Open Access Open Access  Restricted Access Subscription Access

Advances in Understanding of Structure, Function and Plasticity of HCN Channels


Affiliations
1 Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741 246, India
 

Hyperpolarization-activated, cyclic nucleotide-sensitive, cation non-selective (HCN) channels are gaining attention in recent years. These ion-channels are gated by two factors: voltage, specifically hyperpolarization and some subunits, more than others, by cyclic nucleotides. They conduct both sodium and potassium ions, and regulate many functions in both neuronal and non-neuronal cells. In neurons, HCN channel functions range from setting resting potential, synaptic normalization, gain control, after-hyperpolarization, setting responses in dendrites, mediating cannabinoid role in neuronal plasticity to the gating of plasticity. Emerging properties of circuits expressing HCN channels manifest in the form of various kinds of functions like brain rhythms, perception, spatial learning, executive memory, epilepsy, ataxia, depression, sound localization, anxiety, etc. We are beginning to understand the roles of these channels in non-neuronal cells and we would discover more such roles in the future. Following ischaemia, there is HCN over-expression in the reactive astrocytes. There is an altered expression of HCN channels in few tissues in diabetic animal models. Recent evidence suggests that HCN channels are also involved in uterine contractions and renal functioning. This review focuses on the function, structure, interacting proteins and developmental plasticity that enable the versatility of HCN channels.

Keywords

Developmental Plasticity, HCN Channels, Interacting Proteins.
User
Notifications
Font Size

  • Santoro, B. and Tibbs, G. R., The HCN gene family: molecular basis of the hyperpolarization-activated pacemaker channels. Ann. NY Acad. Sci., 1999, 868, 741–764.
  • Santoro, B., Grant, S. G., Bartsch, D. and Kandel, E. R., Interactive cloning with the SH3 domain of N-src identifies a new brain specific ion channel protein, with homology to eag and cyclic nucleotidegated channels. Proc. Natl. Acad. Sci. USA, 1997, 94, 14815–14820.
  • Ludwig, A., Zong, X., Jeglitsch, M., Hofmann, F. and Biel, M., A family of hyperpolarization-activated mammalian cation channels. Nature, 1998, 393, 587–591.
  • Santoro, B. et al., Identification of a gene encoding a hyperpolarizationactivated pacemaker channel of brain. Cell, 1998, 93, 717–729.
  • DiFrancesco, D., A new interpretation of the pace-maker current in calf Purkinje fibres. J. Physiol., 1981, 314, 359–376.
  • DiFrancesco, D., Characterization of the pace-maker current kinetics in calf Purkinje fibres. J. Physiol., 1984, 348, 341–367.
  • Barbuti, A. and DiFrancesco, D., Control of cardiac rate by ‘funny’ channels in health and disease. Ann. NY Acad. Sci., 2008, 1123, 213–223.
  • Barbuti, A., Baruscotti, M. and DiFrancesco, D., The pacemaker current: from basics to the clinics. J. Cardiovasc. Electrophysiol., 2007, 18, 342–347.
  • DiFrancesco, D., Characterization of single pacemaker channels in cardiac sinoatrial node cells. Nature, 1986, 324, 470–473.
  • Maccaferri, G. and McBain, C. J., The hyperpolarization-activated current (Ih) and its contribution to pacemaker activity in rat CA1 hippocampal stratum oriens-alveus interneurones. J. Physiol., 1996, 497(1), 119–130.
  • Khurana, S., Remme, M. W. H., Rinzel, J. and Golding, N. L., Dynamic interaction of Ih and IK-LVA during trains of synaptic potentials in principal neurons of the medial superior olive. J. Neurosci., 2011, 31, 8936–8947.
  • Cao, D.-N., Song, R., Zhang, S.-Z., Wu, N. and Li, J., Nucleus accumbens hyperpolarization-activated cyclic nucleotide-gated channels modulate methamphetamine self-administration in rats. Psychopharmacology (Berl), 2016, 233, 3017–3029.
  • DiFrancesco, D. and Mangoni, M., Modulation of single hyperpolarizationactivated channels (i(f)) by cAMP in the rabbit sinoatrial node. J. Physiol., 1994, 474, 473–482.
  • Hagiwara, N. and Irisawa, H., Modulation by intracellular Ca2+ of the hyperpolarization-activated inward current in rabbit single sino-atrial node cells. J. Physiol., 1989, 409, 121–141.
  • Pape, H. C., Queer current and pacemaker: the hyperpolarization-activated cation current in neurons. Annu. Rev. Physiol., 1996, 58, 299–327.
  • Munsch, T. and Pape, H. C., Modulation of the hyperpolarization-activated cation current of rat thalamic relay neurones by intracellular pH. J. Physiol., 1999, 519(Pt 2), 493–504.
  • Leist, M. et al., Two types of interneurons in the mouse lateral geniculate nucleus are characterized by different h-current density. Sci. Rep., 2016, 6, 24904.
  • Williams, S. R., Christensen, S. R., Stuart, G. J. and Häusser, M., Membrane potential bistability is controlled by the hyperpolarizationactivated current I(H) in rat cerebellar Purkinje neurons in vitro. J. Physiol., 2002, 539, 469–483.
  • Robinson, R. B. and Siegelbaum, S. A., Hyperpolarization-activated cation currents: from molecules to physiological function. Annu. Rev. Physiol., 2003, 65, 453–480.
  • Zuniga, R., Gonzalez, D., Valenzuela, C., Brown, N. and Zuniga, L., Expression and cellular localization of HCN channels in rat cerebellar granule neurons. Biochem. Biophys. Res. Commun., 2016, 478, 1429–1435.
  • Magee, J. C., Dendritic hyperpolarization-activated currents modify the integrative properties of hippocampal CA1 pyramidal neurons. J. Neurosci., 1998, 18, 7613–7624.
  • Gravante, B. et al., Interaction of the pacemaker channel HCN1 with filamin A. J. Biol. Chem., 2004, 279, 43847–43853.
  • Beaumont, V. and Zucker, R. S., Enhancement of synaptic transmission by cyclic AMP modulation of presynaptic Ih channels. Nature Neurosci., 2000, 3, 133–141.
  • Beaumont, V., Zhong, N., Froemke, R. C., Ball, R. W. and Zucker, R. S., Temporal synaptic tagging by Ih activation and actin: involvement in long-term facilitation and cAMP-induced synaptic enhancement. Neuron, 2002, 33, 601–613.
  • Narayanan, R. and Johnston, D., The h channel mediates location dependence and plasticity of intrinsic phase response in rat hippocampal neurons. J. Neurosci., 2008, 28, 5846–5860.
  • Narayanan, R. and Johnston, D., Long-term potentiation in rat hippocampal neurons is accompanied by spatially widespread changes in intrinsic oscillatory dynamics and excitability. Neuron, 2007, 56, 1061–1075.
  • Moosmang, S., Biel, M., Hofmann, F. and Ludwig, A., Differential distribution of four hyperpolarization-activated cation channels in mouse brain. Biol. Chem., 1999, 380, 975–980.
  • Santoro, B. et al., Molecular and functional heterogeneity of hyperpolarizationactivated pacemaker channels in the mouse CNS. J. Neurosci., 2000, 20, 5264–5275.
  • Brewster, A. L. et al., Quantitative analysis and subcellular distribution of mRNA and protein expression of the hyperpolarizationactivated cyclic nucleotide-gated channels throughout development in rat hippocampus. Cereb. Cortex, 2007, 17, 702–712.
  • Nolan, M. F. et al., A behavioral role for dendritic integration: HCN1 channels constrain spatial memory and plasticity at inputs to distal dendrites of CA1 pyramidal neurons. Cell, 2004, 119, 719–732.
  • Herrmann, S., Stieber, J. and Ludwig, A., Pathophysiology of HCN channels. Pflügers Arch. – Eur. J. Physiol., 2007, 454, 517–522.
  • Stieber, J. et al., The hyperpolarization-activated channel HCN4 is required for the generation of pacemaker action potentials in the embryonic heart. Proc. Natl. Acad. Sci. USA, 2003, 100, 15235–15240.
  • Zong, X., Stieber, J., Ludwig, A., Hofmann, F. and Biel, M., A single histidine residue determines the pH sensitivity of the pacemaker channel HCN2. J. Biol. Chem., 2001, 276, 6313–6319.
  • Hussaini, S. A., Kempadoo, K. A., Thuault, S. J., Siegelbaum, S. A. and Kandel, E. R., Increased size and stability of CA1 and CA3 place fields in HCN1 knockout mice. Neuron, 2011, 72, 643–653.
  • Kim, C. S., Chang, P. Y. and Johnston, D., Enhancement of dorsal hippocampal activity by knockdown of HCN1 channels leads to anxiolytic- and antidepressant-like behaviors. Neuron, 2012, 75, 503–516.
  • Kim, C. S., Brager, D. H. and Johnston, D., Perisomatic changes in h-channels regulate depressive behaviors following chronic unpredictable stress. Mol. Psychiatry, 2017; doi:10.1038/mp.2017.28.
  • Lewis, A. S. et al., Deletion of the hyperpolarization-activated cyclic nucleotide-gated channel auxiliary subunit TRIP8b impairs hippocampal Ih localization and function and promotes antidepressant behavior in mice. J. Neurosci., 2011, 31, 7424–7440.
  • Kole, M. H. P., Hallermann, S. and Stuart, G. J., Single Ih channels in pyramidal neuron dendrites: properties, distribution, and impact on action potential output. J. Neurosci., 2006, 26, 1677–1687.
  • Simeone, T. A., Rho, J. M. and Baram, T. Z., Single channel properties of hyperpolarization-activated cation currents in acutely dissociated rat hippocampal neurones. J. Physiol., 2005, 568, 371–380.
  • Khurana, S. et al., An essential role for modulation of hyperpolarizationactivated current in the development of binaural temporal precision. J. Neurosci., 2012, 32, 2814–2823.
  • Wahl-Schott, C. and Biel, M., HCN channels: structure, cellular regulation and physiological function. Cell. Mol. Life Sci., 2009, 66, 470–494.
  • Pian, P., Bucchi, A., Robinson, R. B. and Siegelbaum, S. A., Regulation of gating and rundown of HCN hyperpolarization-activated channels by exogenous and endogenous PIP2. J. Gen. Physiol., 2006, 128, 593–604.
  • Zolles, G. et al., Pacemaking by HCN channels requires interaction with phosphoinositides. Neuron, 2006, 52, 1027–1036.
  • Stevens, D. R. et al., Hyperpolarization-activated channels HCN1 and HCN4 mediate responses to sour stimuli. Nature, 2001, 413, 631–635.
  • Wahl-Schott, C., Baumann, L., Zong, X. and Biel, M., An arginine residue in the pore region is a key determinant of chloride dependence in cardiac pacemaker channels. J. Biol. Chem., 2005, 280, 13694–13700.
  • Zong, X. et al., A novel mechanism of modulation of hyperpolarizationactivated cyclic nucleotide-gated channels by Src Kinase. J. Biol. Chem., 2005, 280, 34224–34232.
  • Li, C.-H. et al., Src tyrosine kinase alters gating of hyperpolarizationactivated HCN4 pacemaker channel through Tyr531. Am. J. Physiol. – Cell Physiol., 2008, 294, C355–C362.
  • Poolos, N. P., Bullis, J. B. and Roth, M. K., Modulation of hchannels in hippocampal pyramidal neurons by p38 mitogenactivated protein kinase. J. Neurosci., 2006, 26, 7995–8003.
  • Abbott, G. W. et al., MiRP1 forms IKr potassium channels with HERG and is associated with cardiac arrhythmia. Cell, 1999, 97, 175–187.
  • Tinel, N. et al. M-type KCNQ2-KCNQ3 potassium channels are modulated by the KCNE2 subunit. FEBS Lett., 2000, 480, 137–141.
  • Zhang, M., Jiang, M. and Tseng, G. N., MinK-related peptide 1 associates with Kv4.2 and modulates its gating function: potential role as beta subunit of cardiac transient outward channel? Circ. Res., 2001, 88, 1012–1019.
  • Yu, H. et al. MinK-related peptide 1: a beta subunit for the HCN ion channel subunit family enhances expression and speeds activation. Circ. Res., 2001, 88, E84–E847.
  • Qu, J. et al., MiRP1 modulates HCN2 channel expression and gating in cardiac myocytes. J. Biol. Chem., 2004, 279, 43497–43502.
  • Decher, N., Bundis, F., Vajna, R. and Steinmeyer, K., KCNE2 modulates current amplitudes and activation kinetics of HCN4: influence of KCNE family members on HCN4 currents. Pflugers Arch. – Eur. J. Physiol., 2003, 446, 633–640.
  • Kimura, K., Kitano, J., Nakajima, Y. and Nakanishi, S., Hyperpolarizationactivated, cyclic nucleotide-gated HCN2 cation channel forms a protein assembly with multiple neuronal scaffold proteins in distinct modes of protein-protein interaction. Genes Cells, 2004, 9, 631–640.
  • Barbuti, A. et al., Localization of pacemaker channels in lipid rafts regulates channel kinetics. Circ. Res., 2004, 94, 1325–1331.
  • Santoro, B., Wainger, B. J. and Siegelbaum, S. A., Regulation of HCN channel surface expression by a novel C-terminal protein–protein interaction. J. Neurosci., 2004, 24, 10750–10762.
  • Lewis, A. S. et al., Alternatively spliced isoforms of TRIP8b differentially control h channel trafficking and function. J. Neurosci., 2009, 29, 6250–6265.
  • Santoro, B. et al., TRIP8b splice variants form a family of auxiliary subunits that regulate gating and trafficking of HCN channels in the brain. Neuron, 2009, 62, 802–813.
  • Zolles, G. et al., Association with the auxiliary subunit PEX5R/Trip8b controls responsiveness of HCN channels to cAMP and adrenergic stimulation. Neuron, 2009, 62, 814–825.
  • Santoro, B. et al., TRIP8b regulates HCN1 channel trafficking and gating through two distinct C-terminal interaction sites. J. Neurosci., 2011, 31, 4074–4086.
  • Bankston, J. R. et al., Structure and stoichiometry of an accessory subunit TRIP8b interaction with hyperpolarization-activated cyclic nucleotide-gated channels. Proc. Natl. Acad. Sci. USA, 2012, 109, 7899–7904.
  • Han, Y. et al., Trafficking and gating of hyperpolarizationactivated cyclic nucleotide-gated channels are regulated by interaction with tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b) and cyclic AMP at distinct sites. J. Biol. Chem., 2011, 286, 20823–20834.
  • Huang, Z. et al., TRIP8b-independent trafficking and plasticity of adult cortical presynaptic HCN1 channels. J. Neurosci., 2012, 32, 14835–14848.
  • Wilkars, W. et al., Regulation of axonal HCN1 trafficking in perforant path involves expression of specific TRIP8b isoforms. PLOS ONE, 2012, 7, e32181.
  • Gisselmann, G. et al., Molecular and functional characterization of an I (h)-channel from lobster olfactory receptor neurons. Eur. J. Neurosci., 2005, 21, 1635–1647.
  • Surges, R. et al., Regulated expression of HCN channels and cAMP levels shape the properties of the h current in developing rat hippocampus. Eur. J. Neurosci., 2006, 24, 94–104.
  • Vasilyev, D. V. and Barish, M. E., Postnatal development of the hyperpolarization-activated excitatory current Ih in mouse hippocampal pyramidal neurons. J. Neurosci., 2002, 22, 8992–9004.
  • Hogg, R. C., Harper, A. A. and Adams, D. J., Developmental changes in hyperpolarization-activated currents I(h) and I(K(IR)) in isolated rat intracardiac neurons. J. Neurophysiol., 2001, 86, 312–320.
  • Westermann, A. J., Gorski, S. A. and Vogel, J., Dual RNA-seq of pathogen and host. Nature Rev. Microbiol., 2012, 10, 618–630.
  • Ram, P. T., Mendelsohn, J. and Mills, G. B., Bioinformatics and systems biology. Mol. Oncol., 2012, 6, 147–154.
  • Tibbs, G. R., Posson, D. J. and Goldstein, P. A., Voltage-gated ion channels in the PNS: novel therapies for neuropathic pain? Trends Pharmacol. Sci., 2016, 37, 522–542.
  • Emery, E. C., Young, G. T. and McNaughton, P. A., HCN2 ion channels: an emerging role as the pacemakers of pain. Trends Pharmacol. Sci., 2012, 33, 456–463.
  • Ding, W. et al., Inhibition of HCN channel activity in the thalamus attenuates chronic pain in rats. Neurosci. Lett., 2016, 631, 97–103.
  • Zhang, S. et al., Neuropeptide S modulates the amygdaloidal HCN activities (Ih) in rats: implication in chronic pain. Neuropharmacology, 2016, 105, 420–433.
  • Liu, H., Zhou, J., Gu, L. and Zuo, Y., The change of HCN1/HCN2 mRNA expression in peripheral nerve after chronic constriction injury induced neuropathy followed by pulsed electromagnetic field therapy. Oncotarget, 2016, 8, 1110–1116.
  • Huang, H. and Trussell, L. O., Presynaptic HCN channels regulate vesicular glutamate transport. Neuron, 2014, 84, 340–346.
  • Huang, Z., Li, G., Aguado, C., Lujan, R. and Shah, M. M., HCN1 channels reduce the rate of exocytosis from a subset of cortical synaptic terminals. Sci. Rep., 2017, 7, 40257.
  • Hegle, A. P. et al., The Ih channel gene promotes synaptic transmission and coordinated movement in Drosophila melanogaster. Front. Mol. Neurosci., 2017, 10, 41.
  • Yi, F. et al., Autism-associated SHANK3 haploinsufficiency causes Ih channelopathy in human neurons. Science, 2016, 352, aaf2669.
  • Han, Y. et al., HCN-channel dendritic targeting requires bipartite interaction with TRIP8b and regulates antidepressant-like behavioral effects. Mol. Psychiatry, 2017, 22, 458–465.
  • Heuermann, R. J. et al., Reduction of thalamic and cortical Ih by deletion of TRIP8b produces a mouse model of human absence epilepsy. Neurobiol. Dis., 2016, 85, 81–92.
  • Shah, M. M., Huang, Z. and Martinello, K., HCN and KV7 (M-) channels as targets for epilepsy treatment. Neuropharmacology, 2013, 69, 75–81.
  • Shah, M. M., Hyperpolarization-activated cyclic nucleotide-gated channel currents in neurons. Cold Spring Harbor. Protoc., 2016; pdb.top087346.
  • Li, S., Kalappa, B. I. and Tzounopoulos, T., Noise-induced plasticity of KCNQ2/3 and HCN channels underlies vulnerability and resilience to tinnitus. Elife, 2015, 4.
  • Kopp-Scheinpflug, C., Pigott, B. M. and Forsythe, I. D., Nitric oxide selectively suppresses IH currents mediated by HCN1-containing channels. J. Physiol., 2015, 593, 1685–1700.
  • Ko, K. W., Rasband, M. N., Meseguer, V., Kramer, R. H. and Golding, N. L., Serotonin modulates spike probability in the axon initial segment through HCN channels. Nature Neurosci., 2016, 19, 826–834.
  • Carlson, A. E., Rosenbaum, J. C., Brelidze, T. I., Klevit, R. E. and Zagotta, W. N., Flavonoid regulation of HCN2 channels. J. Biol. Chem., 2013, 288, 33136–33145.
  • Cao, D.-N., Song, R., Zhang, S.-Z., Wu, N. and Li, J., Nucleus accumbens hyperpolarization-activated cyclic nucleotide-gated channels modulate methamphetamine self-administration in rats. Psychopharmacology, 2016, 233, 3017–3029.
  • Santos-Vera, B. et al., Cocaine sensitization increases I h current channel subunit 2 (HCN2) protein expression in structures of the mesocorticolimbic system. J. Mol. Neurosci., 2013, 50, 234–245.
  • Huang, X. et al., Reduced expression of HCN channels in the sinoatrial node of streptozotocin-induced diabetic rats. Can. J. Physiol. Pharmacol., 2017, 95, 586–594.
  • Dong, X. et al., Interaction of caveolin-3 and HCN is involved in the pathogenesis of diabetic cystopathy. Sci. Rep., 2016, 6, 24844.
  • Alotaibi, M., Kahlat, K., Nedjadi, T. and Djouhri, L., Effects of ZD7288, a hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker, on term-pregnant rat uterine contractility in vitro. Theriogenology, 2017, 90, 141–146.

Abstract Views: 284

PDF Views: 73




  • Advances in Understanding of Structure, Function and Plasticity of HCN Channels

Abstract Views: 284  |  PDF Views: 73

Authors

Sukant Khurana
Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741 246, India

Abstract


Hyperpolarization-activated, cyclic nucleotide-sensitive, cation non-selective (HCN) channels are gaining attention in recent years. These ion-channels are gated by two factors: voltage, specifically hyperpolarization and some subunits, more than others, by cyclic nucleotides. They conduct both sodium and potassium ions, and regulate many functions in both neuronal and non-neuronal cells. In neurons, HCN channel functions range from setting resting potential, synaptic normalization, gain control, after-hyperpolarization, setting responses in dendrites, mediating cannabinoid role in neuronal plasticity to the gating of plasticity. Emerging properties of circuits expressing HCN channels manifest in the form of various kinds of functions like brain rhythms, perception, spatial learning, executive memory, epilepsy, ataxia, depression, sound localization, anxiety, etc. We are beginning to understand the roles of these channels in non-neuronal cells and we would discover more such roles in the future. Following ischaemia, there is HCN over-expression in the reactive astrocytes. There is an altered expression of HCN channels in few tissues in diabetic animal models. Recent evidence suggests that HCN channels are also involved in uterine contractions and renal functioning. This review focuses on the function, structure, interacting proteins and developmental plasticity that enable the versatility of HCN channels.

Keywords


Developmental Plasticity, HCN Channels, Interacting Proteins.

References





DOI: https://doi.org/10.18520/cs%2Fv114%2Fi03%2F462-467