You are on page 1of 9

HCN channels : Function and clinical implications

Eduardo E. Benarroch
Neurology 2013;80;304
DOI 10.1212/WNL.0b013e31827dec42

This information is current as of January 16, 2013

The online version of this article, along with updated information and services, is
located on the World Wide Web at:
http://www.neurology.org/content/80/3/304.full.html

Neurology ® is the official journal of the American Academy of Neurology. Published continuously
since 1951, it is now a weekly with 48 issues per year. Copyright © 2013 by AAN Enterprises, Inc. All
rights reserved. Print ISSN: 0028-3878. Online ISSN: 1526-632X.
CLINICAL
IMPLICATIONS OF
NEUROSCIENCE
HCN channels
RESEARCH
Function and clinical implications
Section Editor
Eduardo E.
Benarroch, MD

Eduardo E. Benarroch, The hyperpolarization-activated cyclic nucleotide-gated contains the cyclic nucleotide-binding domain (CNBD),
MD (HCN) channels belong to the superfamily of pore-loop which mediates modulation by cyclic nucleotides. A
cation channels. In mammals, the HCN channel family highly conserved asparagine residue in the extracellular
comprises 4 members (HCN1–4) that are expressed in loop between S5 and the pore loop is glycosylated; this
Correspondence to
Dr. Benarroch: heart and nervous system. HCN channels are activated posttranslational channel modification is crucial for nor-
benarroch.eduardo@mayo.edu by membrane hyperpolarization, are permeable to Na1 mal cell surface expression.
and K1, and are constitutively open at voltages near the
Heterogeneity and distribution of HCN channels. There are
resting membrane potential. In many cases, activation is
4 homologous HCN channel subunits (HCN1–4) in
facilitated by direct interaction with cyclic nucleotides,
mammals. These subunits form different homotetramers
particularly cyclic adenosine monophosphate (cAMP).
or heterotetramers with distinct biophysical properties
The cation current through HCN channels is known as
(table 1).1,2 HCN1 is expressed in the neocortex, hippo-
Ih; opening of HCN channels elicits membrane depo-
campus, brainstem, spinal cord, and dorsal root ganglion
larization toward threshold for action potential genera-
(DRG); HCN2 is distributed nearly ubiquitously, with
tion, and reduces membrane resistance and thus the
the highest expression in the thalamus, brainstem nuclei,
magnitude of excitatory and inhibitory postsynaptic po-
and small nociceptive DRG neurons; HCN3 is expressed
tentials. HCN channels have a major role in controlling
at very low levels in the CNS, except the olfactory bulb
neuronal excitability, dendritic integration of synaptic
and hypothalamus; and HCN4 is strongly expressed in
potentials, synaptic transmission, and rhythmic oscilla-
thalamic nuclei and the olfactory bulb.11 In neurons,
tory activity in individual neurons and neuronal net-
HCN channels are expressed in dendrites and axon
works. These channels participate in mechanisms of
terminals, in some cases with a heterogeneous distribu-
synaptic plasticity and memory, thalamocortical rhythms,
tion. For example, in neocortical and CA1 hippocampal
and somatic sensation. Experimental evidence indicates
pyramidal and thalamic reticular neurons, there is a gra-
that HCN channels may also contribute to mechanisms
dient of HCN1 expression, with higher expression in the
of epilepsy and pain. The physiologic functions of HCN
distal dendrites than in the soma12,13; in layer 5 neocor-
channels and their implications for neurologic disorders
tical pyramidal neurons, HCN1 is predominantly ex-
have been recently reviewed.1–10
pressed in dendrites, whereas HCN2 is expressed in the
soma. All 4 HCN channel isoforms, particularly HNC4
HCN CHANNELS AND IH CURRENT Structure of HCN
and to a lesser extent HCN2, are expressed in the heart.
channels. The HCN channels form the subgroup of
cyclic nucleotide-gated cation channels within the large Activation of HCN channels and Ih current. The HCN
superfamily of the pore-loop cation channels. Like other channels are permeable to Na1 and K1 and conduct a
pore-loop channels, HCN channels are complexes con- mixed cation current, Ih. The ratio of the Na1 to K1
sisting of 4 subunits that are arranged around the central permeability of the channel ranges from 1:3 to 1:5, yield-
pore. Each HCN channel subunit consists of 3 principal ing values for the reversal potential between 225 and
structural modules: the transmembrane core and the 240 mV. As a consequence, activation of Ih at resting
cytosolic amino (N)-terminal and carboxy (C)-terminal membrane potentials depolarizes the membrane toward
domains (figure).1 The transmembrane core is com- threshold for firing of action potentials.1,7 An increase in
posed of 6 transmembrane segments (S1–S6) including extracellular K1 concentration strongly increases current
a positively charged voltage sensor (S4) and the ion- amplitude.1 Membrane hyperpolarization is necessary
conducting pore region between S5 and S6. The prox- and sufficient to activate the HCN channels; cyclic
imal portion of the cytosolic C-terminal domain nucleotides such as cAMP act as costimulatory modu-
GLOSSARY
cAMP 5 cyclic adenosine monophosphate; CNBD 5 cyclic nucleotide-binding domain; DRG 5 dorsal root ganglion; EPSP 5
excitatory postsynaptic potentials; HCN 5 hyperpolarization-activated cyclic nucleotide-gated; LTP 5 long-term potentia-
tion; PGE2 5 prostaglandin E2; PIP2 5 phosphatidylinositol 4,5-bisphosphate; SA 5 sinoatrial.

From the Department of Neurology, Mayo Clinic, Rochester, MN.


Go to Neurology.org for full disclosures. Funding information and disclosures deemed relevant by the authors, if any, are provided at the end of the article.

304 © 2013 American Academy of Neurology

ª 2013 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.


Figure Structure and functions of hyperpolarization-activated cyclic nucleotide-gated channels

The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels belong to the superfamily of pore-loop cation channels.
HCN channels are complexes consisting of 4 subunits that are arranged around the central pore. Each subunit consists of a trans-
membrane core and the cytosolic amino (N)-terminal and carboxy (C)-terminal domains. The transmembrane core is composed of 6
transmembrane segments (S1–S6) including a positively charged voltage sensor (S4) and the ion-conducting pore region between
S5 and S6. The proximal portion of the cytosolic C-terminal domain contains the cyclic nucleotide-binding domain (CNBD), which
mediates modulation by cyclic nucleotides, such as cyclic adenosine monophosphate (cAMP). The HCN channels are permeable to
Na1 and K1 and conduct a mixed cation current, Ih. Membrane hyperpolarization is necessary and sufficient to activate the HCN
channels; cAMP act as costimulatory modulators that facilitate voltage-dependent HCN channel activation.

lators that facilitate voltage-dependent HCN channel of cAMP or PIP2. For example, in cardiac sinoatrial (SA)
activation by shifting the voltage dependence of activa- cells, norepinephrine, acting via b1 adrenoceptor,
tion to more positive voltages. Binding of cAMP to the increases cAMP production and Ih, leading to increased
CNBD induces a conformational change that removes a rate of spontaneous depolarization (and thus heart rate).
tonic inhibition conferred by this domain on channel In contrast, in dendritic spines prefrontal pyramidal neu-
opening. Within the HCN channel family, HCN1 is rons, norepinephrine acting via a2A receptors inhibits
the subtype with the fastest kinetics and weakest mod- cAMP production and HCN activity.15
ulation by cAMP; both HCN2 and HCN4 are strongly HCN channels are macromolecular protein com-
modulated by cAMP.1 plexes that contain auxiliary proteins required for the
fine-tuning of their electrophysiologic properties, func-
Regulation of HCN channels. HCN channels are tightly tional coupling to signaling pathways, and trafficking
regulated by interacting proteins and ions in the cytosol to specific cellular compartments.1 These proteins
and the extracellular space.1 HCN channel activity and include the MinK-related protein MiRP1 and several
regulation occurs at multiple levels, dependent on the scaffold proteins such as TPR-containing Rab8b inter-
number and subtype of HCN subunits expressed on the acting protein (TRIP8b), which colocalizes with HCN1
plasma membrane, distribution of HCN channel sub- in dendrites of cortical and hippocampal pyramidal cells.1
units in distinct subcellular compartments, and biophys-
ical properties of the channel (such as voltage-activation
PHYSIOLOGIC ROLE OF HCN CHANNELS AND IH
profile and kinetics).1 Intracellular HCN channel mod-
CURRENT The unique properties of HCN channels
ulators include phosphatidylinositol 4,5-bisphosphate
and Ih make them versatile regulators of neuronal excit-
(PIP2), which acts as an allosteric activator,14 and intra-
ability and activity of neuronal networks1,6,7 (table 2).
cellular protons, which slow down the speed of channel
opening. The protective action of carbonic anhydrase Setting of the membrane potential. HCN channels are par-
inhibitors in generalized seizures has been attributed to tially open at rest at subthreshold potentials; Ih depolar-
the high sensitivity of HCN channels to intracellular izes the cell and maintains the membrane potential near
pH. The HCN channels are also the target of phospho- the resting membrane potential. Because Ih is partially
rylation by different protein kinases. Many neurotrans- activated at physiologic, “resting” conditions, it serves as
mitters affect HCN channel function by affecting levels a feedback mechanism that counteracts both membrane

Neurology 80 January 15, 2013 305

ª 2013 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.


Table 1 HCN channel subtypes

HCN1 HCN2 HCN3 HCN4

Inactivation kinetics Fast Intermediate Slow Slow

cAMP modulation Weakest Strong Weak Strongest

CNS expression Neocortex Ubiquitous Olfactory bulb Thalamus

Hippocampus Hypothalamus Basal ganglia

Brainstem Retinal cones Olfactory bulb

DRG expression Large neurons Nav1.8 expressing — —


nociceptive neurons

Cold-sensitive neurons

Cardiac expression SA node Ubiquitous Cardiac muscle SA bode

AV node AV node

Purkinje system

Knockout mouse Enhanced hippocampal-dependent Ataxia Increased T-wave Embryonic lethal phenotype
phenotype learning and memory amplitude (bradycardia and absent
pacemaker cells)

Impaired motor learning Absence epilepsy Sinus pauses, bradycardia,


and heart block

Epilepsy Reduced inflammatory


and neuropathic pain

Altered color vision Sinus arrhythmia

Reduced cold allodynia

Clinical correlations Increased level and dendric HCN2 mutation causing Ih HCN4 mutation:
expression in granule cells of augmentation associated bradycardia, SA
the dentate gyrus in resected with GEFS1 and AV conduction
hippocampi in TLE impairment, QT
prolongation, syncope

Oxaliplatin-induced cold allodynia?

Abbreviations: AV 5 atrioventricular; cAMP 5 cyclic adenosine monophosphate; DRG 5 dorsal root ganglion; GEFS 5 generalized epilepsy with febrile
seizures plus; HCN 5 hyperpolarization-activated cyclic nucleotide-gated; SA 5 sinoatrial; TLE 5 temporal lobe epilepsy.

hyperpolarization and depolarization. Thus, HCN chan- Dendritic integration. Dendritic integration is crucial for
nels may have excitatory, inhibitory, or modulatory func- signal processing in most neurons. Since individual EPSP
tions.7 HCN channels interact with other ion channels are too small to reach the action potential threshold,
that influence the membrane potential. For example, the generation of action potentials at the soma requires the
delayed-rectifier M-type K1 channels (Kv7 or KCNQ integration of multiple excitatory synaptic inputs. The
channel) have a large effect on HCN function, as they presence of constitutively open Ih channels in dendrites
determine whether HCN channels act in an excitatory or influences the passive propagation of EPSP by lowering
inhibitory fashion.16 The tonic depolarizing effect of Ih membrane resistance and reducing their amplitude.
increases the steady-state inactivation of low-threshold HCN1 channels participate in dendritic integration of
voltage-gated calcium (Ca21) channels (T-channels) EPSP in neocortical and CA1 pyramidal neurons.17 In
and thus restrict the genesis of dendritic Ca21 spikes.17 these neurons, the density of Ih is higher in distal than in
the proximal dendrites, and therefore temporal summa-
Dampening of synaptic potentials. According to Ohm’s tion is more dampened for distal than for proximal
law, the magnitude of voltage change of the membrane inputs propagating to the soma; this somatoden-
is proportional to its input resistance. As HCN opening dritic gradient of HCN expression allows for the effects
reduces the input resistance of the membrane, Ih damp- of excitatory synaptic inputs to becomes independent of
ens the amplitude and duration of both excitatory post- their location.12 Activated HCN channels also participate
synaptic potentials (EPSP) and inhibitory postsynaptic in the detection of coincident synaptic inputs, suppress-
potentials.6,17 In some neurons, the depolarizing effects ing the summation of EPSP that occur at low frequency
of Ih may counterbalance its dampening effect on EPSP and improving the selectivity for synchronous synaptic
triggered by glutamate receptor activation. In many cells, inputs.18
such as those in the subthalamic nucleus, the main effect
of Ih is to antagonize the hyperpolarizing inhibitory in- Neuronal and network oscillations. Sustained rhythmic
puts mediated by GABAA receptors.17 oscillations are a hallmark feature of neuronal circuits

306 Neurology 80 January 15, 2013

ª 2013 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.


Table 2 Physiologic effects of HCN channel and Ih

Function Ih property Physiologic implications

Maintenance of Constitutively open at voltages near RMP Voltage-dependent Ih activation and deactivation act as negative feedback that
RMP opposes Vm deviations from RMP

Elicit membrane depolarization Reduces amplitude of EPSP or IPSP

Lower membrane resistance

Dendritic Somatodendritic gradient of Ih expression in hippocampal and Affects temporal summation of EPSP
integration neocortical neurons

Increase spatial Interactions of HCN1 with a2A adrenoreceptors in dendritic a2A Receptor-mediated inhibition of cAMP reduces Ih and increases efficacy of
working memory spines of prefrontal neurons excitatory inputs to prefrontal neurons

Impaired High HCN1 expression in distal dendrites of CA1 neuron Ih in distal dendrites inhibits Ca21 spikes triggered by activation of glutamate
hippocampal LTP receiving inputs from the entorhinal cortex receptors, thus reducing LTP at the perforant pathway–CA1 synapse

Motor learning HCN1 mediates a depolarizing current in spontaneously By counteracting the hyperpolarizing effects of inhibitory inputs, Ih enables
spiking Purkinje cells Purkinje cells to maintain a high-frequency input–output relationship

Neuronal HCN1 expression in hippocampus Promotes hippocampal u oscillations (4–6 Hz)


oscillations

HCN2 expression in TC neurons Ih promotes burst-firing mode in TC neurons through interactions with T currents

HCN2 deficiency leads to spike-and-wave discharge by increasing number of


closed T channels available to activation by depolarizing stimuli

Thalamocortical GABAergic inputs from the thalamic reticular nucleus elicit Via interactions with T currents, Ih promotes rhythmic burst firing in TC neurons
network oscillations rhythmic IPSP that activate Ih in TC neurons and spindle oscillation during non-REM sleep

Persistent activation of Ih in TC neurons responsible for the silent period


between spindle waves

Abbreviations: EPSP 5 excitatory postsynaptic potentials; HCN 5 hyperpolarization-activated cyclic nucleotide-gated; IPSP 5 inhibitory postsynaptic
potentials; LTP 5 long-term potentiation; RMP 5 resting membrane potential; TC 5 thalamocortical; Vm 5 membrane potential.

in various brain regions including the thalamus, hippo- of Ih and a low-threshold Ca21 current (T-current). The
campus, and neocortex. HCN channels have an impor- activation of Ih by hyperpolarization beyond 265 mV
tant role in oscillatory properties of individual neurons slowly depolarizes the membrane potential until a
and networks. Typical examples are u oscillations in rebound low-threshold Ca21 spike is generated by
hippocampal circuits19 and slow and spindle oscillation T-channel activation, leading to the discharge of a burst
in thalamocortical circuits.20 There are also several of action potentials. Depolarization inactivates both
studies that demonstrated the importance of Ih for g HCN and T channels, leading to termination of the
oscillations in the hippocampus, synchronized oscilla- spike. This is followed by a hyperpolarizing “overshoot”
tions in the inferior olive, and subthreshold oscillations that activates Ih, leading to repetition of the cycle and
in the entorhinal cortex.1 continuous rhythmic burst firing. These single-cell oscil-
lations are synchronized in large neuronal circuits and
PHYSIOLOGIC IMPLICATIONS HCN channels may can be recorded in the EEG as d waves during non-
have an important role in mechanisms of thalamocorti- REM sleep. Electrophysiologic recordings in thalamic
cal oscillations, hippocampal plasticity, spatial working slice preparations of HCN2-deficient mice revealed that
memory, motor learning, and nociception (table 2). the current produced by HCN2 constitutes the major
component underlying thalamocortical Ih.21 Deletion of
Generation of thalamic rhythms. Ih is involved in slow (d) HCN2 results in hyperpolarizing shift of the resting
frequency single-cell oscillations in thalamic relay (thal- membrane potential in thalamocortical neurons, result-
amocortical) neurons; synchronized thalamocortical net- ing in higher susceptibility of these neurons to fire in the
work oscillations at d and spindle frequency during burst mode in response to excitatory inputs as compared
non-REM sleep; and generalized spike-and-wave dis- with wild-type neurons. This altered thalamic firing
charges associated with absence seizures.1,6 Thalamocor- behavior of HCN2-deficient mice manifests with
tical neurons fire in 2 distinct firing modes: transmission spike-and-wave discharges, the hallmark of absence
mode and burst mode. During wakefulness and REM epilepsy.20
sleep, thalamocortical neurons are depolarized by affer- Ih is also involved in the generation of the synchro-
ent inputs and fire in a transmission of “single spike” nized thalamocortical network oscillations that underlie
mode; burst firing mode occurs when thalamocortical spindle waves on surface EEG during N2 stage of non-
neurons are hyperpolarized and is characteristically seen REM sleep. In this state, burst firing in thalamic retic-
during non-REM sleep or absence seizures. Thalamic ular neurons induces rhythmic inhibitory postsynaptic
neurons fire in the “burst mode” through the interaction potentials in thalamocortical cells; this hyperpolarization

Neurology 80 January 15, 2013 307

ª 2013 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.


activates Ih and removes inactivation of the low-threshold mediators such as prostaglandin E2 (PGE2), which
Ca21 currents, resulting in burst firing in thalamocor- increase cAMP production.26
tical neurons, which re-excites the thalamic reticular
Cardiac rhythm. Although not directly related to neural
neurons and stimulates cortical pyramidal cells. Net-
work synchronization between the cortex and the function, control of cardiac rhythm is critical to maintain
thalamus leads to nearly simultaneous occurrence of cardiac output and cerebral perfusion and is controlled
spindle waves over widespread areas of the cortex.22 by autonomic influences on HCN channels expressed
by SA node cells and the cardiac conduction system.
Learning and memory mechanisms. Mice with selective
Ih was initially described as “pacemaker current,” as it
HCN1 gene deletions have better hippocampal- has a prominent role in the progressive diastolic depo-
dependent spatial learning and memory than do wild- larization of SA cells that drives their membrane toward
type mice; this reflects enhanced long-term poten- threshold of Ca21 channel activation following repo-
tiation (LTP) at the synapse between the entorhinal larization.1 HCN4 makes up ;80% and HCN2
cortex and CA1 and increased excitability of CA1 den- ;20% of SA node Ih. Sympathetic stimulation acceler-
drites.23 This suggests that HCN1 channels inhibit den- ates heart rate via b-adrenoceptor-triggered cAMP pro-
dritic integration and synaptic long-term plasticity at the duction and Ih activation, whereas low level vagal
perforant path inputs to the distal dendrites of CA1 stimulation lowers heart rate via inhibition of cAMP
pyramidal neurons; the high HCN1 expression at this synthesis. Adult HCN4 knockout mice have cardiac
level may interfere with generation of Ca21 spikes, a arrhythmia characterized by recurrent sinus pauses.
process that is critical for the induction of LTP at den-
dritic synapses.1 CLINICAL CORRELATIONS Only 4 inherited HCN
HCN channels are also involved in mechanisms of channel mutations have been described; all are localized
spatial working memory in the prefrontal cortex. In den- in the HCN4 gene and are associated with sinus brady-
dritic spines of prefrontal neurons, HCN channels coloc- cardia. So far, disease-causing mutations in human
alize and functionally interact with a2A adrenoceptors, HCN1–3 genes have not been reported. Acquired
which are activated by relatively low levels of norepi- HCN1 and HCN2 channels dysfunction has been
nephrine released during alert and wakefulness.15 Acti- linked with epilepsy, necropathic pain, and subarach-
vation of a2A adrenoceptors inhibits the production of noid hemorrhage.
cAMP and thus reduces the open probability of HCN
channels, leading to increased membrane resistance; this Epilepsy. Given the complex effects of Ih in fine-tuning
would increase the efficiency of excitatory synaptic cellular excitability, synaptic responses, and network
inputs to the dendrites of prefrontal neurons engaged activity, it is not surprising that abnormal regulation of
in a specific task.1 HCN expression or function has been implicated in
HCN1 channels also participate in cerebellar mecha- epilepsy.6 However, the role of HCN dysregulation in
nisms of motor learning. HCN1 deletion causes pro- epilepsy is complex. Studies in experimental models
found deficits in tests that require complex and indicate that both upregulation and downregulation of
repeated coordination of motor output by the cere- HCN channels can be associated with seizures; different
bellum.24 The HCN1 channels are expressed in Pur- HCN types may contribute to different extents to focal
kinje cells; in these spontaneously spiking neurons, or generalized epilepsy syndromes; and the role of HCN
the depolarizing effects of Ih counteract the effects of channels strongly depends on cellular localization and
inhibitory (hyperpolarizing) inputs. This mechanism physiologic context.5,6,27 Furthermore, the results on
would allow Purkinje to maintain a constant input- experimental epilepsy models may not necessarily be
output relationship independently of the effects of applicable to human epilepsy syndromes.
previous inputs to the cerebellum. HCN channel abnormalities in the epileptic brain
can manifest as altered mRNA and protein expression,
Somatic sensation. Many DRG neurons express HCN subcellular distribution, biophysical properties, and spa-
channels. Large, non-nociceptive DRG neurons with tial selectivity and temporal pattern of dysregulation fol-
large myelinated (Ab) axons and a small population lowing seizures.6 Altered HCN1 or HCN2 expression
of cold-sensitive DRG neurons express HCN1, which (i.e., transcriptional channelopathy) has been demon-
is relatively insensitive to cAMP. In contrast, nocicep- strated in hippocampus of patients with severe temporal
tive DRG neurons with small myelinated (Ad) or lobe epilepsy as well as in animal models of temporal
unmyelinated (C) axons express HCN2, which is lobe and absence epilepsies.6,28–30 These changes may not
responsible for a slower and cAMP-sensitive in Ih cur- necessarily be the cause but rather represent compensa-
rent.25 In nociceptive neurons, HCN2 expression has tory responses to abnormal excitability during seizures.
an important role in modulating the generation of In organotypic hippocampal slice cultures, seizure-
action potentials in response to inflammatory like events selectively reduced HCN1 and increased

308 Neurology 80 January 15, 2013

ª 2013 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.


HCN2 mRNA levels.31 Loss of HCN1 expression is a contributory role in cold hyperalgesia and allo-
associated with higher seizure severity and higher dynia in the setting of nerve injury or oxaliplatin-
seizure-related mortality in mouse models of limbic induced neuropathy.36
seizures.28 For example, adult HCN1 null mice are more
Subarachnoid hemorrhage. Disruption of ionic homeosta-
susceptible to kainic acid–induced seizures; this is
sis and neuronal hyperexcitability contribute to early
accompanied by increased spontaneous activity,
brain injury after subarachnoid hemorrhage. Whole-cell
increased dendric excitability, and imbalance between
recordings in rat brain slices indicate that perfusion of
normal excitatory and inhibitory synaptic activity.29
hemoglobin-containing artificial CSF produced neuro-
Electrophysiologic recording in vitro from layers II/III of
nal hyperexcitability and inhibited HCN currents in
neocortex resected from patients with temporal or fron-
CA1 pyramidal neurons by scavenging nitric oxide.37
tal lobe epilepsy suggests a deficit of HCN1 subunits in
the human epileptogenic neocortex, which in turn may
PERSPECTIVE HCN channels are important regula-
increase excitability and probability of seizure activity.30
tors of neuronal excitability and network activity in the
However, there is also evidence for enhanced HCN1
nervous system. Given the evidence of their potential role
expression and dendritic localization in surviving granule
in epilepsy and pain, these channels provide an attractive
cells of the dentate gyrus in resected hippocampi from
therapeutic target. Whereas HCN4 agonists, such as
patients with temporal lobe epilepsy and in experimental
ivabradine, provide an alternative to b-blockers or calci-
temporal lobe seizure models7; in these conditions,
um channel blockers for treatment of tachycardia,2 ago-
increased Ih in the dentate gyrus may represent a com-
nists or antagonists of HCN1 and HCN2 are not yet
pensatory mechanism to reduce neuronal
available for clinical use. Whereas selective HCN2 block-
hyperexcitability.32
ers could be used to treat neuropathic pain, the complex
Inherited cortical HCN1 channel loss also amplifies
and diverse role of HCN1 and HCN2 in epilepsy may
dendritic Ca21 electrogenesis and burst firing in a rat
prove challenging, as both inhibition and activation of Ih
absence epilepsy model.33 In contrast, increased HCN1
may be beneficial depending on the type of epilepsy.
activity in thalamocortical neurons, leading impaired
Some currently available antiepileptic drugs such as lam-
control of the shift from burst to tonic firing, has been
otrigine increase Ih in dendrites of pyramidal cells.38
found in a genetic rat model of absence epilepsy.22
The role of HCN2 channels in epilepsy is also incom-
DISCLOSURE
pletely understood. Spontaneous HCN2 mutations lead- The author reports no disclosures relevant to the manuscript. Go to
ing to markedly reduced HCN2 expression has been Neurology.org for full disclosures.
linked to generalized spike-wave absence seizures in
mice.21,34 A recessive loss-of-function mutation of the REFERENCES
HCN2 has also been linked to human generalized epi- 1. Biel M, Wahl-Schott C, Michalakis S, Zong X. Hyperpolar-
ization-activated cation channels: from genes to function.
lepsy.35 However, screening in subjects with febrile seiz-
Physiol Rev 2009;89:847–885.
ures and genetic epilepsy with febrile seizures plus 2. Postea O, Biel M. Exploring HCN channels as novel drug
revealed that 2.4% (as opposed to 0.2% of controls) targets. Nat Rev Drug Discov 2011;10:903–914.
carried a common triple proline deletion HCN2, result- 3. Baruscotti M, Bottelli G, Milanesi R, DiFrancesco JC,
ing in a gain of function of HCN2-mediated Ih.32 DiFrancesco D. HCN-related channelopathies. Pflugers Arch
2010;460:405–415.
4. Reid CA, Phillips AM, Petrou S. HCN channelopathies:
Pain. Several findings in experimental models of inflam-
pathophysiology in genetic epilepsy and therapeutic impli-
matory or neuropathic pain suggest HCN channels, par- cations. Br J Pharmacol 2012;165:49–56.
ticularly HCN2, have a prominent role in both 5. Poolos NP. Hyperpolarization-activated cyclic nucleotide-gated
inflammatory and neuropathic pain.1,2,25 Experimental (HCN) ion channelopathy in epilepsy. In: Noebels JL,
nerve injury results in increased Ih currents; pharmaco- Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV,
logic blockade of HCN channels by the pan-HCN eds. Jasper’s Basic Mechanisms of the Epilepsies, 4th ed. Be-
thesda: National Center for Biotechnology Information; 2012.
inhibitor, ZD-7288, reverses both pain behavior and
6. Noam Y, Bernard C, Baram TZ. Towards an integrated view
the spontaneous discharges in injured nerve fibers and of HCN channel role in epilepsy. Curr Opin Neurobiol 2011;
gives significant analgesia in a variety of pain models.2,25 21:873–879.
There is evidence that HCN2 has a prominent role in 7. Kase D, Imoto K. The role of HCN channels on membrane
mediating enhanced sensitivity to heat, cold, or mechan- excitability in the nervous system. J Signal Transduct 2012;
ical stimuli in both inflammatory and neuropathic 2012:619747.
pain.5,27 Selective HCN2 deletion restricted to the 8. Lewis AS, Chetkovich DM. HCN channels in behavior and
neurological disease: too hyper or not active enough? Mol Cell
Nav1.8-expressing nociceptive DRG neurons pre-
Neurosci 2011;46:357–367.
vents heat hyperalgesia triggered by proinflamma- 9. Shah MM, Huang Z, Martinello K. HCN and K(V)7 (M-)
tory stimuli such as PGE2. Selective knockout channels as targets for epilepsy treatment. Neuropharmacology
studies indicate that HCN1 channels may also have Epub 2012 Mar 15.

Neurology 80 January 15, 2013 309

ª 2013 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.


10. Herrmann S, Stieber J, Ludwig A. Pathophysiology of HCN 24. Nolan MF, Malleret G, Lee KH, et al. The hyperpolari-
channels. Pflugers Arch 2007;454:517–522. zation-activated HCN1 channel is important for motor
11. Notomi T, Shigemoto R. Immunohistochemical localization learning and neuronal integration by cerebellar Purkinje
of Ih channel subunits, HCN1-4, in the rat brain. J Comp cells. Cell 2003;115:551–564.
Neurol 2004;471:241–276. 25. Emery EC, Young GT, McNaughton PA. HCN2 ion chan-
12. Berger T, Senn W, Luscher HR. Hyperpolarization-activated nels: an emerging role as the pacemakers of pain. Trends
current Ih disconnects somatic and dendritic spike initiation Pharmacol Sci 2012;33:456–463.
zones in layer V pyramidal neurons. J Neurophysiol 2003;90: 26. Emery EC, Young GT, Berrocoso EM, Chen L,
2428–2437. McNaughton PA. HCN2 ion channels play a central role
13. Abbas SY, Ying SW, Goldstein PA. Compartmental distribu- in inflammatory and neuropathic pain. Science 2011;333:
tion of hyperpolarization-activated cyclic-nucleotide-gated 1462–1466.
channel 2 and hyperpolarization-activated cyclic-nucleotide- 27. Noam Y, Zha Q, Phan L, et al. Trafficking and surface expres-
gated channel 4 in thalamic reticular and thalamocortical relay sion of hyperpolarization-activated cyclic nucleotide-gated
neurons. Neuroscience 2006;141:1811–1825. channels in hippocampal neurons. J Biol Chem 2010;285:
14. Pian P, Bucchi A, Decostanzo A, Robinson RB, 14724–14736.
Siegelbaum SA. Modulation of cyclic nucleotide-regulated 28. Santoro B, Lee JY, Englot DJ, et al. Increased seizure severity
HCN channels by PIP(2) and receptors coupled to phospho- and seizure-related death in mice lacking HCN1 channels.
lipase C. Pflugers Arch 2007;455:125–145. Epilepsia 2010;51:1624–1627.
15. Wang M, Ramos BP, Paspalas CD, et al. Alpha2A- 29. Huang Z, Walker MC, Shah MM. Loss of dendritic
adrenoceptors strengthen working memory networks by HCN1 subunits enhances cortical excitability and epilep-
inhibiting cAMP-HCN channel signaling in prefrontal cor- togenesis. J Neurosci 2009;29:10979–10988.
tex. Cell 2007;129:397–410. 30. Wierschke S, Lehmann TN, Dehnicke C, Horn P, Nitsch R,
16. George MS, Abbott LF, Siegelbaum SA. HCN hyper- Deisz RA. Hyperpolarization-activated cation currents in
polarization-activated cation channels inhibit EPSPs by human epileptogenic neocortex. Epilepsia 2010;51:404–
interactions with M-type K(1) channels. Nat Neurosci 414.
2009;12:577–584. 31. Richichi C, Brewster AL, Bender RA, et al. Mechanisms
17. Atherton JF, Kitano K, Baufreton J, et al. Selective participa- of seizure-induced ’transcriptional channelopathy’ of
tion of somatodendritic HCN channels in inhibitory but not hyperpolarization-activated cyclic nucleotide gated (HCN)
excitatory synaptic integration in neurons of the subthalamic channels. Neurobiol Dis 2008;29:297–305.
nucleus. J Neurosci 2010;30:16025–16040. 32. Dibbens LM, Reid CA, Hodgson B, et al. Augmented
18. Hutcheon B, Yarom Y. Resonance, oscillation and the intrin- currents of an HCN2 variant in patients with febrile sei-
sic frequency preferences of neurons. Trends Neurosci 2000; zure syndromes. Ann Neurol 2010;67:542–546.
23:216–222. 33. Strauss U, Kole MH, Brauer AU, et al. An impaired neo-
19. Hu H, Vervaeke K, Storm JF. Two forms of electrical reso- cortical Ih is associated with enhanced excitability and
nance at theta frequencies, generated by M-current, h-current absence epilepsy. Eur J Neurosci 2004;19:3048–3058.
and persistent Na1 current in rat hippocampal pyramidal 34. Chung WK, Shin M, Jaramillo TC, et al. Absence epilepsy in
cells. J Physiol 2002;545:783–805. apathetic, a spontaneous mutant mouse lacking the h channel
20. Ludwig A, Herrmann S, Hoesl E, Stieber J. Mouse models subunit, HCN2. Neurobiol Dis 2009;33:499–508.
for studying pacemaker channel function and sinus node 35. DiFrancesco JC, Barbuti A, Milanesi R, et al. Recessive loss-
arrhythmia. Prog Biophys Mol Biol 2008;98:179–185. of-function mutation in the pacemaker HCN2 channel caus-
21. Ludwig A, Budde T, Stieber J, et al. Absence epilepsy and ing increased neuronal excitability in a patient with idiopathic
sinus dysrhythmia in mice lacking the pacemaker channel generalized epilepsy. J Neurosci 2011;31:17327–17337.
HCN2. EMBO J 2003;22:216–224. 36. Momin A, Cadiou H, Mason A, McNaughton PA. Role of
22. Budde T, Caputi L, Kanyshkova T, et al. Impaired regu- the hyperpolarization-activated current Ih in somatosensory
lation of thalamic pacemaker channels through an imbal- neurons. J Physiol 2008;586:5911–5929.
ance of subunit expression in absence epilepsy. J Neurosci 37. Li B, Luo C, Tang W, et al. Role of HCN channels in neu-
2005;25:9871–9882. ronal hyperexcitability after subarachnoid hemorrhage in rats.
23. Nolan MF, Malleret G, Dudman JT, et al. A behavioral J Neurosci 2012;32:3164–3175.
role for dendritic integration: HCN1 channels constrain 38. Poolos NP, Migliore M, Johnston D. Pharmacological upre-
spatial memory and plasticity at inputs to distal dendrites gulation of h-channels reduces the excitability of pyramidal
of CA1 pyramidal neurons. Cell 2004;119:719–732. neuron dendrites. Nat Neurosci 2002;5:767–774.

310 Neurology 80 January 15, 2013

ª 2013 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.


HCN channels : Function and clinical implications
Eduardo E. Benarroch
Neurology 2013;80;304
DOI 10.1212/WNL.0b013e31827dec42
This information is current as of January 16, 2013

Updated Information & including high resolution figures, can be found at:
Services http://www.neurology.org/content/80/3/304.full.html
References This article cites 36 articles, 11 of which can be accessed free
at:
http://www.neurology.org/content/80/3/304.full.html#ref-list-1
Permissions & Licensing Information about reproducing this article in parts (figures,
tables) or in its entirety can be found online at:
http://www.neurology.org/misc/about.xhtml#permissions
Reprints Information about ordering reprints can be found online:
http://www.neurology.org/misc/addir.xhtml#reprintsus

You might also like