Cardiac AP Notes-2020

Total Page:16

File Type:pdf, Size:1020Kb

Cardiac AP Notes-2020 DeWayne Townsend PHSL 4702/5702 Cell Physiology Fall, 2020 Cardiac Action Potential Cardiac Conduction Voltage Gated Ca2+ Channels Anatomical Regions Structure and isoforms Pacemaking Cells Gating properties Role in Automaticity Anatomy of the Cardiac Action Potential Role in Contractile Muscle Diastolic Depolarization Important Currents Cardiac K+ Channels Transient Outward Current Hyperpolarization-Cyclic Nucleotide-Gated Delayed Rectifiers Channel G-Protein Linked Inward Rectifying K+ Channel Structure and isoforms ATP Activated K+ Channel GENESIS AND REGULATION OF THE HEART AUTOMATICITY 921 Gating properties Cardiac Cl- Channels Voltage Gated Na+ Channels Cardiac CFTRregulation of pacemaker activity have been obtained: the venosus (227, 228) and mammalian SAN (44) has been Structure and isoforms Rectifyingrelevance Cl- Channels of f-channels in the regulation of heart rate at recently highlighted, and the contribution of spontaneous 2 Gating properties Ca2+ Activatedlow parasympatheticCl- Channels tone (124) and the physiological intracellular Ca ϩ release in the generation of automatic- Role in Automaticity significance of the heterogeneity of ion channel expres- ity has been emphasized (510). Role in Contractile Muscle Connexins sion in the SAN (58). The importance of the Naϩ-Ca2ϩ New important insights into the pacemaker mecha- Structureexchanger and isoforms (NCX) in pacemaking in the amphibian sinus nism are now coming from the study of genetically mod- Gating properties ified mouse strains in which specific ion channels have been inactivated or modified. This genetic approach con- Cardiac Conduction stitutes a necessary implementation to electrophysiologi- cal and pharmacological evidence, since selective inhibi- Anatomical Regions tors of ion channels are not always available. Gene tar- In healthy hearts, the cardiac action geting in the mouse has generated interesting models of dysfunction of pacemaker activity (393, 467, 522). The potential originates in specialized possibility to study pacemaker activity in mouse models is Downloaded from pacemaking cells in the sinoatrial node (Fig. very recent (312, 313) and has provided exciting insights 1). This depolarization is spread from these into the specific functional role of L-type Cav1.3 (309, 546) pacemaker cells to the atrial cardiac and T-type Cav3.1 (314) channels in the generation of SAN myocytes. This depolarization reaches a automaticity. Because of the tiny size of the dominant group of cells at the border of the atria and pacemaker region in the mouse heart (498), isolation of mouse SAN cells is technically challenging. Our group has http://physrev.physiology.org/ ventricular chambers called the been the first to obtain recordings of automaticity and ion atrioventricular node. These cells have a channels from isolated mouse SAN cells (312, 313). Other slowed conduction time resulting in a delay groups have successfully employed this new preparation of transmission of the cardiac action to study pacemaker activity in wild-type (86, 93, 279) and potential from the atria to the ventricule. A genetically modified mouse strains (196, 294, 309, 314, Figure 1. Diagram of the cardiac conduction pathways. 546). Limitations of the use of the mouse for the study of SCV- superior vena cava, SAN- sinoatrial node, CFB- fibrous band at the level of mitral and cardiac automaticity are correlated with the very high tricuspid valves, prevents direct spreading central fibrous body, AVN- atrioventricular node, AVB- atrioventricular bundle, BB - bundle branch, PFN- Purkinje basal heart rate of this species. We can thus expect that of the action potential to the ventricular fiber network, ICV- inferior vena cava. From Mangoni et pacemaker mechanisms may assume a different role in by 10.220.33.5 on July 18, 2017 muscle. After passing through the AV-node, al., 2008. mice than in larger mammals and humans. Nevertheless, the depolarization enters the Purkinje fiber the new possibility of isolating mouse SAN tissue and primary pacemaker cells has created a new interest into network, which rapidly transmits the depolarization toward the apex of the heart. At this the study of the ionic basis of pacemaker activity gener- point the depolarization comes in contact with the contractile cells of the ventricle. This ation and regulation. arrangement results in the synchronized contraction of both the right and left ventricules Until the last decade, research on the physiology of from the apex toward the heart valves. Importantly, each of these cell types has a heart automaticity was limited to the domain of basic unique action potential signature that reflects the role of the action potential in each of science. However, there is currently a renewal of interest these cells along the conduction system (Fig. 2). on cardiac pacemaking, and an increasing number of laboratories are now focusing their efforts on the regula- tion of heart rate. Indeed, the pharmacological control of cardiac automaticity is now becoming an important issue in the management of ischemic heart diseases (123). Iden- FIG. 1. A: the mammalian heart with the cardiac conduction system. The sinoatrial node (SAN) is located at the entry of the superior vena cava (SCV) in the right atrium (RA). The atrioventricular node (AVN) extends in a region delimited by the inferior vena cava (ICV), the central fibrous body (CFB), and the tricuspid valve (TV). The atrioventricular bundle (AVB) divides in the bundle branches (BB) and originates the left and right Purkinje fibers network (PFN). Other abbreviations are: LA, left atrium; PV, pulmonary veins; MV, mitral valve; RV, right ventricle; LV, left ventricle. [Adapted from Moorman and Christoffels (341).] B: recordings of automaticity and action potential waveforms of isolated mouse SAN, AVN, and PFN cells. Cl, cycle length; APD, action potential duration; Eth, action potential threshold; LDD, linear part of the diastolic depolarization; EDD, exponential part of the diastolic depolarization. (From Marger Nargeot and Matteo Mangoni, unpublished observations.) Physiol Rev • VOL 88 • JULY 2008 • www.prv.org DeWayne Townsend PHSL 4702/5702 Cell Physiology Fall, 2020 Pacemaking Cells There are several cells within the heart capable of generating rhythmic action potentials. Each of these cells display diastolic depolarization, a steady increase in membrane potential following the hyperpolarization of proceeding action potential. These cells are highly concentrated in the sinoatrial (SA) and atrioventricular (AV) nodes, although the Purkinje fibers are also capable of diastolic depolarization. The cells in the SA node display the most rapid diastolic depolarization. Therefore, they are the first to reach the threshold potential for initiating an action potential and are Figure 2. Action potential tracings from different regions of the cardiac conduction system. Color coded to demonstrate which responsible for producing normal cardiac portions of the traveling depolarization is responsible for which action potentials. aspect of the surface ECG tracing. Anatomy of the Cardiac Action Potential In many respects the cardiac action potential is quite similar to those of other excitable tissues that have been previously discussed in this course. The following section will focus on several of the unique features of the cardiac action potential. Diastolic Depolarization One of the defining feature of the cardiac action potential is the periodicity of its initiation. This phenomenon is the result of the presence of a diastolic depolarization in specific cells that generate cardiac action potentials. These pacemaker cells are influenced by the inputs from the nervous system, especially both Figure 3. A generic cardiac action potential. Upper tracing measures branches of the autonomic nervous the membrane potential (y-axis) versus time (x-axis). Phase 4 occurs system. The depth of diastolic during diastole and is sloped in pacemaker cells (left side), but relatively hyperpolarization and the slope of flat in contracting myocardium (right side). Individual currents and their activation kinetics are shown in the lower portion of the figure. FUNCTION OF HCN CHANNELS 851 ϳ0.5% (440, 441). The functional relevance of Ca2ϩ entry 333, 341, 344, 400) but are missing in Caenorhabditis through Ih channels is not clear at the moment. It was elegans, yeast, and prokaryotes. assumed that in dorsal root ganglion neurons, Ca2ϩ influx In all mammals investigated so far, four homologous through Ih channels at negative potentials contributes to HCN channel subunits (HCN1-4) exist. HCN1-4 are also activity-evoked secretion (441). present in the genome of fishes (e.g., Tetraodon nigro- There is an ongoing controversy on the size of the viridis and Takifugu rubripes). Analysis of genomic se- DeWayne Townsend PHSL 4702/5702 Cell Physiology Fall, 2020 single-channel conductance of Ih. Originally, single-chan- quences suggests that HCN2–4 genes underwent duplica- nel conductance was found to be very low, in the range of tions in the fish lineage increasing the number of potential ϳ1 pS (110). This estimate is in good agreement with very HCN species (194). HCN homologs have also been cloned diastolicrecent data depolarization (210). However, are single-channel the chief conductances determinantsfrom of the several heart invertebrates rate. including arthropods [e.g., that are 10–30 times higher have been reported for cloned spiny lobster (157), insects (158, 211, 251)] and sea ur- ImportantHCN channels Currents (267) as in well the
Recommended publications
  • Ion Channels 3 1
    r r r Cell Signalling Biology Michael J. Berridge Module 3 Ion Channels 3 1 Module 3 Ion Channels Synopsis Ion channels have two main signalling functions: either they can generate second messengers or they can function as effectors by responding to such messengers. Their role in signal generation is mainly centred on the Ca2 + signalling pathway, which has a large number of Ca2+ entry channels and internal Ca2+ release channels, both of which contribute to the generation of Ca2 + signals. Ion channels are also important effectors in that they mediate the action of different intracellular signalling pathways. There are a large number of K+ channels and many of these function in different + aspects of cell signalling. The voltage-dependent K (KV) channels regulate membrane potential and + excitability. The inward rectifier K (Kir) channel family has a number of important groups of channels + + such as the G protein-gated inward rectifier K (GIRK) channels and the ATP-sensitive K (KATP) + + channels. The two-pore domain K (K2P) channels are responsible for the large background K current. Some of the actions of Ca2 + are carried out by Ca2+-sensitive K+ channels and Ca2+-sensitive Cl − channels. The latter are members of a large group of chloride channels and transporters with multiple functions. There is a large family of ATP-binding cassette (ABC) transporters some of which have a signalling role in that they extrude signalling components from the cell. One of the ABC transporters is the cystic − − fibrosis transmembrane conductance regulator (CFTR) that conducts anions (Cl and HCO3 )and contributes to the osmotic gradient for the parallel flow of water in various transporting epithelia.
    [Show full text]
  • The Example of Short QT Syndrome Jules C
    Hancox et al. Journal of Congenital Cardiology (2019) 3:3 Journal of https://doi.org/10.1186/s40949-019-0024-7 Congenital Cardiology REVIEW Open Access Learning from studying very rare cardiac conditions: the example of short QT syndrome Jules C. Hancox1,4* , Dominic G. Whittaker2,3, Henggui Zhang4 and Alan G. Stuart5,6 Abstract Background: Some congenital heart conditions are very rare. In a climate of limited resources, a viewpoint could be advanced that identifying diagnostic criteria for such conditions and, through empiricism, effective treatments should suffice and that extensive mechanistic research is unnecessary. Taking the rare but dangerous short QT syndrome (SQTS) as an example, this article makes the case for the imperative to study such rare conditions, highlighting that this yields substantial and sometimes unanticipated benefits. Genetic forms of SQTS are rare, but the condition may be under-diagnosed and carries a risk of sudden death. Genotyping of SQTS patients has led to identification of clear ion channel/transporter culprits in < 30% of cases, highlighting a role for as yet unidentified modulators of repolarization. For example, recent exome sequencing in SQTS has identified SLC4A3 as a novel modifier of ventricular repolarization. The need to distinguish “healthy” from “unhealthy” short QT intervals has led to a search for additional markers of arrhythmia risk. Some overlap may exist between SQTS, Brugada Syndrome, early repolarization and sinus bradycardia. Genotype-phenotype studies have led to identification of arrhythmia substrates and both realistic and theoretical pharmacological approaches for particular forms of SQTS. In turn this has increased understanding of underlying cardiac ion channels.
    [Show full text]
  • Nitric Oxide Modulates HCN Channels in Magnocellular Neurons of the Supraoptic Nucleus of Rats by an S-Nitrosylation-Dependent Mechanism
    11320 • The Journal of Neuroscience, November 2, 2016 • 36(44):11320–11330 Cellular/Molecular Nitric Oxide Modulates HCN Channels in Magnocellular Neurons of the Supraoptic Nucleus of Rats by an S-Nitrosylation-Dependent Mechanism X Melina Pires da Silva,1 Davi Jose´ de Almeida Moraes,1 Andre´ de Souza Mecawi,2 Jose´ Antunes Rodrigues,1 and X Wamberto Antonio Varanda1 1Department of Physiology, Ribeira˜o Preto Medical School, University of Sa˜o Paulo, 14049-900 Ribeira˜o Preto, Sa˜o Paulo, Brazil, and 2Department of Physiological Sciences, Biology Institute, Federal Rural University of Rio de Janeiro, 23890-000, Serope´dica, Rio de Janeiro, Brazil The control of the excitability in magnocellular neurosecretory cells (MNCs) of the supraoptic nucleus has been attributed mainly to synapticinputsfromcircunventricularorgans.However,nitricoxide(NO),agaseousmessengerproducedinthisnucleusduringisotonic and short-term hypertonic conditions, is an example of a modulator that can act directly on MNCs to modulate their firing rate. NO inhibits the electrical excitability of MNCs, leading to a decrease in the release of vasopressin and oxytocin. Although the effects of NO on MNCs are well established, the mechanism by which this gas produces its effect is, so far, unknown. Because NO acts independently of synaptic inputs, we hypothesized that ion channels present in MNCs are the targets of NO. To investigate this hypothesis, we used the patch-clamptechniqueinvitroandinsitutomeasurecurrentscarriedbyhyperpolarization-activatedandnucleotide-gatedcation(HCN) channelsandestablishtheirroleindeterminingtheelectricalexcitabilityofMNCsinrats.OurresultsshowthatblockadeofHCNchannels by ZD7288 decreases MNC firing rate with significant consequences on the release of OT and VP, measured by radioimmunoassay. NO induced a significant reduction in HCN currents by binding to cysteine residues and forming S-nitrosothiol complexes.
    [Show full text]
  • HCN Channels—Modulators of Cardiac and Neuronal Excitability
    Int. J. Mol. Sci. 2015, 16, 1429-1447; doi:10.3390/ijms16011429 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Review HCN Channels—Modulators of Cardiac and Neuronal Excitability Stefan Herrmann *, Sabine Schnorr and Andreas Ludwig * Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; E-Mail: [email protected] * Authors to whom correspondence should be addressed; E-Mails: [email protected] (S.H.); [email protected] (A.L.); Tel.: +49-9131-85-26979 (S.H.); +49-9131-85-2220 (A.L.); Fax: +49-9131-85-22774 (S.H. & A.L.). Academic Editor: Jens Schlossmann Received: 28 November 2014 / Accepted: 31 December 2014 / Published: 8 January 2015 Abstract: Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels comprise a family of cation channels activated by hyperpolarized membrane potentials and stimulated by intracellular cyclic nucleotides. The four members of this family, HCN1–4, show distinct biophysical properties which are most evident in the kinetics of activation and deactivation, the sensitivity towards cyclic nucleotides and the modulation by tyrosine phosphorylation. The four isoforms are differentially expressed in various excitable tissues. This review will mainly focus on recent insights into the functional role of the channels apart from their classic role as pacemakers. The importance of HCN channels in the cardiac ventricle and ventricular hypertrophy will be discussed. In addition, their functional significance in the peripheral nervous system and nociception will be examined. The data, which are mainly derived from studies using transgenic mice, suggest that HCN channels contribute significantly to cellular excitability in these tissues.
    [Show full text]
  • TRPC3 and NALCN Channels Drive Pacemaking in Substantia Nigra Dopaminergic Neurons
    bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.447013; this version posted June 4, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. TRPC3 and NALCN channels drive pacemaking in substantia nigra dopaminergic neurons Ki Bum Um1, Suyun Hahn1, So Woon Kim1, Yoon Je Lee1, Lutz Birnbaumer3, Hyun Jin Kim1,2*, Myoung Kyu Park1,2,* 1Department of Physiology, Sungkyunkwan University School of Medicine, 2066, Seoburo, Jangan-gu, Suwon, 16419, Korea 2Samsung Biomedical Research Institute, Samsung Medical Center, 81 Irwon-Ro Gangnam-gu, Seoul, 06351, Korea 3Neurobiology Laboratory. National Institute of Environmental Health Sciences, North Carolina 27709, USA; and Institute of Biomedical Research (BIOMED), Catholic University of Argentina, C1107AFF, Autonomous City of Buenos Aires, Argentina Abbreviated title: Pacemaker channels for nigral dopamine neurons *Corresponding authors: Professor Myoung Kyu PARK Department of Physiology, Sungkyunkwan University School of Medicine, 2066, Seoburo, Jangan-gu, Suwon 440-746, Korea TEL: +82-31-299-6101 e-mail: [email protected] or Associate Professor Hyun Jin Kim TEL: +82-31-299-6105 e-mail: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.447013; this version posted June 4, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.
    [Show full text]
  • Co-Application of Eugenol and QX-314 Elicits the Prolonged Blockade of Voltage-Gated Sodium Channels in Nociceptive Trigeminal Ganglion Neurons
    biomolecules Article Co-Application of Eugenol and QX-314 Elicits the Prolonged Blockade of Voltage-Gated Sodium Channels in Nociceptive Trigeminal Ganglion Neurons 1, 1, 1,2, 1 1, Sung-Min Hwang y , Kihwan Lee y, Sang-Taek Im y, Eun Jin Go , Yong Ho Kim * and Chul-Kyu Park 1,* 1 Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Korea; [email protected] (S.-M.H.); [email protected] (K.L.); [email protected] (S.-T.I.); [email protected] (E.J.G.) 2 Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Seoul National University Hospital, Seoul 03082, Korea * Correspondence: [email protected] (Y.H.K.); [email protected] (C.-K.P.) These authors contributed equally to this work. y Received: 5 October 2020; Accepted: 2 November 2020; Published: 5 November 2020 Abstract: Local anesthetics (LAs) can completely block nociception by inhibiting voltage-gated sodium channels (VGSCs), and thus, blocking action potentials (APs) within sensory neurons. As one of the several LAs, eugenol is used for dental pain treatment. It reportedly features multiple functions in regulating diverse ion channels. This study aimed to investigate the long-lasting analgesic effect of eugenol alone, as well as that of the combination of eugenol as a noxious-heat-sensitive transient receptor potential vanilloid 1 (TRPV1) channel agonist and a permanently charged sodium channel blocker (QX-314), on neuronal excitability in trigeminal ganglion (TG) neurons. Eugenol alone increased inward current in a dose-dependent manner in capsaicin-sensitive TG neurons. Eugenol also inhibited the VGSC current and AP.
    [Show full text]
  • Dissection of the Components for PIP2 Activation and Thermosensation in TRP Channels
    Dissection of the components for PIP2 activation and thermosensation in TRP channels Sebastian Brauchi*†, Gerardo Orta*, Carolina Mascayano‡, Marcelo Salazar*, Natalia Raddatz*, Hector Urbina‡, Eduardo Rosenmann*, Fernando Gonzalez-Nilo‡, and Ramon Latorre*§ *Laboratory of Biophysics and Molecular Physiology, Centro de Estudios Cientificos, Valdivia 509-9100, Chile; ‡Centro de Bioinformatica y Simulacion Molecular Simulation Center, Universidad de Talca, Talca 346-0000, Chile Contributed by Ramon Latorre, April 17, 2007 (sent for review March 26, 2007) Phosphatidylinositol 4,5-bisphosphate (PIP2) plays a central role in In this article, we address the problem of PIP2 binding and its the activation of several transient receptor potential (TRP) chan- relationship with the temperature-dependent properties of ther- nels. The role of PIP2 on temperature gating of thermoTRP channels mally sensitive TRP (thermoTRP) channels. This is an important has not been explored in detail, and the process of temperature problem to be solved because, first there is no direct evidence activation is largely unexplained. In this work, we have exchanged that positive charges present in the TRPV1 TRP domain are different segments of the C-terminal region between cold- involved in PIP2 activation; second, the process of temperature sensitive (TRPM8) and heat-sensitive (TRPV1) channels, trying to activation remains obscure; and third, the role of PIP2 in such understand the role of the segment in PIP2 and temperature process has not been explored in detail. activation. A chimera in which the proximal part of the C-terminal of TRPV1 replaces an equivalent section of TRPM8 C-terminal is Results activated by PIP2 and confers the phenotype of heat activation.
    [Show full text]
  • Neuronal HCN Channel Function and Plasticity
    Accepted Manuscript Title: Neuronal HCN channel function and plasticity Author: Mala M. Shah PII: S2468-8673(18)30001-4 DOI: https://doi.org/doi:10.1016/j.cophys.2018.01.001 Reference: COPHYS 37 To appear in: Received date: 30-10-2017 Revised date: 1-1-2018 Accepted date: 2-1-2018 Please cite this article as: Mala M. Shah, Neuronal HCN channel function and plasticity, Current Opinion in Physiology (2018), https://doi.org/10.1016/j.cophys.2018.01.001 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. Neuronal HCN channel function and plasticity Mala M. Shah Department of Pharmacology, UCL School of Pharmacy, University College London, London, UK. Short title: HCN channel function Corresponding Author: Mala M. Shah, Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London, UK. e-mail: [email protected] Accepted Manuscript 1 Page 1 of 13 Abstract : The hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channel is a voltage-gated cation channel that is activated with hyperpolarization. Four subunits, HCN1 -4, have thus far been identified. All four subunits are expressed in the central nervous system (CNS), though their expression pattern varies considerably.
    [Show full text]
  • Ligand Discrimination and Gating in Cyclic Nucleotide-Gated Ion
    RESEARCH ARTICLE Ligand discrimination and gating in cyclic nucleotide-gated ion channels from apo and partial agonist-bound cryo-EM structures Jan Rheinberger1, Xiaolong Gao1, Philipp AM Schmidpeter1, Crina M Nimigean1,2,3* 1Departments of Anesthesiology, Weill Cornell Medical College, New York, United States; 2Department of Physiology and Biophysics, Weill Cornell Medical College, New York, United States; 3Department of Biochemistry, Weill Cornell Medical College, New York, United States Abstract Cyclic nucleotide-modulated channels have important roles in visual signal transduction and pacemaking. Binding of cyclic nucleotides (cAMP/cGMP) elicits diverse functional responses in different channels within the family despite their high sequence and structure homology. The molecular mechanisms responsible for ligand discrimination and gating are unknown due to lack of correspondence between structural information and functional states. Using single particle cryo- electron microscopy and single-channel recording, we assigned functional states to high-resolution structures of SthK, a prokaryotic cyclic nucleotide-gated channel. The structures for apo, cAMP- bound, and cGMP-bound SthK in lipid nanodiscs, correspond to no, moderate, and low single- channel activity, respectively, consistent with the observation that all structures are in resting, closed states. The similarity between apo and ligand-bound structures indicates that ligand-binding domains are strongly coupled to pore and SthK gates in an allosteric, concerted fashion. The different
    [Show full text]
  • Investigating Developmental and Epileptic Encephalopathy Using Drosophila Melanogaster
    International Journal of Molecular Sciences Review Investigating Developmental and Epileptic Encephalopathy Using Drosophila melanogaster Akari Takai 1 , Masamitsu Yamaguchi 2,3, Hideki Yoshida 2 and Tomohiro Chiyonobu 1,* 1 Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; [email protected] 2 Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 603-8585, Japan; [email protected] (M.Y.); [email protected] (H.Y.) 3 Kansai Gakken Laboratory, Kankyo Eisei Yakuhin Co. Ltd., Kyoto 619-0237, Japan * Correspondence: [email protected] Received: 15 August 2020; Accepted: 1 September 2020; Published: 3 September 2020 Abstract: Developmental and epileptic encephalopathies (DEEs) are the spectrum of severe epilepsies characterized by early-onset, refractory seizures occurring in the context of developmental regression or plateauing. Early infantile epileptic encephalopathy (EIEE) is one of the earliest forms of DEE, manifesting as frequent epileptic spasms and characteristic electroencephalogram findings in early infancy. In recent years, next-generation sequencing approaches have identified a number of monogenic determinants underlying DEE. In the case of EIEE, 85 genes have been registered in Online Mendelian Inheritance in Man as causative genes. Model organisms are indispensable tools for understanding the in vivo roles of the newly identified causative genes. In this review, we first present an overview of epilepsy and its genetic etiology, especially focusing on EIEE and then briefly summarize epilepsy research using animal and patient-derived induced pluripotent stem cell (iPSC) models. The Drosophila model, which is characterized by easy gene manipulation, a short generation time, low cost and fewer ethical restrictions when designing experiments, is optimal for understanding the genetics of DEE.
    [Show full text]
  • Ion Channels
    RESEARCH HIGHLIGHTS ION CHANNELS These studies add HCN channels to the list of ion channels that are regulated by auxiliary subunits and A regulatory sidekick show that specific TRIP8b isoforms can differentially regulate Ih and thus The trafficking and function of changes in HCN channel surface control intrinsic neuronal excitability. many ion channels are regulated by expression resulting from alterations Katherine Whalley subunits that do not contribute to the in HCN subunit trafficking. pore-forming core of the channel. TRIP8b also altered HCN ORIGINAL RESEARCH PAPERS Lewis, A. S. et al. Alternatively spliced isoforms of TRIP8b Three recent papers reveal a role for channel gating. Santoro et al. and differentially control h channel trafficking and cytoplasmic tetratricopeptide repeat- Zolles et al. showed that TRIP8b function. J. Neurosci. 29, 6250–6265 (2009) | containing Rab8b-interacting protein slowed channel opening and acceler- Santoro, B. et al. TRIP8b splice variants form a family of auxiliary subunits that regulate gating (TRIP8b, also known as PEX5R and ated channel closing. Furthermore, and trafficking of HCN channels in the brain. PEX5Rp) as a regulatory auxiliary Lewis et al. and Santoro et al. found Neuron 62, 802–813 (2009) | Zolles, G. et al. Association with the auxiliary subunit PEX5R/ subunit of hyperpolarization- that co-expressing TRIP8b with Trip8b controls responsiveness of HCN channels activated cyclic nucleotide-gated HCN subunits shifted the membrane to cAMP and adrenergic stimulation. Neuron 62, (HCN) channels. potential threshold for HCN channel 814–825 (2009) FURTHER READING Santoro, B. et al. The HCN channel pore, consist- activation to more hyperpolarized Regulation of HCN channel surface expression ing of HCN1–HCN4 subunits, opens potentials, regardless of the isoform by a novel C-terminal protein-protein at negative membrane potentials and tested.
    [Show full text]
  • Fine Tuning HCN Channel Activity Tural Basis of This Coupling Has Only Been Well Characterized in Kcsa
    Wednesday, February 15, 2017 465a Symposium: Protein Dynamics and Allostery example, in-silico models of human cardiac electrophysiology are being considered by the FDA for prediction of proarrhythmic cardiotoxicity as a 2285-Symp core component of the preclinical assessment phase of all new drugs. How- Coupled Residue-Residue Dynamics in Protein Allosteric Mechanisms ever, one issue that exists with current models is that they each respond Donald Hamelberg. differently to insults such as drug block of ion channels or mutation of cardiac Department of Chemistry, Georgia State University, Atlanta, GA, USA. ion channel genes. Clearly this poses a problem in relation to the utility of Although the relationship between structure and function in biomolecules is these models in making quantitative predictions that are physiologically or well established, it is not always adequate to provide a complete understanding clinically meaningful. To examine this in detail we tested the ability of three of biomolecular function. The dynamical fluctuations of biomolecules can also models of the human ventricular action potential, the O’hara-Rudy the Grandi- play an essential role in function. Detailed understanding of how conforma- Bers and the Ten Tusscher models, to reproduce the clinical phenotype of tional dynamics orchestrates function in allosteric regulation of recognition different subtypes of the long QT syndrome. All models, in their original and catalysis at atomic resolution remains ambiguous. The overarching goal form, produce markedly different and unrealistic predictions of QT prolonga- is to understand how biomolecular dynamics are coupled to function by using tion. To address this, we used a global optimization approach to constrain ex- atomistic molecular simulations to complement experiments.
    [Show full text]