8 abolished
abnormal transformation of as well as their targeting to neuronal compartments118 may repetitive firing of these neurons in response to an eleva-
thalamocortical oscillations.
be crucial for inducing aberrant neuronal firing patterns. tion in cAMP. Inflammation did not cause hyperalgesia
Temporal lobe epilepsy The complexity and diversity of the mechanisms con- to heat stimuli in these mice. Moreover, after a nerve
A form of epilepsy in which necting impaired HCN channel activity with epilepsy lesion, HCN2‑deficient mice displayed no neuropathic
seizures typically involve the make it very challenging to develop a generally applica- pain in response to thermal or mechanical stimuli.
hippocampus and adjacent
ble rationale for the design of anticonvulsant drugs based Together, these findings indicate that HCN channels
cortices. This form of epilepsy
is not thought to have a strong on HCN channels. Indeed, there are reports that either are promising targets for the treatment of peripheral pain
genetic component. pharmacological block or activation may be beneficial disorders. Indeed, low concentrations of the Ih blocker
in the context of epilepsy, depending on the clinical cir- ZD7288 reversed both pain behaviour and spontaneous
Febrile seizure syndromes cumstances. For example, the HCN channel inhibitor discharges in injured nerve fibres in rats87,128,129. Moreover,
Seizures that take place in
children at the onset of or
ZD7288 was shown to reduce the generation of hipppo­ intraperitoneal administration of ivabradine reversed
during fever. They are the most campal epileptic discharges in rabbits121. Conversely, two oxaliplatin-mediated cold hypersensitivity in mice125.
common types of seizures in well-established anticonvulsant drugs — lamotrigine HCN1 seems to be a promising HCN channel subtype to
humans, and usually short and (Lamictal; GlaxoSmithKline) and gabapentin (Gabapen; target in the context of peripheral pain disorders because
benign. However, when they
Pfizer) (FIG. 4) — that act by blocking voltage-gated sodium this channel is broadly expressed in sensory neurons and
are long (especially longer than
30 minutes), these seizures and calcium channels, respectively, were shown to upregu- it is transcriptionally upregulated in neuropathic pain.
are associated with a high late the activity of HCN channels122,123. The contribution In addition, a recent study 127 has indicated that HCN2 is
probability of the development of HCN channel upregulation to the global clinical effect an attractive target for the development of analgesics to
of hippocampal epilepsy of these compounds is currently unclear. However, it may combat both inflammatory and neuropathic pain.
(temporal lobe epilepsy) later in
life. There are both genetic and
be speculated that the action of both drugs is directed The ideal analgesic compound should be strongly
environmental contributions to primarily at HCN1, which is the main HCN subtype in selective for HCN1 and/or HCN2 over HCN4 to prevent
febrile seizures. the cortex and hippocampus. effects on cardiac rhythmicity. Moreover, it should not

910 | DECEMBER 2011 | VOLUME 10 www.nature.com/reviews/drugdisc

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

C D 
O
O
O O O

N 
R
O
N
O

'%=µ/?
N
/'.# 
O

O
N N
S O
O 
O
O
+XCDTCFKPG

O 
O /'.# +XCDTCFKPG '%
N   
O
O N %QORQWPF
O
'% *%0 *%0 *%0
Figure 5 | Development of derivatives of ivabradine with increased subtype specificity. a | Chemical structure
of ivabradine and two of its derivatives (MEL57A and EC18). b | EC50 (effector concentration for half-maximum
response) values for ivabradine, MEL57A and EC18 are shown, as determined for hyperpolarization-activated cyclic
nucleotide-gated channel 1 (HCN1), HCN2 and HCN4 expressed in human embryonic kidney 0CVWTG4GXKGYU^&TWI&KUEQXGT[
293 (HEK293) cells.
The ratios of the EC50 values of HCN4 versus HCN1 are given in parentheses for each compound. Ivabradine does not
differentiate between HCN1 and HCN4. By contrast, MEL57A is strongly selective for HCN1 over HCN4, whereas EC18
displays an approximately five times greater affinity for HCN4 than for HCN1 (data from REFS 106, 112).

cross the blood–retina barrier to circumvent effects on evidence that neuronal Ih is also inhibited by clinically
vision. An agent fulfilling these criteria is not yet available. relevant concentrations of general anaesthetics133–135.
However, the study by Melchiorre et al.106 indicates that For halothane and isofluorane, the inhibitory effect
HCN channel subtype-selective agents can be designed on Ih was shown to comprise at least two components:
based on the ivabradine backbone (FIG. 5). Interestingly, namely, a hyperpolarizing shift in the activation curve
the μ‑opioid receptor agonist loperamide (FIG. 4) was and a decrease in the peak current amplitude 135,136.
also shown to block Ih in dorsal root ganglion neurons Studies in heterologous systems136 and HCN1‑knockout
in the low micromolar range36. It was speculated that this mice137 indicate that HCN1 is the major channel con-
block contributes to the analgesic action of loperamide. ferring the anaesthetic action of the drugs mentioned
Thus, loperamide may serve as another lead structure for above. Interestingly, despite being structurally unre-
developing HCN channel blockers that are not structur- lated, two other well-established intravenous anaes-
ally related to the bradines. Similarly, clonidine (FIG. 4), thetics — propofol and ketamine — also inhibit the
which blocks Ih in rat dorsal root ganglion neurons, may activity of HCN1 by shifting the voltage-dependence to
serve as a starting point for the development of novel more hyperpolarized voltages and decreasing the peak
analgesic compounds 130. Finally, the only clinically current 137,138 (FIG. 4). The importance of HCN1 as a tar-
approved HCN channel blocker — ivabradine — might get of anaesthetics is corroborated by the finding that
also be beneficial in the therapeutic management of neu- HCN1‑knockout mice are markedly less sensitive to the
ropathic pain. Although ivabradine is not selective for hypnotic actions of propofol and ketamine137. A report
any of the four HCN channel subtypes, it could be used has also suggested that part of the mechanism of action
in a dose range that keeps heart rate under physiological of local anaesthetics such as lidocaine may be caused by
parameters125. HCN channel inhibition139.

HCN channel blockers in anaesthesia. General anaes- New compounds: challenges and future directions
thetics (for example, halothane and isofluorane; FIG. 4) The successful therapeutic use of ivabradine in patients
exert their pharmacological effects by modulating the with angina pectoris has served as a bona fide proof of
Background potassium activity of ion channels in the CNS. Among the identi- principle that interference with HCN channel function
channels fied targets of these compounds are ionotropic recep- can be beneficial in a clinical setting. However, to explore
Potassium channels that are
constitutively open or possess
tors (for example, the GABAA (γ-aminobutyric acid the full spectrum of possible indications covered by HCN
high basal activity (sometimes type A) and NMDA (N-methyl-d-aspartate) receptors) channels, second-generation compounds will have to
also called ‘leaky’ channels). and several background potassium channels131,132. There is be designed. Three crucial points should be considered

NATURE REVIEWS | DRUG DISCOVERY VOLUME 10 | DECEMBER 2011 | 911

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

in future studies. First, compounds with higher affinity of nonspecific HCN channel blockers. Similarly, com-
have to be developed. The currently available HCN pounds that selectively inhibit HCN1 would have no
channel blockers (ivabradine) and agonists (lamotrigine major effects on cardiac pacemaker function but could
and gabapentin) discussed in this article display consid- be used for the treatment of neuropathic pain or epi-
erably modest IC50 values in the low micromolar range. lepsy. Subtype specificity has not yet been achieved in a
Ideal compounds should operate in the nanomolar range satisfactory manner; however, a recent study 106 indicates
to lower the risk of off-target and toxic side effects. It has that it is in principle feasible to synthesize selective com-
been shown that nicotine blocks native neuronal HCN pounds (FIG. 5). Recently, a series of novel HCN channel
channels at remarkably low concentrations 140 (with blockers has been reported. The new compounds have
an EC50 (effector concentration for half-maximum an indane backbone and display selectivity for HCN1
response) value of around 60 nM). Although nicotine over HCN4 (REF. 146). Finally, it is important to note
itself is clearly not useful as a therapeutic HCN channel that HCN channels also assemble into heterotetramers,
blocker because it activates the nicotinic acetylcholine besides forming homomers. There is growing evidence
receptor as well, analogues of this drug may interact with that an increase in the formation of HCN1–HCN2
HCN channels with greater selectivity. heteromers has an important role in the pathology of
The second issue to consider is the localization of the temporal lobe epilepsies4. It is tempting to speculate
drug binding side within HCN channels. Ivabradine, whether altered hetero­merization of HCN subunits
like most other blockers, targets the intracellular side could also be relevant in other types of epilepsies and
of the HCN channel, which requires drug penetration neuronal diseases. Thus, it will be necessary to search for
through the plasma membrane. Moreover, most blockers compounds that are selective for heteromeric channels
are trapped within the ion-conducting pore, which over homomeric channels.
results in very slow off-kinetics and, in some cases, (for An important drawback in the design of specific
example, with ZD7288) an almost irreversible block. HCN channel blockers is the lack of a high-resolution
It would be desirable to design compounds that bind to structure of the transmembrane core. So far, only parts
HCN channels in a reversible manner from the extracell­ of the C terminus of HCN channels have been crystal-
ular side of the membrane. However, such compounds lized. As most ligands interact with the transmembrane
are not yet available. Clonidine could serve as a template core (including the ion-conducting region) of the chan-
for such a drug because it exerts its action by binding to nel, structural information on this part of the channels
an external side of HCN channels (M.B. and X. Zong, is urgently needed. Currently, several laboratories are
unpublished observations). seeking to crystallize full-length HCN channels. Once
Probably the most important goal in the development a three-dimensional structure is available, molecular
of drugs targeting HCN channels will be the design of modelling approaches could be applied to identify drug
subtype-specific blockers. For example, a specific HCN4 binding sites and optimize drugs targeting HCN channels
blocker would effectively lower heart rate and probably in terms of affinity, selectivity and mode of interaction
lack the side effects associated with vision that are typical (for example, state-dependence of binding).

1. Biel, M., Wahl-Schott, C., Michalakis, S. & Zong, X. 11. Gauss, R., Seifert, R. & Kaupp, U. B. Molecular 18. Santoro, B. et al. Identification of a gene encoding
Hyperpolarization-activated cation channels: from identification of a hyperpolarization-activated a hyperpolarization-activated pacemaker channel
genes to function. Physiol. Rev. 89, 847–885 (2009). channel in sea urchin sperm. Nature 393, 583–587 of brain. Cell 93, 717–729 (1998).
2. Robinson, R. B. & Siegelbaum, S. A. (1998). This was the first study to report the cloning and
Hyperpolarization-activated cation currents: This was the first study to report the cloning of an functional expression of HCN1.
from molecules to physiological function. invertebrate HCN channel. 19. Santoro, B., Grant, S. G., Bartsch, D. & Kandel, E. R.
Annu. Rev. Physiol. 65, 453–480 (2003). 12. Ishii, T. M., Takano, M., Xie, L. H., Noma, A. & Interactive cloning with the SH3 domain of N‑src
3. Baruscotti, M., Bucchi, A. & Difrancesco, D. Physiology Ohmori, H. Molecular characterization of the identifies a new brain specific ion channel protein,
and pharmacology of the cardiac pacemaker (“funny”) hyperpolarization-activated cation channel in with homology to Eag and cyclic nucleotide-gated
current. Pharmacol. Ther. 107, 59–79 (2005). rabbit heart sinoatrial node. J. Biol. Chem. 274, channels. Proc. Natl Acad. Sci. USA 94,
4. Noam, Y., Bernard, C. & Baram, T. Z. Towards an 12835–12839 (1999). 14815–14820 (1997).
integrated view of HCN channel role in epilepsy. 13. Krieger, J., Strobel, J., Vogl, A., Hanke, W. & 20. Shi, W. et al. Distribution and prevalence of
Curr. Opin. Neurobiol. 20 Jun 2011 (doi:10.1016/j. Breer, H. Identification of a cyclic nucleotide- and hyperpolarization-activated cation channel (HCN)
conb.2011.06.013). voltage-activated ion channel from insect antennae. mRNA expression in cardiac tissues. Circ. Res. 85,
5. Lewis, A. S. & Chetkovich, D. M. HCN channels in Insect Biochem. Mol. Biol. 29, 255–267 (1999). e1–e6 (1999).
behavior and neurological disease: too hyper or not 14. Ludwig, A., Zong, X., Jeglitsch, M., Hofmann, F. & 21. Mannikko, R., Elinder, F. & Larsson, H. P.
active enough? Mol. Cell. Neurosci. 46, 357–367 Biel, M. A family of hyperpolarization-activated Voltage-sensing mechanism is conserved among ion
(2011). mammalian cation channels. Nature 393, 587–591 channels gated by opposite voltages. Nature 419,
6. Dunlop, J., Vasilyev, D., Lu, P., Cummons, T. & (1998). 837–841 (2002).
Bowlby, M. R. Hyperpolarization-activated cyclic This was the first study to identify three members 22. Zagotta, W. N. et al. Structural basis for modulation
nucleotide-gated (HCN) channels and pain. of the HCN channel family in a mouse brain. and agonist specificity of HCN pacemaker channels.
Curr. Pharm. Des. 15, 1767–1772 (2009). 15. Ludwig, A. et al. Two pacemaker channels from human Nature 425, 200–205 (2003).
7. Brown, H. F., DiFrancesco, D. & Noble, S. J. heart with profoundly different activation kinetics. This study reported the crystal structure of the
How does adrenaline accelerate the heart? EMBO J. 18, 2323–2329 (1999). CNBD–C‑linker of HCN2.
Nature 280, 235–236 (1979). 16. Marx, T., Gisselmann, G., Stortkuhl, K. F., 23. Xu, X., Vysotskaya, Z. V., Liu, Q. & Zhou, L.
8. Halliwell, J. V. & Adams, P. R. Voltage-clamp analysis Hovemann, B. T. & Hatt, H. Molecular cloning of a Structural basis for the cAMP-dependent gating
of muscarinic excitation in hippocampal neurons. putative voltage- and cyclic nucleotide-gated ion in the human HCN4 channel. J. Biol. Chem. 285,
Brain Res. 250, 71–92 (1982). channel present in the antennae and eyes of 37082–37091 (2010).
9. Pape, H. C. Queer current and pacemaker: Drosophila melanogaster. Invert. Neurosci. 4, 24. DiFrancesco, D. & Tortora, P. Direct activation of
the hyperpolarization-activated cation current in 55–63 (1999). cardiac pacemaker channels by intracellular cyclic
neurons. Annu. Rev. Physiol. 58, 299–327 (1996). 17. Seifert, R. et al. Molecular characterization of a slowly AMP. Nature 351, 145–147 (1991).
10. Yu, F. H., Yarov-Yarovoy, V., Gutman, G. A. & gating human hyperpolarization-activated channel 25. Wainger, B. J., DeGennaro, M., Santoro, B.,
Catterall, W. A. Overview of molecular relationships predominantly expressed in thalamus, heart, and Siegelbaum, S. A. & Tibbs, G. R. Molecular mechanism
in the voltage-gated ion channel superfamily. testis. Proc. Natl Acad. Sci. USA 96, 9391–9396 of cAMP modulation of HCN pacemaker channels.
Pharmacol. Rev. 57, 387–395 (2005). (1999). Nature 411, 805–810 (2001).

912 | DECEMBER 2011 | VOLUME 10 www.nature.com/reviews/drugdisc

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

26. Mistrik, P. et al. The murine HCN3 gene encodes a 48. Piskorowski, R., Santoro, B. & Siegelbaum, S. A. 71. Jiang, Y. Q., Sun, Q., Tu, H. Y. & Wan, Y.
hyperpolarization-activated cation channel with slow TRIP8b splice forms act in concert to regulate the Characteristics of HCN channels and their
kinetics and unique response to cyclic nucleotides. localization and expression of HCN1 channels in CA1 participation in neuropathic pain. Neurochem. Res.
J. Biol. Chem. 280, 27056–27061 (2005). pyramidal neurons. Neuron 70, 495–509 (2011). 33, 1979–1989 (2008).
27. Ulens, C. & Tytgat, J. Functional heteromerization of 49. Lipscombe, D. & Pan, J. Q. Tripping the HCN breaker. 72. Poolos, N. P. The yin and yang of the H‑channel and its
HCN1 and HCN2 pacemaker channels. J. Biol. Chem. Neuron 62, 747–750 (2009). role in epilepsy. Epilepsy Curr. 4, 3–6 (2004).
276, 6069–6072 (2001). 50. Yu, H. et al. MinK-related peptide 1: a β subunit 73. Schulze-Bahr, E. et al. Pacemaker channel dysfunction
28. Moosmang, S., Biel, M., Hofmann, F. & Ludwig, A. for the HCN ion channel subunit family enhances in a patient with sinus node disease. J. Clin. Invest.
Differential distribution of four hyperpolarization- expression and speeds activation. Circ. Res. 88, 111, 1537–1545 (2003).
activated cation channels in mouse brain. Biol. Chem. e84–e87 (2001). 74. Ueda, K. et al. Functional characterization of a
380, 975–980 (1999). 51. Michels, G. et al. K+ channel regulator KCR1 trafficking-defective HCN4 mutation, D553N,
29. Monteggia, L. M., Eisch, A. J., Tang, M. D., suppresses heart rhythm by modulating the associated with cardiac arrhythmia. J. Biol. Chem.
Kaczmarek, L. K. & Nestler, E. J. Cloning and pacemaker current If. PLoS ONE 3, e1511 (2008). 279, 27194–27198 (2004).
localization of the hyperpolarization-activated 52. Gravante, B. et al. Interaction of the pacemaker 75. Milanesi, R., Baruscotti, M., Gnecchi-Ruscone, T.
cyclic nucleotide-gated channel family in rat brain. channel HCN1 with filamin A. J. Biol. Chem. 279, & DiFrancesco, D. Familial sinus bradycardia
Brain Res. Mol. Brain Res. 81, 129–139 (2000). 43847–43853 (2004). associated with a mutation in the cardiac
30. Santoro, B. et al. Molecular and functional 53. Kimura, K., Kitano, J., Nakajima, Y. & Nakanishi, S. pacemaker channel. N. Engl. J. Med. 354, 151–157
heterogeneity of hyperpolarization-activated Hyperpolarization-activated, cyclic nucleotide-gated (2006).
pacemaker channels in the mouse CNS. J. Neurosci. HCN2 cation channel forms a protein assembly with 76. Nof, E. et al. Point mutation in the HCN4 cardiac ion
20, 5264–5275 (2000). multiple neuronal scaffold proteins in distinct modes channel pore affecting synthesis, trafficking, and
31. Notomi, T. & Shigemoto, R. Immunohistochemical of protein–protein interaction. Genes Cells 9, functional expression is associated with familial
localization of Ih channel subunits, HCN1–4, in the 631–640 (2004). asymptomatic sinus bradycardia. Circulation 116,
rat brain. J. Comp. Neurol. 471, 241–276 (2004). 54. Barbuti, A., Terragni, B., Brioschi, C. & DiFrancesco, D. 463–470 (2007).
This is an extensive immunohistochemistry study Localization of f‑channels to caveolae mediates specific 77. Schweizer, P. A. et al. cAMP sensitivity of HCN
examining the expression pattern of all four HCN β2-adrenergic receptor modulation of rate in sinoatrial pacemaker channels determines basal heart rate
channels in the rat brain. myocytes. J. Mol. Cell. Cardiol. 42, 71–78 (2007). but is not critical for autonomic rate control.
32. Moosmang, S. et al. Cellular expression and functional 55. Pian, P., Bucchi, A., Robinson, R. B. & Siegelbaum, S. A. Circ. Arrhythm. Electrophysiol. 3, 542–552 (2010).
characterization of four hyperpolarization-activated Regulation of gating and rundown of HCN hyper­ 78. Laish-Farkash, A. et al. A novel mutation in the HCN4
pacemaker channels in cardiac and neuronal tissues. polarization-activated channels by exogenous and gene causes symptomatic sinus bradycardia in
Eur. J. Biochem. 268, 1646–1652 (2001). endogenous PIP2. J. Gen. Physiol. 128, 593–604 moroccan jews. J. Cardiovasc. Electrophysiol. 21,
33. Fenske, S. et al. HCN3 contributes to the ventricular (2006). 1365–1372 (2010).
action potential waveform in the murine heart. 56. Zolles, G. et al. Pacemaking by HCN channels requires 79. Harzheim, D. et al. Cardiac pacemaker function of
Circ. Res. 109, 1015–1023 (2011). interaction with phosphoinositides. Neuron 52, HCN4 channels in mice is confined to embryonic
34. Muller, F. et al. HCN channels are expressed 1027–1036 (2006). development and requires cyclic AMP. EMBO J. 27,
differentially in retinal bipolar cells and concentrated 57. Fogle, K. J., Lyashchenko, A. K., Turbendian, H. K. & 692–703 (2008).
at synaptic terminals. Eur. J. Neurosci. 17, Tibbs, G. R. HCN pacemaker channel activation 80. Herrmann, S., Stieber, J., Stockl, G., Hofmann, F. &
2084–2096 (2003). is controlled by acidic lipids downstream of Ludwig, A. HCN4 provides a ‘depolarization reserve’
35. Cho, H. J., Staikopoulos, V., Furness, J. B. & diacylglycerol kinase and phospholipase A2. and is not required for heart rate acceleration in mice.
Jennings, E. A. Inflammation-induced increase in J. Neurosci. 27, 2802–2814 (2007). EMBO J. 26, 4423–4432 (2007).
hyperpolarization-activated, cyclic nucleotide-gated 58. Frace, A. M., Maruoka, F. & Noma, A. Control of the 81. Hoesl, E. et al. Tamoxifen-inducible gene deletion in
channel protein in trigeminal ganglion neurons and hyperpolarization-activated cation current by external the cardiac conduction system. J. Mol. Cell. Cardiol.
the effect of buprenorphine. Neuroscience 162, anions in rabbit sino-atrial node cells. J. Physiol. 453, 45, 62–69 (2008).
453–461 (2009). 307–318 (1992). 82. Cerbai, E. & Mugelli, A. If in non-pacemaker cells:
36. Vasilyev, D. V. et al. Direct inhibition of Ih by analgesic 59. Munsch, T. & Pape, H. C. Modulation of the role and pharmacological implications. Pharmacol.
loperamide in rat DRG neurons. J. Neurophysiol. 97, hyperpolarization-activated cation current of rat Res. 53, 416–423 (2006).
3713–3721 (2007). thalamic relay neurones by intracellular pH. J. Physiol. 83. Mangoni, M. E. & Nargeot, J. Genesis and regulation
37. Tellez, J. O. et al. Differential expression of ion 519 (Pt 2), 493–504 (1999). of the heart automaticity. Physiol. Rev. 88, 919–982
channel transcripts in atrial muscle and sinoatrial 60. Zong, X., Stieber, J., Ludwig, A., Hofmann, F. & (2008).
node in rabbit. Circ. Res. 99, 1384–1393 (2006). Biel, M. A single histidine residue determines the 84. Stillitano, F. et al. Molecular basis of funny current (If) in
38. Liu, J., Dobrzynski, H., Yanni, J., Boyett, M. R. & pH sensitivity of the pacemaker channel HCN2. normal and failing human heart. J. Mol. Cell. Cardiol.
Lei, M. Organisation of the mouse sinoatrial node: J. Biol. Chem. 276, 6313–6319 (2001). 45, 289–299 (2008).
structure and expression of HCN channels. 61. Stevens, D. R. et al. Hyperpolarization-activated 85. Campbell, J. N. & Meyer, R. A. Mechanisms of
Cardiovasc. Res. 73, 729–738 (2007). channels HCN1 and HCN4 mediate responses to sour neuropathic pain. Neuron 52, 77–92 (2006).
39. Stieber, J. et al. The hyperpolarization-activated stimuli. Nature 413, 631–635 (2001). 86. Bender, R. A. & Baram, T. Z. Hyperpolarization
channel HCN4 is required for the generation of 62. Wahl-Schott, C., Baumann, L., Zong, X. & Biel, M. activated cyclic-nucleotide gated (HCN) channels in
pacemaker action potentials in the embryonic heart. An arginine residue in the pore region is a key developing neuronal networks. Prog. Neurobiol. 86,
Proc. Natl Acad. Sci. USA 100, 15235–15240 (2003). determinant of chloride dependence in cardiac 129–140 (2008).
40. Baruscotti, M. et al. Deep bradycardia and heart pacemaker channels. J. Biol. Chem. 280, This report shows that developmental HCN
block caused by inducible cardiac-specific knockout of 13694–13700 (2005). channel dysregulation has a role in the generation
the pacemaker channel gene Hcn4. Proc. Natl Acad. 63. Zong, X. et al. A novel mechanism of modulation of of epilepsies.
Sci. USA 108, 1705–1710 (2011). hyperpolarization-activated cyclic nucleotide-gated 87. Chaplan, S. R. et al. Neuronal hyperpolarization-
41. Ludwig, A. et al. Absence epilepsy and sinus channels by Src kinase. J. Biol. Chem. 280, activated pacemaker channels drive neuropathic pain.
dysrhythmia in mice lacking the pacemaker channel 34224–34232 (2005). J. Neurosci. 23, 1169–1178 (2003).
HCN2. EMBO J. 22, 216–224 (2003). 64. Arinsburg, S. S., Cohen, I. S. & Yu, H. G. Constitutively This study links upregulation of HCN channels
This report shows that deletion of HCN2 leads active Src tyrosine kinase changes gating of HCN4 with peripheral neuropathies.
to absence epilepsy. channels through direct binding to the channel proteins. 88. Kitagawa, J. et al. Mechanisms involved in modulation
42. Cho, H. J., Furness, J. B. & Jennings, E. A. J. Cardiovasc. Pharmacol. 47, 578–586 (2006). of trigeminal primary afferent activity in rats with
Post-natal maturation of the hyperpolarisation- 65. Poolos, N. P., Bullis, J. B. & Roth, M. K. Modulation of peripheral mononeuropathy. Eur. J. Neurosci. 24,
activated cation current, Ih, in trigeminal sensory h‑channels in hippocampal pyramidal neurons by p38 1976–1986 (2006).
neurons. J. Neurophysiol. 106, 2045–2056 (2011). mitogen-activated protein kinase. J. Neurosci. 26, 89. Doan, T. N. et al. Differential distribution and function
43. Huang, X., Yang, P., Du, Y., Zhang, J. & Ma, A. 7995–8003 (2006). of hyperpolarization-activated channels in sensory
Age-related down-regulation of HCN channels in rat 66. Jung, S. et al. Downregulation of dendritic HCN neurons and mechanosensitive fibers. J. Neurosci. 24,
sinoatrial node. Basic Res. Cardiol. 102, 429–435 channel gating in epilepsy is mediated by altered 3335–3343 (2004).
(2007). phosphorylation signaling. J. Neurosci. 30, 90. Dyhrfjeld-Johnsen, J., Morgan, R. J. & Soltesz, I.
44. Surges, R. et al. Regulated expression of HCN 6678–6688 (2010). Double trouble? Potential for hyperexcitability
channels and cAMP levels shape the properties 67. Hammelmann, V., Zong, X., Hofmann, F., Michalakis, S. following both channelopathic up- and
of the h current in developing rat hippocampus. & Biel, M. The cGMP-dependent protein kinase II is an downregulation of Ih in epilepsy. Front. Neurosci.
Eur. J. Neurosci. 24, 94–104 (2006). inhibitory modulator of the hyperpolarization-activated 3, 25–33 (2009).
45. Zolles, G. et al. Association with the auxiliary subunit HCN2 channel. PLoS ONE 6, e17078 (2011). 91. Benetos, A., Rudnichi, A., Thomas, F., Safar, M. &
PEX5R/Trip8b controls responsiveness of HCN 68. Baruscotti, M., Bottelli, G., Milanesi, R., Guize, L. Influence of heart rate on mortality in a
channels to cAMP and adrenergic stimulation. DiFrancesco, J. C. & DiFrancesco, D. HCN-related French population: role of age, gender, and blood
Neuron 62, 814–825 (2009). channelopathies. Pflugers Arch. 460, 405–415 pressure. Hypertension 33, 44–52 (1999).
This was a proteomic study on neuronal HCN (2010). 92. Yusuf, S. & Camm, A. J. Sinus tachyarrhythmias and
channel complexes. This paper provides an excellent overview of HCN the specific bradycardic agents: a marriage made in
46. Santoro, B. et al. TRIP8b splice variants form a channelopathies. heaven? J. Cardiovasc. Pharmacol. Ther. 8, 89–105
family of auxiliary subunits that regulate gating and 69. DiFrancesco, D. The role of the funny current in (2003).
trafficking of HCN channels in the brain. Neuron 62, pacemaker activity. Circ. Res. 106, 434–446 (2010). 93. Borer, J. S. Drug insight: If inhibitors as specific
802–813 (2009). 70. Cohen, I. S. & Robinson, R. B. Pacemaker current heart‑rate‑reducing agents. Nature Clin. Pract.
47. Lewis, A. S. et al. Alternatively spliced isoforms of and automatic rhythms: toward a molecular Cardiovasc. Med. 1, 103–109 (2004).
TRIP8b differentially control h channel trafficking and understanding. Handb. Exp. Pharmacol. 171, 41–71 94. Levine, H. J. Rest heart rate and life expectancy.
function. J. Neurosci. 29, 6250–6265 (2009). (2006). J. Am. Coll. Cardiol. 30, 1104–1106 (1997).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 10 | DECEMBER 2011 | 913

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

95. Doesch, A. O. et al. Heart rate reduction after heart 112. Romanelli, M. N. et al. Isoform-selective HCN 131. Rudolph, U. & Antkowiak, B. Molecular and neuronal
transplantation with β-blocker versus the selective If blockers. WO Patent 2011/000915 (2011). substrates for general anaesthetics. Nature Rev.
channel antagonist ivabradine. Transplantation 84, 113. Savelieva, I. & Camm, A. J. If inhibition with Neurosci. 5, 709–720 (2004).
988–996 (2007). ivabradine: electrophysiological effects and safety. 132. Linden, A. M. et al. TASK‑3 knockout mice exhibit
96. Bois, P., Guinamard, R., Chemaly, A. E., Faivre, J. F. & Drug Saf. 31, 95–107 (2008). exaggerated nocturnal activity, impairments in
Bescond, J. Molecular regulation and pharmacology 114. Knaus, A. et al. Direct inhibition of cardiac cognitive functions, and reduced sensitivity to
of pacemaker channels. Curr. Pharm. Des. 13, hyperpolarization-activated cyclic nucleotide-gated inhalation anesthetics. J. Pharmacol. Exp. Ther. 323,
2338–2349 (2007). pacemaker channels by clonidine. Circulation 115, 924–934 (2007).
97. Bucchi, A., Baruscotti, M. & DiFrancesco, D. 872–880 (2007). 133. Sirois, J. E., Pancrazio, J. J., Iii, C. L. & Bayliss, D. A.
Current-dependent block of rabbit sino-atrial node If This is a report on the effects of clonidine on Multiple ionic mechanisms mediate inhibition of rat
channels by ivabradine. J. Gen. Physiol. 120, 1–13 HCN channels. motoneurones by inhalation anaesthetics. J. Physiol.
(2002). 115. Huang, Z., Walker, M. C. & Shah, M. M. Loss of 512 (Pt 3), 851–862 (1998).
This study examines the molecular mechanism dendritic HCN1 subunits enhances cortical excitability 134. Sirois, J. E., Lynch, C. & Bayliss, D. A. Convergent and
of action of ivabradine. and epileptogenesis. J. Neurosci. 29, 10979–10988 reciprocal modulation of a leak K+ current and Ih by an
98. Bucchi, A., Tognati, A., Milanesi, R., Baruscotti, M. (2009). inhalational anaesthetic and neurotransmitters in rat
& DiFrancesco, D. Properties of ivabradine-induced 116. Santoro, B. et al. Increased seizure severity and brainstem motoneurones. J. Physiol. 541, 717–729
block of HCN1 and HCN4 pacemaker channels. seizure-related death in mice lacking HCN1 channels. (2002).
J. Physiol. 572, 335–346 (2006). Epilepsia 51, 1624–1627 (2010). 135. Budde, T. et al. Reciprocal modulation of Ih and ITASK
99. Riccioni, G., Vitulano, N. & D’Orazio, N. Ivabradine: 117. Shah, M. M., Anderson, A. E., Leung, V., Lin, X. & in thalamocortical relay neurons by halothane.
beyond heart rate control. Adv. Ther. 26, 12–24 Johnston, D. Seizure-induced plasticity of h channels Pflugers Arch. 456, 1061–1073 (2008).
(2009). in entorhinal cortical layer III pyramidal neurons. 136. Chen, X. et al. HCN subunit-specific and cAMP-
100. Cerbai, E. et al. Characterization of the Neuron 44, 495–508 (2004). modulated effects of anesthetics on neuronal
hyperpolarization-activated current, If, in ventricular 118. Shin, M., Brager, D., Jaramillo, T. C., Johnston, D. pacemaker currents. J. Neurosci. 25, 5803–5814
myocytes from human failing heart. Circulation 95, & Chetkovich, D. M. Mislocalization of h channel (2005).
568–571 (1997). subunits underlies h channelopathy in temporal lobe 137. Chen, X., Shu, S., Kennedy, D. P., Willcox, S. C. &
101. Hoppe, U. C. et al. Effect of cardiac resynchronization epilepsy. Neurobiol. Dis. 32, 26–36 (2008). Bayliss, D. A. Subunit-specific effects of isoflurane on
on the incidence of atrial fibrillation in patients with 119. Dibbens, L. M. et al. Augmented currents of an HCN2 neuronal Ih in HCN1 knockout mice. J. Neurophysiol.
severe heart failure. Circulation 114, 18–25 (2006). variant in patients with febrile seizure syndromes. 101, 129–140 (2009).
102. Manz, M., Reuter, M., Lauck, G., Omran, H. & Jung, W. Ann. Neurol. 67, 542–546 (2010). This study shows that general anaesthetics target
A single intravenous dose of ivabradine, a novel If 120. Richichi, C. et al. Mechanisms of seizure-induced HCN channels.
inhibitor, lowers heart rate but does not depress left ‘transcriptional channelopathy’ of hyperpolarization- 138. Cacheaux, L. P. et al. Impairment of hyperpolarization-
ventricular function in patients with left ventricular activated cyclic nucleotide gated (HCN) channels. activated, cyclic nucleotide-gated channel function by
dysfunction. Cardiology 100, 149–155 (2003). Neurobiol. Dis. 29, 297–305 (2008). the intravenous general anesthetic propofol.
103. Fox, K., Ford, I., Steg, P. G., Tendera, M. & Ferrari, R. 121. Kitayama, M. et al. Ih blockers have a potential of J. Pharmacol. Exp. Ther. 315, 517–525 (2005).
Ivabradine for patients with stable coronary artery antiepileptic effects. Epilepsia 44, 20–24 (2003). 139. Meng, Q. T., Xia, Z. Y., Liu, J., Bayliss, D. A. & Chen, X.
disease and left-ventricular systolic dysfunction 122. Poolos, N. P., Migliore, M. & Johnston, D. Local anesthetic inhibits hyperpolarization-activated
(BEAUTIFUL): a randomised, double-blind, placebo- Pharmacological upregulation of h‑channels reduces cationic currents. Mol. Pharmacol. 79, 866–873
controlled trial. Lancet 372, 807–816 (2008). the excitability of pyramidal neuron dendrites. Nature (2011).
This study reported the results of a large clinical Neurosci. 5, 767–774 (2002). 140. Griguoli, M. et al. Nicotine blocks the hyper­
trial (BEAUTIFUL) of ivabradine. 123. Surges, R., Freiman, T. M. & Feuerstein, T. J. polarization-activated current Ih and severely impairs
104. Fox, K. et al. Heart rate as a prognostic risk factor Gabapentin increases the hyperpolarization-activated the oscillatory behavior of oriens-lacunosum moleculare
in patients with coronary artery disease and cation current Ih in rat CA1 pyramidal cells. Epilepsia interneurons. J. Neurosci. 30, 10773–10783 (2010).
left-ventricular systolic dysfunction (BEAUTIFUL): 44, 150–156 (2003). 141. Knop, G. C. et al. Light responses in the mouse retina
a subgroup analysis of a randomised controlled trial. 124. Luo, L. et al. Role of peripheral hyperpolarization- are prolonged upon targeted deletion of the HCN1
Lancet 372, 817–821 (2008). activated cyclic nucleotide-modulated channel channel gene. Eur. J. Neurosci. 28, 2221–2230
105. Swedberg, K. et al. Ivabradine and outcomes in pacemaker channels in acute and chronic pain models (2008).
chronic heart failure (SHIFT): a randomised placebo- in the rat. Neuroscience 144, 1477–1485 (2007). 142. Moroni, A. et al. Hyperpolarization-activated cyclic
controlled study. Lancet 376, 875–885 (2010). 125. Descoeur, J. et al. Oxaliplatin-induced cold nucleotide-gated channel 1 is a molecular determinant
This study reported the results of a clinical trial hypersensitivity is due to remodelling of ion channel of the cardiac pacemaker current If. J. Biol. Chem.
(SHIFT) of ivabradine. expression in nociceptors. EMBO Mol. Med. 3, 276, 29233–29241 (2001).
106. Melchiorre, M. et al. Design, synthesis, and 266–278 (2011). 143. Marionneau, C. et al. Specific pattern of ionic channel
preliminary biological evaluation of new isoform- This study demonstrates that HCN channels are gene expression associated with pacemaker activity in
selective f‑current blockers. J. Med. Chem. 53, potential targets for the treatment of oxaliplatin- the mouse heart. J. Physiol. 562, 223–234 (2005).
6773–6777 (2010). induced cold hypersensitivity. 144. Hurtado, R., Bub, G. & Herzlinger, D. The pelvis-
This was the first report on the attempts to design 126. Momin, A., Cadiou, H., Mason, A. & McNaughton, P. A. kidney junction contains HCN3, a hyperpolarization-
subtype-specific HCN channel blockers. Role of the hyperpolarization-activated current Ih activated cation channel that triggers ureter
107. Bois, P., Bescond, J., Renaudon, B. & Lenfant, J. in somatosensory neurons. J. Physiol. 586, peristalsis. Kidney Int. 77, 500–508 (2010).
Mode of action of bradycardic agent, S 16257, 5911–5929 (2008). 145. Dobrzynski, H. et al. Site of origin and molecular
on ionic currents of rabbit sinoatrial node cells. 127. Emery, E. C., Young, G. T., Berrocoso, E. M., Chen, L. & substrate of atrioventricular junctional rhythm in the
Br. J. Pharmacol. 118, 1051–1057 (1996). McNaughton, P. A. HCN2 ion channels play a central rabbit heart. Circ. Res. 93, 1102–1110 (2003).
108. DiFrancesco, D. & Camm, J. A. Heart rate lowering role in inflammatory and neuropathic pain. Science 146. McClure, K. J. et al. Discovery of a novel series of
by specific and selective If current inhibition with 333, 1462–1466 (2011). selective HCN1 blockers. Bioorg. Med. Chem. Lett.
ivabradine: a new therapeutic perspective in 128. Lee, D. H., Chang, L., Sorkin, L. S. & Chaplan, S. R. 21, 5197–5201 (2011).
cardiovascular disease. Drugs 64, 1757–1765 Hyperpolarization-activated, cation-nonselective,
(2004). cyclic nucleotide-modulated channel blockade Acknowledgements
109. Stieber, J. Ivabradine: pharmacodynamic aspects of alleviates mechanical allodynia and suppresses This work was supported by the German Research Foundation
its clinical use. Methods Find. Exp. Clin. Pharmacol. ectopic discharge in spinal nerve ligated rats. J. Pain (the Deutsche Forschungsgemeinschaft).
30, 633–641 (2008). 6, 417–424 (2005).
Competing interests statement
110. Barrow, A. J. & Wu, S. M. Low-conductance HCN1 ion 129. Sun, Q., Xing, G. G., Tu, H. Y., Han, J. S. & Wan, Y.
The authors declare no competing financial interests.
channels augment the frequency response of rod and Inhibition of hyperpolarization-activated current by
cone photoreceptors. J. Neurosci. 29, 5841–5853 ZD7288 suppresses ectopic discharges of injured
(2009). dorsal root ganglion neurons in a rat model of FURTHER INFORMATION
111. Cervetto, L., Demontis, G. C. & Gargini, C. Cellular neuropathic pain. Brain Res. 1032, 63–69 (2005). Martin Biel’s homepage:
mechanisms underlying the pharmacological induction 130. Yagi, J. & Sumino, R. Inhibition of a hyperpolarization- http://cup.uni-muenchen.de/ph/aks/biel
of phosphenes. Br. J. Pharmacol. 150, 383–390 activated current by clonidine in rat dorsal root ganglion ALL LINKS ARE ACTIVE IN THE ONLINE PDF
(2007). neurons. J. Neurophysiol. 80, 1094–1104 (1998).

914 | DECEMBER 2011 | VOLUME 10 www.nature.com/reviews/drugdisc

© 2011 Macmillan Publishers Limited. All rights reserved

You might also like